• Biotechnology
  • Healthcare
Moderna, Inc. logo
Moderna, Inc.
MRNA · US · NASDAQ
82.9
USD
+1.61
(1.94%)
Executives
Name Title Pay
Ms. Shannon Thyme Klinger Chief Legal Officer & Corporate Secretary 1.39M
Dr. Jerh Collins Ph.D. Chief Technical Operations & Quality Officer --
Ms. Lavina Talukdar CFA Senior Vice President & Head of Investor Relations --
Dr. Melanie Ivarsson M.B.A., Ph.D. Chief Development Officer --
Mr. Stephane Bancel Chief Executive Officer & Director 4.55M
Colleen Hussey Senior Director of Corporate Communications --
Ms. Tracey Franklin Chief Human Resources Officer --
Dr. Stephen Hoge M.D. President 1.92M
Mr. Brad Miller Chief Information Officer --
Mr. James M. Mock Chief Financial Officer 1.4M
Insider Transactions
Date Name Title Acquisition Or Disposition Stock / Options # of Shares Price
2024-08-09 Hoge Stephen President A - M-Exempt Common Stock 522 0
2024-08-12 Hoge Stephen President D - S-Sale Common Stock 254 84.1047
2024-08-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 522 0
2024-08-05 Rubenstein David M. director A - A-Award Stock Option (Right to Buy) 5051 83.75
2024-08-05 Rubenstein David M. director A - A-Award Stock Option (Right to Buy) 4065 83.75
2024-08-05 Rubenstein David M. director A - A-Award Restricted Stock Units 850 0
2024-08-05 Rubenstein David M. director A - A-Award Restricted Stock Units 677 0
2024-08-05 Rubenstein David M. - 0 0
2024-07-31 AFEYAN NOUBAR director D - S-Sale Common Stock 4599 118.9622
2024-07-31 AFEYAN NOUBAR director D - S-Sale Common Stock 5664 119.8365
2024-07-31 AFEYAN NOUBAR director D - S-Sale Common Stock 3428 120.8802
2024-07-31 AFEYAN NOUBAR director D - S-Sale Common Stock 1309 121.6229
2024-07-24 AFEYAN NOUBAR director D - S-Sale Common Stock 2668 116.3275
2024-07-24 AFEYAN NOUBAR director D - S-Sale Common Stock 3500 117.2504
2024-07-24 AFEYAN NOUBAR director D - S-Sale Common Stock 6832 118.2974
2024-07-24 AFEYAN NOUBAR director D - S-Sale Common Stock 2000 119.013
2024-07-17 AFEYAN NOUBAR director D - S-Sale Common Stock 2601 120.8564
2024-07-17 AFEYAN NOUBAR director D - S-Sale Common Stock 2100 121.5699
2024-07-17 AFEYAN NOUBAR director D - S-Sale Common Stock 2900 122.8614
2024-07-17 AFEYAN NOUBAR director D - S-Sale Common Stock 6699 123.8212
2024-07-17 AFEYAN NOUBAR director D - S-Sale Common Stock 700 124.6143
2024-07-15 Hoge Stephen President D - S-Sale Common Stock 15000 121.12
2024-07-10 AFEYAN NOUBAR director D - S-Sale Common Stock 6121 116.0086
2024-07-10 AFEYAN NOUBAR director D - S-Sale Common Stock 4279 117.1812
2024-07-10 AFEYAN NOUBAR director D - S-Sale Common Stock 3000 117.8276
2024-07-10 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 119.2829
2024-07-10 AFEYAN NOUBAR director D - S-Sale Common Stock 500 119.952
2024-07-05 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 1452 0
2024-07-05 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1452 0
2024-07-08 Mock James M Chief Financial Officer D - S-Sale Common Stock 689 118.2371
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 2460 122.6887
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1600 123.7363
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1500 124.9015
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 125.7064
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 127.5035
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1435 128.7789
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 2300 129.8397
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 2705 130.756
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 700 131.5557
2024-06-26 AFEYAN NOUBAR director D - S-Sale Common Stock 100 134.05
2024-06-18 AFEYAN NOUBAR director D - S-Sale Common Stock 1700 132.2245
2024-06-18 AFEYAN NOUBAR director D - S-Sale Common Stock 6160 133.2609
2024-06-18 AFEYAN NOUBAR director D - S-Sale Common Stock 6840 134.0932
2024-06-18 AFEYAN NOUBAR director D - S-Sale Common Stock 200 135.015
2024-06-18 AFEYAN NOUBAR director D - S-Sale Common Stock 100 136.92
2024-06-17 Hoge Stephen President D - S-Sale Common Stock 15000 138.16
2024-06-14 Bancel Stephane Chief Executive Officer A - J-Other Common Stock 26840 0
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 700 142.9696
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 408 144.4619
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 1263 145.5748
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 3031 146.5483
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 6397 147.5498
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 1601 148.613
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 1400 149.3927
2024-06-12 AFEYAN NOUBAR director D - S-Sale Common Stock 200 150.24
2024-06-11 AFEYAN NOUBAR director D - S-Sale Common Stock 163649 148.2944
2024-06-11 AFEYAN NOUBAR director D - S-Sale Common Stock 6420 149.4038
2024-06-11 AFEYAN NOUBAR director D - S-Sale Common Stock 31531 148.2944
2024-06-11 AFEYAN NOUBAR director D - S-Sale Common Stock 1232 149.4038
2024-06-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 9108 0
2024-06-10 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 5178 148.0851
2024-06-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1139 0
2024-06-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1139 0
2024-06-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 9108 0
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 900 145.9144
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 700 147.7754
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 1800 148.571
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 300 149.35
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 2840 151.0679
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 3902 151.7612
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 2437 152.8954
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 400 153.71
2024-06-05 AFEYAN NOUBAR director D - S-Sale Common Stock 1721 154.8577
2024-05-31 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1326 0
2024-06-03 Mock James M Chief Financial Officer D - S-Sale Common Stock 691 140.7198
2024-05-31 Mock James M Chief Financial Officer A - A-Award Performance Stock Units 5304 0
2024-05-31 Mock James M Chief Financial Officer D - M-Exempt Performance Stock Units 1326 0
2024-05-31 Hoge Stephen President A - M-Exempt Common Stock 651 0
2024-06-03 Hoge Stephen President D - S-Sale Common Stock 339 140.7198
2024-05-31 Hoge Stephen President D - M-Exempt Restricted Stock Units 651 0
2024-05-31 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 232 0
2024-06-03 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 122 140.7198
2024-05-31 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 232 0
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 1000 141.01
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 3013 141.725
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 142.82
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 1500 143.942
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 2400 144.9858
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 5581 146.0198
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 2229 146.8197
2024-05-29 AFEYAN NOUBAR director D - S-Sale Common Stock 3177 148.0575
2024-05-28 Mock James M Chief Financial Officer A - M-Exempt Common Stock 328 0
2024-05-29 Mock James M Chief Financial Officer D - S-Sale Common Stock 183 144.5047
2024-05-28 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 328 0
2024-05-28 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 328 0
2024-05-29 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 183 144.5047
2024-05-28 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 328 0
2024-05-28 Hoge Stephen President A - M-Exempt Common Stock 610 0
2024-05-29 Hoge Stephen President D - S-Sale Common Stock 341 144.5047
2024-05-28 Hoge Stephen President D - M-Exempt Restricted Stock Units 610 0
2024-05-24 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1326 0
2024-05-28 Mock James M Chief Financial Officer D - S-Sale Common Stock 648 162.4736
2024-05-24 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 1326 0
2024-05-24 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 2652 0
2024-05-28 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 1296 162.4736
2024-05-24 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 2652 0
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 100 142.95
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 200 144.395
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 100 145.03
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 300 147.0633
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 600 148.2067
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 300 149.4533
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 400 150.595
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 700 151.7329
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 800 152.92
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 200 153.84
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 600 155.66
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 1802 157.4117
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 1923 158.2529
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 2200 159.5683
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 825 160.2355
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 1005 162.1918
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 2223 163.0236
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 625 163.9212
2024-05-22 AFEYAN NOUBAR director D - S-Sale Common Stock 97 164.67
2024-05-15 AFEYAN NOUBAR director D - S-Sale Common Stock 4488 125.5251
2024-05-15 AFEYAN NOUBAR director D - S-Sale Common Stock 2001 126.6208
2024-05-15 AFEYAN NOUBAR director D - S-Sale Common Stock 2600 127.7063
2024-05-15 AFEYAN NOUBAR director D - S-Sale Common Stock 5711 128.7466
2024-05-15 AFEYAN NOUBAR director D - S-Sale Common Stock 200 129.15
2024-05-15 Hoge Stephen President D - S-Sale Common Stock 15000 127.49
2024-05-09 Hoge Stephen President A - M-Exempt Common Stock 522 0
2024-05-10 Hoge Stephen President D - S-Sale Common Stock 255 120.9573
2024-05-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 522 0
2024-05-08 AFEYAN NOUBAR director D - S-Sale Common Stock 2000 120.3705
2024-05-08 AFEYAN NOUBAR director D - S-Sale Common Stock 5004 121.586
2024-05-08 AFEYAN NOUBAR director D - S-Sale Common Stock 7996 122.0689
2024-05-03 Berenson Stephen director A - M-Exempt Common Stock 732 0
2024-05-06 Berenson Stephen director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-03 Berenson Stephen director D - M-Exempt Restricted Stock Units 732 0
2024-05-06 Nader Francois director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-06 SAGAN PAUL director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-06 Tallett Elizabeth E director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-06 Horning Sandra director A - A-Award Stock Option (Right to Buy) 5668 122.13
2024-05-06 Horning Sandra director A - A-Award Restricted Stock Units 979 0
2024-05-06 LANGER ROBERT director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-06 NABEL ELIZABETH G director A - A-Award Stock Option (Right to Buy) 5668 122.13
2024-05-03 NABEL ELIZABETH G director A - M-Exempt Common Stock 732 0
2024-05-06 NABEL ELIZABETH G director A - A-Award Restricted Stock Units 979 0
2024-05-03 NABEL ELIZABETH G director D - M-Exempt Restricted Stock Units 732 0
2024-05-06 AFEYAN NOUBAR director A - A-Award Stock Option (Right to Buy) 7557 122.13
2024-05-01 AFEYAN NOUBAR director D - S-Sale Common Stock 3982 109.6974
2024-05-01 AFEYAN NOUBAR director D - S-Sale Common Stock 4572 110.4243
2024-05-01 AFEYAN NOUBAR director D - S-Sale Common Stock 4246 111.552
2024-05-01 AFEYAN NOUBAR director D - S-Sale Common Stock 1500 112.7673
2024-05-01 AFEYAN NOUBAR director D - S-Sale Common Stock 700 113.3986
2024-04-24 AFEYAN NOUBAR director D - S-Sale Common Stock 3926 107.1573
2024-04-24 AFEYAN NOUBAR director D - S-Sale Common Stock 5821 108.2955
2024-04-24 AFEYAN NOUBAR director D - S-Sale Common Stock 4153 109.041
2024-04-24 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 109.91
2024-04-17 AFEYAN NOUBAR director D - S-Sale Common Stock 8243 103.0983
2024-04-17 AFEYAN NOUBAR director D - S-Sale Common Stock 6475 103.6908
2024-04-17 AFEYAN NOUBAR director D - S-Sale Common Stock 282 104.4896
2024-04-15 Hoge Stephen President D - S-Sale Common Stock 15000 105.02
2024-04-10 AFEYAN NOUBAR director D - S-Sale Common Stock 6351 106.5945
2024-04-10 AFEYAN NOUBAR director D - S-Sale Common Stock 7512 107.3131
2024-04-10 AFEYAN NOUBAR director D - S-Sale Common Stock 748 108.5631
2024-04-10 AFEYAN NOUBAR director D - S-Sale Common Stock 389 109.3628
2024-04-05 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 1453 0
2024-04-05 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1453 0
2024-04-08 Mock James M Chief Financial Officer D - S-Sale Common Stock 705 101.9281
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 200 97.47
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 1499 99.3996
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 3167 100.6552
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 3832 101.328
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 3500 102.6433
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 2702 103.6431
2024-04-03 AFEYAN NOUBAR director D - S-Sale Common Stock 100 104.15
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 1817 105.7269
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 1100 106.7057
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 1466 107.7898
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 3100 109.0889
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 4544 110.2401
2024-03-27 AFEYAN NOUBAR director D - S-Sale Common Stock 2973 110.7486
2024-03-20 AFEYAN NOUBAR director D - S-Sale Common Stock 2113 101.048
2024-03-20 AFEYAN NOUBAR director D - S-Sale Common Stock 7109 102.1464
2024-03-20 AFEYAN NOUBAR director D - S-Sale Common Stock 5778 103.1753
2024-03-15 Hoge Stephen President D - S-Sale Common Stock 15000 103.17
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 6281 106.5956
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 2300 107.6595
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 600 108.515
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 412 109.4976
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 3803 110.8001
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 1404 111.6851
2024-03-13 AFEYAN NOUBAR director D - S-Sale Common Stock 200 112.435
2024-03-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1138 0
2024-03-08 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 544 99.0509
2024-03-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1138 0
2024-03-01 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 233 0
2024-03-04 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 117 94.3383
2024-03-01 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 233 0
2024-03-01 Hoge Stephen President A - M-Exempt Common Stock 652 0
2024-03-04 Hoge Stephen President D - S-Sale Common Stock 309 94.3383
2024-03-01 Hoge Stephen President D - M-Exempt Restricted Stock Units 652 0
2024-02-28 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1315 0
2024-02-29 Mock James M Chief Financial Officer D - S-Sale Common Stock 647 94.572
2024-02-28 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 1315 0
2024-02-28 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1315 0
2024-02-29 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 670 94.572
2024-02-28 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1315 0
2024-02-28 Hoge Stephen President A - M-Exempt Common Stock 2442 0
2024-02-29 Hoge Stephen President D - S-Sale Common Stock 2388 94.572
2024-02-28 Hoge Stephen President A - M-Exempt Common Stock 2411 0
2024-02-28 Hoge Stephen President D - M-Exempt Restricted Stock Units 2442 0
2024-02-28 Hoge Stephen President D - M-Exempt Restricted Stock Units 2411 0
2024-02-27 Mock James M Chief Financial Officer A - A-Award Stock Option (Right to Buy) 60216 96.2
2024-02-27 Mock James M Chief Financial Officer A - A-Award Stock Option (Right to Buy) 23635 96.2
2024-02-27 Mock James M Chief Financial Officer A - A-Award Restricted Stock Units 12377 0
2024-02-27 Mock James M Chief Financial Officer A - A-Award Restricted Stock Units 5304 0
2024-02-27 Klinger Shannon Thyme Chief Legal Officer A - A-Award Stock Option (Right to Buy) 60216 96.2
2024-02-27 Klinger Shannon Thyme Chief Legal Officer A - A-Award Stock Option (Right to Buy) 23635 96.2
2024-02-27 Klinger Shannon Thyme Chief Legal Officer A - A-Award Restricted Stock Units 12377 0
2024-02-27 Klinger Shannon Thyme Chief Legal Officer A - A-Award Restricted Stock Units 10609 0
2024-02-27 Hoge Stephen President A - A-Award Stock Option (Right to Buy) 43894 96.2
2024-02-27 Hoge Stephen President A - A-Award Restricted Stock Units 22987 0
2024-02-27 Bancel Stephane Chief Executive Officer A - A-Award Stock Option (Right to Buy) 151943 96.2
2024-02-22 Hoge Stephen President D - S-Sale Common Stock 15000 100
2024-02-14 Hoge Stephen President A - A-Award Common Stock 22694 0
2024-02-16 Hoge Stephen President D - S-Sale Common Stock 10542 88.8808
2024-02-14 Bancel Stephane Chief Executive Officer A - A-Award Common Stock 56736 0
2024-02-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 26351 88.8808
2024-02-09 Hoge Stephen President A - M-Exempt Common Stock 522 0
2024-02-12 Hoge Stephen President D - S-Sale Common Stock 304 86.2292
2024-02-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 522 0
2024-01-16 Hoge Stephen President D - S-Sale Common Stock 15000 103.9
2024-01-05 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 1452 0
2024-01-05 Mock James M Chief Financial Officer A - M-Exempt Common Stock 1452 0
2024-01-08 Mock James M Chief Financial Officer D - S-Sale Common Stock 772 109.7546
2023-12-27 Hoge Stephen President D - S-Sale Common Stock 45000 100.01
2023-12-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1139 0
2023-12-08 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 563 79.0854
2023-12-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1139 0
2023-12-05 Garay Arpa Chief Commercial Officer D - M-Exempt Restricted Stock Units 1141 0
2023-12-05 Garay Arpa Chief Commercial Officer A - M-Exempt Common Stock 1141 0
2023-12-06 Garay Arpa Chief Commercial Officer D - S-Sale Common Stock 564 78.3368
2023-12-01 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 232 0
2023-12-04 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 111 79.3134
2023-12-01 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 232 0
2023-12-01 Hoge Stephen President A - M-Exempt Common Stock 651 0
2023-12-04 Hoge Stephen President D - S-Sale Common Stock 309 79.3134
2023-12-01 Hoge Stephen President D - M-Exempt Restricted Stock Units 651 0
2023-11-28 Hoge Stephen President A - M-Exempt Common Stock 2411 0
2023-11-29 Hoge Stephen President D - S-Sale Common Stock 1136 79.8112
2023-11-28 Hoge Stephen President D - M-Exempt Restricted Stock Units 2411 0
2023-11-09 Hoge Stephen President A - M-Exempt Common Stock 522 0
2023-11-10 Hoge Stephen President D - S-Sale Common Stock 263 68.76
2023-11-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 522 0
2023-11-01 AFEYAN NOUBAR director D - S-Sale Common Stock 1900 75.2979
2023-11-01 AFEYAN NOUBAR director D - S-Sale Common Stock 8100 76.1686
2023-10-25 AFEYAN NOUBAR director D - S-Sale Common Stock 6500 76.7283
2023-10-25 AFEYAN NOUBAR director D - S-Sale Common Stock 3100 77.3399
2023-10-25 AFEYAN NOUBAR director D - S-Sale Common Stock 400 78.415
2023-10-11 AFEYAN NOUBAR director D - S-Sale Common Stock 8700 102.8798
2023-10-11 AFEYAN NOUBAR director D - S-Sale Common Stock 5700 103.758
2023-10-11 AFEYAN NOUBAR director D - S-Sale Common Stock 500 104.816
2023-10-11 AFEYAN NOUBAR director D - S-Sale Common Stock 100 105.58
2023-10-05 Mock James M Chief Financial Officer D - M-Exempt Restricted Stock Units 5810 0
2023-10-05 Mock James M Chief Financial Officer A - M-Exempt Common Stock 5810 0
2023-10-06 Mock James M Chief Financial Officer D - S-Sale Common Stock 2858 102.5027
2023-10-04 AFEYAN NOUBAR director D - S-Sale Common Stock 1030 100.0176
2023-10-04 AFEYAN NOUBAR director D - S-Sale Common Stock 1800 100.9389
2023-10-04 AFEYAN NOUBAR director D - S-Sale Common Stock 2700 102.0317
2023-10-04 AFEYAN NOUBAR director D - S-Sale Common Stock 5604 103.2201
2023-10-04 AFEYAN NOUBAR director D - S-Sale Common Stock 3866 104.0368
2023-09-27 AFEYAN NOUBAR director D - S-Sale Common Stock 5204 98.4564
2023-09-27 AFEYAN NOUBAR director D - S-Sale Common Stock 4596 99.4877
2023-09-27 AFEYAN NOUBAR director D - S-Sale Common Stock 200 100.165
2023-09-20 AFEYAN NOUBAR director D - S-Sale Common Stock 4614 103.8719
2023-09-20 AFEYAN NOUBAR director D - S-Sale Common Stock 9380 105.0556
2023-09-20 AFEYAN NOUBAR director D - S-Sale Common Stock 1006 105.769
2023-09-15 Hoge Stephen President D - S-Sale Common Stock 15000 112.99
2023-09-15 Bancel Stephane Chief Executive Officer A - J-Other Common Stock 17893 0
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 4281 108.2059
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 4174 108.7667
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 1400 110.2593
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 3895 111.2306
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 900 112.1744
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 250 113.22
2023-09-13 AFEYAN NOUBAR director D - S-Sale Common Stock 100 114.32
2023-09-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1138 0
2023-09-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1138 0
2023-09-08 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 557 107.221
2023-09-06 AFEYAN NOUBAR director D - S-Sale Common Stock 9011 107.2698
2023-09-06 AFEYAN NOUBAR director D - S-Sale Common Stock 5689 108.0702
2023-09-06 AFEYAN NOUBAR director D - S-Sale Common Stock 300 108.9133
2023-09-05 Garay Arpa Chief Commercial Officer D - M-Exempt Restricted Stock Units 1140 0
2023-09-05 Garay Arpa Chief Commercial Officer A - M-Exempt Common Stock 1140 0
2023-09-06 Garay Arpa Chief Commercial Officer D - S-Sale Common Stock 574 108.3961
2023-09-01 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 233 0
2023-09-05 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 115 111.436
2023-09-01 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 233 0
2023-09-01 Hoge Stephen President A - M-Exempt Common Stock 652 0
2023-09-05 Hoge Stephen President D - S-Sale Common Stock 320 111.436
2023-09-01 Hoge Stephen President D - M-Exempt Restricted Stock Units 652 0
2023-08-30 AFEYAN NOUBAR director D - S-Sale Common Stock 5847 114.6411
2023-08-30 AFEYAN NOUBAR director D - S-Sale Common Stock 4717 115.4251
2023-08-30 AFEYAN NOUBAR director D - S-Sale Common Stock 4336 116.5736
2023-08-30 AFEYAN NOUBAR director D - S-Sale Common Stock 100 117.21
2023-08-28 Hoge Stephen President A - M-Exempt Common Stock 2411 0
2023-08-29 Hoge Stephen President D - S-Sale Common Stock 1135 115.0146
2023-08-28 Hoge Stephen President D - M-Exempt Restricted Stock Units 2411 0
2023-08-23 AFEYAN NOUBAR director D - S-Sale Common Stock 311 112.6116
2023-08-23 AFEYAN NOUBAR director D - S-Sale Common Stock 854 113.8344
2023-08-23 AFEYAN NOUBAR director D - S-Sale Common Stock 3657 115.1558
2023-08-23 AFEYAN NOUBAR director D - S-Sale Common Stock 9878 115.8446
2023-08-23 AFEYAN NOUBAR director D - S-Sale Common Stock 300 116.46
2023-08-16 AFEYAN NOUBAR director D - S-Sale Common Stock 1600 95.7025
2023-08-16 AFEYAN NOUBAR director D - S-Sale Common Stock 2900 96.4387
2023-08-16 AFEYAN NOUBAR director D - S-Sale Common Stock 1200 98.0658
2023-08-16 AFEYAN NOUBAR director D - S-Sale Common Stock 4000 98.8222
2023-08-16 AFEYAN NOUBAR director D - S-Sale Common Stock 300 99.5
2023-08-17 Hoge Stephen President D - S-Sale Common Stock 15000 100
2023-08-09 Hoge Stephen President A - M-Exempt Common Stock 523 0
2023-08-10 Hoge Stephen President D - S-Sale Common Stock 247 102.5314
2023-08-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 523 0
2023-08-02 AFEYAN NOUBAR director D - S-Sale Common Stock 9725 110.5876
2023-08-02 AFEYAN NOUBAR director D - S-Sale Common Stock 4175 111.4608
2023-08-02 AFEYAN NOUBAR director D - S-Sale Common Stock 900 112.5867
2023-08-02 AFEYAN NOUBAR director D - S-Sale Common Stock 200 113.675
2023-07-26 AFEYAN NOUBAR director D - S-Sale Common Stock 2400 119.6054
2023-07-26 AFEYAN NOUBAR director D - S-Sale Common Stock 11499 120.5002
2023-07-26 AFEYAN NOUBAR director D - S-Sale Common Stock 1101 121.2937
2023-07-19 AFEYAN NOUBAR director D - S-Sale Common Stock 847 124.1409
2023-07-19 AFEYAN NOUBAR director D - S-Sale Common Stock 9004 125.2576
2023-07-19 AFEYAN NOUBAR director D - S-Sale Common Stock 5149 126.0799
2023-07-17 Hoge Stephen President D - S-Sale Common Stock 15000 120.6
2023-07-12 AFEYAN NOUBAR director D - S-Sale Common Stock 1990 124.5941
2023-07-12 AFEYAN NOUBAR director D - S-Sale Common Stock 5865 125.6058
2023-07-12 AFEYAN NOUBAR director D - S-Sale Common Stock 6845 126.2697
2023-07-12 AFEYAN NOUBAR director D - S-Sale Common Stock 300 127.15
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 3533 123.4556
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 6097 124.1735
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 2639 125.145
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 1012 126.5364
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 1549 127.5514
2023-07-05 AFEYAN NOUBAR director D - S-Sale Common Stock 170 128.1712
2023-06-28 AFEYAN NOUBAR director D - S-Sale Common Stock 1700 119.8371
2023-06-28 AFEYAN NOUBAR director D - S-Sale Common Stock 3131 120.7674
2023-06-28 AFEYAN NOUBAR director D - S-Sale Common Stock 8073 121.8721
2023-06-28 AFEYAN NOUBAR director D - S-Sale Common Stock 2096 122.825
2023-06-28 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 27155 0.99
2023-06-28 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2278 120.39
2023-06-28 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11417 121.4
2023-06-28 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13094 122.01
2023-06-28 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 366 123.05
2023-06-28 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 27155 0.99
2023-06-21 AFEYAN NOUBAR director D - S-Sale Common Stock 7810 121.5956
2023-06-21 AFEYAN NOUBAR director D - S-Sale Common Stock 6790 122.2644
2023-06-21 AFEYAN NOUBAR director D - S-Sale Common Stock 400 123.475
2023-06-22 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-06-21 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 20711 121.77
2023-06-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 24829 119.13
2023-06-21 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 19289 122.21
2023-06-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 15171 119.87
2023-06-21 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-22 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-14 AFEYAN NOUBAR director D - S-Sale Common Stock 5471 125.4302
2023-06-14 AFEYAN NOUBAR director D - S-Sale Common Stock 5997 126.1728
2023-06-14 AFEYAN NOUBAR director D - S-Sale Common Stock 3002 127.1895
2023-06-14 AFEYAN NOUBAR director D - S-Sale Common Stock 530 128.1464
2023-06-15 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-06-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4788 125.61
2023-06-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 5377 127.01
2023-06-14 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 10642 125.32
2023-06-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8443 127.99
2023-06-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7115 129.12
2023-06-14 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 20848 126.1
2023-06-14 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8510 127.25
2023-06-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 14277 129.77
2023-06-14 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-15 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-15 Hoge Stephen President D - S-Sale Common Stock 15000 125.93
2023-06-07 AFEYAN NOUBAR director D - S-Sale Common Stock 12298 125.756
2023-06-07 AFEYAN NOUBAR director D - S-Sale Common Stock 2702 126.5929
2023-06-08 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-06-08 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 22215 123.58
2023-06-07 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 35872 125.73
2023-06-08 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 17396 124.53
2023-06-07 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4128 126.51
2023-06-08 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 389 125.22
2023-06-07 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-08 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1139 0
2023-06-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1139 0
2023-06-08 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 565 124.0899
2023-06-05 Garay Arpa Chief Commercial Officer D - M-Exempt Restricted Stock Units 4562 0
2023-06-05 Garay Arpa Chief Commercial Officer A - M-Exempt Common Stock 4562 0
2023-06-06 Garay Arpa Chief Commercial Officer D - S-Sale Common Stock 2290 125.7518
2023-06-01 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 232 0
2023-06-02 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 110 130.3232
2023-06-01 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 232 0
2023-06-01 Hoge Stephen President A - M-Exempt Common Stock 651 0
2023-06-02 Hoge Stephen President D - S-Sale Common Stock 309 130.3232
2023-06-01 Hoge Stephen President D - M-Exempt Restricted Stock Units 651 0
2023-05-31 AFEYAN NOUBAR director D - S-Sale Common Stock 2949 125.6921
2023-05-31 AFEYAN NOUBAR director D - S-Sale Common Stock 3373 126.7241
2023-05-31 AFEYAN NOUBAR director D - S-Sale Common Stock 5637 127.8391
2023-05-31 AFEYAN NOUBAR director D - S-Sale Common Stock 2441 128.5456
2023-05-31 AFEYAN NOUBAR director D - S-Sale Common Stock 600 129.6783
2023-06-01 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-06-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1410 125.96
2023-05-31 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9478 125.38
2023-06-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8611 127.53
2023-05-31 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 14636 126.71
2023-05-31 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11036 127.84
2023-05-31 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4850 128.42
2023-06-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 29979 128.18
2023-05-31 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-06-01 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-26 Hoge Stephen President A - M-Exempt Common Stock 2410 0
2023-05-30 Hoge Stephen President D - S-Sale Common Stock 1181 124.9524
2023-05-26 Hoge Stephen President D - M-Exempt Restricted Stock Units 2410 0
2023-05-25 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-05-25 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 10367 126.32
2023-05-24 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 28144 131.13
2023-05-25 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 20641 127.45
2023-05-24 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9820 131.98
2023-05-25 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8654 128.33
2023-05-24 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2036 132.93
2023-05-25 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 338 128.99
2023-05-24 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-25 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-18 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-05-17 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2333 123.22
2023-05-17 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9350 124.24
2023-05-17 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11547 125.34
2023-05-18 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 28476 123.53
2023-05-18 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 10653 124.31
2023-05-17 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16248 126.17
2023-05-17 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 522 126.86
2023-05-18 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 871 125.24
2023-05-17 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-18 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-11 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-05-10 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16954 131.08
2023-05-11 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 35330 127.17
2023-05-10 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 21247 131.89
2023-05-10 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1499 132.97
2023-05-11 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4470 127.92
2023-05-10 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 300 133.71
2023-05-11 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 200 128.84
2023-05-10 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-11 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-09 Hoge Stephen President A - M-Exempt Common Stock 522 0
2023-05-10 Hoge Stephen President D - S-Sale Common Stock 250 133.3743
2023-05-09 Hoge Stephen President D - M-Exempt Restricted Stock Units 522 0
2023-05-03 Tallett Elizabeth E director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-03 SAGAN PAUL director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-03 Nader Francois director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-03 NABEL ELIZABETH G director A - A-Award Stock Option (Right to Buy) 4601 130.14
2023-05-03 NABEL ELIZABETH G director A - A-Award Restricted Stock Units 732 0
2023-05-03 LANGER ROBERT director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-03 Horning Sandra director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-03 Berenson Stephen director A - A-Award Stock Option (Right to Buy) 4601 130.14
2023-05-03 Berenson Stephen director A - A-Award Restricted Stock Units 732 0
2023-05-03 AFEYAN NOUBAR director A - A-Award Stock Option (Right to Buy) 6134 130.14
2023-05-04 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 600 132.98
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2846 134.32
2023-05-03 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13570 130.6
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13468 135.06
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16552 136.01
2023-05-03 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 21854 131.54
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 5427 136.97
2023-05-03 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4576 132.12
2023-05-04 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1107 137.76
2023-05-03 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-05-04 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-28 NABEL ELIZABETH G director A - M-Exempt Common Stock 670 0
2023-04-28 NABEL ELIZABETH G director D - M-Exempt Restricted Stock Units 670 0
2023-04-28 Horning Sandra director A - M-Exempt Common Stock 670 0
2023-04-28 Horning Sandra director D - M-Exempt Restricted Stock Units 670 0
2023-04-28 Berenson Stephen director A - M-Exempt Common Stock 670 0
2023-04-28 Berenson Stephen director D - M-Exempt Restricted Stock Units 670 0
2023-04-27 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-04-26 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7600 130.27
2023-04-26 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16898 131.1
2023-04-27 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 29600 130.41
2023-04-26 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9002 132.55
2023-04-27 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9900 131.16
2023-04-26 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 6500 133.93
2023-04-27 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 500 131.9
2023-04-26 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-27 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-20 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-04-20 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 6752 141.35
2023-04-19 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11900 141.94
2023-04-20 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8780 142.24
2023-04-19 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 14038 142.95
2023-04-20 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16495 143.53
2023-04-20 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7973 144.08
2023-04-19 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 14062 143.61
2023-04-19 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-20 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-13 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-04-12 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 12634 155.67
2023-04-12 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 16650 156.69
2023-04-13 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 30456 160.63
2023-04-12 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8334 157.67
2023-04-13 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9444 161.16
2023-04-12 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2382 158.56
2023-04-13 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 100 161.98
2023-04-12 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-13 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-06 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-04-06 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2739 154.79
2023-04-05 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 6666 152.74
2023-04-05 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2934 153.66
2023-04-06 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7602 155.58
2023-04-06 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1200 157
2023-04-05 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 22533 155.01
2023-04-06 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 25343 158.19
2023-04-05 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7754 155.54
2023-04-05 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 113 156.5
2023-04-06 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 3116 158.81
2023-04-05 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-04-06 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-30 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-03-30 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13900 147.13
2023-03-30 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2979 148.24
2023-03-29 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 31349 148.21
2023-03-30 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 15965 149.39
2023-03-29 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8300 148.83
2023-03-29 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 351 149.72
2023-03-30 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7156 149.99
2023-03-29 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-30 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-23 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-03-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 3879 149.13
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 6138 147.71
2023-03-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 18321 149.84
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 18458 148.62
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 5906 149.61
2023-03-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 14962 150.76
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7243 150.66
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 996 151.53
2023-03-22 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2838 151.67
2023-03-23 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1259 152.55
2023-03-22 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-23 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-16 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-03-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 4200 148.63
2023-03-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7500 149.81
2023-03-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 15472 148.33
2023-03-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7600 150.39
2023-03-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 17032 151.75
2023-03-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 22481 149.03
2023-03-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1847 150
2023-03-15 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 200 150.72
2023-03-16 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 3668 152.49
2023-03-15 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-16 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-09 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11461 137.25
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 7739 137.97
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 3896 139.24
2023-03-08 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 26018 141.18
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 5563 140.23
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 9232 141.3
2023-03-09 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 2109 142.26
2023-03-08 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13982 141.76
2023-03-08 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-09 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-07 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 1138 0
2023-03-07 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 1138 0
2023-03-08 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 561 141.5109
2023-03-01 AFEYAN NOUBAR director D - S-Sale Common Stock 10000 139
2023-03-01 Hoge Stephen President A - M-Exempt Common Stock 2606 0
2023-03-02 Hoge Stephen President D - S-Sale Common Stock 1177 136.4339
2023-03-01 Hoge Stephen President D - M-Exempt Restricted Stock Units 2606 0
2023-03-01 Klinger Shannon Thyme Chief Legal Officer A - M-Exempt Common Stock 930 0
2023-03-02 Klinger Shannon Thyme Chief Legal Officer D - S-Sale Common Stock 459 136.4339
2023-03-01 Klinger Shannon Thyme Chief Legal Officer D - M-Exempt Restricted Stock Units 930 0
2023-03-02 Bancel Stephane Chief Executive Officer A - M-Exempt Common Stock 40000 0.99
2023-03-02 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 8700 136.44
2023-03-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 11319 134.34
2023-03-02 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 17541 137.42
2023-03-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 21938 135.26
2023-03-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 5593 136.17
2023-03-02 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 13659 138.27
2023-03-01 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 1150 137.03
2023-03-02 Bancel Stephane Chief Executive Officer D - S-Sale Common Stock 100 139.03
2023-03-01 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-03-02 Bancel Stephane Chief Executive Officer D - M-Exempt Stock Option (Right to Buy) 40000 0.99
2023-02-28 Klinger Shannon Thyme Chief Legal Officer A - A-Award Stock Option (Right to Buy) 20040 138.81
2023-02-28 Klinger Shannon Thyme Chief Legal Officer A - A-Award Restricted Stock Units 5260 0
2023-02-28 Hoge Stephen President A - M-Exempt Common Stock 2410 0
2023-03-01 Hoge Stephen President D - S-Sale Common Stock 1072 138.0254
2023-02-28 Hoge Stephen President A - A-Award Stock Option (Right to Buy) 37217 138.81
2023-02-28 Hoge Stephen President A - A-Award Restricted Stock Units 9768 0
Transcripts
Operator:
Good day and thank you for standing by. Welcome to the Moderna second quarter 2024 conference call. At this time, all participants are in listen-only mode. After the speakers’ presentation, there will be a question and answer session. To ask a question during the session, you’ll need to press star-one-one on your telephone. You will then hear an automated message advising your hand is raised. To withdraw your question, please press star-one-one again. Please be advised today’s conference is being recorded. I would now like to hand the conference over to your speaker today, Lavina Talukdar. Please go ahead.
Lavina Talukdar:
Thank you Kevin. Good morning everyone and thank you for joining us on today’s call to discuss Moderna’s second quarter 2024 financial results and business update. You can access the press release issued this morning, as well as the slides that we will be reviewing, by going to the Investors section of our website. On today’s call are Stéphane Bancel, our Chief Executive Officer; Jamey Mock, our Chief Financial Officer, and Stephen Hoge, our President. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you Lavina. Good morning or good afternoon everyone. Thank you for joining us today. I will start with a review our business, Jamey will present our financial results for the quarter and take you through our basic financial framework. Stephen will then review our clinical progress. I will then close by sharing our commercial progress and our outlook for major upcoming milestones. In the second quarter of 2024, our respiratory franchise has shown remarkable progress and is poised to positively impact millions of lives globally each year. This is why we started Moderna, to impact patients, and that is why the Moderna team is so focused on execution because of that profound impact on so many lives. Our mRNA-1273 vaccine, Spikevax, continues to play a critical role in combating COVID. Based on CDC data for the season just ending, October 23 to June 24, COVID continues to result in higher hospitalization than other respiratory viruses. We are pleased that our RSV vaccine, whose brand name is mRESVIA, our second respiratory product, has launched in the U.S. and has now shipped to U.S. customers. It is poised to impact public health in the U.S. this year and soon in many other countries for many years to come. Our flu vaccine candidate, mRNA-1010, has demonstrated positive Phase III results, meeting all immunogenicity end points in adults 18 and older. Our flu + COVID combo vaccine candidate, mRNA-1083, has also shown positive results in Phase III, underscoring our ability to innovate and [indiscernible] respiratory illnesses effectively. These achievements highlight the strength of our mRNA platform and our commitment to public health. We have now five out of five vaccines with positive Phase III data
Jamey Mock:
Thanks Stéphane, and hello everyone. Today I will walk you through our financial performance for the second quarter and also update you on our financial outlook for the remainder of 2024. Let me start with our commercial performance on Slide 9. Net product sales for Q2 were $184 million, down 37% year-over-year, mainly driven by lower sales volumes of our COVID-19 vaccine in regions outside of the United States compared to the second quarter of 2023, when we fulfilled orders from prior year contracts. Q2 sales were above our guidance of approximately $100 million primarily due to stronger than expected sales in the United States. We recognized sales from a number of other countries, including a small portion of the Brazil contract we announced last quarter. Year-to-date sales were $351 million, down 83% year-over-year, largely driven by the same Q2 year-over-year trends I just mentioned a moment ago. Moving to Slide 10, as I just explained, net product sales were $184 million. In Q2, we recognized $30 million of licensing revenue which is included in the other revenue line of $57 million. This revenue comes from a non-exclusive intellectual property out-licensing agreement with a leading pharmaceutical company in Japan announced in our Q1 earnings call. The deal includes an upfront payment and low double-digit royalties on net sales of their COVID-19 product in Japan. For the second quarter of 2024, our cost of sales was $115 million, which included $10 million of third party royalties, $55 million related to unutilized manufacturing capacity and wind down costs, and $14 million of inventory write-downs. This resulted in our cost of sales representing 62% of net product sales. As a result of our initiative to resize our manufacturing cost structure, cost of sales was down 84% from Q2 last year when cost of sales was 249%. R&D expenses were $1.2 billion in Q2, reflecting a slight increase of $73 million or 6% year-over-year. We purchased a priority review voucher during the second quarter which is included in our Q2 results. With first half R&D spending at $2.3 billion, we are tracking towards a full year expected spend of approximately $4.5 billion. SG&A expenses for Q2 were $268 million, reflecting a 19% decrease year-over-year. This reduction is a result of our continued strong focus on cost discipline and strategic investments driving productivity, on which I will provide additional detail in the upcoming slide. We reported zero income tax expense for the second quarter of 2024 compared to an income tax benefit of $369 million in the same period last year. This shift is primarily due to the continued application of a valuation allowance on a majority of our deferred tax assets, which we first established in the third quarter of 2023. Our net loss for the period was $1.3 billion, an improvement from the net loss of $1.4 billion recorded last year. Loss per share was $3.33 compared to a loss of $3.62 in the second quarter of 2023. We ended Q2 with cash and investments totaling $10.8 billion, down from $12.2 billion at the end of Q1, primarily due to ongoing research and development expenses and operating activities. Moving to Slide 11 and similar to Q1, I want to provide additional detail on the cost reductions we are driving across the company. You can see in our Q2 results that we had a 19% year-over-year reduction in SG&A spend due to efficiency gains. One of the main drivers for the year-over-year reduction in SG&A is from our commercial and medical affairs group. Over the past year, we have built our internal capabilities, which has allowed us to drive cost efficiencies by reducing our use of external consultants and other purchased services. We’ve also been more focused and targeted on how we invest in these areas to drive the strongest possible return on investment. As the endemic market has been more seasonal, we have shifted more of our commercial spend to the second half of the year. Additionally, our procurement team has successfully driven company-wide cost reductions in the first half of 2024. Their primary focus has been on reducing third party supplier rates, and we have seen strong progress in contract rates for raw materials, components, clinical, travel, and consulting services. We continue to see strong adoption in artificial intelligence by our employees which will allow us to scale the business in an efficient manner with a digital-first mindset. For example, in the second quarter our HR team launched benefits and equity GPTs. These AI-driven assistants are designed to handle frequently asked questions previously directed to the HR operations team and allow us to scale efficiently. Overall, we have built a solid foundation and made purposeful investments in people, processes and technology. We’ve highlighted some of the significant drivers of the Q2 SG&A savings, but we are also seeing additional efficiency savings in R&D and manufacturing. I’m very pleased with the cost savings results in the first half of the year and want to thank our Moderna teams. Now let’s turn to our 2024 financial framework on Slide 12. We are revising our expectation for 2024 net product sales to a range of $3.0 billion to $3.5 billion. There are three primary drivers for the updated outlook. First, we are now expecting very low sales in 2024 from EU member states based on recent feedback and discussions with country health officials. Second, in the U.S. we are seeing increased competitive pressures for our respiratory vaccines. While this has led to a slower RSV ramp than previously anticipated, we continue to believe and mRESVIA’s long term potential. Third, in the rest of the world we have provided for the potential risk of revenue deferrals from 2024 into 2025. We remain committed in our attempt to mitigate these risks, but believe it’s appropriate to adjust our guidance at this time. Finally, this revenue framework assumes a U.S. COVID vaccination rate similar to last season. Our second half sales mix will be dependent on timing of regulatory approvals across the world and the number of days available in the third quarter to ship. We currently expect a remaining sales lift of 40% to 50% in Q3, with the balance in Q4. We expect cost of sales as a percentage of product sales for the full year to be in the range of 40% to 50%, based upon our updated sales range. For R&D, we continue to expect full year expenses to be approximately $4.5 billion, down from $4.8 billion in 2023. For SG&A, we continue to expect full year expenses to be approximately $1.3 billion, down from $1.5 billion in 2023. Note that we expect SG&A to be higher in the second half versus the first half, primarily due to increased commercial activity, but still expect the second half to be down on a year-over-year basis. We continue to expect taxes to be negligible in 2024 and capital expenditures to be approximately $0.9 billion. Finally, we continue to expect that we will end 2024 with approximately $9 billion in cash. We have made strong progress in improving our working capital management, which is offsetting the change in our product sales outlook. With that, I will now hand the call over to Stephen.
Stephen Hoge:
Thank you Jamey. Today I’ll review updates from our clinical programs in three of the four development areas in our portfolio. In the second quarter, we had important updates in respiratory vaccines, oncology and rare diseases. Starting with respiratory vaccines, we are very pleased by the approval and ATIP recommendation for our RSV vaccine, mRESVIA, in the U.S. for all unvaccinated adults 75 years and older and in unvaccinated adults 60 to 74 years of age who are at increased risk from RSV. This recommendation was the same as for the other two previously approved RSV vaccines. We also recently received a positive CHMP opinion from the European Medicine Agency for mRESVIA, and as Stéphane mentioned earlier, we are working towards approvals in additional countries. Turning to flu, we are in discussions with multiple regulatory authorities and intend to file in 2024. For our next-generation COVID vaccine, mRNA-1283, we announced positive Phase III efficacy results demonstrating non-inferior efficacy against COVID-19 compared to Spikevax in all trial participants 12 years of age and older. Efficacy was higher than Spikevax in adults 18 years and older. We are excited by the data and are sharing the results with regulators and intend to file for approval beginning in 2024. Rounding out the news in respiratory vaccines development, in the quarter we shared positive results from our Phase III trial with our combination flu and COVID vaccine. The trial met its primary immunogenicity end points with the combination vaccine eliciting higher immune responses against flu and SARS-CoV-2 than the licensed flu and licensed COVID vaccines given separately in adults 50 years of age and older in the trial. In the subset of participants 65 years of age and older, our combination vaccine also elicited higher immune responses than an enhanced flu vaccine that is recommended in the 65 and older age group in many countries, including the United States. These results are exciting, and we have begun sharing them with regulators and planning for the next steps. Turning now to oncology, in the quarter we share an update on our mRNA-4157 program, also known as INT, which elicits anti-tumor T-cell responses by targeting a patient’s unique tumor mutations, or neoantigens. INT is in multiple large randomized trials, including two Phase III trials, one in adjuvant melanoma and the other in adjuvant non-small cell lung cancer; one Phase II/III trial in adjuvant cutaneous squamous cell carcinoma, and two randomized Phase II studies, one in adjuvant kidney cancer and the other in bladder cancer. We and our partner, Merck expect to start additional studies in new tumor types. The development program was launched on the back of impressive randomized Phase II trial results in adjuvant melanoma. We recently shared the three-year follow-up data from that trial at ASCO this past June, and in the next few slides, I will quickly summarize the highlights from that presentation. The primary clinical end point for the Phase II adjuvant melanoma study is recurrence-free survival, or RFS. As presented at ASCO, there is a sustained improvement in RFS with the combination of INT plus Keytruda, versus Keytruda alone through three years of follow-up. 74.8% of patients receiving the combination treatment of INT plus Keytruda were alive and tumor-free at three years, which was 19 percentage points higher than Keytruda alone, and resulted in an impressive hazard ratio of 0.51. The combination of INT plus Keytruda also showed sustained improvement in distant metastasis-free survival at three years. 89.3% of patients in combination INT plus Keytruda treatment group were alive and without metastases or distant spread of their tumor at three years of follow-up versus 68.7% of patients in the Keytruda monotherapy group. This is an equally impressive and remarkable hazard ratio of 0.38. Lastly, we were pleased to share data showing an early favorable trend in overall survival at three years of follow-up. Finally on the safety side, INT continues to demonstrate a remarkable profile for a novel cancer therapy. Immune-related adverse events were not higher in the combination INT plus Keytruda arm despite the benefits than in the Keytruda monotherapy arm. This highlights the impressive emerging benefit-risk profile for our INT program. Moving now to rare diseases, I’m happy to share that our MMA candidate, mRNA-3705, was selected for the FDA START program. MMA is a rare disease in which patients cannot properly break down proteins from the food they digest; as a result, toxins build up in the bloodstream and cause recurrent episodes of life-threatening metabolic decompensation. The FDA START program is a program designed to accelerate development of new and promising therapies in rare diseases where there is a high unmet need. Our MMA candidate, mRNA-3705 is being evaluated in patients in a Phase I/II study with encouraging early results that we’ve previously shared, and we look forward to working with the FDA to accelerate the development of this promising potential medicine. With that, I’ll now turn it back to Stéphane.
Stéphane Bancel:
Thank you Stephen and Jamey. Let’s start with COVID. With the start of the COVID season only a few weeks away, contracting for the season is almost completed as we speak. In the U.S., we have seen increased competitive pressure during the contracting season compared to last year, and we have not finalized most of the contracts. We are working closely with public health officials, healthcare providers, pharmacies [indiscernible] with these stakeholders are critical in driving widespread vaccination adoption. In the EU, we are in advanced discussions on the tender framework to provide market access; however, based on recent feedback from several large governments, we are now expecting very low sales to EU member states in 2024. Some large countries will not buy Spikevax for the ’24 - ’25 season. In the rest of the world, we have multiple signed contracts in place, some of which could be deferred into 2025. Contracting discussions are ongoing with some additional countries, but we do not expect this will have a significant impact on our updated sales framework. Moving now to launch preparation for the ’24 - ’25 season, our organization is fully prepared for the upcoming season thanks to the substantial efforts made by the Moderna teams to ensure the global availability of COVID vaccines. In North America, we are ready to supply Spikevax targeting the KP2 strain, as asked by the FDA, demonstrating our ability to quickly pivot and manufacture Spikevax for selected strains. In the rest of the world, we are supplying vaccine targeting the GA1 strain as requested by health authorities in those regions. Let me now take you through details on the U.S. COVID market. We’ll be ready to supply millions of Spikevax doses to all segments of the U.S. market upon regulatory approval. Our goal is to ensure that Spikevax is available to all customers - large retail pharmacies, independent pharmacies, healthcare professionals, [indiscernible] hospital network and public health entities at the same time, facilitating early and widespread vaccination. [Indiscernible] the world healthcare system to be ready to vaccinate is a key priority for us this season. To this end, we have implemented a real-time order tracking system which is available to all of our customers and provides detailed information on every order, including [indiscernible] timing, package size, and temperature status. Our supply chain is using AI tools to estimate the optimal order of distribution to locations within the U.S. market. These preparations highlight our commitment to ensuring the broad availability of Spikevax and meeting market demand. Let me now turn to our marketing efforts. We believe education and awareness campaigns will be important drivers to increase vaccination rates. We have started these efforts early. Ahead of the season, we have launched a back-to-basics campaign. This initiative is designed to raise awareness about how the SARS-CoV-2 virus works and why it is critical to get a new booster each year, like for flu. The virus mutates over time and we need to have a new tool to teach your immune system the new mutation to reduce infection, reduce hospitalization, and reduce deaths. Additionally, we are working with public health authorities to educate the public on the impact of long COVID, highlighting how vaccines can help reduce risk for young and middle age adults. Soon, we will start our in-season outreach to high risk groups. By collaborating with major retail pharmacies, we aim to effectively market our products during the season. This targeted approach ensures that our efforts are focused on those who are the most vulnerable, maximizing the benefits of vaccination for those groups. By leveraging media channels, we aim to reach a broader audience and emphasize the critical need for vaccination to mitigate [indiscernible] outcomes. Let me now turn to our RSV launch. We are very excited about the launch of our second product, mRESVIA. Following approval, [indiscernible] vaccine recommendation on par with competitor vaccines. Specifically, ACIP [indiscernible] recommended a single dose of RSV vaccine for all unvaccinated adults age 75 and above. ACIP also recommended a single dose for unvaccinated adults between the age of 60 and 74 who are at increased risk. Based on these recommendations, approximately 40 million people in the U.S. are eligible for vaccination. The RSV market size beyond the ’24 - ’25 season will depend on re-vaccination recommendations which are anticipated to be discussed by ACIP in future meetings. In RSV, our focus is to direct our efforts to the segments where the vast majority of vaccinations occur. In the U.S., we’re targeting the pharmacy segment, which includes both large retail chains as well as independent pharmacies. Together, the total pharmacy segment accounted last year for around 95% of RSV vaccine administration. We began shipping product in pharmacy in July, and as Jamey mentioned earlier, we are seeing a highly competitive environment. Additionally, larger competitor contracts will negotiate prior to approval and launch, resulting in lower 2024 share than we would have liked. Long term, we continue to believe our [indiscernible] will resonate well with customers, offering them the ability to use their time, maximize efficiency during the busy fall vaccination season, and also reduce potential medical errors. We look forward to some major upcoming milestones in the near term. In respiratory vaccine, we expect a COVID approval and will be ready to ship in the U.S. and rest of the world in the August-September time frame. In RSV, we expect to release Phase III data for high risk individuals 18 years of age and above. We are in discussions with regulators also on our flu program and intend to file in 2024. We also expect to present immunogenicity data in older adults for our flu vaccine, Fluzone HD. For our next-gen COVID vaccine, mRNA-1283, we are engaging with regulators and intend to file in 2024. In our combination flu -COVID vaccine, mRNA-1083, we are engaging regulators and hope to be able to give an update soon. [Indiscernible] fully enrolled and [indiscernible] we look forward to potentially Phase III vaccine efficacy readout in 2024. As Stephen just talked about, we are very excited about the INT program and we are looking forward to the completion of enrollment of our Phase III in adjuvant melanoma, which is of course a major milestone. We are keen to discuss with regulators the possibility of accelerated approval based on the Phase II study data. As we shared before, they are treating the review as necessary before we could consider pursuing accelerated approval for INT
Operator:
Thank you. [Operator instructions] Our first question comes from Salveen Richter with Goldman Sachs. Your line is open.
Salveen Richter:
Good morning. Thanks for taking my question. A couple from me here. Can you help us understand the factors contributing to maintenance of the year-end cash balance guidance following the lowered product revenue guidance range? Then with regard to competitive pressures noted regarding contracts for COVID into second half, can you be more specific on the factors here, whether it’s contracting logistics or the clinical profile competitive dynamics, or what that might be? Third, how much visibility do you have on second half demand for the RSV vaccine based on contracting to date? Thank you.
Jamey Mock:
Hi Salveen, I’ll take the first question regarding the year-end cash balance. I would say there’s a few factors in there. Number one, on the deferrals of revenue, some of which we have already collected the cash and ultimately we will collect the remaining balance that might push into 2025, but some of that is already a prepayment. Number two, as I mentioned in my prepared remarks, we have been working on working capital. We’ve got a whole team focused on it, and the teams are doing a great job from accounts payable, inventory balances, receivables and our collections progress. You can see our receivables balance on the balance sheet is relatively small at this point, so the team’s done a great job, which helps offset it. Then third, just coming into the year, we had a little bit of cushion to the $9 billion, so overall we remain confident in the $9 billion and are pleased with that kind of ending balance heading into 2025.
Stéphane Bancel:
And Salveen, I’ll take the two or the question--it’s Stéphane. On the competitive pressure, I think on both products, COVID and RSV, we are seeing similar things, which is [indiscernible] from larger competitors with a portfolio of products, an ability, if you want, to also be much more aggressive on activities, whether it’s supply chain, whether it’s pricing, whether it’s co-marketing funding and dollars activity, so it’s really a mix at the customer level. We’ve just seen much more intensity and pressure on COVID versus last year, and on RSV, as you know, we’re entering a market where there’s two large established players, one with very large market share where of course [indiscernible], the other one who, as indicated, they want to gain market share because they are not pleased with their market share last year, and so we have been working really actively on that. In terms of visibility for RSV, we have some contracts that have already been signed and that we are actively supplying customers now that the product has shipped. We have some contracts that are being finalized with ability in season to demonstrate to the customer the value of PFS and to be able to move that into season, so we are working through all of those things. We’ll provide updates as the season goes, but we know this is very important for patients, it’s very important for the company, so we’re all hands on deck on COVID and RSV.
Operator:
Thank you. Our next question comes from Terence Flynn with Morgan Stanley. Your line is open.
Terence Flynn:
Great, thanks for taking the questions. Maybe two for me as well. Was just wondering on the guidance cut, if you can quantify how much was from COVID versus RSV, and then on the seasonal flu and the seasonal flu plus COVID combo, I might be parsing words here, but on seasonal flu, I notice the press release said intend to file in 2024, the combo said engaging with regulators on next steps, so maybe you could just help us think through the gating factors for each of those and the difference in language there. Thank you.
Jamey Mock:
Yes, thanks Terence. I’ll take the first one related to guidance and what’s the split between COVID and RSV. What I would say is if you look at the three drivers, all of them are, first, similar in size. Then if I break them down, the deferrals are all related to COVID, the EU is all related to COVID, and then that last category of competitiveness in RSV vaccine--or in respiratory vaccines, there’s a split between COVID and RSV, so that should give you some understanding of the general split between COVID and RSV.
Stephen Hoge:
Great, and for the question on the language, good pick-up. The 1083 flu COVID program is the most recent Phase III results, really quite fresh, and we have just begun the process of engaging with regulators, meaning sharing that data and discussing with them what their expectations would be on submission. For the other programs that you referenced, obviously we’ve had more time, had some of those discussions, and we’re obviously preparing for submissions, as we said in our prepared remarks.
Operator:
Thank you. Our next question comes from Michael Yee with Jefferies. Your line is open.
Michael Yee:
Thank you guys. Two questions from us. Looking forward on guidance, you obviously have revenue guidance this year, you have opex guidance which we see is around $6 billion of R&D and SG&A, and as you project out to the following years, you have already given cash flow guidance, so if things are generally staying along the same trajectory, I would think that cash guidance would be lower. Can you speak to the math there, and perhaps Jamey, who has said you could flex opex, is that of heightened importance and heighted priority more aggressively as you think about where things stand today in the change of guidance and talk to that, given cash guidance is a big concern for investors? The second question is going back to RSV - I appreciate the comments you just said, that was super helpful in breaking it down. Is that to say that the RSV projections you have are going to be perhaps in half, and is that due to lower share and lower price, and speak to that? Thank you.
Jamey Mock:
Yes, thanks Mike. We are still--as I mentioned, we’re still expecting $9 billion in cash next year and don’t currently have any different change in our outlook for the $6 billion to $7 billion that we said we’d end 2025 with, and let me break that down. First, this year we went from the start of the year, $13 billion to $9 billion, so a $4 billion loss in cash. That included many prepayments coming into the year that will no longer repeat, so we’re not actually getting the cash from operations and that won’t be a drag year-over-year. Second, we plan to return to growth in 2025 and, as I mentioned earlier, some of that will also be buoyed by the fact that we have some deferrals heading into 2025, but more so we’ll have an entire year and operating experience on RSV, we hope to bring new products to market, and so we’re still expecting to return to growth. But yes, I mean, we continue to look at opex and understand what we want to do there, and we are always laser focused on our cash balance, and at this time we don’t think the $6 billion to $7 billion will change.
Michael Yee:
Okay, and then on RSV, breaking it down, just a little more specifics on the impact of market share contracting and price, and how to think about that?
Jamey Mock:
Yes, it’s a bit of both. We’re not breaking it down, nor have we given guidance on RSV, but I would say this year is not turning out as we expected, and I answered Terence’s questions in terms of how much is really split between COVID and RSV, so you can kind of get an understanding there. But again, we were third to market this year, some of the contracts were already negotiated, we only are participating in the second half, but I think our hope is that we get access across many of the retail chains, they start to get comfortable with us with the second product so that when we head into 2025, we can get a more fair market share on RSV.
Michael Yee:
Perfect, thank you.
Operator:
Our next question comes from Eliana Merle with UBS. Your line is open.
Elliott Bosco :
Hi, this is Elliott Bosco from UBS on for Eli Merle. Two from us. Can you elaborate on the latest timing you expect for the Phase III CMV study, based on how event rates are tracking, and based on just the necessary 12 months of median safety follow-up, from a purely protocol perspective, what is the earliest we could potentially get the interim? Then second on international COVID revenues in future years, what do you see as the likelihood of potential deferral again in 2025?
Stephen Hoge:
Great, thanks for the questions. I’ll take it first. First, we have enough data on case numbers for CMV. We may have an update at R&D day - I’m not sure, but we don’t have any change to our prior guidance, which is we do think that the interim analysis of efficacy could happen this year, and that would account for also median safety follow-up from a timing perspective, and so at this point, no new update, but we continue to stand behind the belief that the interim analysis for efficacy on the CMV program could happen this year.
Jamey Mock:
Yes, and Elliott, on the deferrals, could they happen in 2025, the answer is yes, they can always happen. That said, some of this will push into next year, so, and if some of those push into the following year, those two would offset, but we’re not expecting that at that point. The only thing I’d say is we will come to market with our resilience contracts in the U.K., Australia and Canada, that provides us additional growth in those areas as well as potentially participating in additional public tenders as well, so there’s a little bit of risk but I think we can mitigate it with the deferrals from this year, as well as additional resilience contracts.
Operator:
Thank you. Our next question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
Thank you. I just have one question regarding the guidance for this year, particularly regarding the COVID revenue in the U.S. It seems contracting is mostly down, and you already also expect similar vaccination rates versus last year, and we know there is mainly only two players, so what could lead to the lower COVID revenue this year from prior [indiscernible] guidance?
Stephen Hoge:
Yes, thanks Gena. We define competitiveness both in terms of share and in price, so we’re relatively pleased with our market share, there’s potentially some price pressure in there. But generally speaking, we think that the U.S. will perform pretty well compared to last year, but we are seeing a bit of a competitive--a more competitive market this year after a year where in the U.S., we had nearly or approximately 50% market share, so. We’re seeing the pressure but still confident in the overall performance of the U.S. business.
Gena Wang:
Thank you.
Operator:
Our next question comes from Luca Issi with RBC Capital. Your line is open.
Luca Issi:
Oh, great. Thanks so much for taking my questions. Maybe Stephen, quick question - the recent ACIP meeting, it feels to me that the debate before that ACIP meeting was whether RSV was going to be either an annual vaccine or an every other year vaccine. However, ACIP clearly chose neither of them and simply said, this is one and done, so two questions. Were you surprised by that decision, and two, how should we think about the market potential for RSV long term, in light of the fact this is maybe a one and done vaccine versus obviously COVID and flu are annual vaccines? Any color there would be much appreciated, and then maybe quickly walk us through what’s the latest thinking on the opportunity for bird flu. Thanks so much.
Stephen Hoge:
Great, thank you for both questions. Obviously first and foremost, our focus at the ACIP meeting was we were quite pleased with the parity recommendation, and I think we were all pleased with the clear recommendation for those over the age of 75 and those with higher risk from RSV, that they really should get vaccinated. As its only a year old is the market, the most important thing we can do is cover the large number of people who are now recommended, the 40 million-plus who are not vaccinated, and that’s quite a large increase in the number of folks who we hope to cover soon. Now, on the question--the specific question of revaccination, I think it’s also important to say that we are--you know, the ACIP is really just looking at one year of real-world data, and all three of the vaccines, but including mRESVIA, show significant second season protection, although it does decrease, and so there is the decline from season one into season two from a clinical trial perspective, but there’s still protection there in that year two. While you’re only one year into that public experience, public health experience, I think ACIP took a prudent choice and said there’s still a benefit, and maybe the focus should be on increasing vaccination coverage rates for those who are current unprotected because there clearly is some benefit, even still at that second year. I think if you fast forward a year, and this would be my perception, but if you fast forward a year, if you look at the rates of waning for the other two vaccines than ours, there is clearly a decline from year one to two, and from year two to three. The pace of that decline suggests that by year three, there really won’t be much protection for those people who have received vaccines in the first year. I think that’s when you probably need to start asking the question of, do we continue to leave those people unprotected or boost them again? Now, from a scientific perspective, you asked my view, you will be infected with RSV almost 20 times in your life, you will repeatedly get ill. In fact, many people who are at risk of severe complications over the age of 75 or otherwise with medical co-morbidities, they’ve seen RSV before. It’s the waning protection from that infection that ultimately is the reason why you need a vaccine, and because you get infected multiple times by that virus over life, even if you’ve seen it before, you probably will benefit from a booster in future. We continue to believe that as the public health story evolves, that recommending bodies and CDC and ACIP will look at the waning efficacy, the potential to boost people again and hopefully provide additional public benefit, and that eventually there will be recommendations for revaccination for those who are at highest risk of RSV, but it is not our choice. We are providing that data to obviously the regulators but principally to groups like CDC, and then they will determine the right moment, if ever, to recommend that revaccination in the United States. As it relates to bird flu, we continue to follow that very closely. It’s drifted a little bit out of the news more recently - I think that’s a good thing, but the most important news for us has been in the quarter, we executed an agreement with BARDA to advance into Phase III with our pandemic bird flu vaccine, which we’ll provide updates on as we move forward in the months and the year ahead. But we will be sure to be partnering with public health entities across the world, including the United States government specifically. We’re grateful for working with BARDA again in the event that bird flu does emerge as a pandemic or epidemic threatening this country.
Luca Issi:
Thanks so much.
Operator:
Our next question comes from Alexandria Hammond with BofA. Your line is open.
Alexandria Hammond:
Hi, thanks for taking my question. Ahead of results for VX-522 in collaboration with Vertex for CF, if results provide positive proof of concept for the ability of mRNA and liquid nanoparticles to be aerosolized and cross the mucus barrier in the lung, can you walk us through additional indications you may be interested in, and should we expect you to develop these indications in partnership with Vertex? Thank you.
Stephen Hoge:
Thanks for the question. We have--as you highlight, there is a large number of pulmonary diseases for which respiratory delivery could be quite impactful. We have not provided any updates on the preclinical programs that we’ve moved into development, and so I would describe those activities right now as still research and discovery phase, and not something for competitive reasons and for scientific reasons that we’ll talk about. They’re in the earlier stages. As it relates to whether we would partner with Vertex on it, we would of course always welcome partnering with Vertex given their expertise, but the partnership we have right now, the deal we have with them is limited to the cystic fibrosis program at this point.
Operator:
Thank you. Our next question comes from Edward Tenthoff with Piper Sandler. Your line is open.
Edward Tenthoff:
Great, thank you. Most of my questions have been answered, but I appreciate all the detail. I wanted to ask a little bit about the orphan disease program, and maybe you can provide a little bit of color on what the regulatory path might look like in MMA and PA. Thank you.
Stephen Hoge:
Thanks Ted. They are somewhat different, and they are in early stages of those regulatory conversations, so. We just received the FDA START designation, but if you look at the data that we shared last R&D day, and we’ll obviously provide updates on this going forward, one of the clearer pictures that emerged was that in MMA, we are seeing good movement of a biomarker of methylmalonic acid that’s pathognomonic for the disease, and so you can imagine of the challenges very quickly when you have a good biomarker is how do you validate that biomarker and show that moving that biomarker is reasonably likely to predict benefit, and that’s a scientific question we will work on and ultimately engage with regulators, including FDA, as a key step for moving forward. We are evaluating in the clinical trial folks movement in their biomarker from baseline, so it’s a clear study to run. In the case of proprionic acidemia, there is not as clear a biomarker in the field because of the structure of that particular part of the metabolic pathway, and so there we’ve been following events, and as you know, we’ve previously shared in a single-arm study the pre and post treatment rates of metabolic decompensations. We have seen a favorable trend in that - we’re actually quite excited by that. That takes a little more time than a biomarker, but we obviously have been working on that a little bit longer, and so we’ll be engaging with regulators on that single arm approach and understanding the difference between internal controls or other controls to evaluate that. In any event, you may end up with a randomized study, you may be able to use a single arm study, and in the case of MMA, you may be able to use a biomarker, so those are the kinds of conversations we’re having, but as I said, they’re early days. The FDA discussion on MMA just started, and so we’ll provide updates as we have more clarity on those timelines.
Edward Tenthoff:
Great, thanks. Looking forward to the R&D day.
Operator:
Our next question comes from Jessica Fye with JP Morgan. Your line is open.
Jessica Fye:
Hey guys, good morning. Thanks for taking my questions. For RSV, I’m curious your take on Pfizer’s comment that customers want to carry an RSV vaccine that could address both maternal and the older adult population. Then for INT, I saw it on the near term milestone slide, but to clarify, what’s your latest projection on when you expect to complete enrolment of the Phase III adjuvant melanoma trial, and what’s the latest you can share on manufacturing progress for that product? Are there any updates to share on how that’s going? Thank you.
Stephen Hoge:
Sure, I’ll take that. Obviously the majority of the recommended population in this country continues to be the older adults. I wouldn’t comment on Pfizer’s competitive positioning on our quarterly call except to say that in the retail channel overwhelmingly and in many physicians’ offices overwhelmingly, the focus is on protecting older Americans. What’s happening with the younger population, maternal population is obviously smaller in terms of an overall market opportunity. We do recognize the need to try and provide the broadest potential label and, as we’ve previously updated, we are pleased that we’ve conducted our Phase III study in 18 to 59-year-old high risk populations, and we intend to file that this year with a goal of expanding that label for those who are at high risk for medical reasons and may benefit from an RSV vaccine, even if they are younger. I would say we agree that a broad label is valuable, but I think it’s important to cover as many people as possible, and that’s what we’re excited by moving forward with our 18 to 59-year-old filing. On the question of INT, we have not with our partner updated yet on where we are in enrolment. Obviously as we had said at ASCO, and there was quite some coverage of risk, we are pleased, very pleased with the pace of enrolment, and we do expect that to conclude quickly, but we have not yet provided any updates and we’ll obviously do so at the appropriate time with our partner, Merck. As it relates to manufacturing, we actually have made great progress there establishing our Marlborough facility, as well as demonstrating through the clinical trial our ability to manufacture at high volume, frankly commercially relevant volumes, and so we’re--we feel like we’re in a good place in terms of the manufacturing trajectory. It’s still a few months of work to go this year to get to where we want to be, but we’re starting to feel quite optimistic that the manufacturing will be online as and when we hope.
Jessica Fye:
Thank you.
Operator:
Our next question comes from Evan Wang with Guggenheim Securities. Your line is open.
Evan Wang:
Hey guys, thanks for taking the questions. A few from me. Just first, just given the comments on the higher competitive environment, how are you thinking longer term about the outlook for RSV and COVID versus some of the [indiscernible] you made earlier in the year and historically, just given some of the pricing and market share dynamics there? Second, with COVID and the EU tender, what’s your level of confidence in the EU being a meaningful contributor in 2025, and what changes between now and then? You know, does having the combo or potentially having a combo add to competitiveness there? Lastly, I saw that you guys purchased a PRV - is that for use on the combo or the standalone flu, and are you comfortable in getting ahead of the potential June recommendation next year? Thanks.
Stéphane Bancel:
Thanks, this is Stéphane. On the high competitive environment on COVID and RSV, what we believe is on RSV, our customers are going to experience the PFS product, and that’s going to be ability to really have full season, to be able to try and get the products to a channel that will have lots, so we anticipate in ’25 to have better share than in ’24 for the U.S. We’re going to be launching probably outside U.S. markets in ’25, that’s of course we’ll not have sales in ’24, so that’s all going to be important for growth. In COVID, we believe the portfolio in terms of next-gen COVID and then flu plus COVID, it’s going to be an important driver to kind of reset the expectation moving forward and the market dynamics. In terms of the EU, the current contract between Pfizer and the EU ends in 2026, so we think 2025 and 2026 are still going to be low. Some countries have actually used a lot of our vaccine and so we could see some countries in ’25 and more in ’26 needing COVID vaccine and then wanting to diversify their supply base [indiscernible] vaccine as well, given its performance, so as you mentioned the combo, it’s something that we have been discussing quite a lot with governments in Europe. They see the value of a combo given its strong performance, as we’ve shared, to be an important tool for public health in terms of compliance. In a world where governments are worried about people getting their flu shot and their COVID shot and their RSV shot, that’s just a lot of shots and nobody likes needles, nobody likes to go to the doctor or to the pharmacist, and so the combo is something that customers are really valuing, so that could be another opportunity in countries when this product is available to go back into growing Europe. The key priority for us in Europe now for 2025 is really launching RSV and growing the business on RSV until we see the flu mono or flu plus COVID launch.
Operator:
Thank you. Our next question comes from Tyler Van Buren with TD Securities. Your line is open.
Greg:
Hi there, this is Greg [indiscernible] from TD Securities. I am more interested in your thoughts on the magnitude of the RSV market. [Indiscernible] small in relation to last year during earnings, so do you agree, and what is your latest thinking on the overall sizes of the RSV market in elderly patients versus the prior that you built you’re your expectations?
Lavina Talukdar:
You broke up a little bit during your question. I think we caught the tail end of your question, which is what is the size of the RSV market relative to what we though previously?
Greg:
Yes, thank you. Just to reiterate quickly, I believe that GSK discussed the RSV market being smaller this year relative to last year [indiscernible]. Just didn’t know if you agree with that and what was your latest thinking on the overall size versus the prior $6 billion to $8 billion [indiscernible].
Stéphane Bancel:
Yes, so let me try to take a stab at it. As Stephen stated, in the long term we believe that as real-world evidence data is gathered by public health leaders, that they will most probably be in for boosting. The current recommendation is what it is, but as Stephen said, we believe it will potentially evolve over time. For this year, I think what is interesting is on paper, it seems that it could be a smaller market. The thing that’s going to be interesting to see how it plays out in terms of market size and number of doses in arms is the guidelines are much more clear than last year, and as you know, sometimes in vaccinations, you get a better reaction from the doctors and pharmacists and consumers with clear guidelines versus not as clear guidelines with larger population potential. Because of those two factors, it’s going to be interesting to see how the season plays out.
Greg:
Thank you.
Stephen Hoge:
Then I would just comment on the current year market size as well. I think we talked about what we believe the patient population is, which is still sizeable, and we believe that the market will still be overall sizeable this year and similar to last year, if not a little bit more. I think there are other factors at play with those comments - you know, there was a lot of inventory that was--or a lot of sales that happened in the prior year, that customers may be sitting on some of that inventory, so from a revenue perspective that might be different, but from a vaccination rate perspective, we believe that it will still be similar, if not a little bit bigger this year.
Operator:
Our next question comes from Cory Kasimov with Evercore. Your line is open.
Adi :
Hi, good morning. This is Adi on for Cory. I have a question on has the recent summer COVID surge caused any concern for fall vaccinations as the infected population is usually recommended not to get vaccinated for six months post infection, and is the base case for the U.S. COVID revenue similar vaccination as last year, which I think per CDC tracking was 22.5 overall population and 14% for 65 and above?
Stephen Hoge:
Yes, I’ll take it. First, the question on the summer wave and the epidemiology, we’ve seen this multiple years in a row now, and so in some ways, there is a move in certain geographies towards a little bit of a summer wave. It just highlights the fact that this virus is incredibly effectively at spreading, incredibly effective in creating disease even for folks many years after vaccination and other protection, and why we need to protect people for the winter season, because whatever we see in the summer, you see a dramatically higher wave in the November through February time horizon, and so that’s really where we’re trying to protect people for. But in terms of does the summer wave impact, the small summer wave impact the view of the fall wave, it really doesn’t change year-over-year that perspective, and that’s why we’re confident vaccination coverage rates will still be there, and as Jamey said, we see them the same. We hope to do better, but we see them minimally as similar to last year in the United States.
Operator:
Thank you. Ladies and gentlemen, this does conclude the Q&A portion of today’s call. I’d like to turn it back to Stéphane Bancel for any closing remarks.
Stéphane Bancel:
Well, thank you everybody for joining us, and we look forward to speaking to many of you in the coming days and weeks, and if not, seeing you for R&D day on September 12. Have a great day, bye.
Operator:
Ladies and gentlemen, this does conclude today’s presentation. You may now disconnect and have a wonderful day.
Operator:
Good day, and thank you for standing by. Welcome to Moderna's First Quarter 2024 Conference Call. [Operator Instructions] Please be advised, this conference being recorded.
I would now like to hand the conference over to the speaker today, Lavina Talukdar. Please go ahead.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's First Quarter 2024 financial results and business updates. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website.
On today's call are Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; and Jamey Mock, our Chief Financial Officer. Before we begin, please note, this conference call will include forward-looking statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thanks, Lavina. Good morning or good afternoon, everyone. Thank you for joining us today. I will start with a review of our business. Jayme ill then present our financial results. Stephen will review our late-stage clinical programs, and I will close by sharing our 2024 commercial priorities and major upcoming milestones.
Our COVID vaccine has already impacted hundreds of millions of people. I'm excited by the progress we've made with our pipeline that has the potential to impact many more people. During the first quarter, we presented substantial clinical progress during our Vaccine Day, with exciting data on EBV, VZV and Norovirus. In addition, along with our partner, Merck, we expanded studies for Individualized Neoantigen Therapy, INT into 3 new indications. In addition, through ongoing Phase III studies in adjuvant melanoma and adjuvant non-small cell lung cancer, a Phase II/III study has started in neoadjuvant, adjuvant, cutaneous squamous cell carcinoma, another form of skin cancer. Phase II clinical trials have started in adjuvant bladder and adjuvant kidney cancer. Together, our vaccines and therapeutic portfolio have a potential to impact hundreds of millions of people each year. I am pleased with our Q1 performance. Since the beginning of the year, we announced 4 important business agreements and collaboration. We entered into a nonexclusive IP out licensing agreement with a leading pharmaceutical company in Japan. The agreement includes an upfront payment and low double-digit royalty to Moderna on net sales of our COVID key products marketed in Japan by this company. It is nice to see a company recognizing our IP and our figures for our license. Second, we recently announced a contract to provide 4.5 million royalty of COVID-19 vaccine to the Ministry of Health in Brazil. I am very pleased with this partnership as it is the very first time that Moderna works with a broad environment, and we look forward to providing these doses to protect people in Brazil as they go into their winter season. We announced the project financing program for up to $750 million in funding to Blackstone to further develop our flu program. We also made public our collaboration with OpenAI to use AI as a transformative tool to increase speed and efficiency and ultimately to improve patient outcomes across our business. Finally, we agreed with Metagenomi to terminate our gene editing collaboration. All rights granted under the collaboration will be returned to Metagenomi. This is a good proof point of Moderna's continually aim to prioritize our investment for our best opportunities to drive returns. Turning to Q1 financial results. In revenues, we were ahead of our plans at $167 million, reflecting a highly seasonal nature of our respiratory vaccine business. The net growth was $1.2 billion. We ended the quarter with $12.2 billion of cash and investments. We communicated during our November call, our focus on financial discipline. I am pleased with what the team has achieved with our operating expenses, cost of manufacturing expenses plus cost of R&D expenses plus cost of SG&A expenses were down almost $800 million in Q1 2024 versus Q1 2023. Jamey will elaborate on this in his section. With that, I will now hand over to Jamey.
James Mock:
Thanks, Stéphane, and hello, everyone. Today, I will walk you through our financial performance for the first quarter and provide commentary on our 2024 financial framework.
Let me start with our commercial performance on Slide 8. Net product sales for Q1 were $167 million, down 91% year-over-year, mainly driven by lower sales volumes of our COVID-19 vaccine in regions outside the United States. This decline aligns with the anticipated transition of the COVID-19 vaccine market or it's a seasonal pattern, whereas in the first quarter of 2023, we primarily delivered doses that were deferred from 2022. Q1 was driven by sales in the U.S. and the Rest of the World, largely Latin America markets. For Q2, we expect about $100 million in sales for a total of approximately $300 million in the first half of 2024. Q2 will include a portion of our recently announced contract with Brazil. Moving to Slide 9. Net product sales were $167 million, as I just explained. For the first quarter of 2024, our cost of sales was $96 million, which included third-party royalties of $8 million, inventory write-downs of $30 million and $27 million related to unutilized manufacturing capacity and wind out costs. This resulted in our cost of sales representing 58% of net product sales, up from 43% in the same quarter last year. The increase in cost of sales percentage was primarily due to the lowest level of sales in the quarter. We continue to expect the full year cost of sales to be approximately 35% of product sales. However, due to the strong seasonality of our business, we expect a higher percentage in the first half. Moving to our R&D efforts. Q1 R&D expenses were $1.1 billion, reflecting a decrease of 6% year-over-year. This reduction was primarily due to the absence of upfront collaboration payments being made this quarter. The upfront payments made in the first quarter of 2023 were related to our strategic collaborations with Generation Bio and Life Edit Therapeutics. With the Q1 spend of $1.1 billion, we are tracking towards the full year expected spend of approximately $4.5 billion. Q1 SG&A expenses were $274 million, marking a 10% decrease year-over-year. Importantly, this decrease was driven by all functions in SG&A, and it is a result of our strong focus on cost discipline and strategic investments, driving productivity. I will provide additional color on the next page. We reported an income tax expense of $10 million for the first quarter of 2024 compared to an income tax benefit of $384 million in the same period last year. The shift is primarily due to the continued application of valuation allowance on the majority of our deferred tax assets, which we first established in the third quarter of 2023. Net loss for the period was $1.2 billion compared to net income of $79 million last year. Diluted loss per share was $3.07 compared to diluted earnings per share of $0.19 in 2023. We ended the first quarter with cash and investments totaling $12.2 billion, down from $13.3 billion at year-end 2023, largely attributable to research and development expenses and operating activities. Moving to Slide 10. I want to take a moment to elaborate on the efficiencies we are now seeing across the company. As a platform company, we have the opportunity to build a unique operating model. And over the last few years, we have invested purposefully into people, processes and technologies to build foundational capabilities that will allow us to scale efficiently. First, we ended 2023 with nearly 6,000 employees, up from 1,300 at the end of 2020. Every function scaled capabilities to enable the increasing product launches we expect over the coming years. Additionally, as you know, Moderna has always led with a digital-first mindset. Over the past 3 years, we have nearly doubled our built-for-purpose software applications to digitally enable our teams. As an example, we recently went live with the newly implemented rebuilt ERP system. SAP S4/Hana is our new digital backbone for all our operational activities. We have used SAP in the past, however, it was built for a research and development-focused company. And now we have implemented an entirely revised version, supporting our end-to-end business processes more effectively and efficiently. Another example is our rapid adoption of artificial intelligence. Over the past year, we have built over 750 GPTs. One example in the legal space, our contract companion GPT streamlines the task of reviewing and summarizing contracts across the business. With the GPT providing step-by-step guidance to craft a tailored and insightful salary. This enables any function to extract critical insights on contracts whenever needed, minimizing bottlenecks and freeing up Moderna's legal department to focus on work of higher strategic value, thus enhancing operational efficiency and decision making. Another example in G&A is a big purchase of paid GPT for all questions around our procurement and payment processes. Instead of our employees having to find and read policies and procedures, they can easily query to GPT. It also saves time for our procurement and payables teams from answering numerous questions. AI has already been a good chance for win in short period of time. In general, we see the areas helping incredible speed that allows for an unprecedented impact on productivity and many more. We have rolled out a comprehensive training program and are committed to driving this technology breakthrough. 1 As a result of these strategic investments in the people, processes and technology, we were able to significantly reduce purchase services and our use of external consultants, which contributed heavily to the 10% year-over-year reduction in SG&A spend. We are also seeing similar benefits in R&D and manufacturing. In general, we now have a solid foundation with our operating model. As we continue to grow our commercial activities, we will need to further invest, however we will be able to do that more efficiently. Now let's turn to the 2024 financial framework on Slide 11, which is in line with what I shared on our last earnings call in February. We continue to expect net sales for 2024 of approximately $4 billion, which we think will be a low point as we expect to return to growth in 2025. Sales in the first half of the year are now expected to be approximately $0.3 billion. We continue to expect cost of sales of approximately 35% of product sales for the full year. For R&D we continue to expect full year expenses to be approximately $4.5 billion, down from $4.8 billion in 2023. For SG&A, we continue to expect full year expenses to be approximately $1.3 billion down from $1.5 billion in 2023, and we expect taxes to be negligible in 2024 and capital expenditures in 2024 to be approximately $0.9 billion. Finally, we expect to end 2024 with approximately $9 billion in cash after touching a low point of approximately $8 billion at the end of Q3, due to the seasonality of collections. Finally, let me also touch on our recently announced project financing deal with Blackstone, which we are excited about. In March, we entered into a development and commercialization funding arrangement, which commits Blackstone to providing us with up to $750 million of funding for our flu program, so that we can strengthen the product label and fulfill our remaining regulatory applications. Subject to the regulatory approval in the United States, which depends on data from the funded activities, Blackstone will be entitled to receive up to $750 million in sales milestone payments. These milestone payments are contingent upon achieving specified cumulative net sales targets for our future influenza and combination vaccines. Additionally, Blackstone will earn royalties on applicable net sales at a low single-digit percentage rate. This funding will offset our R&D expenses and is factored into our R&D framework for the year of approximately $4.5 billion. Overall, we are excited that this deal enables us to accelerate the advancement of our pipeline. And with that, I will now hand the call over to Stephen.
Stephen Hoge:
Thank you, Jamey. Today, I'll review updates from our clinical programs that were shared during our recent Vaccines Day, as well as new developments in our therapeutics portfolio.
Starting with respiratory vaccines. We shared updates to many of our respiratory programs at Vaccines Day in March. Our RSV vaccine in Canada is undergoing regulatory review in multiple countries. And pending approval, we expect to launch the product in the United States following the June ACIP meeting and recommendations this year. At Vaccines Day, we shared updates from co-administration studies of RSV, confirming the ability to administer our vaccine and other vaccines given during the respiratory season. With our flu program, we recently presented data from our Phase III P303 study at ECCMID and continued discussions with regulators globally towards the goal of filing this year. For our next-generation COVID vaccine, mRNA-1283, we have presented positive Phase III safety and immunogenicity data and are engaging with regulators on the path to approval for that product. Our combination flu and COVID vaccine, mRNA-1083 is in Phase III, and we look forward to sharing those clinical data in the current quarter. Turning now to our leading and other vaccines. As shared at Vaccines Day, we've had -- we've made significant progress in this portfolio. Our CMV vaccine, mRNA-1647, has fully enrolled its Phase III trial, and we have the potential for an interim analysis of efficacy this year. We announced positive Phase I immunogenicity and safety data from our EBV vaccine candidate, mRNA-1189, and we are now advancing towards pivotal trials with that program. A second therapeutic EBV candidate, mRNA-1195 is in a separate ongoing Phase I study. mRNA-1468, our vaccine against varicella-zoster virus showed strong immunogenicity, including strong T cell responses, and we are preparing to move that program forward towards a pivotal Phase III study as well. And our HSV vaccine against Herpes simplex mRNA-1608, is now fully enrolled in its Phase I/II study, and we look forward to sharing clinical data updates when that's available. Now rounding out this portfolio, we presented the positive clinical data from our norovirus vaccine candidate, mRNA-1403 and shared that we are advancing that program towards its pivotal Phase III trial. Turning now to Oncology Therapeutics. We are happy to report our ongoing Phase III studies are enrolling well. We were excited to announce 3 new INT trials, including a randomized Phase II/III study in neoadjuvant, adjuvant cutaneous squamous cell carcinoma; a randomized Phase II trial in adjuvant high-risk muscle invasive bladder cancer; and lastly, a randomized Phase II trial in an adjuvant renal cell carcinoma. Now recently at AACR, we presented Phase I data from our INT program in advanced unresectable HPV-negative head and neck cancer in the metastatic setting. At AACR, we also presented Phase I translational data from another oncology therapeutic program, mRNA-2752 in various tumor types. Links to both of these presentations are provided on the slide. Now as a final note, at ASCO, we'll be hosting another Moderna oncology event on the evening of June 3, and we look forward to seeing you there or having you join us virtually. With that, I'll turn it back over to Stéphane.
Stéphane Bancel:
Thank you, Stephen and Jamey. Slide 18 is an overview of our COVID-19 strategy for 2024, which is focused on the need of each 2 region.
In the U.S., our focus is working with public health officials, health care providers and pharmacies to increase vaccination coverage rates. In Europe, we are actively participating in the 2024 tender process. The tender allows for up to 36 million doses per year for up to 4 years. And in the Rest of the World, we have reoriented our commercial teams to prioritize markets for greater commercial focus and impact. As mentioned earlier, the Brazil contract is an example of how this is working. In the fall of 2023, U.S. COVID vaccination rates lagged behind flu-vaccination rates. U.S. COVID vaccination rates were 11%, with flu vaccination rates at 4x that. And yes, COVID continues to show a higher [indiscernible] dieses. U.S. operation for COVID began October 2023. And last week, we have 424,000 people, which is markedly higher demonstration from either flu or RSV infection. Actually, COVID operations were around the same level as [indiscernible] of flu plus RSV combined. In addition, long COVID continues to be a serious risk to many healthy young adults in their 20s, their 30s, their 40s are losing lung capacity and the mental capacity due to long COVID. The data shows that COVID-19 vaccine reduced the risk of long COVID by 70%. We believe education and awareness will be very important. We are working on educating consumers about the need for an annual COVID vaccine, just like flu. Too many people are getting hurt when we have safe and effective vaccines available. Our job will not stop until these vaccination numbers come down significantly. As you know, strength selection by health authorities received approval and launch of COVID vaccine. Health authorities including the WHO and EMA in Europe, have recently selected the JN.1 strain for the 2024-25 formula. The FDA will launch the [ real fast ] meeting on May 16 to select the strength for the U.S. market. Moderna has already manufactured JN.1 drug substance to support the potential August launch. We have also prepared for backups in case the FDA does not select JN.1. In 2023, COVID vaccines were available 5 weeks later after 2 vaccines. In the recent channel alone, more than 3 million flu vaccines were administered before the updated COVID vaccines were available. As we look into the fall 2024 season, we see the potential to align the timing of flu and COVID vaccine approvals. We are encouraged by the earlier [indiscernible] meeting for this year's COVID trend selection versus last year. And we're working here and [indiscernible] for timely COVID approval. We expect higher vaccination uptake if COVID vaccines are available sooner. Turning now to the anticipated launch of our second respiratory vaccine, or RSV vaccine, which is expected to launch into a large market. In its first year, the older adult RSV market was $2.5 billion in sales, and analysts expect the older adults' market to grow between $6 billion and $8 billion per year. With marketing application filed in markets globally, we are anticipating approval beginning in the first half of 2024. In the U.S., we're targeting a launch after the June ACIP meeting. We are very excited to bring a product from a strong differentiated profile to market. Our vaccine has a strong efficacy and safety in clinical trials, and we'll be the only product available in the pre-lled syringe or PFS presentation. Let me now double-click on what we believe are the benefit of PFS. We recently published a time and motion study that shows faster preparation time for PFS relative to vaccines that require constitution. We call that both RSV combination vaccines on the market require multiple steps to prepare their vaccines for administration. One vaccine requires process and the overall next steps to prepare. Our PFS presentation is ready to use vaccine straight out of the box. The study from the PFS presentation to be 3x to 4x more efficient as measured by preparation time. Details from the study can be found through the link on the slide. We believe our PFS presentation for RSV vaccine has the potential to ease the personal burden on pharmacies during the fall respiratory season. The pharmacy chains [indiscernible] independent pharmacies and we're looking forward to the launch. Let me close with major upcoming pipeline milestones. While we're excited about the commercial prospects for the year, we're even more excited about the upcoming pipeline milestone and the effect they will have on our commercial outlook for the next several years serving patients. In respiratory vaccines, we are eagerly awaiting the approval of RSV and the [indiscernible]. We are also waiting for data for RSV in the age group 18 and above. We are in discussion with regulators on our flu program and intend to file in 2024. We won next-gen COVID vaccine and on [indiscernible], we are pleased with a positive Phase III immunogenicity data and are engaging with regulators. Our flu plus COVID vaccine combo should get this Phase III data soon. In latent with CMV fully enrolled and at current cases, we look forward to potential for Phase III data efficacy data in 2024. In our INT program, we're looking forward to completion of enrollment for Phase III adjuvant melanoma study. In addition, we are keen to discuss the possibility of accelerated approval with regulators based on Phase II study data. As we shared before, there are 3 things we view as necessary before we could consider pursuing fed approval for INT. First, durability data from our Phase II study, which we announced in December last year; second, a substantially enrolled Phase III adjuvant melanoma study; and third, manufacturing readiness at the Marlborough site. And last but not the least, our revenue portfolio, we look forward to initiating pivotal study for PMMA. This milestone will represent continued progress towards our mission to deliver the greatest possible impact to people for mRNA medicines. And asModerna, we are dedicated to achieving them all. Every data continues to confirm the power of our platform and its breadth in the service of patients. Two important save a date for your calendars. We will discuss our COVID program in Chicago on June 1, and our annual R&D Day will be held in New York the morning of September 1. Thank you for listening, and we look forward to taking your questions. Operator?
Operator:
[Operator Instructions] Our first question comes from Salveen Richter with Goldman Sachs.
Salveen Richter:
Firstly, could you discuss your strategy for pursuing contracts for the RSV vaccine given 2 approved vaccine sort of have a head start, timing-wise? And help us understand, as you've communicated with large retail pharmacy, how significant the PFS formulation is to them?
And then secondly, with regard to moving into the 3 new indications for the INT program. Maybe help us understand signals or specific data points that support that?
Stéphane Bancel:
On the RSV contract. So as you know, we are not allowed to contract until the product is approved by the regulators. But what we are doing, because we can do that, is our medical teams are actively engaged with retail pharmacies, but also IV and hospital networks in terms of making sure the data on the efficacy profile of the product, on safety, and of course, in the PFS and the benefit of PFS in terms of productivity. Those discussions are ongoing literally on a daily basis, including with leadership of those pharmacies. And the next step, of course, is to wait for the FDA approval. Stephen?
Stephen Hoge:
Yes, sure. So thanks for the question. So on the 3 additional INT indications, I think the short version of it is they are all adjuvant settings. Similar to our melanoma Phase II results have been so encouraging, where KEYTRUDA has a known benefit and where we still believe that there's an opportunity to improve on that by driving a specific T cell response with INT.
And so as you know, in Phase I, we looked across a range of different indications. That was more in the metastatic setting. But as we've announced since we first saw that positive Phase II results from melanoma, we have been aggressively pursuing adjuvant indications where IO is approved and where we see an opportunity. And all 3 of these fits squarely in that space.
Operator:
Our next question comes from Michael Yee with Jefferies.
Michael Yee:
Two questions as well. On RSV, I guess, the competitor GSK as well this week was commenting about how they're expecting you to be in the mix and contracting is ongoing. I know you have some RSV in your guidance. I think the math implies maybe hundreds of millions of dollars. Can you just perhaps comment on how you adjusted, or probability adjusted or thought about how much is there in your guidance and your confidence on that for this year?
And then secondly, on INT as well. I know you had some data at ASCO. I know you have breakthrough therapy and prime designation. How important is the Phase III enrollment progress, the confirmatory study? I feel like that's always an important discussion with FDA. So how important is that progress before you can really engage with FDA?
James Mock:
Yes. Maybe I'll take the first one. Thanks, Mike, for the question.
So as you may know, we haven't guided any specific guidance or number for RSV. In the past, we did break down the $4 billion into 3 different segments around the U.S. market, the APAs we walked into the year with and then another category of other COVID sales that didn't have any APAs across the rest of the world, a good example is Brazil that we just signed, as well as RSV. So no specific guidance for RSV from a financial perspective.
Stephen Hoge:
And your question on INT. I think you nailed it, it's a really important topic, and in particular right now, as we think about accelerated approval that we demonstrate the diligence and substantially enroll the confirmatory study. So really all you're waiting for is for that study to mature, it could be several years.
We think it's really important. To be fair, we have not consulted with the agency on that yet. As we've said, we're waiting until we crossed our own threshold. And also at this point, until we've established the manufacturing facility with that line of sight to that. But we do feel that, as we've said, substantial enrollment demonstrating that essentially all you're waiting for is the readout on that confirmatory study is our obligation before we even want to go forward with that question. We are making great progress this year, and we're optimistic that both that and the facility will be available in short order. And then, of course, we'll want to start engaging with our data, including the FDA on the question of accelerated approval.
Operator:
Our next question comes from Terence Flynn with Morgan Stanley.
Terence Flynn:
Great. Maybe two parts for me. Just on the RSV vaccine. Can you provide your latest perspective on what the most likely ACIP recommendation will be? Will you get a parity recommendation to the competitors? Or do you think there's a potential for a differential recommendation here?
And then just wondering any update on your ongoing conversations regarding filing your seasonal flu vaccine? I know you mentioned in your prepared remarks, but just any more insight in terms of what the gating steps are here.
Unknown Executive:
Thanks for both questions. So first on RSV, caveat by saying we have to complete the approval process with FDA. And then at the end of the day, the recommendation really falls to ACIP and the committee members, so I defer to them.
Our expectation, our hope is that when they review the data package that we already have as well as additional data that we expect to be able to share at the ACIP meeting on durability through a second season, on immunogenicity across other populations, we expect a parity recommendation. We certainly think the data supports that. But again, I'll defer to the committee members on the ultimate decision.
Stephen Hoge:
On the question of flu, we are actively engaged right now on the -- with regulators on the process for submission of the flu vaccine. As I mentioned a moment ago, we are also closing in on clinical data from our combination flu COVID vaccine, mRNA-1083. And that obviously has an important role in our engagement with regulators, generally on flu versus flu COVID combination.
And so those discussions are ongoing. I won't provide any other update on it except to say, as we've said today, and we will continue to say we expect to file the flu product this year, but it will be dependent upon a number of considerations plus we also including the [indiscernible] data that we expect to see soon.
Operator:
Our next question comes from Eli Merle with UBS.
Eliana Merle:
On CMV, how are you thinking about the need or benefits of potentially boosting both from a clinical as well as a commercial perspective? And if you would study those? And then second, just on CMV, if you don't meet the interim, the analysis there, would you disclose that?
Stephen Hoge:
So first, on the question of boosting. So far, what we have -- we obviously don't have the efficacy readout. That's the Phase III study is ongoing. But we do expect to have quite substantial durability data on immunogenicity. And it's quite possible that the efficacy data will give us a signal what the core of production could be.
And so we don't, right now, have any evidence they're not good, durable, multiyear, possibly as long as 5 years, we continue to track the immunogenicity protection. It's possible that will extend out to 10 years and then some boosting is necessary. It's also possible that we decide that a booster might be necessary shorter term in that, let's say, 5 years or 10 years. We just don't know at this time. And so at present, the data we do have on the durability of the immunogenicity, it looks quite strong. And so we do think our 3-dose series will likely be protective for a very long period of time, all subject to the efficacy data that you just referenced. So on the interim analysis for efficacy, as we've said before, we're making great progress in that study in accruing cases, and we do expect to be able to provide an update on -- or conduct at least an initial interim efficacy analysis this year. Because of the rate of case accrual and also because the protocol calls for us to cross a median of 1-year follow-up, the timing may be sets -- by the time we get to that first interim analysis, we are also have enough cases for a final analysis or that a final analysis is imminent. Let's say, it's a very short period of time away. And so because of that uncertainty, until we see that data and understand how close we are to that final analysis, I don't think we can commit, one way or the other, whether we're going to be updating, it will really depend upon the data we see and then how quickly we expect to get that final analysis. At the end of the day, if we have news to share both on interim and the final, we, of course, will, but I don't think we can commit at this stage because we haven't seen the data yet.
Operator:
Our next question comes from Hartaj Singh with Oppenheimer.
Hartaj Singh:
Great. I just have a question on -- you're developing a refrigerator stable vaccine and flu vaccine, I believe. And I'd just like to kind of understand how you think about that. When could that get approved? And then will the combo vaccine also be refrigerator stable?
Stephen Hoge:
Great. [indiscernible]
Operator:
[Technical Difficulty] Ladies and gentlemen, please stand by, your conference will resume momentarily. Once again, ladies and gentlemen, please stay on the line.
And pardon me, can you all hear me now? Could you try speaking again? Your line was muted.
Unknown Executive:
Yes, I'm here. Can you hear me?
Operator:
Yes, we can hear you now. So your line got muted, but you can go ahead and continue.
Stephen Hoge:
Right. Sorry for that brief interruption. So Hartaj, thank you for the question. Just to quickly restate what I was saying. The -- all of our respiratory portfolio, RSV, flu, COVID and the flu COVID combo are being developed towards refrigerator stable PFS. And so our mRNA-1083 program, the flu COVID program as well as the flu program are intended to be a refrigerator stable prefilled syringes. As Stéphane mentioned a moment ago, we really view that as the ideal presentation, the helpful presentation for health care providers really around the world to facilitate their delivery of the vaccine to patients.
Operator:
Our next question comes from Gena Wang with Barclays.
Huidong Wang:
I have two. One is regarding COVID. So for the EU, you said up to 36 million doses every year in EU. What could be the scenario you can get 36 million doses in EU? And also the price in Brazil and the EU, should we use pandemic price of $25 to $30 per doses at the benchmark?
Quickly on R&D, accelerated approval path. Based on, say, today's comments and the prior discussion, our impression is you could achieve all the 3 key components by the end of this year. Is Merck is also fully on board to submit for the accelerated approval in melanoma?
Stéphane Bancel:
Gena. It's Stéphane. I'll take the COVID question and then Stephen will talk about INT. So the tender is up to 36 million doses, will depend on a number of countries that apply to the tender through the EU. So this, we'll know at the end of the process. And as it's a tender process so there is no dialogue, we're just entering all the files and all the data, and that's really ongoing. The team is obviously very active on it.
And on price, for obvious competitive reasons, we're not going to share price in any market to -- because competitors will know the price right away. Stephen, INT?
Stephen Hoge:
Yes. So we haven't specifically guided to when we expect to complete, obviously, the second and third parts of our 3-part criteria. That being manufacturing readiness, as you can imagine, work is going on around the clock as well as the enrollment we've made great progress, but we have to sustain that progress.
On your question of where is Merck on this. I think you'll have to direct it to them. Our view is that if we're able to get to the point where ancillary approval is appropriate and regulators are supportive of that, we can't imagine why ourselves and Merck wouldn't want to make the product available to help people suffering from cancer right now. But the contingencies, there are, obviously, we have to do our work and our doses this year. And then ultimately, we have to speak to regulators, and they get to decide whether that pathway is available to us. And so I think for both ourselves and our partner Merck, we want to defer to regulators ultimately on that choice.
Operator:
Our next question comes from Luca Issi with RBC Capital.
Luca Issi:
Maybe a very quick one on RSV. I think the last press release actually cited May 12 as the PDUFA date. While today, you're simply saying the initial regulatory approvals in the first half of '24. So is there anything to read to it? Can you just confirm that the PDUFA date is still May 12, which is actually the end of next week?
And then maybe second, on IP, can you just comment on the recent decision by Judge Goldberg to rule out, obviously, [indiscernible] or you're looking on a particle. Our understanding, this can have pretty material impact on both prior and future sales of COVID. So again, any thoughts there, much appreciated. And then super quickly on INT. Stephen, what's holding you back on starting the randomized trial intended to cancer into metastatic static? I thought the data they see that was pretty impressive. So any thoughts there, much appreciated.
Stephen Hoge:
Great. Perfect question. I'll take this first from the third very quickly. So on the function of RSV, we continue working towards the same PDUFA date, and there's no change to that. As you know, there's a lot of work and around the clock work by ourselves, obviously, and folks at the agency.
And so we're hopeful that, that happens as planned, but if it takes a little bit longer, at the end of the day, what really matters is the duty for [indiscernible] meeting, but there's been no change to report. So don't read anything into that. As far as the INT question, on head and neck, I appreciate the question because we are also obviously enthusiastic about that data. However, our partner, Merck and ourselves, we have not yet decided where that fits in the priority of other indications, pathologies and opportunities we're pursuing. As you've already seen in the past year, we've stood up a very large number of study. And we're just trying to pace ourselves. And so it will take us a little bit of time with our partner, Merck, to determine what the next steps are in head and neck. And at this point, we do not have an update on.
Stéphane Bancel:
And I'll take the IP questions. I mean as you know, our COVID-19 vaccine technology, including our lipid nanoparticle delivery system is the result of independent research and development. We have a strong belief that our technology does not impact on the patent asserted by [indiscernible]. We are confident in our position, and we look forward to presenting our case after next year.
Operator:
Our next question comes from Jessica Fye with JPMorgan.
Jessica Fye:
I had a few here. So for INT, I know you mentioned you can't comment on when you expect to complete the manufacturing scale up. But can you provide a status update on where you stand with that today, and the number of patients you can support right now as well as where you want to take that capacity once you get to the end of this 3-phase scale-up process, even if you don't put a timeline on when you'll get there?
Next one is coming back to flu. Can you just refine a little bit when the 1083 immunogenicity data will be available? And maybe elaborate on how the combo data play a role in the regulatory talks on 1010. I thought you previously said these products could stand on their own. And that you might not need 1010 approved to get 1083 approval. So now wondering kind of why 1083 might factor in for 1010. And then lastly, on CMV, can you remind me how the risk of CMV and pregnancy compares in seropositive versus seronegative individuals. I'm thinking from a commercial standpoint, how you weigh the strategy of pursuing vaccinating everyone versus just seronegative people.
Stéphane Bancel:
Great. I'll start with the INT question on manufacturing, then Stephen can add around [indiscernible]. So we have not provided some capacity numbers of the factory in Marlborough. But of course, as you can assume, we now the size of melanoma market. And we know our stronger data. We [indiscernible] people benefiting from the Phase II data.
So we've sized the plant accordingly, as you can imagine. But also, we are building the plant for care because, as you know, Stephen and his team and his colleagues are running a lot of studies. So this is not a platform in [indiscernible] a plant for INT. Again, from a manufacturing standpoint, we don't care which cancer is in terms of organ because we use genetic information to design an individual product for every human being. So basically, we bought a plant last year that was kind of -- is a big building finished, saves us time to market. Obviously, we don't have to get the permitting and build the building. And so the team since then, that is now more than a year ago, has been working actively to get the plant ready. And the plant is on the building in modules inside the building. So basically, we're going to launch with the first module of manufacturing capacity. And then -- and one day maybe we do an opening of a facility like we did for [indiscernible]. You will see that there's another empty space left behind, which has also a very quick ramp-up because the HVAC system and all utility system has been set up for the entire plant. And so then you just add modules as you go. So we'll be able to scale very quickly as we get more indication available. But again, we're aware of the melanoma number of cases. And so the plant will be sized so we make sure we can provide products to patients. Stephen?
Stephen Hoge:
Thank you for the clarifying question on flu. So let me just start by saying that independently, we are looking to submit both the flu program and the flu COVID combo program. So that's 1010 and 1083. Obviously, we need to see the 1083 data, and we'll announce that when we have it.
The question on the timing of that data, it's imminent. And so in the coming quarter, we expect to be able to share that update. The point about interdependency, I suppose, is just more about sequencing of those submissions and in some places and some regulatory geographies, obviously, you can't stack them on the same day if you will. There's a logical sequence. And what we want to assess once we see the 1083 data is our regulatory strategy as well as our preparation and delivery of data for the submissions to determine which one will go first or second. But at this point, we're trying hard to make sure that we can do both products across all of our major markets, if the data is filed in this year. And so we'll go more on that as we move forward. But for now, we are proceeding without independently. Sorry for the confusion model. On the question of CMV and seropositivity. So it's a really important point. Thank you for raising it. But while the majority -- well, the risk of vertical transmission of CMV to pregnancy to the fetus is highest in seronegative. It does happen in seropositives as well. So congenital CMV is a disease that's seen in -- particularly in reactivation or sometimes reinfection, even in the seropositive context. And so we do believe that there's a potential for benefit for a vaccine even in seropositive population. We are evaluating the study right now in seronegative because the rate of that transmission and obviously, the potential to prevent against infection is more enriched and therefore the study size, primary are focused on seronegatives. But we are looking. We have studied the vaccine from a safety perspective in seropositives. And we are looking at things like [ Sheng ]. If you draw a little bit of an analogy to correlate to the EVB data that we've already put out there in a different virus, but we've been able to show that we can really control the rate setting even in seropositives [indiscernible] qualifiers. And so we have some reasons for optimism and believe that when we pull together the totality of the data, there will both be the obvious potential benefit, which is that there is still vertical transmission in seropositives and some -- potentially some data on the rate of [indiscernible] that would be supportive to that. Ultimately, though, we're studying all the way down to 16-year-olds. And our goal will be a label that 16 plus, with the goal going into a population that is not as highly seropositive as it is later in life and therefore, we see a very large opportunity, improvement, and then primary infection CMV with the vaccine and the potential for [indiscernible] seropositives [indiscernible].
Operator:
Our next question comes from Geoff Meacham with BofA.
Alexandria Hammond:
This is Alex Hammond for Geoff Meacham. So on your [ zoster ] vaccine candidate, when should we receive updates on your pivotal strategy? And is there any color you can provide today in terms of your current thinking on the Phase III design?
And then our second question is on the PA and MMA programs that are advancing into pivotal trials, can you provide any thoughts on the nature of editorial and the comments on safety?
Stephen Hoge:
Yes. Could you describe about the first part of the question was on which program? The zoster?
Alexandria Hammond:
The zoster, the shingle.
Stephen Hoge:
Shingle zoster. So on the [indiscernible] program, we have -- we're obviously very excited by the Phase I data, which was compared against a licensed product, and we saw really strong T cell response in immunogenicity. And generally, we've been in that across our programs. But in that one, it was very encouraging.
We're in the process right now of trying to find the pivotal strategy. That will include, obviously, dose selection, the number of doses in that study and then how we're going to expect that study. We do not have an update till today on what that will look like in addition to our own thoughts on it, we obviously want to consult with regulators before we finalize that [indiscernible]. But we are moving for forward -- towards a pivotal study in PCV. We do not have a [indiscernible] on that update yet. As it relates to MMA and PA, the clinical data that we have continues to show a compelling benefit risk profile, good safety profile. In fact, in the PA studies, we have many folks who've been in those -- in the study on drug for well over a year. And over 30 years, I think, from our last update in overall patient dosing experience. So we are starting to get a very clear perspective on the safety profile. The editorial question, I don't have a view on editorials or opinions based on the preclinical data. I think we stand behind the clinical data that we have and are quite encouraged by that profile, and we'll continue to watch it closely in our ongoing Phase I studies, but we do not have any specific or new concerns based on the clinical data today.
Operator:
Our next question comes from Evan Wang with Guggenheim Securities.
Boran Wang:
Two for me. First, on the combo 1083 program, so data, it sounds like this quarter, I believe enrollment was completed a few months ago. So I guess how comprehensive will the top line update be in terms of follow-up? And then with submission, is longer-term vault needed there? And are there parallels from 1010 that we can take in terms of regulatory filing speed for 1083? Or is that more impacted by the decision for buying one or the other first?
And then second, on RSV, it's kind of early ahead of approval, but with some international markets, it seems that's more nascent in terms of established some reimbursement there. So I guess how are you thinking about positioning internationally?
Stephen Hoge:
Great. So for the 1083 data, yes, on this quarter, and I would say that we're -- we enrolled the majority of the 1083 studies you know last fall. And the 1010 second-generation study. So we talked about the P303 study. The first part of that enrolled over last summer, just a few months before the combo study.
And the second part of it, there was a part B and C, as you know, looking head-to-head against [indiscernible], that actually enrolled the same time as the 1083 study last fall. And so they've been actually kind of tracking right on top of each other. I think we're going to wait to see the data before we can provide growth guidance on timing. But obviously, we're -- we've been working towards that flu COVID combination product for a while, and we will want to make sure that we get that filed, if it is positive as fast as possible. I wouldn't draw too many, because of the difference in the structures of the study between the P303 study, which has a part A and the B and a C, and the 1083 study, which was done very quickly. I wouldn't draw too many correlations between [indiscernible] reading out and the timing for submission on [indiscernible]. Stéphane, do you want to take the RSV question?
Stéphane Bancel:
Sure. So with internal markets, obviously, very important, the U.S. is very important, the U.S. and Russia also super important. As we shared before, we found in all the major geographies already. Of course, EU, U.K., Canada, Australia, some countries in Asia, some countries in the Middle East.
RSV is well known by public health leaders like it is in the U.S. So I think that there's a very strong desire, again, to protect the elderly or what we are doing here in terms of true COVID RSV, that's becoming very kind of a standard that probably [indiscernible] across at least the developed world. But even in developing countries, there's more and more interest as you see aging population everywhere. So it's not only the U.S. approval that we expect coming soon is we have several geographies that would start being approved so.
Operator:
Our next question comes from Simon Baker with Redburn Atlantic.
Simon Baker:
Two quick ones, if I may. Just in terms of the timing on the CMV interim data. You said this quarter, it could be as early as the end of '24. That sounds slightly later than you previously said. I just wondered if that's me over-interpreting the semantics or whether there is a slight delay there?
And then the second question is on the HSV vaccine. Previous quarters, we talked about the EBV vaccine and the potential utility in multiple sclerosis. So I just wondered what your thoughts were about HSV and it's the hypothesis that implicates it's role in Alzheimer's disease?
Stephen Hoge:
So first on the clarification. There's no change to our expectations on when the CMV readout will happen. I think we previously tried to be careful in saying that we expect it to happen this year. And so obviously, by the end of this year, it is meant to say the same thing, but there's no change in our expectations at this point.
On the HSV Alzheimer's hypothesis, it's a very interesting -- there's a lot of neuroinflammatory questions that go with the herpes simplex virus infection across a range of different mutations, Alzheimer's one of them. At this point, the studies that we expect to move forward with HSV will be for seropositive to improve outcomes. So shedding days, for instance, or lesion based, and then eventually, we will want to consider whether we want to go at prevention of infection, which is obviously a different standard of different indication. That might be more relevant for them, how you think about some of the neuroinflammatory or long-term supply. I think you asked my opinion on the -- I think it's incredibly interesting and exciting. I do think it's early for us to start drawing connection from a vaccine perspective in terms of our potential impact for it. I hope over time, there is an opportunity to intervene and things like that. Obviously, in the EBV vaccine with multiple sclerosis, that science has firmed up to the point where there's reasonably high conviction that there's a potential for benefit there. We have to go prove that. But at this point, it's still earlier days, I think, with HSV and Alzheimer's.
Operator:
Our next question comes from Edward Tenthoff with Piper Sandler.
Edward Tenthoff:
Congrats on everything. Actually, most of my questions have been answered. But I wanted to ask with respect to the cancer efforts, are you able to break out what the actual R&D cost is for that program and that's still a cause some profit share with Merck? And how many indications do you guys ultimately plan on pursuing?
Stephen Hoge:
Maybe I'll take the first one, Ed. So we obviously know what we're spending, [indiscernible] but at this point, that we are not prepared to disclose. So maybe someday, but not at this time.
Edward Tenthoff:
Understood. And then -- with the expansion...
Stephen Hoge:
Yes. On the expansion indication. Look, at the joint decision, our partnership with Merck has been really strong. We've been building this out. We do like to review those strategically and then bring them forward once we've started them. And so I don't want to get ahead of that because those are our private strategic competition with Merck. But we are not done yet. We will keep adding in the years ahead.
Edward Tenthoff:
Very exciting. Look forward to seeing you in Chicago.
Operator:
Ladies and gentlemen, this does conclude the Q&A portion of today's conference. I'd like to turn the call back over to Stéphane for any closing remarks.
Stéphane Bancel:
Well, thank you, everybody, for joining in today for a great session. We look forward to seeing you at latest ASCO. Have a great day.
Operator:
Ladies and gentlemen, this does conclude today's presentation. You may now disconnect, and have a wonderful day.
Operator:
Good day, and thank you for standing by. Welcome to the Moderna Fourth Quarter 2023 Conference Call. At this time, all participants are in a listen-only mode. After the speaker's presentation, there will be a question-and-answer session. [Operator Instructions]. Please be advised, today's conference is being recorded. I would now like to hand the conference over to your speaker today, Lavina Talukdar. Please go ahead.
Lavina Talukdar :
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's fourth quarter and full year 2023 financial results and business updates. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the investor section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; and Jamey Mock, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see slide two of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I'll turn it over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Thank you for joining us today. I will start with a review of 2023. Jamey will then present our financial results. Stephen will then review our late-stage clinical programs, and I will close by sharing our 2024 priorities. Let me start with our mission, Moderna's commitment to deliver the greatest possible impact to people through mRNA medicine. In 2023, every member of the Moderna team helped to advance our mission, which is a driving force that motivates our team every single day. We impacted more than 100 million people around the world and we advanced our development pipeline across all of our franchises, including infectious disease, oncology, and rare disease. 2023 was a difficult year as we transitioned from a pandemic to a seasonal endemic market. We reported sales of $6.1 billion for 2023, which was at the low end of our financial framework range. These sales exclude the recognition of $600 million of deferred revenue from Gavi. In the U.S., the vaccination rate was down year-over-year. We were pleased that our U.S. commercial team drove an increase in our retail market share from 37% to 48%. Outside of the U.S., we were not able to compete in the EU market in the second half of 2023 due to a competitor contract, and our performance led to a low market share in Japan. We did have a strong performance in Israel, Switzerland and Taiwan. We took several important actions in 2023 that we set our commercial COVID business up for success. We resized our manufacturing footprint, which has been established to support capacity at pandemic levels. We exited contract manufacturing relationships and reduced inventory levels. These initiatives will improve cash flow from our COVID business moving forward. We also flattened the commercial structure. All regions report directly to me now for more targeted sales execution and also a better integration with global teams. We focused on R&D spending and our SG&A expenditures towards near-term growth and higher return on investment projects. While sales were challenging in 2023, our development team had a great year with excellent progress across many of our late-stage pipeline programs. In 2023, we advanced our pipeline and now have nine late-stage programs. Let's start with Respiratory vaccine. For RSV, we filed for approvals around the world. We reported positive data from a flu P303 study, and we are fully enrolled in the Phase 3 studies for our next-gen COVID, mRNA-1283, and flu plus COVID combination vaccine, mRNA-1083. In our latent franchise, we are very excited that our Phase 3 CMV trial is now fully enrolled. In our Individualized Neoantigen Therapy Program or INT, where we partner with Merck, Phase 3 studies in adjuvant melanoma and non-small cell lung cancer are enrolling. We purchased and are currently building out a manufacturing site in Marlborough, Massachusetts to enable commercialization of our INT program. Our rare disease therapeutic programs continue to progress well. We are in dose selection of the restrictional study of our Propionic Acidemia Program. In MMA, we are pleased to see improvement in biomarkers and clinical outcomes. In research, we made six estimated [ph] investments, including one acquisition, which we expect will increase the strategic reach and also the breadth of our mRNA platform. Now turning to our 2023 financial summary, we reported GAAP revenue of $6.8 billion, a net loss of $4.7 billion, primarily driven by mostly non-cash charges of $3.7 billion related to resizing of manufacturing and the tax valuation allowance. We are pleased to end the year with cash and cash investment of $13.3 billion. Let me turn to Jamey for more color on our financials.
Jamey Mock:
Thank you, Stéphane, and hello everyone. Today, I will review our financial performance for both the fourth quarter and the full year of 2023. I'll also provide our financial framework for 2024. Let me start with a review of our commercial performance this year. In the first half of 2023, we reported product sales of $2.1 billion, with the majority of sales from advanced purchase agreements signed for delivery in 2022 that were deferred into 2023. We do not expect these sales to repeat in 2024. In the second half of 2023, we recorded $4 billion in sales from seasonal endemic demand and an additional $600 million from deferred revenue related to Gavi. Sales in the fourth quarter were $2.8 billion, with $0.8 billion in sales in the U.S. and $0.6 billion in Europe and $1.4 billion in the rest of the world, including the deferred revenue from Gavi. For the full year, 2023, product sales were $6.7 billion, comprised of $1.7 billion in sales in the U.S., $1.4 billion in Europe, and $3.6 billion in the rest of the world, again, including deferred revenue from Gavi. Moving to side 10. As mentioned, net product sales were $2.8 billion this quarter, a 43% decrease from last year. This was largely attributable to the anticipated reduction in sales volume, which was partially offset by a higher average selling price. This decrease is indicative of the evolving market dynamics as we navigate the transition of the COVID-19 vaccine market towards a more predictable seasonal pattern like traditional flu vaccines. Cost of sales was $929 million, down from 39% of net product sales in the previous year to 33% this year. This is a demonstration of our strategic efforts in Q3 to resize our manufacturing footprint, which as expected, led to additional charges in Q4 of $169 million, primarily related to the wind down of certain contract manufacturing operations. Additionally, faster sales also includes an inventory write-down of $322 million, reflecting revised demand forecasts. R&D expenses increased by 16% to $1.4 billion. This uptick reflects our commitment to advancing our late-stage clinical development programs, particularly with our RSV vaccine, CMV vaccine, combination vaccine against flu and COVID-19, as well as our INT program. The increase also included an upfront payment of $120 million associated with the strategic research and development collaboration with Immatics. SG&A expenses were $470 million, up 25% year-over-year. The increase in spending was primarily due to the expansion of our commercial operations, particularly in the U.S. market. Income tax was a benefit of $147 million for the fourth quarter of 2023, largely attributable to the tax benefits as part of finalizing our 2022 U.S. tax return. Net income for the quarter was $217 million, compared to $1.5 billion in the fourth quarter last year. Diluted earnings per share was $0.55 compared to $3.61 in 2022. We closed the quarter with a strong cash position of $13.3 billion, which is slightly higher than the $12.8 billion we had at the end of the prior quarter. Now, let's turn to our annual performance on Page 11. Net product sales for the full year of 2023 were $6.7 billion, a decrease of 64% from the previous year, mainly due to lower sales volume of our COVID-19 vaccine. As mentioned earlier, this includes the recognition of $0.6 billion from the deferred revenue related to Gavi. Excluding this item, our sales of $6.1 billion were still in line with the framework we provided for the full year. Cost of sales for the full year represented 70% of net product sales, a substantial increase from 29% of product sales in 2022. This shift is largely attributable to our strategic efforts to optimize our manufacturing operations, resulting in charges of $1.6 billion, and other manufacturing and distribution costs over reduced sales volume. Overall, cost of sales came in at $4.7 billion, slightly below the $5 billion we provided in our latest framework. Research and development spend was $4.8 billion, and SG&A was $1.5 billion, both in line with our expectations. Our income tax provision was $772 million for the full year 2023. During our Q3 earnings call, we discussed the requirement under GAAP to establish a valuation allowance against deferred tax assets, when the current year and cumulative income projection for the next three years is in a loss position. It's important to note that future income from products not yet approved by regulators are excluded from these income projections, which restricts us to just our COVID vaccine, and it does not include expected future launches. This valuation allowance does not impact cash flows, future tax returns or the company's ability to utilize deferred tax assets in future periods. Net loss for the year was $4.7 billion, compared to net income of $8.4 billion last year. The decrease in profit was primarily due to lower product sales and higher R&D expenses in 2023. Diluted loss per share was $12.33, compared to diluted earnings per share of $20.12 in 2022. So now let's move to slide 12. We wanted to provide you additional perspective on our full-year financial results by presenting them alongside a summarized version that excludes the impact of the Gavi deferred revenue recognition, our resizing charges, and the tax valuation allowance. Our total GAAP net loss for the full year was $4.7 billion. However, when excluding these primarily non-cash items, the net loss is reduced to $1.6 billion. Now let's turn to our 2024 financial framework on slide 13, which is mostly in line with what I shared on our Q3 call. We expect net sales for 2024 of approximately $4 billion, which we think will be a low point, as we expect to return to growth in 2025. Sales in the first half of the year are expected to be approximately $100 million, reflecting the strong seasonality of respiratory vaccines. We expect cost of sales of approximately 35% of product sales, in line with our cost of sales framework which we introduced in our Q3 earnings call last year. For R&D we expect full year expenses to be approximately $4.5 billion, down from $4.8 billion in 2023, and for SG&A, we expect full year expenses to be approximately $1.3 billion, down from $1.5 billion in 2023. We also expect taxes to be negligible in 2024. In our Q3 earnings call, I provided our Moderna operating principles, which largely centered around a very disciplined approach to capital allocation. Our number one priority has been, and will continue to be, reinvesting in the business. In addition to the investment into our pipeline, we expect capital expenditures in 2024 to be approximately $0.9 billion, as we mostly complete the construction of our facilities across the globe. Our teams are laser-focused on operational improvements for both expense management and working capital. As a result, we expect to end 2024 with approximately $9 billion in cash. I will now turn the call over to Stephen.
Stephen Hoge :
Thank you, Jamey. Good morning or good afternoon, everyone. Today I'll do a quick review of our clinical highlights from our late-stage programs in 2023, and then look ahead to anticipated milestones in 2024. While we have over 40 programs in development, they'll focus on our late-stage pipeline, which consists of nine programs across four franchises, all made significant progress in 2023. Four of these programs are in our respiratory vaccine franchise. We hope to launch all of these potential products by the end of 2025. I'll discuss these further in a moment. The other five late-stage programs are spread across latent vaccines, oncology therapeutics, and rare disease therapeutics. We hope to begin launching these beginning in 2026, and I'll discuss our progress in these areas as well. In infectious disease vaccines, we've achieved many important clinical milestones in 2023. Within our respiratory vaccine franchise, we filed for RSV vaccine approvals in many countries around the world. We recently presented follow-up data from our Phase 3 study at the RSVVW meeting that I'll recap in a moment. In our mRNA-1010 flu program, our Phase 3 P303 study met its primary safety endpoints and hit all eight co-primary immunogenicity endpoints against all strains of influenza. We're also excited that we fully enrolled the Phase 3 immunogenicity and safety study of our Next-Gen COVID vaccine and the Phase 3 immunogenicity safety and safety study of our flu and COVID combination vaccine. In our latent and other vaccines franchise, we're proud of the great progress our team made to complete enrollment in the Phase 3 study of our CMV vaccine in 2023. That study is now accruing cases towards its first interim analysis of efficacy. On slide 17, I want to briefly review the primary analysis for our RSV vaccine, which was first shared in January of 2023. The study met its primary and key secondary endpoints, leading the DSMB to recommend unblinding. Vaccine efficacy was 83.7% against RSV with two or more symptoms of lower respiratory tract disease at a median follow-up of 3.7 months with a wide range of two weeks to 12 months of total follow-up. The primary analysis also showed 82.4% and 68.4% vaccine efficacy against three or more symptoms and acute respiratory distress respectively. These data were recently published in the New England Journal of Medicine in December of last year. We've continued to follow the participants in the trial and announced follow-up data at the RSVVW conference earlier this month. An additional analysis showed sustained vaccine efficacy against RSV with VE of 63.3% against RSV, lower respiratory tract disease with two or more symptoms through a medium follow-up of 8.6 months and a maximum follow-up of 17.7 months. The vaccine efficacy was 74.6% against RSV, lower respiratory tract disease associated with shortness of breath, which has been shown to be a key driver of seeking a higher level of medical care and the associated burdens and costs. Now, slide 19 summarizes the overall timing of enrollment, primary efficacy analysis, and subsequent analysis overlaid against the epidemiology of RSV over the two seasons that have now contributed to the efficacy in the trial. Our trial enrolled steadily over 13 months between November 2021 and December 2022. As a result, the primary efficacy analysis included cases from both the smaller RSV season of 2021 and 2022 and the much more significant RSV season of 2022 and 2023. Due to enrollment throughout the year, the median follow-up of the primary analysis was 3.7 months, but as I noted, the maximum follow-up was 12 months. The primary analysis met its success criteria leading to a study on blinding. We continued a pre-plan additional analysis on April 30, 2023. At that analysis, the medium follow-up was 8.6 months, with a maximum follow-up of 17.7 months. Or put another way, approximately 17,000 participants were between nine and 18 months of study follow-up at the time of the analysis, with many completing their second RSV season on the study. Per protocol, we are continuing to follow cases through one year, but as is evident from the epidemiology, very few cases would be expected in the six months after the April 30th cutoff date from the last analysis. In summary, we are pleased that the data shows sustained efficacy through two seasons, including the large RSV season of 2022, 2023, and we look forward to providing further updates from this ongoing study. We also achieved clinical milestones across our therapeutics franchises in 2023. In oncology, our Individualized Neoantigen Therapy developed in partnership with Merck, began Phase 3 clinical studies in both adjuvant melanoma and non-small cell lung cancer. Both phase three trials are now actively enrolling. Our primary analysis from the Phase 2 study was recently published in The Lancet, giving greater detail on the two-year follow-up data that we had released in 2022. Now in December of 2023, we shared top-line follow-up data from that same Phase 2 study in adjuvant melanoma patients, confirming the durability of the initially reported responses. At a medium follow-up of now three years, the recurrence-free survival and distant metastasis-free survival remained extremely favorable, with a reduction of the risk of recurrence or death by 49% and a reduction of the risk of distant metastasis or death of 62%. Both results were highly statistically significant at three years. And in PA and MMA, our most advanced rare-disease therapeutic programs, we continued to see positive clinical data in our Phase 1/2 studies, including improvements in biomarkers and clinical outcomes, such as metabolic decompensations. Now, turning to slide 21. While we're proud of the progress in 2023, we have much more ahead in 2024. Let me take you through some of the late-stage milestones we anticipate for this year. I'll start with our respiratory franchise, where we are targeting the first approval for our RSV vaccine, beginning in the first half of 2024, with commercial launches shortly thereafter. With our flu vaccine, we're in discussions with regulators about potential submissions for approvals, and we expect to begin filing this year. Phase 3 data from our Next-Gen COVID vaccine is expected in the first half of 2024, which will inform the next steps. And we expect Phase 3 data for our flu and COVID combination vaccine this year. In latent vaccines, we are looking forward to potential efficacy data from our CMV Phase 3 study. In oncology, we expect continued progress enrolling our two Phase 3 studies in INT for adjuvant melanoma and non-small cell lung cancer. We also expect to expand into additional tumor types this year. And finally in rare diseases, we expect to move into registrational studies for both PA and MMA in 2024. It'll be a very busy year, and we look forward to sharing progress with you as the year progresses. With that, I'll turn the call over to Stéphane.
Stéphane Bancel :
Thank you, Stephen and Jamie. For Moderna and the patients we serve, 2024 is all about execution. Execution in commercial, execution of our late-stage pipeline, and execution with financial discipline. Starting with commercial, first our COVID vaccine. We continue to work with health authorities to increase vaccination rates and improve public health by reducing the substantial burden of disease from COVID in this upcoming 2024-2025 season. While I am pleased with the U.S. market share outcome of the current season, I believe we can and must do better on vaccination rates. Our teams are actively working on it already. A few weeks ago, the EU published a new mRNA COVID vaccine tender, up to 36 million doses per year for up to four years. Our team is actively working to respond to this standard. We are prioritizing our commercial focus on specific markets around the world to deliver where it matters the most. Moving to our RSV vaccine candidates, we are very excited about launching the RSV vaccine this year. That will be the launch of our second product. Our mRNA platform is delivering. The FDA PDUFA date is May 12. If the outcome is positive, we anticipate that ACIP will include mRNA-1345 on the agenda in late June. In Europe, we expect Germany to launch in 2024. We also expect Australia to launch this year, while other markets will likely launch in 2025. In many markets around the world, we need to secure regulatory approval before we can participate in tenders. As communicated last year, given expected approval outside the U.S., Germany and Australia will anticipate launching RSV in this market in 2025. The RSV market is estimated to be a $10 billion opportunity, consisting roughly of $6 billion to $8 billion in older adults and $2 billion to $4 billion in the pediatric and maternal setting. In 2023, the first-year RSV vaccine launches, consumer awareness of RSV, and demand for RSV vaccine was strong. The 2023 adult RSV vaccine market was around $2.5 billion. This is quite impressive, given the first year RSV vaccines were available, they were not even available for a full year, and the products were not approved in many countries. With 2023 RSV vaccination rate as a small percentage of a total addressable market, we are quite excited to launch our product into this large and growing market. Let me now turn to our RSV vaccine profile. We believe we have the best profile to serve patients and completing the RSV market, efficacy, safety, and ease of use. Our clinical data shows strong vaccine efficacy. We have a well-established safety and tolerability profile that leverages the same mRNA technology that has been delivered in over 1 billion COVID vaccines. Additionally, we have not seen any case of Guillain-Barre Syndrome or GBS in our Phase 3 trials. We expect to be the only company to offer an RSV vaccine in ready-to-use prefilled syringe or PFS. Our one-step administration compares well relative to competitive products and requires multiple preparation steps by pharmacists and clinicians. As you know, one of our competitors’ products requires nine steps of preparation for each consumer needing an RSV vaccine, and the other competitors’ product requires four steps of preparation. With over 90% of U.S. RSV vaccines given to-date in the pharmacy setting, PFS presentations offer ease of use, time saving and the potential to reduce medical errors. Given the labor shortage in retail pharmacy channels, we anticipate our PFS presentation will be welcomed by pharmacists in a very busy respiratory vaccine season, where pharmacists need, in addition to their regular tasks, to administer flu vaccine, COVID vaccine and RSV vaccine. Today we talked about how much progress we made in the late-stage pipeline in 2023. This year, we look forward to continue the execution and reporting milestones in each of these programs. If you look at this slide, it is going to be a very busy and very exciting year with multiple Phase 3 readouts like the COVID Next-Gen, also Flu plus COVID Phase 3 data, and potentially CMV Phase 3 data. But also we're going to be filing products like flu, which will be our product file, and of course approvals in many countries around the world for RSV. Finally, we are all committed to exercise financial discipline across the business. While we have resized our manufacturing footprint in 2023, we will continue to find ongoing cost improvements in manufacturing. We will reduce operating expenses in R&D and SG&A and prioritize programming R&D with near-term commercial potential in areas of unmet medical needs. Overall, our CapEx will be up modestly in 2024 compared to 2023, and will mostly complete construction of several important plans in Marlborough, Massachusetts for INT, and Canada, UK and Australia. In 2025, we expect CapEx to be down significantly. We're also targeting working capital improvements. And importantly as you know, we are adopting AI across the business, which we expect to save time, increase productivity, and drive scalability in addition to cost saving. Our use of AI is increasing by the week, and new use cases are exciting to see how our teams are embracing this new tool across the business. In summary, 2024 is an important year of execution across our company, one that will set the stage for the next several years. I am very excited by how our company is positioned. I believe 2024 will be a year where many observers of Moderna go from thinking of us as a COVID vaccine company, to seeing Moderna as an mRNA platform company, with several products approved and more approvals on the way for 2025 and beyond. Over the next few years, our ability to deliver on our mission will increase significantly and be very meaningful for our teams. With that, Operator, we'll be now happy to take questions.
Operator:
Thank you. [Operator Instructions] Our first question comes from Michael Yee with Jeffries. Your line is open.
Michael Yee:
Hey guys. Good morning and thanks for the question. Focusing on RSV, I know you made a number of nice comments about comparing the data. Maybe you could talk to two or three points. One is perhaps how it will work in the commercial market in the U.S. with contracting. Is that contracting season or do you have to have contracts and how does that work between the two different other competitors? And secondly, how would that work at the pharmacy level when either patients or doctors are making the selection, given the fact that Pfizer and GSK both had similar sales but different profiles? Maybe talk to those two different things and how you expect that to play out for this year. Thank you.
Stéphane Bancel:
Great. Thank you, Michael. It’s Stéphane. So in terms of contracting, obviously we cannot start contracting now, because the product is not approved. But our medical team has been quite engaged across the board at medical conferences, talking to healthcare professionals and sharing the great data that was published in December, as you know, for a Phase 3 in the New England Journal of Medicine. So we have active discussions. There is quite high excitement about the possibility, again, if a product was to be approved to get a pre-filled syringe product. As we discussed in my remarks, as you know, labor shortages are a big issue in pharmacy. For any of us that has gone to a pharmacy in the fall, you could see it was very busy, sometimes a bit too hectic, and so the leadership of the big retail pharmacies is very engaged to think about COVID versus RSV and how to simplify the workflow, how to reduce medical errors, which is why those medical discussions that we're having so far give me significant hope that our products will be meaningful tools for our customers.
Operator:
Thank you. Our next question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
Thank you. I have two very quick questions. First one is regarding the RSV vaccine. What portion of 35,000 to 36,000 participants that completed two seasons and regarding February 29 ACIP meeting, what additional data you will be presenting? And very quickly on 2024 guidance, now we have a better understanding of both COVID and RSV market size for 2023 and 2024 season. What could be the upside or downside for your 2024 revenue guidance of $4 billion?
Stephen Hoge:
Great. Thanks Gena. I'll take the first part of that. So I actually can't – I don't know off the top of my head the proportion, because there are both Southern and Northern Hemisphere participants that were enrolled in the study. And so counting the two RSV seasons will depend upon that. It is a sizable proportion, because as I said, the maximum follow-up at the additional analysis is about 18 months, medium was nine months. And so by definition, about half, as you just said, about 17,000 participants are between it. And we did enroll pretty continuously over 13 months, not exactly evenly, but pretty close to it. So I don't want to give you an inaccurate number, but I would suspect it is a pretty sizable proportion because of that steady, pretty consistent enrollment over the course of 13 months. It's not exactly equal, but it trended in that direction. As far as the digital data for the ACIP meeting, we will obviously continue to provide updates from any additional analysis we're doing on durability. We obviously have immunogenicity data, as well as data across different subpopulations. And if we're fortunate enough to have the opportunity to present at the ACIP, we will of course listen to the committee on anything they would like to see as well on the performance of mRNA 1345 as a vaccine, and the performance of the vaccines in general in terms of durability, as it guides decision-making around repeat dosing or boosting on some schedule and the public health.
Stéphane Bancel:
Thanks Stephen. And Gena, its Stéphane, on the upside and downside on the sales for this year. I think on the upside obviously, the COVID in Europe, as I just mentioned, we couldn't participate in the market last fall. The new tender is an opportunity for us to participate. Quite a number of doctors, hospitals, public health leaders have actually complained that the Moderna vaccine, given the higher efficacy reported for reduction of hospitalization was not available, especially for the elderly, for immunocompromised people, so that's an interesting upside. Of course, the vaccination rate in the U.S. as we reported, the vaccination rate in the current ending season was lower than last year. As I said in my remarks, we need to do better to protect more people. And our team is actively already working in a cross-functional matter to address the VCR and increase the vaccination rate. And the other upside could be the RSV, both for market growth, as well as our share. How quickly can we get share from the current two solutions available. On the downside, of course, the vaccination rate could be a downside. And the other one is of course timing of RSV launch, given we rely on regulators for the approval of products, and then the public health recommendations like CDC over different NITAGs in the different countries, that could of course delay launches and of course impacting sales. Thank you.
Operator:
Thank you. Our next question comes from Eli Merle with UBS. Your line is open.
Eli Merle :
Hey guys, thanks so much for taking the question. Just turning to the CMV Phase 3, can you talk a little bit about what you would view as clinically meaningful or commercially relevant on vaccine efficacy? And if successful, also, how are you thinking about use in seronegative versus seropositive patients? Thanks.
Stephen Hoge :
For sure. So thank you for the question. So in CMV, obviously there's currently no vaccine that can prevent infection against CMV. And as it is a source of devastating birth defects, anything that provides a statistically significant reduction in the rate of infection and therefore vertical transmission would be, we think terrific. Now, the minimum bar that we are powering in our study would support is a vaccine efficacy of approximately 50% as we've shared previously. Anything above that would obviously beat our expectations and be incredibly exciting for the field. We are in the Phase 3 study, enrolling both seropositive and seronegative participants. And part of the reason for that is that there's currently no broadly used diagnostic. And so we want to demonstrate benefit or safety in both populations, because we do believe that the most likely use of the vaccine could be that it's given regardless of serostatus to both seronegatives and seropositives. And there are potential benefits for seropositives that could include control of shedding or viremia or other long-term sequelae of CMV, but we would have to prove those. And again, those are not something we're exploring explicitly in the current Phase 3 study. So as a practical matter, from a labelling perspective and a launch perspective, our goal is to try and launch the product for both seropositives and seronegatives, so that no diagnostic would be needed and it can be broadly used across populations to try and prevent the devastating effects of CMV on vertical transmission to newborn babies.
Operator:
Thank you. Our next question comes from Hartaj Singh with Oppenheimer. Your line is open.
Hartaj Singh :
Great. Thank you. You had indicated that you are going to complete the enrollment for CMV this year and maybe have a readout. Can you just walk us through the steps or how that would look like, and maybe just give us some ideas on powering statistical assumptions? Thank you.
Stephen Hoge :
Yeah, of course. Thanks Hartaj. So we're currently fully enrolled and we're accruing cases in that study. I think as we've shared before, we've actually made substantial progress in a number of cases. It is a case-driven endpoint and we'll need to see approximately 80 cases before we'll do the first interim analysis for efficacy. That, its 81 cases to be specific. That interim analysis actually will look a lot like our other vaccine efficacy studies. DSMB will evaluate that data and if we meet the statistical threshold, which is for early efficacy, meaning we're doing better than our minimum of 49.5% vaccine efficacy, then they will at that moment tell us to unblind and share the results, and of course, we will share them broadly with the world. If for whatever reason we don't quite have the statistical power in that first interim analysis efficacy, the study is powered to continue on and continue to accrue cases towards a final analysis of efficacy. Now, given the rate of the final analysis at 112 cases, now given the rate of a case accrual that we're currently seeing in the study, we do expect that we will have more than enough cases this year. We are therefore pretty confident that we're going to be seeing a readout from the interim analysis, possibly even a final analysis for efficacy in 2024. But again, since its case and event driven, we just have to bide our time. And ultimately, we will depend upon the DSMB to tell us whether or not we've met that statistical threshold.
Operator:
Thank you. Our next question comes from Luca Issi with RBC Capital. Your line is open.
Luca Issi :
Oh, great. Thanks so much for taking my question. Maybe Stephen, at INT, maybe can you remind us what's your latest thinking in terms of potential for accelerated approval there for melanoma? And then maybe on the additional tumor types that you guys and Merck are thinking about it, can you just maybe talk about what are the tumor types that you are contemplating and whether those tumor types are going to be in the adjuvant settings or in the metastatic settings? And then maybe on RSV quickly, can you just maybe expand on durability in the context of your competitor? Is there a scenario where ACIP recommends the GSK vaccine for every other year versus your vaccine for every year? Any color there is much appreciated. Thanks so much.
Stephen Hoge :
Quite a few questions there. I'll try and get them all. I apologize if I forget any one. So first on the question of INT and accelerated approval in the adjuvant melanoma setting. So as we've said before, we continue to be really excited about the data and are excited to start looking to talk to regulators about it. There have been three things that we've tried to say that had to be true for us to believe it was appropriate to even ask about accelerated approval. The first was we had to see durability. And clearly the data that we just saw from December, just two months ago, shows that durability and really a clear statistically significant result where the comparator arm, the control arm looks really just like the labeled data, and so we're incredibly encouraged by that durability, that was criteria one. But the second and third are still there and really important. The second is that we have to substantially enroll the confirmatory Phase 3 study. For an accelerated approval in this space, we do believe we have to show we've really already done the diligence to allow that confirmatory data to come in, so that three or four years from now, that further readout would confirm anything that would happen in accelerated approval context. And then the third and perhaps now increasingly important criteria, was that we had to establish the commercial manufacturing facility. And so as we've announced, we've been building a facility in Marlborough, Massachusetts. It'll be a purpose-built, personalized, individualized neoantigen therapy facility that is ultimately what will be licensed to create this product for the world, whether it's in an accelerated context or in the future with full approval. That facility is coming online. We look forward to hosting many of you and others in tours as we bring it online. But without that facility, there isn't really a product here to talk about. And so all three of them are essential. We're making progress on all three. And I think the most exciting thing is what you were just alluding to, which is the durability of the benefit we've been seeing really causes us to now lean into completing, working hard to complete the enrollment of that confirmatory study criteria too, and finish the build out of that Marlborough facility, which is the third criteria. Now, on the point of other indications that we're going after, I will defer to our partners, Merck, on the specifics. We will do it together at the right time, opening up additional Phase 3 and confirmatory studies. We do expect to open multiple this year. Those include some additional adjuvant indications. They also include some potential metastatic indications, and we are looking at monotherapy indications. And that can be either in places where PD-1s, KEYTRUDA, may not be indicated or even earlier lines of therapy. And so all of those are under consideration, and as soon as we start those studies up and begin enrolling, of course, we'll make announcements about them with our partner, Merck. Now lastly, on the question of RSV, we continue to be really enthusiastic about the data that our product has, and I think the durability now shown through this second large season is quite encouraging. We'll be sharing that data with the ACIP. Well, first we have to get to the regulatory process and approval in this country. And Stéphane mentioned our PDUFA date in May. And if we have the opportunity, we'll be sharing the data with the ACIP, which includes, in our case, booster data on immunogenicity from ongoing work that has actually already previously been presented publicly at meetings. And so just like our competitors, we have shared data of what a second dose looks like in terms of boosting neutralizing antibody titers back up, both at one year, and we're also going to be looking at two years. And all of the data, ourselves, as well as that similar booster data from the competitor products, will likely be brought together to inform the ACIP's recommendation of how they think RSV vaccines should be readministered when a booster might be necessary. It really falls to the committee to make that determination, not us. Your question was about whether we expect there to be any difference or distinction in terms of how they treat the vaccine. You know at this point, given the immunogenicity data and booster data that has been shared across all three products, which is remarkably consistent, as well as the consistent picture in terms of efficacy, including some waning efficacy for all products in the second year, we would suspect that they will continue to view the products as more similar than not and therefore continue with consistent recommendations. But it's really up to the committee to make that determination. At least from my perspective, I certainly think the science would support that.
Operator:
Thank you. Our next question comes from Salveen Richter with Goldman Sachs. Your line is open.
Elizabeth Webster :
Hey, good morning, and thank you for taking our questions. This is Elizabeth on for Salveen. Two questions from us. First, can you just remind us of where you stand with evaluating the RSV vaccine in the paediatric population? And then for the Phase 3 CMV vaccine, can you walk us through what you've seen on durability to-date and how you are thinking about the durability of that vaccine, particularly as it relates to the potential commercial opportunity in a younger adult women/adolescent population? Thank you.
Stéphane Bancel :
Hey. So I'll take the question. So on RSV vaccines [ph], we have not started the study yet. We are of course considering taking this into a Phase 3. It is in the clinic with Phase 1/2. We are waiting for the data to be able to move at the right time into the paediatric setting. In terms of CMV, we don't have data yet on durability. Again, like we've done for other programs, when we share the data, we share the data, including durability, because it's very important. As you know, the benefit of young women is they have a very strong immune system. As we've shown in our INT programs, our T-cells work very well in terms of the vaccination technology of Moderna. So we really expect to have good durability over time. But again, we have to wait for the data to make such a determination. Thank you.
Operator:
Thank you. Our next question comes from Geoff Meacham with Bank of America. Your line is open.
Alex Hammond:
Hi, this is Alex Hammond on for Geoff Meacham. Thank you for taking our question. So a great way to equalize vaccine efficacy results is to use a correlative of protection. So for RSV, when do you think we could have a line of sight into those type of metric? And how important could this be for competitive dynamics? Thank you.
Stéphane Bancel :
Good morning. This is Stéphane.
Stephen Hoge :
Hi. Sorry about that -- small technical snafu. So I'm back. I believe I caught the end of the question, which is when do we think we'll have a correlative of protection that can inform dynamics, both between products and boosting, if that's correct. We and all the other manufacturers have been sharing publicly our work on a correlative protection. We do believe that we've identified a strong candidate in neutralizing antibodies, not surprisingly, from our clinical study. We've been sharing that data with regulators, and we've been sharing the preliminary analysis with public health officials, including advisory groups and ITAGs like the ACIP. We will be publishing that data and ultimately submitting that to our regulatory submissions as a correlate through the balance of this year. And if we and I think the other competitors are successful in establishing neutralizing antibodies against RSV as a correlative protection in RSV, then it really will probably be the primary way that public health officials make determinations about revaccination and boosting, and ultimately how we maintain durable protection against RSV for high-risk populations like older adults. From a competitive dynamics perspective, once each product has established their correlate, their correlate will relate to them. But we do think there's probably going to be more commonality than not in the correlates of protection, which makes sense, because at the end of the day we're still talking about the same virus and vaccination against it for all three products.
Operator:
Thank you. Our next question comes from Terence Flynn with Morgan Stanley. Your line is open.
Terence Flynn :
Great. Thanks for taking the question. I was just wondering if you could provide an update on your discussions with the FDA for 1010, your seasonal flu vaccine, and what's gating to filing here. Thank you.
Stephen Hoge :
Thanks. So we are speaking to the FDA and regulators around the world about what would be – what they would like to see from a submission perspective for – in first-generation or influenza program, our mRNA 1010 program, as you referenced. I don't have any specific updates right now. We're in those conversations as we speak. I really don't want to get ahead of them. The kinds of things we're talking about are what's the total submission data package, what's the duration of follow-up in some of these studies, and what additional studies or data might be supportive to the application. Those conversations are ongoing. We will provide updates as and when we have them, but I have nothing further to add right now.
Operator:
Thank you. Our next question comes from Jessica Fye with J.P. Morgan. Your line is open.
Jessica Fye :
Hey guys. Good morning. Thanks for taking my question. For INT, can you talk about what's driving your confidence to go into metastatic settings? How is enrollment going in the Phase 3 melanoma trial? And I know you touched on manufacturing being important here. What's the status of that manufacturing scale of work, and when is that facility going to be ready to go live?
Stephen Hoge :
Great questions all. So, I'll take the first part of that. So first of all, metastatic. So we have not formally decided or announced that we're going into a metastatic indication. We do have data from our Phase I study, our initial Phase 1 study in metastatic patients, including non-small-cell lung cancer, but we have not yet made a determination that we're going into the metastatic indication. And I think behind your question is a view that I would agree with, which is to the extent that INT is going to provide a really substantial benefit, we think it is probably in earlier lines of therapy. So not just adjuvant, but perhaps even Stage I disease or a Stage II disease, depending on the indication, because the safety and tolerability profile is we think incredibly favorable, and the benefits we're seeing are pretty remarkable from an immune perspective. That said, there is still a really high unmet need in the metastatic space, and even immunotherapies like the PD-1s, like KEYTRUDA, provide a substantial benefit there. And so at the right time, we may well choose to study the metastatic indication, but as I said or metastatic indications and settings, but as I said, we have not yet formally decided to do that today, and we are really focused on adjuvant and earlier and monotherapy principally. On the manufacturing process and enrollment, we have substantially scaled up our ability to enroll patients in those Phase 3 studies. I can assure you that with our partner, Merck, we wouldn't have opened a second Phase 3 study and be talking about the third if we didn't have confidence in our ability to rapidly meet the demand for the substantial demand from those clinical research sites for INT manufacturing. We haven't specifically put out numbers, but suffice it to say, we are rapidly enrolling in those studies, and we would expect to make substantial progress this year and even perhaps getting close to completing enrollment in at least one of those studies if it continues trajectory. So we're excited about the progress we've made in scaling up the manufacturing for clinical supply. We're excited about the progress we're making right now in enrolling patients and the demand that we're seeing from clinical sites. And we do believe that we've solved a lot of the, for clinical research, for clinical development, the manufacturing requirements. The question then becomes commercial, and as we alluded to a few minutes ago, the Marlboro site would really become the purpose-built commercial site, which needs to not only be able to deliver high-quality products at high volumes, but also do it at a valuable price and cost point and all of that work is ongoing. We've made great progress in building that site. Our goal is to establish that site for at least clinical supply this year, but we haven't provided further guidance on when we will have that fully operational for potential commercial use. And ultimately, it depends upon discussions with regulators as well.
Lavina Talukdar:
Kevin, we'll take our last question.
Operator:
Okay. Our last question comes from Evan Wang with Guggenheim Securities. Your line is open.
Evan Wang:
Great. Thanks guys. Two from me. First, on RSV, I know there are some additional studies to expand the initial population. Can you comment on when we may see data from the Phase 3 and 50-plus in the higher-risk younger adults, and when you could potentially supplement the filing if everything's positive? And then, I know you commented on Australia. So just, as you were thinking about or as you've talked about Australia as a proxy for COVID sales in 2023. Now can you comment on how interpretable Australia would be? Will it be through your RSV launch, and as we look for trends in COVID vaccination rates? Thanks.
Stephen Hoge :
Great. I'll quickly take the first part of that and then hand it over to Stéphane for the second. So the additional data on 50-plus, we have obviously immune bridging data and co-administration data. We'll be sharing that at the appropriate time with public health officials and others. It could be as soon as the ACIP. It just depends on when the data comes in, as well as the 18-plus high-risk populations. In your question about getting those in the label, it's important to note that public health officials can recommend use even beyond the label and may choose to do that, but our responsibility would be to get it in the label, and we will need to complete the initial BLA before then we could submit the SBLA to get that data in the label. And so obviously it would follow shortly after our hopefully successful PDUFA outcome in May.
Stéphane Bancel :
Thanks, Stephan. And on Australia, so a few things. First, I mean, our team has delivered a strong performance on COVID in Australia. We've been helping the government since the beginning of the pandemic. As you recall, we have announced a long-term 10-year partnership with the Australian government, and we are currently building a plant in Melbourne that is advancing quite successfully, and we are in active discussions with regulators around Australia for RSV approval. The point I will make is that Australia, as you know is a quite different market commercially to the U.S. It's really mostly driven by the government, so I will compare Australia more to a European market than to the U.S. market. I don't think we can draw any positive or negative correlation in terms of what happened in Australia, COVID or RSV, to what will happen in the U.S. in the fall ‘24, winter ‘25.
Stéphane Bancel:
Thank you so much for the questions today. Thank you for taking the time to be with us. We look forward to talking and seeing many of you in the coming days and weeks. I hope that you will have on March 27, Annual Vaccine Day in your calendar. We'll start the presentation at 9 a.m. Eastern Time. So have a great day and thank you for joining.
Operator:
Ladies and gentlemen, this does conclude today's presentation. You may now disconnect and have a wonderful day.
Operator:
Good morning. My name is Kevin. And welcome to Moderna's Third Quarter 2023 Earnings Call. At this time, all participants are in a listen-only mode. Following the formal remarks, we'll open the call up for your questions. [Operator Instructions] Please be advised, today's call is being recorded. At this time, I'd like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's third quarter 2023 financial results and business updates. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Arpa Garay, our Chief Commercial Officer; and Jamey Mock, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I'll turn it over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Thank you for joining us today. I will start with a quick review of our business for third quarter. Arpa will then give you an update of our commercial progress and plans. Jamey will present our financial results and will explain in detail the one-time charges we announced this morning in our press release. Stephen will then review our clinical programs. And I will share our key priorities for 2024 and 2025 to return Moderna to sales growth and profitability. We delivered $1.8 billion in Spikevax sales of COVID vaccine in the third quarter. Based on trend we are seeing in the U.S. COVID market in recent weeks, we expect our sales for 2023 to be at least $6 billion. We have been preparing for the 2023 fall COVID launch throughout the year, because the U.S. market was pivoting from a pandemic government purchase market to a commercial market. I am very pleased to report that according to IQVIA market data, we have a market share in the U.S. of 45% season to date, compared to 36% for 2022. [And Arpa] (ph) will show you, we even achieved 51% market share last week in the U.S. This commercial performance in the U.S. market shows that Moderna can compete commercially with large established players, [that will prove] (ph) important as we launch RSV 2024 and combo of flu-COVID in 2025. On the cost side of the company, we informed you at R&D Day that it was important for us to resize our manufacturing footprint as the world has moved from a pandemic to an endemic setting. I am pleased that our manufacturing and finance team were able to move fast and resize our manufacturing, so that we can go back to 75% to 80% gross margin levels. This resizing resulted in a charge of $1.6 billion, which Jamey will explain in detail in his section. Now let me turn over to Arpa to walk you through our progress in the U.S. market.
Arpa Garay:
Thank you, Stéphane, and good morning or good afternoon to everyone. Today, I will provide an update on our third quarter performance, our U.S. commercial launch progress and our preparation for our RSV launch next year. Our total sales in the third quarter came in at $1.8 billion, which includes approximately $800 million in international sales and $900 million in U.S. sales. Total sales for the first three quarters of the year were $3.9 billion. Turning now to our expectations for the fourth quarter and full year 2023 outlook. In our international business, we expect an additional $1.1 billion in sales. These sales are based on government contracts and once vaccine doses are shipped and accepted by the customer, they are not returnable. More than half of these sales have already shipped in the fourth quarter. In the U.S., we expect at least $1 billion in sales in the fourth quarter, which would bring U.S. sales to approximately $2 billion for the second half of 2023. This assumes approximately 50 million doses administered in the U.S., which would be similar to the fall season of 2022. With $3.9 billion in sales recorded as of the end of the third quarter, expected fourth quarter sales of $1.1 billion internationally and at least $1 billion in the U.S., our updated sales outlook for 2023 is at least $6 billion. Now turning to the U.S. launch. The data shown here are from IQVIA. As a reminder, IQVIA data only captures the retail channel in the U.S., which includes retail pharmacies and long-term care. I'm first going to share what our weekly market share trends look like and then discuss our cumulative share since launch and how it compares to last year. On the fifth week post launch, for the week ending October 20th, what you can see on the slide here is that Moderna's market share is 51%. As we look over the season to date, Moderna's cumulative market share for this season is now 45%, which is higher than the 36% market share we had in 2022. As Stéphane mentioned, this progress demonstrates our ability to compete effectively in the commercial endemic market. Turning now to Slide 8, which shows cumulative vaccinations in the U.S. retail pharmacy channel. This year, COVID vaccines were launched two weeks later than in 2022. As a result, we analyze the data on a launch-adjusted basis as shown in the graph on the slide. To do this, we look at the data based on the number of weeks post launch rather than simply on a calendar basis. This helps us compare vaccines uptake in 2023 versus 2022. On the graph, the blue line represents cumulative vaccinations post launch in '22 and the red line represents cumulative vaccination post launch this year. And on a launch-adjusted basis, the total market is tracking ahead of last year's vaccination levels, both on a weekly basis but also on a cumulative basis. As a reminder, the retail channel is typically the largest segment, so these trends in the first five weeks are encouraging given the later launch and slightly shorter season in 2023. Let's now turn to look at the channel mix in the market on Slide 9. In 2023, we expect retail and non-retail mix to evolve as the season progresses. The non-retail segment includes independent networks, health systems, U.S. government entities, clinics, and other providers. Last year, this non-retail segment made up approximately 16 million doses in the season, or 33% of the total market. The gray line on the graph charts CDC-reported vaccinations, which capture both retail and non-retail channels in 2022. The blue line is vaccinations at retail pharmacies, as reported by IQVIA, in 2022. The difference between these two lines represents the non-retail segment. Looking at the data on the graph, you can see that the retail channel captures the majority of vaccinations early in the season. This can be seen in the narrow spread between the CDC and IQVIA data represented by the gray and blue lines, respectively. Now, if you look in the seventh week of launch in 2022, the current week that we are in, in '23, you can see that the slope of the non-retail channel is increasing. We know that distributors this year have recently increased shipments to the non-retail channel and, as such, we expect non-retail as a percentage of market to grow between now and year-end. Importantly, we expect our market share to be consistent across channels and higher in 2023 than in 2022. We are committed to focus on public health efforts to increase vaccination rates. In the United States, we are taking a multi-pronged approach to educate all stakeholders and increase the urgency to get vaccinated. Across the medical community, pharmacies and clinics, and advocacy groups, we are providing patient education resources to help our partners encourage their patients to get their COVID vaccine this fall. Earlier this week, we also launched our branded direct-to-consumer campaign, both on TV as well as across digital channels. For the remainder of the year, we will be amplifying education on the need for vaccination prior to gatherings, particularly with at-risk populations such as families, and traveling during the holiday season. We are also launching a focused education effort for consumers who are infected with COVID this summer on the importance of getting vaccinated in November and December. To summarize our COVID outlook, we expect at least $6 billion of sales this year based on the U.S. vaccination trends that signal a market of at least 50 million doses. Moderna's share, both in retail and non-retail, is expected to be consistent across channels and higher than in 2022. Vaccine administrations in the retail channel are tracking ahead of last year on a launch-adjusted basis, signaling strong early consumer demand. Last year, only about 55% of COVID vaccinations were given by the end of October, with an additional 23 million vaccinations given in November and December. We expect the non-retail channel to increase as a percentage of the market, which will provide additional sites of vaccination for consumers to allow for a strong November and December. This year, given the later launch of vaccines, the non-retail channels are just now beginning their vaccination campaigns. To continue the momentum from our launch and supplement our customers' efforts, we will be amplifying our marketing campaigns in November and December. Let me now turn to the upcoming RSV launch in 2024. We believe we have a best-in-class product profile that can make a difference both to patients but also to our customers. Our clinical profile shows strong vaccine efficacy. We have a well-established safety and tolerability profile that leverages the same mRNA technology that has been delivered in over 1 billion COVID vaccines. Additionally, we have not seen any cases of GBS in our Phase 3 trials. An important differentiator for our customers is that we will be the only company with ready-to-use pre-filled syringes, which are preferred by pharmacists and by clinicians. We continue to expect a 2024 launch of our RSV vaccine in the U.S., and are also preparing for launches in several international markets. We're encouraged by the recent RSV launches in the market this year, beating expectations due to robust consumer awareness and demand. And we believe that we are well positioned for our launch in 2024. Our strong clinical profile and ready-to-use pre-filled syringes are key competitive differentiators. My team and I are particularly excited about our ready-to-use pre-filled syringes given the robust demand for our COVID pre-filled syringes this fall. The majority of RSV vaccines in the U.S. will be given in the pharmacy setting. And given the ongoing pharmacy labor shortages, our ready-to-use presentation will save time and also reduce administration errors. We will be the only company with a one-step administration compared to competitive products, which require multiple preparation steps by pharmacists and clinicians. We are very excited for the launch of our RSV vaccine given the strong product profile. And the commercial team is well positioned to bring our RSV vaccine, our second respiratory vaccine, to market. I will now turn it over to Jamey to provide an update on our financials.
Jamey Mock:
Thanks, Arpa, and hello, everyone. Today, I will review our financial performance for the third quarter and provide an updated framework for our full year 2023 financial outlook. Additionally, given we know it's top of mind for investors, we wanted to provide our early thoughts on 2024 and how we're approaching the next couple years. Starting on Slide 15. Total net product sales for the quarter were $1.8 billion, down 44% year-over-year, driven by lower sales volume, and partially offset by a higher average selling price. Product sales were almost evenly distributed between the U.S. market and the rest of the world. We initiated product shipments to customers in mid-September for the fall booster season, following the authorization of our updated COVID-19 vaccine. Cost of sales for the third quarter of 2023 was $2.2 billion compared to $1.1 billion in the prior year. I will provide detailed commentary on the following slides. Research and development expenses were $1.2 billion, which increased by 41% versus the prior year. This increase was driven by our expanded and maturing development pipeline with six products now in Phase 3 studies or pending approval. Selling, general and administrative expenses were $442 million, reflecting an increase of 59% year-over-year. The growth in spending was primarily driven by the buildout of our commercial activities and, in particular, our launch in the U.S. commercial market. The income tax provision in Q3 was $1.7 billion, as we reported evaluation allowance against deferred tax assets of $1.7 billion. Under GAAP accounting rules, we are required to take a reserve, also referred to as evaluation allowance, for deferred tax assets when the current year and cumulative income projection for the next three years is in a loss position. These losses indicate our deferred tax assets may not be fully realized. It's important to note, future income from products not yet approved by regulators are excluded from these income projections, which restricts us to adjust our COVID vaccine, and it does not include expected future launches. In combination with our updated endemic COVID forecast, we determined it was appropriate to record a valuation allowance for our deferred tax assets. This valuation allowance did not impact cash flows, nor future returns, nor the company's ability to utilize deferred tax assets in future periods. Net loss for the period was $3.6 billion compared to net income of $1 billion last year. Diluted loss per share was $9.53 compared to diluted earnings per share of $2.53 in 2022. Finally, we ended the third quarter with $12.8 billion in cash and investments. The decline versus prior quarter was driven by our operating loss and sales to be collected in Q4. Now, let me come back to cost of sales on Slide 16. We now expect full year cost of sales of $5 billion, driven by $1.5 billion of unit-driven expenses, which also includes royalties, and $3.5 billion of inventory write-downs and charges related to CMO purchase commitments, cancellation fees, and wind-down costs. As Stéphane previously highlighted, in our pursuit to optimize the cost structure of our COVID-19 franchise, we undertook a strategic initiative in the third quarter to restructure our manufacturing footprint, which was built for the pandemic. As part of this initiative, we reduced our capacity and commitments with several third-party vendors, we reevaluated our raw material inventory levels, and cut back on our purchase commitments for raw materials not anticipated to be consumed before expiration. As a result, we are recording charges of $1.6 billion, $1.4 billion of which are in Q3 and an expected $0.2 billion in Q4. The $1.4 billion charge in Q3 consists of inventory write-downs of $0.9 billion and CMO wind-down costs and cancellation fees of $0.5 billion. The Q4 charge is related to CMO wind-down costs. Despite the immediate financial impact, we are confident that this strategic move will improve the efficiency of our manufacturing operations and establish a strong foundation for improved margins going forward. As part of the $1.6 billion in total restructuring charges I just mentioned, only the CMO-related costs and cancellation fees are cash restructuring costs. We project this approximately $0.7 billion charge will have a payback in less than two years and a net cash benefit of approximately $1 billion through 2029. As I mentioned, our full year forecast for cost of sales in 2023 is now $5 billion. Before resizing charges of $1.6 billion, our cost of sales for the full year is expected to be at the low end of our previous guidance of $3.5 billion to $4 billion. So, now coming back to my commentary on Q3. In addition to unit-driven manufacturing costs and the cost from this initiative, we also incurred approximately $0.4 billion of inventory charges for excess and obsolete material, demand-related write-downs of our latest Spikevax product, and unutilized manufacturing capacity charges. Moving to Slide 17. In summary, we made substantial progress to resize our COVID cost structure and accelerated our path towards our longer-term target of 20% to 25% of sales. We expect our cost of sales to not only be at a lower level, but also be more predictable in the future. In recent quarters, our cost of sales were highly impacted by write-offs and charges as we just addressed today and in previous calls. Year-to-date write-offs and charges for inventory CMO and supplier-related commitments are 74% of sales. Starting in 2024, we expect those to be less than 10% of sales on an annualized basis. Our capacity is now better positioned to scale with volume. At a $4 billion sales level, we expect cost of sales of approximately 35%, reducing to approximately 30% at $6 billion of sales, and 20% to 25% at even higher sales levels. In other words, with our resized manufacturing footprint, we now expect to achieve significant volume leverage moving forward. Let's now move to Slide 18. While we intend to continue to focus on GAAP results, we wanted to give you a view of our financial results in Q3 with and without the resizing and tax valuation allowance charges. While our total GAAP net loss in the third quarter was $3.6 billion, our loss excluding these charges would have been $0.5 billion. Now, let's turn to our updated 2023 financial framework on Slide 19. As Arpa mentioned earlier, we now expect product sales for 2023 of at least $6 billion for the full year, which is comprised of $3.9 billion of sales through the third quarter, an additional $1.1 billion of international sales in Q4, and at least $1 billion of Q4 sales from the U.S. As explained earlier, we now expect cost of sales for the full year of approximately $5 billion, which includes resizing charges of $1.6 billion. For R&D and SG&A, we now expect full year expenses to be approximately $6.3 billion, with approximately $4.8 billion in research and development. Our R&D spend is slightly higher than the $4.5 billion previously forecasted, which is mainly driven by business development activities as well as additional investments in our late-stage clinical trials. Our forecast for SG&A expenses remain consistent at approximately $1.5 billion. We now expect a full year tax expense of approximately $0.8 billion to $1 billion, driven by the $1.7 billion increase in the deferred tax allowance I referenced earlier. And finally, we now expect capital expenditures of approximately $0.9 billion, down from our previous guidance of $1 billion. Before I get into the specifics on 2024 and 2025, I wanted to share with you our principles on how we are operating the company today and our plans over the next three years. We are laser-focused on making our COVID franchise profitable in 2024 and beyond. We look at our Spikevax product profitability, excluding research and development costs, for our future pipeline, and we believe our recent resizing efforts will ensure that our COVID franchise is a continuous and increasing source of income and cash generation. At the same time, we also recognize the significant and unique opportunity for organic sales growth ahead of us with our late-stage pipeline. We will be disciplined in our investment approach and adjust R&D and SG&A based upon the sales performance of our product lines, which in 2024 is still mostly COVID, but we expect it to also include RSV. We expect this investment in our late-stage pipeline will result in a loss over the next two years, but help us to breakeven starting in 2026. We believe our current balance sheet is more than sufficient to fund our plans without the need to raise equity. We are also not planning to repurchase shares in the intermediate term. Stepping back, we believe this is an unparalleled opportunity to impact the lives of patients while creating shareholder value at the same time. Moving to Slide 21, let's start with our view on sales over the next couple of years. We're expecting sales to hit a low point in 2024 at approximately $4 billion. The biggest change year-over-year is related to our signed APAs. We currently have $1 billion of COVID-related APAs for delivery in 2024. Recall that our first half sales in 2023 of $2.1 billion were mostly deferrals from our 2022 existing contracts. So, we expect minimal sales in the first half of 2024. For the U.S., we expect 2024 to be at least $2 billion and believe it will grow over time. Lastly, we expect approximately $1 billion from RSV and other international COVID sales. And finally, in 2025, we expect a return to growth. Let me finish by giving you a more fulsome view on 2024 and our thinking on 2025. Starting with 2024, as I just explained, we expect sales to be approximately $4 billion. Cost of sales are expected to be approximately 35% of sales. R&D expenses of approximately $4.5 billion in 2024, would be down 6%. In 2024, the majority of our R&D expenses are for registration trials, which are now mostly committed. I will speak to our view on 2025 R&D expenses in a moment. SG&A expenses of approximately $1.3 billion in 2024 would be down 13%. We expect taxes to be negligible in 2024 and capital expenditures to be similar to 2023 at $0.9 billion. In summary for 2024, Spikevax will generate nearly $1 billion of income. When we combine that with our estimated investments in R&D and capital expenditures, our cash balance is projected to be approximately $9 billion at the end of 2024. Now for our preliminary thoughts on 2025. As mentioned earlier, sales will return to growth. Cost of sales will improve with the increased sales growth. R&D will be flat to down, and we have much greater flexibility for reduction, given that only approximately half of our current R&D spending levels for registrational trials are committed for 2025. SG&A will be flat to down. Taxes will continue to be negligible. And capital expenditures will be materially lower after the completion of our facilities in the UK, Canada and Australia in the first half of 2025. In summary, our COVID operating income will grow and our investments will remain flat or lower, leading us to an estimated ending cash balance of approximately $6 billion to $7 billion in 2025. Finally, during this period, we expect to launch five new products to help us breakeven in 2026. So with that, I'll now turn the call over to Stephen.
Stephen Hoge:
Good morning or good afternoon. Today I'll do a quick review of our clinical programs. Many of the details from these programs were shared at our R&D Day in September. I will also review the Phase 3 trial designs for our combination flu and COVID vaccine, mRNA-1083 and the Phase 3 trial design for our INT in non-small cell lung cancer. During R&D Day, we shared the significant progress we've made through the year in advancing our late-stage pipeline, creating the opportunity to have up to 15 product launches in the next five years. Through 2025, and subject to regulatory review and approvals, we anticipate launches for our RSV vaccine, our flu vaccine, a next-generation COVID vaccine, and our combination flu and COVID vaccine. Looking beyond 2025, we have a diverse pipeline of other vaccines, cancer therapies, and rare metabolic disease medicines. We're very excited by the potential benefits these medicines offer across a diverse range of therapeutic areas. Slide 25 is an overview of our respiratory vaccines pipeline. Our leading pipeline includes commercial and Phase 3 programs against COVID, RSV, and flu, as well as earlier-stage next-generation programs in COVID and flu and multiple [combinations] (ph). We recently shared positive topline Phase 1/2 data from our combination flu and COVID vaccine, mRNA-1083. And on the heels of this success, we've started and are rapidly enrolling a Phase 3 study for mRNA-1083 in adults 50 and older. Slide 26 shows the Phase 3 design for mRNA-1083. The Phase 3 study is a randomized, stratified, observer-blind, active-control study evaluating the immunogenicity and safety of 1083 compared to co-administered flu and COVID vaccines. The study will enroll 8,000 participants overall, with two cohorts of 4,000 participants stratified by age 65 years and older and 50 to 64 years of age. Both cohorts will receive mRNA-1083, or in the control arm, an age-recommended licensed quadrivalent flu vaccine plus our approved COVID vaccine. Study participants will be followed for six months. The next, on Slide 27, is an overview of our latent and other vaccines pipeline. As previously announced, our Phase 3 study evaluating vaccine efficacy and safety of our CMV vaccine in women of childbearing age is now fully enrolled, including the adolescent cohort. The study is accruing cases and we look forward to vaccine efficacy data when it becomes available. Earlier-stage clinical programs against EBV, HIV, VZV, HSV, and against the Norovirus and Lyme disease continue to progress. Slide 28 is an overview of our therapeutics pipeline. We're proud of the progress across cancer, rare diseases, and other areas. Many of the details of these programs were highlighted at our recent R&D Day, and I refer you to that presentation on our website. Notable since R&D Day is the continued rapid enrollment of the INT Phase 3 adjuvant melanoma study, which has seen a great deal of interest from investigators and patients since presentation of the Phase 2 data at ASCO earlier this year. As previously announced, before the end of the year, we will conduct another analysis of the Phase 2 study with longer follow-up time at approximately three years, and we are looking forward to sharing that data INT is also moving forward in other types of cancer with the initiation of a second Phase 3 study in adjuvant non-small cell lung cancer. The Phase 3 design for adjuvant non-small cell lung cancer is shown on Slide 29. It is a randomized placebo-blind -- double-blind placebo and active comparator controlled study of a combination of INT plus KEYTRUDA versus placebo plus KEYTRUDA in patients with non-small cell lung cancer. The study will enroll approximately 900 patients with stage II to IIIB tumors who were resected and previously treated with adjuvant chemotherapy. Each patient will receive up to nine doses of INT administered intramuscularly every three weeks with KEYTRUDA administered every six weeks in the active arm, or nine doses of a placebo injection administered every three weeks and KEYTRUDA every six weeks in the comparator arm. The primary endpoint of the study is disease-free survival. Key secondary endpoints include overall survival, distant metastasis-free survival, and patient-reported outcomes. This study marks an important milestone in our collaboration with our partner, Merck, and our shared commitment to rapidly bring INT to patients. With that, I'll turn it over to Stéphane.
Stéphane Bancel:
Thank you, Stephen, Jamey and Arpa. Our focus is to return Moderna to sales growth and profitability. To achieve that, we have three priorities. Priority number one, commercial execution. Our market share in the U.S. demonstrates we can compete. We are focused on launching RSV in 2024. We believe we have best-in-class profile for RSV vaccine, high-efficiency, good safety profile, and the easiest to use in pharmacies or doctor's offices. As you all know, pharmacy chains are having challenges given the workload, especially in the fall season. The two other vaccines on the market today are very complicated to prepare before injection; one is nine steps, and one is four steps. Now we launched pre-filled syringe and, as Arpa said, we are very excited about that. And from the discussions I had with the leadership of pharmacies, I believe that this will be an important differentiation. Our COVID plus flu combo vaccine should launch in 2025. And as you know, flu is a higher volume market than COVID. In the U.S., for example, flu is around 3 times the number of doses compared to the COVID market. With these new product launches in '24 and '25, and the combination of COVID sales in the endemic setting, we believe Moderna will be in sales growth again in 2025. Priority number two, discipline investments. We'll be disciplined in our investments and [cycle] (ph) them based on our sales performance. As you saw, we have taken bold actions to resize our manufacturing footprint in the third quarter. The team is not done and there are many continuous improvement projects to drive a reduction in cost of manufacturing. We'll also look at our R&D costs and will consider partnering some programs, if needed, to allow us to be responsible and disciplined about our costs. For SG&A, as Jamey mentioned, we are currently going through our 2024 budget and our goal is to have a lower spend in SG&A in 2024 than we had in 2023. We still plan to keep SG&A flat in 2025 versus 2024. We're launching respiratory products in 2024 and 2025, and we anticipate the same teams [indiscernible] productivity gains as we improve our commercial operations. As Jamey mentioned, we expect to breakeven in 2026. Priority number three, executing on our late-stage pipeline to drive sales growth. As you all know, we have an exciting late-stage pipeline with six Phase 3 assets. For respiratory program, RSV, flu, next-gen COVID, mRNA-1283, and COVID plus flu combo that Stephen just talked about. One latent program, CMV, which is fully enrolled in Phase 3 and accruing cases. One oncology program, INT in melanoma, and as Stephen mentioned, in lung cancer. We look forward to having and sharing three years of INT data from our melanoma study before the end of this year. If the data are strong, we believe it could be the basis for regulatory discussions for potential accelerated approval. We have been investing in building a factory in Marlborough, Massachusetts, which will enable the commercial launch for INT. Thanks to the mRNA platform we built, we have an exciting pipeline, with up to 15 launches in the next five years. We have the largest late-stage pipeline of any mRNA company in the world and will continue to focus to deliver the greatest possible impact to people for mRNA medicine. I've never been more excited about the potential we have to deliver for patients. The actions we are taking help us to execute on that vision. With this, operator, we'll be happy to take questions now.
Operator:
Thank you. [Operator Instructions] Our first question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
Thank you for taking my questions. I have two questions regarding the commercial questions. First, you did mention 2026, you're looking to have a breakeven. And when we take a quick look based on your 2024 and 2025 outlook, it seems the total cost could be in the $8 billion to $9 billion, that range. Could you give us a sense what could be the additional sales if we're using 2024 guidance as a base point? And the second very quickly regarding the manufacturing resizing. After resizing, what is the full capacity regarding doses and what percentage of the manufacturing will be internal in 2024 and 2025?
Jamey Mock:
Maybe I'll take the first one -- first part of the question. So, in terms of 2026, and thanks for the question, let's talk to you about how we're thinking about it. So, we mentioned $4 billion in 2024, approximately $4 billion. And this is all about our late-stage pipeline coming to fruition. So, in 2024, we'll launch RSV, but it's mostly kind of in the second half year sales, and then we'll have a full year in 2025, so that'll provide growth. We will also come to market with flu in 2025. We'll also come to market with a combination of flu and COVID in 2025. Again, depends on timing, but by 2026, we should have a full portfolio. So, we're not going to say what the exact sales numbers are, but you mentioned $8 billion to $9 billion in costs. I'm not exactly sure where you'd get there unless you're assuming a certain sales line on that, but let me go back to what we tried to lay out here. If you assume, for instance, $6 billion in sales, we should have 30% of cost, or $8 billion in sales, we should have 25% of cost. And then, we've said historically in our R&D Day that we need $25 billion overall to make this investment, which should average $5 billion a year. So, hopefully, we're giving you enough pieces without officially guiding any kind of numbers in 2026. But here's what's also important is, if those sales don't come to fruition, we are telling you that we will adjust our expenditures in our investment. So, that's -- we hope that we are -- they will, we are confident in our pipeline, but should it not happen, then we were prepared to adjust our investment. I missed the second part of the question.
Arpa Garay:
Capacity.
Jamey Mock:
Hey, Gina, can you maybe repeat the second part of your question? I apologize. I missed it.
Gena Wang:
Sure. Basically, after manufacturing resizing, what is the full capacity regarding doses, and what percentage will be internal?
Jamey Mock:
Yeah, thank you for the question. So, we -- as we try to lay out here and are showing you, this capacity is built for volume leverage. So, we at least put $10 billion of sales on that page, and it will require no additional capacity. We will complete, of course, over the next year and a half the UK facility, the Canada facility and Australia facility, but for the respiratory framework, we need no more, at least the $10 billion. I won't project beyond that, but that should answer that question. INT is a little bit different and we are building that and getting that ready for commercial purposes, but we're built for volume leverage moving forward.
Gena Wang:
Thank you.
Operator:
Our next question comes from Salveen Richter with Goldman Sachs. Your line is open.
Salveen Richter:
Good morning. Thanks for taking my questions. Two from me here. One is, you provided guidance of about $4 billion between COVID and RSV on the [forward here] (ph) for 2024. Could you just speak to the contribution from each and how you're thinking about flu monotherapy? And the second question is that your financial framework for 2025 includes the ability to flex R&D and SG&A. Are there any parameters you can share on the range of this flexibility and how you would prioritize R&D programs and development? Thank you.
Jamey Mock:
Yeah, thanks, Salveen. I appreciate the question. So, on the $4 billion, we're not going to break out the $1 billion that we attributed to RSV and COVID. All we can say is we've talked about our PDUFA date and that we filed in certain amount of countries across the globe. I will also say, as Arpa mentioned in her prepared remarks that we're super confident in the product profile. We are encouraged by the market and how it's already started from an uptake perspective, and we think we will compete very well in 2024 and beyond. As it pertains to the flexing on our spending for 2025, obviously, I don't know, 80% of our expenses or investments are in R&D, so $4.5 billion for R&D and $1.3 billion for S&A. So that, as I mentioned, 50% of the current spending levels is not committed. So, we have time to make decisions and watch the market to be able to say what amount of registrational trials and what amount of R&D are we going to spend in 2025. So, hopefully that gives you a sense for how much is still the ability to flex. We also have other levers that we can pull, et cetera. SG&A, we also have some flexibility, probably not to the same magnitude, but there is still some amount of flexibility to bring that down from a variable expense perspective.
Stéphane Bancel:
And Salveen, this is Stéphane. Maybe just to add to Jamey's point on the R&D, as I mentioned in my remarks, if we have to, we will be open, of course, to partnership some of those programs, which is an important way we could flex the R&D number based on where the sales are, as Jamey mentioned, which is if the sales are according to our plan, then we're going to be okay. If the sales are below, we will be very open to partnering. As you know, we've done that in the past. The team knows how to do it. But we will be disciplined about our investments in the business based on where the sales line is.
Operator:
Thank you. Our next question comes from Eliana Merle with UBS. Your line is open.
Unidentified Analyst:
Hi, this is Sarah on for Ellie. Thanks so much for taking our question. First, I guess, can you talk about in 2024 again on that $1 billion RSV international sales number? Are you expecting any contribution from flu in '24, and maybe how you're thinking about it into '25? And then, on CMV, can you talk about where you are in cases and how they're tracking versus R&D Day where I think you said a fourth of them were currently tracked? That would be great. Thanks so much.
Jamey Mock:
So, thanks, Sarah. I'll take the first part and then hand it over to Stephen on CMV. So, there is no flu contribution in our 2024 sales outlook of approximately $4 billion. So, in that $1 billion, that is solely RSV and other COVID international sales. We do, as I mentioned in my -- to answering Salveen's question, we do expect to launch flu in our combination products sometime in 2025, and we'll see what we've projected that time.
Stephen Hoge:
And on the CMV question, thanks for that. So yes, we did update that we're about a quarter of the way through the case accrual back in R&D Day. I think the next -- we continue to accrue cases at a steady pace. I do think the next update will provide is likely our Vaccines Day in the spring.
Unidentified Analyst:
Okay, thanks.
Operator:
Our next question comes from Terence Flynn with Morgan Stanley. Your line is open.
Terence Flynn:
Great. Thanks so much for taking the question. I know GSK has provided an estimate in terms of size of the RSV market, about £5 billion, and Pfizer has given some metrics as well. Given what we're seeing now with these early launches, can you provide us with your assessment of total market size here? And then given some of your comments on competing with larger companies, as you're doing in COVID now, where ultimately do you see your market share shaking out in the RSV space? Thank you.
Arpa Garay:
Thank you for the question. In terms of the total RSV market, as I mentioned earlier, we're excited by the uptake and the consumer awareness of the market overall. And our projections are similar to what both GSK and Pfizer have already guided. In terms of our market share with RSV, we have not yet provided or are ready to provide any forward-looking projections on share, but we are very excited about our strong product profile, both in terms of efficacy, safety, and, as I mentioned, our ready to use pre-filled syringes. So, we will be leveraging the learnings and the success from our COVID commercial launch this year and applying them to RSV next year.
Stéphane Bancel:
Yes. Just to add to Arpa, Terence, it's Stéphane, the point that Arpa and I made about the market share of COVID is what I think is very important. I think some people believe that because we're a new company in commercials we're not able to compete and I think the market share data that Arpa has shared really show that our U.S. team is able to compete and we will continue to improve things that we are doing, because we are not done improving the [indiscernible] culture as you know us. But the share already moving from 36% last year to 45% cumulative so far in the season, I think it's already a demonstration of what the team is able to achieve. And the season is not over, so that's 51%, so let's see where this one finish when the season is over. But basically, the [differential] (ph) we have, as I mentioned, I've been speaking to pharmacist leadership. And they are all, I think, have a very big workload issue, as you know. There's even strikes in some pharmacy chains in the U.S. as we speak. And you think about the season there, [indiscernible] business for the pharmacy for preparations, and then the flu, and then the COVID. And then, as I mentioned, those two other products, if you just download the label of those products from the FDA website and you look at how many steps they have, it's very complicated. And when you talk to a pharmacy leadership, they don't know how they're going to deal with that type of workload. And so, coming with pre-filled syringe is a tremendous differentiator. We have very good efficacy. We have very good safety profile. We really believe that we have the best in-class product in the market. And [indiscernible], it's going to translate, I think, into a very good effect.
Operator:
Thank you. Our next question comes from Jessica Fye with JPMorgan. Your line is open.
Jessica Fye:
Hey, guys, good morning. Thanks for taking my question. Just a couple coming back around to one that I think some others were trying to get at, but maybe a little differently. When we think about breakeven in 2026, what are you contemplating in that sales number embedded in your assumption? Does it reflect just respiratory vaccines? Or are you considering INT could be on the market then? And then second, I know you said the percentage of non-retail jobs would grow as the season progresses from where it has been so far this season. Do you believe that the proportion of COVID shots running through the retail channel has shifted at all, bigger picture in 2023 relative to 2022, or should we think of that proportion as remaining similar year-over-year? Thank you.
Jamey Mock:
Yeah. Thanks, Jess, for the question. Again, without getting into too much detail on 2026 in terms of how we think about it, I mean, the best way to keep going back to that late-stage pipeline that we've been talking to you about, RSV, flu, our combination, our next-gen COVID product as well, will all be very much there for the year 2026. And we are confident in all those product profiles and how we will compete. As I mentioned, at 6%, our cost of sales -- at $6 billion, sorry, our cost of sales would be 30%. At $8 billion, our cost of sales would be 25%. And of course, we'll try to improve on that. And that will give us the envelope for how much we can continue to invest in the future products, which we said we'll launch 15 by 2028. That'll be all of our latent product portfolio, that'll be our INT portfolio, that'll be our rare disease portfolio. So, I think that's as much as we can say right now. I just want everybody to know that we are very committed to breaking even in that year, and we have a lot of flexibility, both from a growth standpoint and a discipline investment standpoint.
Arpa Garay:
Great. And I can take the second question on the percentage of non-retail. So, as expected, in 2023, the retailers have been the majority of the market, with more than 90% of the volume during the first few weeks. However, we're now beginning to see a shift towards more non-retail channels, as I had mentioned. We are seeing increased shipments to IDNs, to clinics, to pediatricians, as of the recent weeks. And as I think about full year 2023, I believe the retail mix will be stronger than in 2022 and could land at about 70% to 80% of total vaccinations, whereas in 2022, we saw that the retail channel was only about two-thirds of the mass.
Operator:
Thank you. Our next question comes from Luca Issi with RBC Capital. Your line is open.
Luca Issi:
Oh, great. Thanks so much for taking my questions. Maybe circling back on the P&L, I appreciate all the effort on resizing manufacturing and the focus on gross margin, but how should we think about OpEx plus CapEx? As COVID numbers continue to come down, we've seen the BioNTech and Pfizer materially realigning OpEx plus CapEx to their top-line. I believe BioNTech lowered by $600 million this year and Pfizer by $1 billion this year and $2.5 billion next year. However, your OpEx plus CapEx is not materially changed this year and you anticipate that the next year it's going to be generally flat to down. So, can you just maybe comment on why you think that's the right strategic decision for the organization? And then maybe second question on RSV. Obviously, impressive initial launch by GSK and Pfizer and appreciate the differentiations of your product. But what's the latest thinking on whether the vaccine is needed every year or less frequently than that? Is there a scenario where Pfizer and GSK penetrate the market pretty aggressively this year and then you face an uphill battle next year as it turns out that we need a vaccine maybe every other year and not every year? Any thoughts there much appreciated. Thanks so much.
Stephen Hoge:
Maybe I'll take the first -- the second part of that question and then hand it over to Jamey for the first. So, on RSV and the need for it, obviously, we're continuing to follow the public health situation in terms of the rate of occurrence of the RSV epidemic this year. At this point, I think we don't have data yet on whether or not it will ultimately be an annual or something less than annual, say every two or three year vaccination regime. I think like everybody else, we'll be looking to our data, the other manufacturers' data, as well as the public health, the epidemiologic data to guide that decision. There are plenty of vaccines for which there is an approach, flu as an example, where there's a seasonal vaccination approach, both because of the benefit offered by the vaccine, but also because of the convenience of just making sure that every season, every year, people are reminded to get that vaccine. So, the ultimate decision on whether this is going to be recommended is not ours, the manufacturer, it will fall to public health officials based on a number of factors, which will include the [indiscernible] and the data we provide, but other factors as well. And we'll work to make sure they have the data they need to make that decision.
Jamey Mock:
And I'll take the first part. Thanks, Luca, for the question. I think the short answer is the opportunity set ahead of us, and we are acting. So, you referenced some of our competitors, so I just want to break that down. I mean, we are super encouraged by the opportunity for additional growth and our ability to impact patients. And we have this 15 products that we think will launch by 2028 or by 2025. We think that's the right thing to do. We have to grow out of our, we have to grow this company and to be able to afford the investment to be able to capture the unparalleled opportunity for this. And I think we are acting, I believe we are acting, and I mentioned everything that we're doing from a cost of sales perspective. And so I think that's very much in line and sized appropriately to have volume leverage when it comes, because it will come. And we are saying in 2024, we can adjust both R&D and SG&A down to a good level, down 6% R&D, down 13%, so, on SG&A. We are largely committed to our registrational trials for 2024. But as I mentioned, we don't have as much flexibility in that particular year, but by 2025, we have even more flexibility. So we're prepared to take action should we need to, but we're very optimistic about the price line that's coming. And hopefully this will just come through growth, and we'll still be able to afford much of this investment.
Stéphane Bancel:
It's Stéphane. Just maybe adding to Jamey, who said it super well. As you know, we have a platform company. And the [indiscernible] success of those programs we feel very good about. If you just look at with COVID and Phase 3 RSV -- sorry, and Phase 3 for RSV and Phase 3 for flu, we have three out of three positive Phase 3. This is not your industry average. So, we think we can create value and create return on capital for shareholders by investing that capital to high-priority projects that are in late-stage pipeline. As I said, we have the largest late-stage pipeline of any mRNA company. We have six programs right now. And as soon as we launch [indiscernible], which is very, very soon, there's going to be seven programs. We believe the best way to create returns for shareholders is to invest that capital to drive sales growth and profitability.
Luca Issi:
That's it. Thanks so much.
Operator:
Our next question comes from Michael Yee with Jefferies. Your line is open.
Unidentified Analyst:
Hi. Thanks for taking our question. This is Dina on for Mike. I just wanted to get a sense of your assumptions for Q4 COVID jabs and what are you seeing in Q4 right now? How much of that is actually jabs and actual injections versus channel fill? And just to follow-up on that. Now that you've seen sort of half of the 2023 fall season play out, what are your assumptions for 2024 and 2025 for COVID? Are you essentially assuming that the same people who got vaccinated this year will continue to get COVID vaccine every year? Thanks so much.
Arpa Garay:
Thank you for the question. In terms of the fourth quarter '23 jabs, what we saw in 2022 is there was a significant portion, about 45% of the total COVID vaccinations happening in November and December. This year, we're expecting a similar split, likely larger, given that we launched two weeks later into the season in 2023 than we did last year. And what we are hearing from our different non-retail customers as well as our retail pharmacy partners is they are planning vaccination campaigns and marketing efforts to really capture on the November and December months. So, in total, we do anticipate getting to at least 50 million doses this year, and we do believe that November and December will be strong months for us. In terms of 2024, our assumption is everyone who has gotten their booster in 2023 will at least get their booster also in 2024 and beyond. Now, given the higher burden of disease with COVID, as consumers become more understanding of the annual recommendations and as the convenience of getting both flu and COVID becomes more normalized, we do believe over time we'll start to see some increase in the overall COVID market.
Operator:
Our next question comes from Hartaj Singh with Oppenheimer. Your line is open.
Hartaj Singh:
Great. Thank you for my question. I just got a question on the combination programs. And just to give a little bit of -- frame the question, in other therapeutic areas, aside from vaccines for infectious diseases, for example, oncology, monotherapy treatments generally tend to be minority of treatments, 10%, 15%, 20%. Currently, monotherapy vaccines dominate the market in COVID-19 flu. So when you get the combination vaccines going, do you imagine -- does your market research tend to suggest that you would -- again, probably a combination approach might dominate that versus a monotherapy approach, singular vaccines going forward? And then secondly, will the cost of goods sold be any different for the combination versus the monotherapy products? Thank you.
Arpa Garay:
Great. Thank you for the question. So, we do anticipate that our combination vaccine will take a substantial share of the monotherapy vaccines that are available. We have seen in the pediatric vaccine market that upon availability of combinations, you see very strong uptake and conversion from monos to combinations, and we expect a similar trend in the adult market. From our market research, we have heard consistently from consumers that they prefer one shot over multiple shots. From a customer perspective, we are hearing, as Stéphane had mentioned, just with workload issues, one shot saves a lot of time and also helps them to get more patients protective. And from a broader healthcare system and government and payer perspective, we are hearing an increased need to help get greater uptake and compliance in adult vaccinations. And our healthcare authorities believe that combinations can help actually boost the vaccination rate. So, we are very excited about our combination products in the future and think this could really be an inflection point for our mandatory vaccines.
Stéphane Bancel:
Yes, it's Stéphane. Just to add to Arpa's comments, during COVID, we've been discussing to a lot of -- with healthcare ministers, and the topic of vaccination combination has come a lot. And as you think, especially outside the U.S., where you have a lot of [indiscernible] taking care of people from birth to death, basically, we are very, very interested in combinations. Because they know that if a participant of a country got the vaccine, they got protection against several viruses which prevent hospitalization. As you know, we've done partnership with some countries like the UK, Canada, and Australia. And through those negotiations, the concept of combination was critical in their decision-making. Because as they see their population getting older, they worry that the number of hospitalizations will just go up over time and the ability to prevent that when you see shortages of healthcare workers and as you project those shortages in the future is a key determination of a decision. So, I only think in integrated healthcare system, the drive the [good combo] (ph) will move even faster than actually in commercial markets like the U.S. market.
Jamey Mock:
And, Hartaj, maybe I'll take the cost of sales question. Thank you for it. So this provides a substantial margin expansion opportunity. So, if you think about it, our cost of sales, the smallest portion is our drug substance, so it's our actual mMRA, and that's a very small portion of our overall cost of sales. Everything from drug product in terms of the cost to finish the product and the presentation type, whether it's PFS or a vial or whatever, that now gets cut in half. So, when we sell two, it's a very limited amount of cost increase versus a single presentation. So, it does provide a significant margin expansion opportunity. So, thank you for the question.
Hartaj Singh:
Thank you all.
Operator:
Our next question comes from Evan Wang with Guggenheim Securities. Your line is open.
Evan Wang:
Hey guys, thanks for taking the question. Appreciate you guys sharing early thoughts on '24 and beyond. For '24 specifically, you talked about some of the contribution from COVID and RSV in terms of split. It sounds like you plan to hit the ground running there in RSV. With international, how are you thinking about the longer-term contribution from COVID as competitor agreements expire? And with flu, with the comments on [marketing] (ph) '25, wondering if you've had any recent conversations with the regulators there in terms of potential approval. Thanks.
Arpa Garay:
Thank you for the question. In terms of our expectations in 2024, we have put about $1 billion across international COVID and RSV. We do anticipate a strong launch in the second half of the year with RSV. And on the international side for COVID, we are continuing to pursue multiple options across a number of countries. In Japan, we will be in a fully commercial market, is our expectation, where we will be competing for the Japanese business. In the EU, we continue to work with countries on agreements to secure our COVID-19 vaccine. As publicly disclosed, the EU has renegotiated their contract with Pfizer earlier this year. So, the EU demand has been substantially satisfied in many markets, but we are hearing from individual member states that they are looking for a second supplier for vaccines. And we are in those discussions right now, both at a country level, but also at a European Commission level to see if a joint procurement agreement can be established in 2024.
Stephen Hoge:
And thank you for the flu question. So, as you referenced, we had really strong data out of our P303 Phase 3 study for flu that we released at R&D Day. We're excited about that. We are engaging right now with multiple regulators about the pathway to licensure. I don't have an update about all those conversations because they're happening as we speak, but we will, once we have clarity across all markets on the pathway licensure, provide an update.
Operator:
Thank you. Ladies and gentlemen, this does conclude the Q&A portion of today's conference -- end of the call itself. You may now disconnect, and have a wonderful day.
Operator:
Good day and thank you for standing by. Welcome to Moderna Second Quarter 2023 Conference Call. At this time, all participants are in a listen-only mode. After the speakers' presentation, there'll be a question-and-answer session. [Operator Instructions]. Please be advised today's conference is being recorded. I would now like to hand the conference over to your speaker today, Lavina Talukdar, Head of Investor Relations. Please go ahead.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone. And thank you for joining us on today's call to discuss Moderna's second quarter 2023 financial results and business updates. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Arpa Garay, our Chief Commercial Officer; and Jamey Mock, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I will turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon. Today, I will start with business review of our second quarter. Stephen will then review our clinical programs before Arpa gives an update on our commercial progress and plans. Jamey will present the financial results and I will come back to share some final thoughts. In Q2, we reported revenues of approximately $300 million, reflecting the seasonal nature of endemic respiratory vaccines. Given our investments in four infectious disease vaccine programs, which are in Phase 3, and our oncology program also in Phase 3, this offseason quarter sales resulted in a GAAP net loss of $1.4 billion, GAAP diluted loss per share of $3.62. We ended the quarter with a cash and investment balance of $14.6 billion. We continue to execute on the capital allocation strategy, prioritizing organic growth of our platform, with investments in our business. If Q2, we invested $1.1 billion in R&D, continuing investments in late stage clinical programs and progressing our pipeline. SG&A costs were approximately $300 million. Capital investments were approximately $200 million. Year-to-date, our external investments were all strategic collaborations, with the exception of OriCiro in Japan, which was an acquisition. OriCiro was renamed Moderna Enzymatics and is being integrated into Moderna manufacturing process development organization. From a capital return perspective, we repurchased 4.4 million shares in Q2 for a total of $628 million. Now turning to commercial and late stage clinical update in the quarter. With the submission of our XBB updated COVID-19 vaccine applications to regulators globally, we are now awaiting approval to start fall of 2023 sales. As you will hear from Arpa shortly, we're updating our COVID-19 sales expectation for 2023 to a range between $6 billion and $8 billion. This range reflects additional contracts in the US commercial market and other countries. This range is wide, given the uncertainty on the US vaccination rate. Our commercial team is also preparing for the 2024 launch of RSV, our next respiratory commercial product. Regulatory applications have been submitted in major markets around the world. We've also started to manufacture mRNA-1345 in preparation for the launch. As a reminder, at launch, these products will be in a prefilled syringe presentation, which combined with the strong efficacy profile will position very well our product to healthcare professionals. In oncology, for mRNA-4157, our individualized neoantigen therapy, or INT, we're continuing to scale up manufacturing to support clinical development and commercial markets. We're very pleased to report our Phase 3 study in adjuvant melanoma has begun enrolling patients in July. We'll plan, on slide 6, our updated company profile. The breadth of our late stage pipeline means we could see multiple launches in 2024, 2025, 2026. Turning now to slide 7. I am proud to share that Moderna has been named one of the world's most innovative companies in 2023 for pioneering AI-driven innovation by the Boston Consulting Group. As many of you know, AI has been part of the foundation of Moderna's research and development programs for several years. We have built our own AI models for protein and mRNA engineering, assay data analysis, regulatory interactions, and many more other use cases, as we continue to be an AI leader in biopharma. One of our key Moderna mindset is that we digitize everything possible as we recognize that this will be central to the impact we have with AI. As of 2023, AI training is required for all Moderna team members, and we facilitate this training at the corporate level through our AI academy. We have challenged everyone across all of our business functions at Moderna to corporate AI into their everyday workflow. I'm happy to share our AI implementation is accelerating throughout Moderna. Our secure large language model is called mChat. As you can see on the graph, our mChat usage has grown rapidly amongst our employees since its introduction on May 18. Around 50% of Moderna employees use it already only 60 days after launch. With that, I will now turn to Stephen for an update on development programs.
Stephen Hoge :
Thank you, Stéphane. Good morning or good afternoon everyone. Today I'll review the progress of our key clinical programs at Moderna. And I'll start with our respiratory vaccines. On the left hand side of the slide, we have our commercial and late stage clinical pipeline program against three important respiratory viruses, COVID-19, flu, and RSV. I'll share some updates on these programs in a moment. Our next generation programs have made substantial progress over the first half of this year, including mRNA-1283, which is enrolling participants in a Phase 3 study, and our next generation influenza vaccines which are both in Phase 2. We currently have six combination vaccine programs addressing adult and pediatric populations. Our combination vaccines are designed to address the largest healthcare burdens caused by respiratory infections, while providing multiple advantages such as increasing compliance and reducing administration costs. Turning to our COVID program. We have submitted our applications for approval and authorization of mRNA-1273.815, our updated monovalent COVID-19 vaccine, targeting the XBB.1.5 variant. Recall that the June VRBPAC meeting, the committee recommended an XBB targeted monovalent vaccine with a preference for the XBB.1.5 strain. This recommendation was adopted by the FDA and is in concordance with EMA and WHO guidances. In the US, the CDC ACIP is expected to publish recommendations and guidelines for the use of updated COVID-19 vaccines following approval and authorization this fall. On slide 11 are the clinical data our team presented during the June VRBPAC meeting. Data from a subset of our Phase 2/3 study with mRNA-1273.815 demonstrated potent neutralization against XBB.1.5 and other variants of the XBB lineage. We're proud that Moderna was the only company that presented clinical data with an XBB.1.5 candidate in advance of the season, which we believe may help uptake of this important vaccine. Moving to RSV. As Stéphane mentioned earlier, we are pleased to be on track for regulatory approvals in 2024. Earlier this month, we announced a rolling submission to the FDA, and we plan to use a priority voucher to accelerate that review. We also filed additional regulatory applications in Europe, Switzerland, Australia, and the UK. We're incredibly encouraged by the profile of mRNA-1345 and look forward to the expected commercial launch next year. Next, on the seasonal influenza program. I'm pleased to announce that our P303 study is fully enrolled, and we look forward to sharing an update this quarter. P303 is testing an update to mRNA-1010 is designed to increase the HAI neutralizing titers against the B antigens. There's a safety and immunogenicity Phase 3 study that we believe will support accelerated approval of the updated mRNA-1010 candidate. Now turning to our latent vaccines on slide 14. Our Phase 3 CMV vaccine study in women of childbearing age is ongoing, and I'm pleased to share that the trial has enrolled more than 80% of participants. We look forward to full enrollment of that study soon. In our early clinical programs, EBV, HIV and VZV vaccine trials are ongoing, and our HSV program is in preclinical. Now let's look at our mRNA therapeutics portfolio on the next slide, and I'll highlight a few programs. We are excited to announce that our Phase 3 melanoma study with INT is now enrolling. And I'll share the Phase 3 design in a moment. Our rare disease programs addressing major unmet medical needs in propionic acidemia, methylmalonic acidemia and GSD 1a are ongoing and we will look forward to sharing updates when the data are mature. Indeed, during the quarter, we presented an update on some of our PA program data at ASGCT. The dose confirmation part of the propionic acidemia study is currently ongoing. And lastly, our collaborators at Vertex are continuing to enroll and dose cystic fibrosis patients in the single ascending dose portion of that Phase 1 study. Now on my last slide today, I want to take a moment to share the exciting Phase 3 trial design for individualized neoantigen therapy, or INT. This Phase 3 study is a randomized, double-blind, placebo controlled study of the combination of INT plus Keytruda against placebo plus Keytruda in patients with resected melanoma at high risk of recurrence. The study will enroll approximately 1,089 resected melanoma patients, stage 2b through 4 with a one-to-one randomization. Each patient will receive up to 9 doses of INT every three weeks and Keytruda every six weeks in the active arm, or 9 doses of placebo every three weeks and Keytruda every six weeks in the comparator arm. The primary endpoint is recurrence free survival and secondary endpoints include distant metastasis free survival and overall survival. I'm incredibly impressed with the collaboration between the Moderna and Merck scientific and clinical teams to rapidly stand up and begin enrolling this Phase 3 study. With that, I'll turn it over to Arpa.
Arpa Garay :
Thank you, Stephen. And good day to everyone. I'll start with a review of sales in the quarter and the first half of 2023. In the second quarter of 2023, we reported approximately $300 million of COVID vaccine sales, which was in line with our expectations given the seasonal nature of endemic respiratory vaccine businesses. Sales of $2.1 billion for the first half of 2023 met our expectations. As a reminder, the sales in the first half were predominantly from mRNA-1273.222, which is our COVID vaccine targeting the BA.4-5 variant. As you heard earlier from Stephen, regulators and health authorities around the world have selected the XBB.1.5 variant for the fall of 2023 season. We have submitted applications to regulators globally for our updated vaccines, and we are now awaiting approvals and authorizations. Subject to regulatory approvals, we expect additional sales in the second half of 2023, primarily from mRNA-1273.815. Turning now to the 2023 COVID sales outlook. We are updating our COVID sales expectation for 2023 to be in the range of $6 billion to $8 billion, with the key variable being vaccination rates from the United States. Our 2023 sales mix consists of $2.1 billion in sales already recorded in the first half of this year, another $2 billion in sales from previously signed advanced purchase agreements for the second-half delivery from several countries, including those listed in the middle column on this slide. Recent discussions with customers around the world have resulted in confirmed contracts of $2 billion for delivery in the second half of this year, updated from the originally expected $3 billion. Approximately $1 billion of the original $3 billion are now expected to be deferred to 2024. Also now included in the total expected 2023 COVID sales range of $6 billion to $8 billion is an additional $2 billion to $4 billion in sales expected from signed and anticipated commercial contracts in the US as well as other markets such as Japan and the EU. The US commercial contracts are a sizable portion of additional new sales expected in the second half of this year, so let me provide some color on that market in particular. As you are aware, the COVID vaccine market in the US has shifted to a commercial market with the transition from the pandemic to the endemic phase. In the US, the commercial vaccines market is made up of many different customer segments, including retail pharmacies, wholesalers, group purchasing organizations, integrated delivery networks, health systems, government entities, employers, and other providers. Today, I am happy to report that we have signed contracts in each of these customer segments and continue to work on additional contracts. We are ready with ample supply to be shipped upon regulatory approval. Our signed contracts give us visibility into the expected US launch in the coming weeks and confidence in the additional sales we expect in the second half of 2023 from new orders. Let me now turn to slide 21 to help frame the fall 2023 US COVID market. As I mentioned earlier, a key determinant for the market size in the US will be vaccine uptake or shots in arms during the upcoming September to December time frame. As I've mentioned before, our expected 2023 sales range of $6 billion to $8 billion will be primarily driven by vaccination rates in the US market. Earlier in the year, we provided parameters that informed our US fall volume forecast for 2023, which included the roughly 50 million doses administered in the US in the fall of 2022; 82 million Americans in the high-risk category, so those who are over 50 with a comorbidity or those over the age of 65; and an average of 150 million doses of flu vaccines given in the US every year for the last nine years, which we believe is a reasonable proxy for a seasonal respiratory vaccine, especially given the higher burden of disease with COVID. These parameters supported a volume forecast of 100 million doses for the US market for fall 2023. But the fact of the matter is, in this first transition year to a commercial endemic market, it is difficult to accurately predict market volumes and predict how many Americans will come in this fall for their shots. As such, we look to Southern Hemisphere countries where the COVID season occurs during their fall/winter months to inform potential vaccination uptake in the US as well. Specifically, in Australia, where they are just completing their 2023 COVID season, the vaccination rate was 19% in populations where the booster vaccine was recommended. The Australian data, combined with prior-year comparisons, indicate US market volumes for fall 2023 to be in the range of 50 million to 100 million doses, and this supports our US sales expectations for the second half of the year. Moving to slide 22. I'm excited to share the fall vaccination campaigns for the updated COVID vaccine launch. Moderna's fall vaccine campaigns are two-pronged and are focused on
Jamey Mock:
Thanks, Arpa. And hello, everyone. This morning, I will cover our second-quarter financial performance and provide a framework for our full-year financial outlook. Starting on slide 25. Total product sales were $293 million, down 94% year-over-year, mainly driven by lower sales volume. We continue to expect 2023 to be a transitional year as we move from a pandemic to an endemic commercial market with significant seasonality. In line with our expectations, sales in the second quarter were relatively low, while we came in at the high end of our communicated range. Cost of sales for the second quarter of 2023 was $731 million. In addition to unit-driven manufacturing costs, this includes the following charges – $464 million for inventory write-downs related to excess and obsolete COVID-19 product, unutilized manufacturing capacity at $135 million, and losses on firm purchase commitments of $75 million. These charges, other than royalties, were primarily driven by a shift in product demand to our latest monovalent XBB.1.5 COVID vaccine candidate and an overall lower market size compared to our expectations at the beginning of the year. In order to have ample supply at the beginning of the 2023 fall season, we prepared for various outcomes of the strain selection, resulting in additional cost to the P&L. The fact that a monovalent was chosen also meant that we were not able to use previously manufactured semi-finished goods. R&D expenses were $1.1 billion, which increased by 62% versus prior year. The increase in R&D continues to be driven by clinical trial-related expenses, particularly with our Phase 3 studies for RSV, seasonal flu, and CMV. The increase in R&D is also attributable to increases in personnel costs due to increased headcount to support our research and late-stage development efforts. SG&A expenses were $332 million, reflecting an increase of 57% year-over-year. The growth in spending was primarily driven by continued investments in personnel and outside services in support of our digital initiatives, marketed products, related commercialization activities, as well as our company expansion. Income tax was a benefit of $369 million for the second quarter, mainly due to a loss from operations. Net loss for the period was $1.4 billion compared to net income of $2.2 billion last year. And diluted loss per share was $3.62 compared to diluted earnings per share of $5.24 in 2022. We ended Q2 with cash and investments of $14.6 billion compared to $16.4 billion at the end of the first quarter. The decrease was driven by our net loss in the period and approximately $600 million of share buybacks. Cash deposits for future product supply declined during the quarter by approximately $100 million to $1.7 billion by the end of the second quarter, which was in line with our expectations. Now turning to slide 27. I want to give an update on the progress we have made on our capital allocation priorities. Our top investment priority has been and will continue to be reinvesting in the base business. R&D spending in the first half of 2023 increased 80% year-over-year to $2.3 billion, and we remain on track to invest $4.5 billion in R&D for the full year. We are also investing in our digital capabilities, the commercial build-out of the organization, as well as expanding our manufacturing footprint. We've accelerated our capital expenditures in 2023 as we expand both our international and US manufacturing footprint. Our second investment priority is to seek attractive external investments and collaboration opportunities that will enable and complement our platform. We remain disciplined in our approach and are in multiple active discussions. After evaluating internal and external investment opportunities, we then assess additional uses of cash. In the first half of 2023, we repurchased 8 million shares for approximately $1.2 billion, and we had $1.7 billion of share repurchase authorization remaining as of June 30, 2023. Now let's turn to our updated 2023 financial framework on slide 28. We would like to share our thinking beyond the advanced purchase agreements. As Arpa mentioned earlier, we now expect product sales for 2023 in the range of $6 billion to $8 billion, comprised of approximately $4 billion from existing APAs and approximately $2 billion to $4 billion from additional sales to the US, Japan, EU, and other countries. As a result of recent discussions with customers around the world, we now expect approximately $1 billion of the original total $5 billion in APAs to be deferred to 2024. Second half sales timing will be dependent on timing of regulatory approvals across the world and the number of days available in the third quarter to ship. We currently expect a sales split of 30% in Q3 and 70% in Q4. We now expect cost of sales for the full year in the range of $3.5 billion to $4 billion. At this point of the year, our production costs are largely fixed, and only a smaller portion is driven by the sales outcome. Therefore, we thought it would be more helpful to provide you an absolute dollar range for your modeling purposes. For R&D and SG&A, we continue to expect full-year expenses to be approximately $6 billion, with approximately $4.5 billion in research and development. We now anticipate a full-year tax benefit in the range of $0.7 billion to $1 billion, driven by an assumed operating loss, R&D credits, international provisions, and non-recurring items. And finally, we continue to expect capital expenditures of approximately $1 billion. That concludes my prepared remarks, and I'll turn the call back over to Stéphane.
Stéphane Bancel:
Thank you, Jamie, Arpa, and Stephen. Moderna has a promising commercial outlook, starting with COVID. While there is uncertainty in vaccination rates as we transition from a pandemic to endemic market, our APAs and US commercial contracts underpin our expected 2023 COVID revenue in the range of $6 billion to $8 billion. We believe this first endemic year will provide visibility to recurring revenue stream. I believe we will be selling COVID vaccines for a very long time, and we are working to combine COVID and flu into a single vaccine. Moving to RSV. RSV vaccine has a strong product profile and a differentiated prefilled syringe presentation that should work to our advantage in the anticipated 2024 launch. In oncology, we are scaling our INT manufacturing capacity to be ready for commercialization. The Phase 2 data are very strong, and we are now in Phase 3 with melanoma indication. With our partner, Merck, we are working to prioritize indications beyond those already announced, which are melanoma and non-small cell lung cancer. Stepping back and looking at the broad portfolio, I'm very excited that we are playing with the high-play [ph] scenario of Moderna with positive clinical data in infectious disease vaccine, in oncology, and in rare genetic disease of liver. For the next three years, from 2024, 2025 to 2026, we anticipate multiple product launches across our vaccine and therapeutics portfolio that will position the company for strong sales growth. This is an incredibly exciting time at Moderna as we enter a new era with a diversified revenue stream and a robust pipeline. The platform is working, and the result will be an unprecedented number of mRNA launches in a very short time. We look forward to updating you further at our upcoming R&D Day on September 30. That event will be live in New York City and, of course, also available online. On December 7, we'll be hosting our second annual ESG Day. This event will be online. The mission of our company is to deliver the greatest possible impact to people through mRNA medicines. This mission is especially relevant now as we approach the launch of multiple new medicines that should extend human lives and alleviate patient suffering. All of our stakeholders are poised to benefit as Moderna continues to deliver on its potential. It is a privilege for all of us to be part of this company. We'll now take questions. Operator?
Operator:
[Operator Instructions]. Our first question comes from Salveen Richter with Goldman Sachs.
Salveen Richter:
With regard to the expected additional sales for second-half deliveries, can you speak to the spectrum of factors in addition to the US vaccination rates that might impact reaching the higher end of that range? And can you discuss confidence that additional contracts wouldn't be pushed to 2024 as the fall season plays out?
Arpa Garay:
In terms of the additional sales of $2 billion to $4 billion, as you mentioned, the key factor of landing within that $6 billion to $8 billion is really around how many Americans this fall come in to get their shot. So vaccination rate is the biggest swing factor within that range. We are confident in our market share and the progress that we've been making with commercial contracts thus far in the US. Additionally, outside of the US, to your question on additional contracts potentially being deferred, we have met with all of the different countries where we have advanced purchase agreements and have already confirmed deliveries for the second half of this year, other than the $1 billion, which was pushed into 2024. So we're confident in staying at that $2 billion range.
Operator:
Our next question comes from Tyler Van Buren with TD Cowen.
Tyler Van Buren:
With the COVID-19 vaccine sales of $6 billion to $8 billion expected for the year, how should we think about what that implies for real demand as we think about the eventual COVID-19 vaccine franchise tail? Is it $2 billion to $4 billion since the $4 billion was already locked in from APAs at the start of the year from prior years? If not, what proportion of the $4 billion of APAs would you consider true annual booster demand? Perhaps, is it the $2 billion expected in the second half? Thoughts there would be helpful.
Arpa Garay:
In terms of how we're thinking about 2024, I would look at the first half of 2023, where we have recently reported $2.1 billion of sales, a portion of that volume is volume that we do not anticipate as a recurring sale in the outer years, given most countries are going towards fall vaccination campaigns with annual shots. For the remainder of the $4 billion to $6 billion, we do believe that vaccine demand will remain and, over time, eventually increase as we think about the flu volumes that we believe the COVID vaccination rates will start trending to. So as I mentioned earlier, for example, in the United States, there are about 150 million flu vaccines given every fall. What we've modeled for 2023 is a range of 50 million to 100 million. And we think over time, given the high disease burden and also as we think about future combinations, we will start trending closer to about 150 million.
Operator:
Our next question comes from Gena Wang with Barclays.
Gena Wang:
Regarding the COVID US commercial contract this time, were there any definitive initial stocking order? And the second regarding the RSV vaccine, given that Pfizer and the GSK label language and a slow initial launch, any learning you could have for your approval and the launch prep?
Arpa Garay:
I'll take the first question around US contracts first. In our contracting progress, we do have minimum volume commitments across many of our commercial contracts, but most of our contracts, we're looking at minimum commitments. And depending on vaccination uptake that we will be monitoring closely with our customers, that volume could go up over time. In terms of the recent RSV ACIP meeting, we think the ACIP recommendation for a shared clinical decision-making will mean a more gradual uptake for the first two vaccines that have been approved. Though as there is more data from both our clinical program as well as GSK and Pfizer's clinical programs, we do believe there is opportunity to potentially update that recommendation, which would, over time, lead to broader uptake of the RSV vaccine.
Operator:
Our next question comes from Michael Yee of Jefferies.
Dina Elmonshed:
This is Dina on for Mike. Just a quick question on the flu update. So I saw that you guys are doing a Q3 update for the second immunogenicity study and then going on to have data in Q3 with that. But how about the efficacy study from that second infection or that – yeah, that previous infection study? What are your timelines for that? Have that shifted? Are you going to share that? And also on PCV, are you going to be sharing any additional updates on the regulatory path moving forward? Or are you sharing any other updates on lung, et cetera?
Stephen Hoge:
First, on the 1010 flu program, as you referenced, we have two ongoing – we have actually three ongoing Phase 3 studies, but two that are continuing to accrue data. We'll be providing updates on the efficacy study, the P302 study you referenced. And as you know, on our Vaccines Day, we talked about an end-of-season update. And we'll also be sharing where we are in the pivotal P303 immunogenicity study. And as I said, we'll be doing that this quarter, and we're excited to continue to update on the progress in influenza. As it relates to INT, now INT, not PCV, that program, we had a great opportunity to provide an update to you all and everyone at ASCO just a month ago. And we're going to be, of course, at our R&D Day reprising that. And also talking to some of the other progress happening in the INT program. We are obviously working hard to get up and running in our non-small cell lung cancer, but we're really – pivotal studies, but we're really excited by the start of the first Phase 3 with melanoma study now, and we'll be providing further updates in a month in New York.
Operator:
Our next question comes from Ellie Merle with UBS. Your line is open.
Unidentified Participant:
This is Sarah [ph] on for Ellie. Just a follow-up on Tyler's earlier question. Thinking about COVID volumes going forward, can you give more color on how you're thinking about first half versus second half? Particularly, any color on your expectation for first half of next year? And then on CMV, any expectation on when you'll complete enrollment? And how we should think about when we could get data and maybe anything you're seeing on a blinded basis on the event rate?
Arpa Garay:
I'm happy to take the first question and then will hand it over to Stephen on CMV. In terms of how we're thinking about the first half versus second half going forward, in the first half of the year, sales that we expect will come from the Southern Hemisphere, where it is their fall and winter season. Additionally, in some countries, in the higher-risk populations, there continue to be spring campaigns and boosters, particularly for immunocompromised, elderly and highest-risk patients. So we do anticipate some sales coming in for the high-risk spring boost for the Southern Hemisphere as well as any carryover from late winter of this year into January. The majority of our sales will continue to be expected in the second half of the year, though, as the majority of our sales will be anticipated from the Northern Hemisphere and the fall vaccine campaign.
Stephen Hoge:
And on the question of CMV enrollment, yes, we're really excited by the recent acceleration toward completion of enrollment there. We're past 80%. We do hope to complete enrollment shortly. We've made good progress in the first half of this year, and so we haven't specifically set a target on that, but we would hope to enroll it for sure this year. Then we will be accruing cases, and I think that's where it gets very interesting in tracking the CMV Phase 3 program. We've already started to accrue cases that we talked about in the Vaccines Day. And as we get to full enrollment, we will have more participants who can contribute cases to our first interim analysis of efficacy, which will be an event-driven analysis, and so not something we can predict the timing of.
Operator:
Our next question comes from Luca Issi with RBC Capital.
Luca Issi:
Maybe one, Jamey, if I can circle back on margins. I think your prior guidance implied 60% to 65% gross margin versus your new guidance today implies 47% gross margin at the midpoint. Can you just maybe expand a little bit more on what's driving that change today? Maybe, Stephen, on flu, I think Sanofi argued that the lower immunogenicity for the B strain could actually be a class effect. So it'd be difficult for any of the mRNA players to actually have strong immunogenicity for B strain. Wondering what are your thoughts there. Thanks so much.
Jamey Mock:
I'll take the first question. So on margins, yes, you're right. We were planning for 60% to 65%. I'd say it's largely volume-driven, $1 billion of APAs pushing out 2024 as an example. We still have the cost for those fixed into our $3.5 billion to $4 billion in addition to other markets as well. So I think our volume expectation prior to this quarter was a little higher, some due to push-out, some just due to overall volume that we anticipate coming through here.
Stephen Hoge:
And I'm aware of the argument that Sanofi made about a class effect. I'm not sure I see it the same way. I think we'll look to the data to answer that question rather than conjecture. We'll look forward to sharing where we are in the P303 Phase 3 study in the next quarter, and I'll leave it at that.
Operator:
Our next question comes from Edward Tenthoff with Piper Sandler. Your line is open.
Edward Tenthoff:
My question is kind of a little bit of a longer-term strategic one. As COVID continues to evolve and then as you ultimately seek approval of 1010 and even RSV, what is the plan for combining these from a regulatory standpoint and then also a commercial standpoint? So, how do we get from where we are today with one approved, maybe two, three approved vaccines next year to one combo-approved or multiple combo-approved vaccines?
Stephen Hoge:
As you know, as I said, we have six combo vaccines. We've got lots of clinical data out there, and we continue to look at new combinations. And if we're in a situation that we expect to be in where we have a flu, RSV, and COVID approved as well as the second-generation COVID moving forward, you can rest assured we'll be looking at multiple different combinations of those, trying to bring forward options that provide the greatest public health flexibility. Those studies, once we have the products approved, the monovalent vaccines, those studies are really just immune bridging studies, demonstrating that we can do the combination and achieve non-inferior immunogenicity and safety in those studies. They can be quite quick. And they also don't need to be run in the season, as you know. And so, our goal is going to be, as we've said throughout this year, is to complete the work to move towards approval, filing, and eventually, hopefully, approval of the three monovalent vaccines and quickly progressing into the pivotal Phase 3s for at least one and multiple combos. Our goal, again, is to be launching those in 2025 and beyond for the obvious reasons, they will improve compliance, deliver more value, and actually decrease the administration cost of healthcare. So the path is actually pretty clear from here, particularly given the strength of the RSV data and where we are in COVID, and we hope to be providing an update very shortly on flu that also provides a clear path for 2024 in the monovalent launches we've guided. And then I think we'll clarify very quickly that we're starting the Phase 3s to allow the combo launches very shortly thereafter.
Edward Tenthoff:
Just to clarify one point, if I may. When you said the pivotal Phase 3s, those would be immuno-bridging studies.
Stephen Hoge:
Correct. They're immunogenicity and safety studies. So very quick studies, just demonstrating – we do not believe nor is it the norm from a regulatory perspective that you have to do subsequent efficacy studies once you've established the efficacy of the monovalent vaccine.
Operator:
Our next question comes from Manos Papadakis with Deutsche Bank.
Emmanuel Papadakis:
Basically, I just wanted to ask in terms of the COGS guidance, does that apply regardless of the revenue range? And if not, what is the sensitivity range that you could provide?
Jamey Mock:
Yes, it does apply regardless, as I mentioned in my prepared remarks. Most of this cost is fixed at this point. We've already ordered the raw materials. We've already started manufacturing most of our – we've got a lot of supply ready to go as soon as we are able to – as soon as we've got regulatory approval. So that $3.5 billion to $4 billion is not really dependent too much on the sales outcome that will happen either way. And maybe just to talk to longer term, so I think what we're suffering with this year is just an unpredictable market. So back to the prior question of the 35% to 40% or 60% to 65% gross margin, as it becomes more predictable, we will be able to order the right amount of material, have the right amount of volume commitments, that might not happen in 2024 as we work through some of our commitments. But we feel very confident in the overall 75% to 80% gross margin range in the long term.
Operator:
Our next question comes from Geoff Meacham with Bank of America.
Alexandria Hammond:
This is Alex on for Geoff. Can you talk about all the other potential indications you may pursue for the INT vaccine? And how will you decide which indications to pursue first? Will this be based on [indiscernible] or an unmet need? Secondly, what commercial hurdles do you expect for flu and RSV, given the entrenched competition and new entrants respectively?
Stephen Hoge:
First on INT, so obviously, we're moving forward in melanoma, as you know. And we're moving forward in non-small cell lung cancer. I think the opportunity there is – the unmet need there is obviously very significant. And as we know, that's one of the largest opportunity for immune therapies generally, and so it's an obvious place for us to go next. As we've said before, we have not yet publicly guided with our partner, Merck, on what the other indications will be. But we have said that we are going to follow the path of all the places where PD-1 Keytruda have been successful, but where we think there is still headroom, still opportunity to significantly improve upon what's been achieved with the PD-1 antibodies. And so, you can follow through the adjuvant settings as well as perhaps some of the stage 4 settings, where there are those benefits and expect us to be moving there in short order. Ourselves and Merck are in the process of finalizing some of those plans. And at the appropriate time, obviously, we'll guide you on the start of those studies. But rest assured, as we've said and as they said before, there is a quite large program of studies that will be ramping up here in very short order, and melanoma and non-small cell lung cancer are really just the first two.
Arpa Garay:
And I can take the question on RSV and flu, we are very excited to be launching RSV next year. So while we will be bringing the third product to market, we're very confident in what we believe is a best-in-class profile with our consistently high effectiveness across high-risk groups, thinking about our well-established safety profile in the only mRNA platform for RSV, and lastly, as I mentioned earlier, being the only product with a ready-to-use prefilled syringe. So we're feeling really good about the RSV launch preparations, and we're ready to execute in 2024. From a flu perspective, we do think one of the advantages of 1010, which would be our first-generation flu vaccine, is our speed to market could give us an opportunity for better strain matching in the future. And as we get enhanced profiles for flu, as Stephen mentioned earlier, from a commercial perspective, we see a tremendous amount of interest in our combination vaccines, both from patients as well as from healthcare systems. So having RSV and flu gives us an incredible opportunity for a combination for the future.
Operator:
Our next question comes from Evan Wang with Guggenheim Securities.
Evan Wang:
I just want to follow up on COVID. And as we're thinking ex-US, any details you can share in terms of size orders and market positioning in some of your markets like Japan? And what could change in 2024? And in flu, just great to see the P303 study fully enrolled, Just want some updated thoughts in terms of confidence on hitting on some of the B strain, given some of the competitor updates.
Arpa Garay:
I'll take the first question. From an ex-US perspective, we have signed a minimum commitment with Japan that has been announced. The minimum commitment that we signed with Japan is just to get product into the country as soon as August, and so we're ready to execute this fall. If their vaccine uptake expands, we do anticipate additional orders coming in from Japan. For the rest of the world, we have advanced purchase agreements that I have outlined already on – in that first column, as well as ongoing conversations with several other international markets, including those in the European Union. In terms of 2024 changes, what we expect outside the US is we expect more and more markets to actually be transitioning from central government procured to more of an endemic commercial market, such as the transition that the US is going through today. So we do anticipate changes. Most of those changes will be around shifting where the procurement is happening as well as changes in pricing and reimbursement going forward.
Stephen Hoge:
The flu question. So, yes, we're fully enrolled in P303, ahead of schedule on that, and we will provide data shortly on the P303 study this quarter, as we said. As far as the B strains goes, we already updated earlier this year that actually in the P302 study, we achieved non-inferiority in the B strain, you'll remember. In the P301, we had missed that. And so, we were learning as we went, and we actually made a series of changes into the P303 study that we were confident and remain very confident will provide a benefit in the B strains that will improve immunogenicity there. So I remain quite optimistic that our understanding of science is really strong, we think, best-in-class and that where we will be and when we look at that P303 data [indiscernible] is in a very strong position as it relates to B strains and continue to be in a very strong position as it relates to the A strains.
Operator:
Our last question comes from Simon Baker with Redburn.
Simon Baker:
Just two questions from me. One was, how has your view of the RSV opportunity changed in light of competitor two-year data and the ASIC recommendation? In what regard do you think that you can achieve best-in-class profile? And the second question was just a little bit more about the CMV Phase 3 enrollment and if you think you'll be able to get full enrollment by this quarter.
Arpa Garay:
I'll start with the RSV question. In light of the ACIP shared decision-making recommendation as well as some of the data that was shared this year around revaccination, we do anticipate a slower vaccine uptake at the start. In terms of our potential in that market, we do continue to be very confident that we have a best-in-class profile. And as we get additional data from our own vaccine program, we are hopeful that the ACIP recommendation will be brought in and could be – will enable a faster uptick in outer years.
Stephen Hoge:
The only thing I'd add to that in terms of the development side is we are obviously looking at multiple different versions of RSV combination vaccines, and one of the advantages there will obviously be increased convenience and compliance. And that's where if we can create healthcare system value, I think it will be – it will continue to evolve, the recommendation that people have around that. As it relates to CMV, so we haven't set out a guidance on when we would expect the enrollment to complete. But we're actually confident, given the trajectory that we're already past 80%, that we will complete enrollment this year. I'm not saying this quarter, but this year. And as I said, what really matters is probably not whether we're at 80% or 99% or 100% enrollment. It's the accrual of cases and events will drive the first interim analysis from here forward. Now, obviously, more patients – more participants enrolled will increase the number of – and pace of those events over time. And so, we are focused very much on that operationally, and we do hope to have that completed this year.
Operator:
And I'd like to turn the call back to Stéphane for any closing remarks.
Stéphane Bancel:
Well, thank you very much for your questions. We look forward to seeing many of you in the next days and weeks, but especially seeing you at the R&D Day on September 13 to see many of you in person. Have a great day. Thank you.
Operator:
Ladies and gentlemen, this does conclude today's presentation. You may now disconnect and have a wonderful day.
Operator:
Good day and thank you for standing by. Welcome to Moderna's First Quarter 2023 Conference Call. At this time, all participants are in a listen-only mode. After the speakers' presentation, there will be a question-and-answer session. [Operator Instructions] Please be advised today's conference is being recorded. I would now like to hand the conference over to your speaker today, Lavina Talukdar. Please go ahead.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's first quarter 2023 financial results and business update. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors Section of our website. On today's call are Stephane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Arpa Garay, our Chief Commercial Officer; and Jamey Mock, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I will turn the call over to Stephane.
Stephane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Today, I will start with a business review Stephen will then review our clinical programs before Arpa gives an update on our commercial progress and plans. Jamie will present our financial results, and I will come back to close. So in the first quarter, we recorded revenues of $1.9 billion, GAAP net income of $79 million and GAAP diluted earnings per share of $0.19, cash and cash investments of $16.4 billion at the end of the quarter. We continued in Q1, executing on our capital allocation strategy, prioritizing investments in our business. In the third quarter, we invested $1.1 billion in R&D, continuing investment in last stage clinical programs and progressing our entire pipeline. SG&A costs were approximately $200 million in the quarter, which continued investment in digital and also AI infrastructure and approximately $100 million in capital investments. In 2023, our team has been very active. This includes the acquisition of OriCiro in Japan, collaboration with CytomX, Life Edit and Generation Bio. All of these investment opportunities further expand the reach of Moderna's mRNA technology. We are growing Moderna mRNA operating system. Probably less than $6 million were returned to shareholders with share purchase of 3.6 million shares in the quarter. Let me turn to Commercial in the first quarter. We continue to expect $5 billion in COVID vaccine deliveries in 2023 from already signed advanced purchase agreements. With $1.8 billion of COVID we are well on our way to achieve $5 billion in APS. The commercial team is actively negotiating to sign new contracts with customers in major markets to finalize additional orders that will add to the $5 billion. The U.S. team is negotiating with pharmacy chains, hospital networks and other customers. As a reminder, the $5 billion of signed APAs does not include any fall 2023 U.S. contracts. So these new U.S. commercial contracts are important to the business as the additional. The team is also making progress in Japan, in Europe and other countries in Asia, Middle East and also Latin America. We recently signed a new contract in Australia for 2023. Our commercial organization is also preparing for the launch of RSV in 2024, our next respiratory commercial products. They're educating the medical community on the health burden of RSV. Our medical team is participating in major infectious disease medical congresses and sharing the new data that shows a strong profile of RSV vaccine. To prepare for RSV launch, our manufacturing team has already begun producing drug substance, the mRNA molecule for RSV vaccine. The vaccine will be supplied as a prefilled syringe. We believe that will help drive adoption by pharmacists and doctors versus some of our RSV products. In oncology, for mRNA-4157 now called individualized neoantigen therapy, or INT, we are scaling up manufacturing to support clinical development and commercial markets. Our commercial team is also working to prioritize tumor types to select for adjuvant Phase 3 studies in addition to melanoma and lung. Turning to the pipeline. At Vaccine Day, we discuss detailed data from our RSV vaccine that were presented at research medical meetings. We believe the profile of RSV vaccine is potentially best-in-class. We shared top line data on the flu programs announcing the initiation of Phase 2 or 3, the Phase 3 study that we intend to use for accelerated approval as well as strategy for our next-generation flu programs. We also announced the start of Phase 3 for next-gen COVID vaccine, mRNA-1283, which should be refrigerator-stable. Today, we're also announcing a new program for pandemic readiness for Influenza H5. In latent vaccine, our CMV Phase 3 trial continued progress and is greater than 50% enrolled. I'm also very pleased to announce progress in our early-stage program, EBV and HIV. EBV, as you know, has two programs. The EBV Phase 1 trials to prevent mononucleosis with mRNA-1189 are fully enrolled in adults and also in adolescents. Our EBV trial for the treatment of long-term complication for EBV infection has now started enrolling. And for HIV, we shall interim analysis from our Phase 1 study at vaccine. We also introduced new programs. We are working on vaccines for norovirus in Lyme disease. Lyme is the first bacterial pathogen we are targeting mRNA platform with vaccine. In therapeutics, we participated in AACR meeting where we presented detailed Phase 2 data from our R&D program partnered with Merck. Later on the call, Stephen will take you through the data presented there. Already this pipeline is also making progress. We're excited to announce a new milestone for our PA program. The Phase 1/2 trial in Propionic academia or PA is now in dose expansion phase. And additional data from this study will be presented at the American Society for Gene and Cell Therapy later this month in May. The Company continues to expand at a rapid pace. We have 47 programs underway which now reflect the inclusion of new program also in the quarter and removes programs that are not continuing to develop. The full of disease pipeline can be seen in the appendix of this presentation or obviously on our website. We now have more than 4,000 team members and 17 commercial subsidiaries across Americas, Europe and Asia Pacific. In addition to our commercial subsidiaries, we also announced we're opening a Seattle office as a technology hub, where we're hiring the data scientists and engineers who will help digitize our business and expand our AI capabilities. I'm also happy to announce Modena was officially recognized in March has a great place to work by the Great Place to Work Institute. Our $16.4 billion of cash at the other quarter is enabling us to scale across our research, development, manufacturing, commercial and G&A. With that introduction, let me now turn to Stephen.
Stephen Hoge:
Thank you, Stephane. Good morning or good afternoon, everyone. Today, I'll review the clinical progress in R&D at Moderna in the first quarter and highlight select data from the past few months has been presented. The core of our respiratory portfolio is made up of vaccines against COVID-19, flu and RSV, which are either commercial or in Phase 3. We're advancing second-generation vaccines, this includes our second-generation refrigerator stable COVID-19 vaccine candidate, mRNA-1283, which is rapidly enrolling in its Phase 3 study and two next-generation influenza vaccines that are in Phase 2. We believe combination vaccines will be the future of our respiratory franchise. And we're pleased that we now have five different combination vaccine candidates in early clinical trials, including two specifically designed for pediatric populations. With 11 programs in clinical trials, including four in Phase 3 and covering five different respiratory viruses, we believe this represents the broadest and most advanced portfolio of respiratory virus vaccine candidates. Now turning specifically to COVID. The FDA recently provided updates on several fronts. Our Omicron-targeting bivalent COVID-19 vaccine targeting the original MBA 45 strains is now our only authorized formulation in the United States with a simplified and streamlined regimen for both children and adults. Individuals, 65 years of age and those with certain kinds of immunocompromise are now also eligible to receive additional doses as needed or recommended by their physicians. As we look to the fall, the strain selection for an updated composition for fall 2023 boosters is now expected to come at the June 15 VRBPAC meeting. Moving to RSV, we're pleased by the profile of our vaccine in older adults with high and consistent efficacy against RSV lower respiratory tract disease across populations in our large Phase 3 study. At two recent medical meetings, we've shared data showing our vaccine's efficacy was consistently high across all age groups, including in the oldest adult and in participants with preexisting comorbidities that put them at higher risk. mRNA-1345 has also shown a favorable tolerability profile with AEs mostly grade 1 or grade 2, mild to moderate. As we shared during Vaccines Day, today, we have not seen any cases of Guillain-Barré syndrome or other severe demyelinating events in the trial. Moving to flu. Our Phase 3 efficacy study in the Northern Hemisphere, P302 is ongoing. And last month, we announced that the study did not accrue sufficient cases to declare early success at the interim analysis and that the DSMB recommended that we continue the study. That trial is still accruing cases through the end of this flu season with an update expected this summer. The preliminary immunogenicity data from that P302 study showed that HAI neutralizing titers were consistent with superiority for both A strains and non-inferior immunogenicity for the B strains when compared to the licensed flu vaccine. Now I'm pleased to announce that we've initiated our Phase 3 P303 flu study with an updated formulation of mRNA-1010. P303 is testing an update that is designed to increase the HAI neutralizing titers against the B antigens. This is an immunogenicity study and is expected to enroll approximately 2,400 adults this spring. We believe this study will support our initial flu filing and the potential for a 2024 launch if approved by regulators. Now turning to our latent vaccines. Our CMV vaccine Phase 3 study in women and childbearing age is ongoing and has enrolled more than 50% of participants. We also have an ongoing Phase 1/2 adolescent dose-ranging study with this vaccine that will expand potential eligible populations. As Stefan mentioned earlier, we continue to make significant progress against all of our latent vaccines in earlier-stage clinical trials, including candidates against EBV, HIV and VZV. As Stephane mentioned, our EBV vaccine includes two vaccine candidates, our EBV program includes two vaccine candidates, mRNA-1189 and mRNA-1195 and are in clinical trials for prevention of infectious mononucleosis and for the prevention of longer-term sequela of EBV infection respectively. Not Vaccines Day, we are pleased to share interim results of our HIV vaccine, mRNA-1644, which continues to advance. Our pipeline in therapeutics targets unmet needs across immuno-oncology, rare disease, cardiovascular disease and autoimmune diseases. All the trials in these therapeutic areas are ongoing. And today, I'll highlight a few of the recent updates. On Slide 16, are the data shared at AACR from our Phase 2 study in adjuvant melanoma with a combination of our individualized neoantigen therapy plus KEYTRUDA versus KEYTRUDA alone. The Kaplan-Meier curve for relapse-free survival solution here, overall, there was a 44% reduction in the rate of relapse or death with a combination of INT and KEYTRUDA compared to KEYTRUDA alone. We are encouraged by the continued separation of the two curves with a follow-up at 18 months. Note that there are very few participants beyond the 140 week cutoff at the right of this slide, less than 10 participants in total. Thus, as the data continues to mature, with additional follow-up time, we remain cautiously optimistic that this picture will get even better. Now on Slide 17, I want to briefly highlight some of the additional data that was shared at AACR. The subgroup analysis confirmed the strength of the treatment effect across two important markers that are known to predict responses to KEYTRUDA. Again, on this slide, we're looking at the relapse-free survival. On the left side, the results are stratified by high tumor mutational burden in red and low tumor mutational burden in blue. In each case, the solid lines are for the INT combination and the dash line is for KEYTRUDA monotherapy control. If you start by looking and comparing the control arms for KEYTRUDA, you will note that the TMB high participants, the red dash line, have a higher relapse-free survival than the TMB low participants in the blue dash line. This is expected because of what we know about KEYTRUDA, and shows that the controls are performing as we expect. Now moving to the INT combination arms, the solid lines and comparing them against the dotted lines of the same color, you can clearly see that the INT combination led to higher relapse-free survival for both TMB high and TMB low patients. This highlights the robustness of the response for INT. It's also quite exciting to note that the improved response rate seen in the blue TMB low population, which historically does not respond well -- as well to KEYTRUDA. On the right, you'll see the result as stratified by PD-L1 status. Now as with the TMB analysis, PD-L1 is known to predict response rates to immunotherapy. As you can see by the dashed lines for KEYTRUDA monotherapy control groups, the relapse-free survival curves look better for PD-L1 positive tumors, these are the red dash lines, and worse for patients unfortunate enough have PD-L1 negative tumors, the blue dashed lines. This isn't surprising as KEYTRUDA monotherapy acts by blocking the PD-1, PD-L1 access. As with TMB on the left, the solid lines on the RFS curves for the combination show the combination of INT, red denotes PD-L1 positive and blue denotes PD-L1 negative. In both cases, there is an increase in the rate of relapse-free survival with the combination treatment. It is encouraging to note again that the response rates are significantly improved for the PD-L1 negative patients. Indeed, the hazard ratio in this subgroup analysis is 0.162. Now pulling back, these data highlights that the observed improvement in relapse-free survival seen in the initial analysis of our Phase 2 study, looks broad-based across subgroups with an indication of a potentially important benefit even in patients who have higher risk of progression, whether due to PD-L1 status or tumor mutational burden compared to KEYTRUDA monotherapy alone. Now these positive data are just the beginning for this ongoing study. We'll be sharing additional data from the Phase 2 study at ASCO, including distant metastasis-free survival which was a secondary endpoint in the primary analysis just conducted and the same data cuts that I just shared. We'll also be sharing important additional biomarker data. In addition, this protocol calls for subsequent analysis at 51 events, which is when we will also be updating the Kaplan-Meier curves for recurrence-free survival with longer follow-up time across the full population, and we eagerly await those updates. INT has received breakthrough therapy in the United States and PRIME designation in Europe, which will facilitate frequent dialogue with regulators as we work to quickly advance this treatment for patients. As Stephane mentioned earlier, we plan to initiate our Phase 3 study in adjuvant melanoma in 2023 and rapidly expand to additional tumor types, including non-small cell lung cancer. We're working closely with our partner, Merck, to explore additional opportunities within KEYTRUDA approved indications and beyond that label. And finally, a quick update on our propionic acidemia program. Our Phase 1/2 study is ongoing and currently enrolling patients in the 0.9 milligrams per kilogram cohort. We have identified a dose for expansion and have moved into that expansion arm in the study. I'm pleased to announce just yesterday that a clinical update through the earlier -- time point earlier this year, including an update on safety and the rate of major metabolic compensations from the higher dose cohorts and including longer-term follow-up from the lower dose cohorts will now be presented at the American Society of Gene and Cell Therapy Meeting on May 18. The abstract for that presentation is now available and can be found in the link on this slide. We look forward to sharing a lot more about the progress of this medicine and our other rare disease programs in the months ahead. With that, I'll turn the call over to Arpa.
Arpa Garay:
Thank you, Stephen, and good day to everyone. I will first start with a review of sales in the quarter. On Slide 22, we summarized the composition of our sales in the first quarter. As you'll see on the chart, our sales to Europe were $0.6 billion and sales to the rest of the world were $1.3 billion. Approximately $1.8 billion of sales from previously announced APAs were delivered in the first quarter of 2023, representing the vast majority of the $2 billion expected in the first half of 2023. As a reminder, U.S. sales for COVID vaccines are expected to begin in the second half of 2023, with updated strain manage. Now as we turn to Slide 23, looking at the 2023 COVID deliveries, today, we are reiterating a minimum of approximately $5 billion in COVID vaccine deliveries from current advanced purchase agreements. And we continue to expect additional orders from key markets. Of the $5 billion in 2023 deliveries from previously announced APAs, $2 billion, as I mentioned earlier, are expected to be delivered in the first half of 2023. We have already delivered $1.8 billion of that $2 billion in the first quarter with substantial fulfillment to Japan and the European Union. We expect to deliver the remaining approximately $3 billion in previously announced APAs in the second half of this year. Additionally, we expect new sales in the U.S., Japan, European Union, Asia and Latin America. I'm happy to announce that the commercial team has signed a contract with the Australian Government for 2023. Our discussion with commercial buyers in the U.S. are positive, and I will elaborate on that shortly. In our discussions with commercial customers in the U.S., it is clear that our customers are aware that COVID is still a substantial health urban. Throughout 2022, COVID continued to be a leading cause of hospitalizations. Data available through September 2022, list COVID as the third leading cost of death in the U.S. only after heart disease and cancer, as shown in the first chart on the left. The chart on the right-hand side of the slide shows the most recent data available for U.S. hospitalizations for COVID, flu and RSV. As you'll see, with current season hospitalizations of over 600,000 per COVID, the hospitalization rate for COVID is almost triple that of flu as well as more than triple that of RSV. There continues to be a clear need to protect against severe COVID infections and our customers recognize that need. For full of 2023, we expect the U.S. market volume to be approximately 100 million doses. As we highlighted earlier in the year, the successful transition of our U.S. COVID business from a government driven to a commercial-driven model is critical. We have made great progress on this front, executing on our action plan to rapidly develop this commercial market. Importantly, the commercial team is in active discussions with customers throughout the U.S. We are contracting with national and regional pharmacies, integrated delivery networks, government health providers, including the VA, the CDC and the Department of Defense, for purchasing, organizations and other providers. In addition, our established national distribution infrastructure and our Moderna Direct e-commerce site is fully operational. Our global supply chain is in place to handle all U.S. customer needs including prefilled syringes and single-dose files at the time of launch. I'm excited to share some of the launch preparation activities for the updated COVID-19 vaccine for fall vaccination campaigns. We believe these activities will drive consumers to get vaccinated. First, we are taking an omnichannel approach via tailored digital messaging to healthcare providers. This multifaceted approach will allow us to drive broad awareness and continue to emphasize the ongoing need for COVID production. Later this year, we will be disseminating customized content to drive physician demand across immunizing physicians, institutional decision-makers, as well as influencers. We are also partnering with our customer base across the different commercial segments to assist with their fall immunization programs. We believe there's an opportunity to continue to provide education, both to staff as well as to patients. And we believe there is an opportunity to harmonize and simplify the vaccination process for patients who are going in to skip their flu vaccine. Consumer promotion will be focused primarily in the fall of this year, driving patients to seek Moderna's COVID vaccine through significant DTC efforts. As I mentioned earlier, we are looking to simplify the experience for our consumers who are already going in together seasonal influenza vaccine this fall. We are energized for this fall season. Moving on to Slide 27, I will update you on our second near-term commercial opportunity, which is RSV. We are excited for the expected 2024 launch of our RSV vaccine and the commercial team is undertaking a number of activities to ensure that we develop what we hope will be a large share of that market as quickly as possible. We are already raising awareness of the health and economic burden of RSV by generating and presenting detailed data from our Phase 3 study at major medical meetings. Our medical team has shared additional data showing that vaccine efficacy is consistently high across all tested age groups as well as in participants with preexisting comorbidities. The figures on the right-hand side of the slide, detail efficacy data in these important subgroups, as we share these data at medical congresses, we are encouraged by and key opinion leader feedback on our data and the application of our mRNA platform to prevent RSV. We're engaging with payers and ITAC to ensure access upon launch. And the commercial team is active in local markets preparing for a commercial launch in 2024. Specifically, we are building our digital capabilities so that we will be able to efficiently educate customers as soon as the vaccine is approved. And as we invest in prelaunch activities, we have already begun manufacturing components of the vaccine and prefilled syringes. We are excited about the profile of the products we will be launching into these large respiratory markets. As we discussed at Vaccines Day, the estimated total addressable markets for our three key respiratory vaccines are substantial. With COVID, RSV and flu offering potential addressable markets of $15 billion, $6 billion to $8 billion and $6 billion to $9 billion, respectively. We believe we can take a sizable share of this roughly $30 billion respiratory market. The commercial team is well underway in preparing for RSV and flu launches in 2024. We believe our opportunity in the respiratory market will continue to expand beyond 2024 with our next-gen vaccines and importantly, with future combination vaccines, positioning us to drive share over time. Now on to Slide 29, I want to share with you how the commercial team is helping identify eligible patient populations in the adjuvant and neoadjuvant settings for various tumor types. Along with our partner, Merck, we have already announced that we will start Phase 3 trials in Adjuvant melanoma and adjuvant non-small cell lung cancer. There is an annual population of over 130,000 new patients in the U.S. and Europe in these two indications. Patient populations for additional potential adjuvant and neoadjuvant tumor types are listed on the right-hand side. Our commercial teams are working closely with our clinical teams to identify the largest unmet need for addressable tumor types for individualized neoantigen therapy. We are excited to be launching into a space where we have one medicine for one patient in areas of great in cancer. Given the individualized approach, we are reimagining our commercial model, along with our partner, along with patient care journey, end-to-end. With that, I will turn it over to Jamie.
Jamey Mock:
Thanks, Arpa, and hello, everyone. This morning, I will cover our Q1 financial performance, review the framework for our 2023 financial outlook and provide a quick recap from our recent Vaccines Day presentation. Moving to our first quarter results, starting on Slide 31. Total product sales decreased 69% year-over-year to $1.8 billion, the decrease in 2023 is consistent with our expectations and mainly driven by lower sales volume compared to the prior year. Cost of sales for the first quarter of 2023 was $792 million, in addition to our unit-driven manufacturing costs. This includes royalties of $86 million and the following charges
Stephane Bancel:
Thank you, Jamie, Arpa and Stephen. Let me share some thoughts before we close into Q&A. Moderna's promising commercial outlook has several development projects come to fruition. In COVID, we are finalizing discussions with customers. And I believe that we'll see significant additional contracts in the U.S., in Japan and around the world for '23. COVID is not going away and governments are getting ready for vaccination campaign in the fall. I'm pleased that we have begun prelaunch market development and as risk manufacturing for RSV, which we expect to launch in '24. And in oncology, the team is making great progress. We expect to deliver key milestones on our development pipeline in the remaining eight months of 2023. We expect regulatory authorities to give us direction on COVID strain with the June VRBPAC meeting, and we expect to launch an updated COVID-19 vaccine for fall of '23. We plan to file for approval for RSV vaccine and of Phase 2/3 to study should be fully enrolled by this summer, and we expect data in Q4. For INT cancer therapy, we expect to make additional Phase 2 update, launch of our Phase 3 study in adjuvant melanoma and expand into additional cancer types. And we'll continue to make progress in our revenues portfolio. We present data for PA on May 18. This is a very exciting time for us at Moderna, and I would like to thank our teams for all their hard work and their commitment to our mission. Our platform is firing on all cylinders. In Infectious Disease vaccine, look at the data and the portfolio of respiratory, latent and now entering a bacterial vaccine. Very excited by where INT is and is going. And rare disease with PA followed closely by MMA and GSD1a. We'll give some key updates this year. On September 13, we have our annual R&D there. We will present new development pipeline data and on December 7, we'll be hosting our second annual ESG day. We believe that the incredible progress we have made across all of our modality, positions our company for long-term financial success. But the mission of our company was motivated for our entire team to come to work every day is to deliver the greatest possible impact to people for mRNA medicine. We believe we have a technology to eliminate or greatly reduced human suffering caused by respiratory viruses, latent viruses, bacteria, cancer, regulating disease and a growing list of diseases. We work to bring a number of our more promising technologies to market in the next several years and have paused to continue to fulfill on our mission. With this, we'll take questions. Operator?
Operator:
[Operator Instructions] Our first question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
I have two quick questions. The first one is regarding the U.S. commercial opportunity in the second half this year. You are in discussion with both, the commercial or government peers, is 110 to 130 still a good benchmark? How is the pricing play out between these two groups? When will you start to see clarity on actual contracts and orders? And then my second quick question is regarding your expectation for ASCO update for your PCV program?
Arpa Garay:
Great. So, I will start with the first two questions on commercial and then hand it over to Stephen to discuss ASCO. In terms of pricing across the U.S. market, we do anticipate our list price when we have our updated vaccine to be in the range of 110 to 130. As you're aware, in the commercial market, we will be providing differentiated discounts across different payer types, from government agencies through to commercial players as well. In terms of the timing of the orders, we are actively in negotiations with U.S. customers. We are very encouraged on two fronts. First and foremost, our customers do appreciate and understand the significant health burden that continued success with COVID and they do want to partner with us to make sure that as many of their patients can get fascinated to protect themselves from potential hospitalizations and severe diseases. The other thing we continue to be very encouraged by in our conversations with different customers, if they are appreciating and recognizing the full real-world evidence behind buyback and the effectiveness and profile of our vaccine. So, we anticipate over the next four to six weeks, we will begin to see some more clarity around contracting, which will continue through the end of Q2 and early into Q3.
Stephen Hoge:
Gena had a question on ASCO? Okay. Sorry. Gena what date was open at ASCO. So it's Stephen. So, as I said, there's two presentations, two sets of data that will be shared. The first will be focused on the distant metastasis-free survival, DMFS. DMFS, as you know, is another surrogate of overall survival and distant metastasis usually, unfortunately, is visceral, and that's obviously a greater concern. The protocol for the Phase 2 study included in the first analysis, primary analysis, looking both at RFS and distant metastasis-free survival, DMFS was a secondary endpoint, and we'll be presenting for the first time that data at ASCO. The second data that will be shared in poster form will be some data on biomarkers and additional data on the performance of INT across populations. That is data that will get more into the basic science for those who are interested in it, but we'll look into the mechanism of action of the product as well as further stratification of risk that we believe provides even more confidence that the signal we're seeing in terms of potential benefit for INT in the Phase 2 study is resilient and really bodes well for the future.
Operator:
Thank you one moment for our next question. Our next question comes from Salveen Richter with Goldman Sachs. Your line is open.
Salveen Richter:
With regard to the PA data that we're going to see at ASGCT, could you just frame that for us? I think in the past, you've talked about 25% being clinically meaningful as you look at relative risk reduction in major decomposition events here and what the translatability is from that to MMA and GSD1 and OTC your overall rare disease franchise? And then a second question for COGS, how are you thinking about beyond 2023? And in the context of your assumed market share of the respiratory franchise revenue as you look out to 2027 which you noted, how do you think about profitability in the context of this OpEx spend, including kind of your R&D and SG&A outlook?
Stephen Hoge:
Thank you for the question. So I'll take the rare disease portion of that on PA first. And so, as we shared last September, we did a data cutoff from the PA study last September after a couple of dose levels. And we were seeing slightly more than 50% reduction in the rate of metabolic decompensation. These are the severe events that really, we believe, will ultimately be the endpoint that we're measuring for this drug in terms of benefit. And what we are going to be sharing at ASGCT is the further update to that. And so this will be a March data cutoff. It's about six more months and that will also include at least six months at the third dose level, 0.5 MPK and some other emerging data at the next dose level. Again, I don't want to get ahead of sharing what that data is. But of course, we will be looking the strength of that benefit, as we go up in dose, we would hope that we would improve from that 50% reduction in the rates of MDs, approximately 50% reduction in MDs. And we'd also want to obviously see that the drug continues to be very well tolerated with no safety concerns in that patient population. We'll also see much more follow-up time in terms of total time on drug across the entire study, which will help to build that case moving forward. Now on the point of relationship to other programs, propionic acidemia, PA is a sister disease to methylmalonic acidemia, MMA. And as we get more and more confident, hopefully, about the dose level in which we're expanding the PA program and the data we're seeing there. We do believe that reads through very directly into the data that we expect to see shortly in MMA. We have started see some of that data from that Phase 1/2 study in populations. I'll remind you that MMA is also chronically dosing in patients and escalating through dose levels. And at the right moment, we will obviously want to provide an update on that data as well in terms of MMA. But we do believe that the PA did really positively through that. And generally I'd say that's true for our liver metabolic rare disease programs including OTC and the others that you referenced.
Jamey Mock:
And Salveen, maybe I'll take the COGS question. So to reiterate, this year is 35% to 40% of sales. And as you mentioned, we'll go to 20% to 25% by 2027. So I'm not sure it's a perfectly straight line, but let me just give you the factors that will improve that over time. First is volume. So increasing volume over time as we add new products, our overall manufacturing footprint should give us better leverage. I think predictability helps that so this is a highly unknown season this year. It will be the first time we're transitioning to an endemic. So that will become more predictable over time. ASP, I think, will continue to go up as well. What might offset that a little bit is a greater single-dose file or PFS presentation over time, and we've got some of that budgeted for 2023. So overall, we feel confident inverting our inventory levels down and overall decreasing our cost as a percent of sales. As it pertains to the 2027 P&L and what does that mean for overall company profitability, we haven't issued any guidance on that. I would say to reiterate for those that heard my prepared remarks, the respiratory vaccine business should generate $4 billion to $9 billion, which we laid out on the page there. And then we'll just have to see, to be honest. We've got an exciting pipeline. We have to understand what's happening with INT. We're quite excited by that, rare diseases, latent diseases as well, and we'll do what's best for all of our stakeholders. So if it makes sense to continue to reinvest some of those profits back into the business, we'll do that. And we're just going to have to wait and see where the pipeline looks like at that time.
Operator:
Thank you one moment for our next question. Our next question comes from Tyler Van Buren with TD Cowen. Your line is open.
Tara Bancroft:
This is Tara on for Tyler. So I know you mentioned a little bit about timing. But specifically, what we're wondering is, once the COVID strain is selected for the fall and winter season next month, how long approximately do you think it will take to finalize the commercial contracts in the U.S. and abroad? And then separately outside of the U.S., does the recent announcement regarding the ongoing negotiation with Pfizer and Europe actually create an opportunity for you guys to perhaps drive a larger contract in Europe than previously anticipated?
Arpa Garay:
Thank you for the question. Your first question around timing of contracting in the U.S. while we are waiting for the final variant strain to be selected in the middle of June, we have already initiated contracting conversations based on an FDA-selected variant. So as I mentioned previously, we do anticipate seeing some of these contracts being signed over the next several weeks, leading into the third quarter. So it will be sort of an evolving time line based on the customer. From an EU perspective, we're encouraged by the news that the EU is in renegotiations with Pfizer. For us, the signals that the EU likely does not want to rely on one sole supplier. And we also are encouraged that the EU does recognize the value of the effectiveness and safety of our COVID-19 vaccine. So we continue to work with them to see how we can help protect the 140 million people or so in the EU, who are at high risk of COVID infection. And as we get updates on EU negotiations, we will be sharing those as well.
Operator:
Thank you one moment for our next question. Next question comes from Michael Yee with Jefferies. Your line is open.
Michael Yee:
Two areas I wanted to ask on PCV or shall I say, INT cancer therapy. Stephen, you've talked and mentioned there in the slides around time as well as breakthrough designation. And I think you mentioned that you do have a discussion with FDA around your recent data. Can you just talk to the scenarios around a potential accelerated approval, the argument that you have to bring this to patients sooner or do we really have to wait years to run the Phase 3? Talk a little bit about how you feel about that this year. And then the second question, I think, is also important with regard to RSV. Obviously, we'd love to see a diversification of revenues. This could be coming next year. How do you see your revenue opportunity in 2024? Is that a payer battle or a couple of competitors out there, where is your advantage? And how do you see yourself with market share and revenues next year?
Stephen Hoge:
Thank you, Mike, for the question. I'll take the INT one. So Look, I think it's obviously still early in this discussion about what it will take to bring this notion forward to patients. And even in the Phase 2 study, while the data is really exciting, it's probably premature to say that it's sufficient for -- at this point for proceeding directly to accelerated approval. But there are conditions we think over the next year -- or couple of years that could lead to that being an appropriate thing for us and regulators to consider. In the short term, look, the data is still maturing. We still haven't put out the distant metastasis free-survival data. We're going to be doing that at ASCO. We're obviously excited to share that data, and it starts to just build that more complete picture and DMFS because it looks at visceral lesions does start to look towards perhaps some of those more -- the severity of those relapses that are happening. We have additional biomarker data that's coming through in that ASCO presentation as well. And both of those data sets are dealing with the initial analysis, which was 40 events. But I'll remind you, we have an analysis at 51 events, which we think will be when these curves are substantially more mature. Obviously, we haven't crossed that yet. And when we do trigger that 51 of that analysis, we will update the RFS curves. We'll update the DMFS curves. We'll obviously look at statistics around that. And it will be a point in time for us to understand, let's say, with more than 2.5, maybe 3.5 years of follow-up in total median follow-ups on that study. What do the overall curves look like? What are the hazard ratios? What do the statistics look around that? And then what more have we learned about the MLA from all of the ongoing biomarker work that we've done. So that richer data set I think, is really what we are waiting for to see. There's a second piece of this, too, though, which is that. Any consideration of an accelerated approval, whether it's for INT or just more generically, increasingly, it's going to depend upon whether or not you have initiated the confirmatory studies and that's appropriate. Because we've all seen how initiating substantially enrolling the confirmatory studies actually helps make sure that, that answer comes quickly and that accelerated approval can either be converted to a full approval or be adjusted as a result of those confirmatory results, and we're very attuned to that. And so our primary focus right now, while we're waiting for the data to mature from the Phase 2 is to make sure that we stand up that Phase 3 study and enroll it as fast as possible. I think the conditions under which we might consider in partnership and discussion with regulators pursuing accelerated approval would really be at some point in the future when that Phase 3 study is well on the path towards being enrolled obviously not read out yet, but well on the path there being enrolled and where a lot of the other data I had described had matured and continued to show a really compelling pace for the potential benefit here, perhaps even in patient populations they are at higher risk from a stratification perspective as we were sharing in some of the data today. And that those -- that complement the factors would trigger an opportunity to say it's time to bring this medicine to patients in an accelerated way while waiting for the confirmatory read out. So premature. All of those things still have to happen. So that's why right now our focus is get that Phase 3 started, get it enrolled and continue to follow this really intriguing story in the randomized Phase 2 study.
Arpa Garay:
And I can take the RSV question. We continue to be very excited about our opportunity to RSV. The profile of our vaccine as we look at the Phase 3 data for tolerability as well as effectiveness, position us at the high end of the competitive landscape. From a safety perspective, to date, we have not seen any neuro adverse events and our serious adverse events were balanced in both arms as well. What this means for 2024 is as we look at RSV as a seasonal business, we do anticipate that negotiations will be happening every year in the United States. The repairs will have an opportunity to continue to review the data across multiple players, and we will be working actively to position ourselves in the U.S. commercial market.
Operator:
Thank you one moment for our next question. Our next question comes from Terence Flynn with Morgan Stanley. Your line is open.
Terence Flynn:
Great. I was just wondering on INT, if you can comment at all about the design of the Phase 3 in lung there. And then just what's driving that confidence to move forward at this point? Can you just remind us of any data you have at this point on that front?
Stephen Hoge:
Yes. So it's a couple of things. So first, we did look in the Phase 1 at non-small cell lung cancer, there were patients in that. Those were not adjuvant patients, but nonetheless we do have some data, biomarker data and other clinical histories, again, not from a controlled study in the Phase 1. I think the other confidence is the strength of the mechanism of action that we're seeing and the translation across risk strata in the Phase 2 study that we've already run, really, we think sets up well as you look at adjuvant indications more broadly, and that's where an adjuvant non-small cell lung cancer, even neoadjuvant non-small cell lung cancer makes a lot of sense from a translatability perspective. And so, it's a combination of a little bit of data from that Phase 1. The breadth and strength of performance we're seeing in the Phase 2 for melanoma and obviously, what's been learned with PD-1, PD-1/L1 therapy in adjuvant settings more broadly. Operator Thank you one moment for our next question. Our next question comes from Jessica Fye with JPMorgan. Your line is open.
Jessica Fye:
A couple of follow-ups on some of the questions that have been asked already. First, on the U.S. COVID contracts for the fall, should we expect updates as those are finalized in real time or more like a combined sort of status report, for example, with 2Q results early on in the third quarter? Second, with the shift to an endemic phase for COVID, do you see any opportunity for improved price for co-vaccines outside the U.S., for example, in Europe? And lastly, on RSV, how soon do you believe RSV needs to be approved for you to participate in contracting for 2024?
Arpa Garay:
Thank you. In terms of your first question on providing updates for U.S. commercial contracts, we are not currently committing to real-time updates per contract. But at a minimum, we will be providing updates at our quarterly calls in terms of where we are with U.S. commercial contracts. The second question around ex-U.S. pricing, we do not comment on our pricing, but what I can tell you is for the majority of the countries outside the U.S., we are still primarily in a centralized government procurement model. So, we have not set endemic or more traditional commercial pricing yet for most of the markets outside of the U.S. And the last question, I believe, is on RSV contracting. We are targeting a 2020 launch for -- we continue to work through the details of the contracting for the U.S. market in particular. But assuming a 2020 for a launch per our current assumptions, we do believe we will be in time for contracting to launch in 2024 in the U.S. market.
Operator:
Thank you one moment for our next question. Our next question comes from Ellie Merle with UBS. Your line is open.
Eliana Merle:
Maybe if you could just elaborate a little bit on your confidence in the updated formulation targeting the B strains in influenza. And if you can comment on the dose level if this is studying a higher absolute dose. And just maybe just in terms of this compound, but also just broader the flu platform as you add additional antigens, just your thoughts on reactogenicity here and then broadly, with the additional balance of these compounds? And then second, just in RSV, just a bit of housekeeping. I guess, do you still plan to file this quarter?
Stephen Hoge:
Yes. Thank you for both. So first on the flu question. So, we obviously have the immunogenicity data that already came out of the P302 study that shows that we've met non-inferiority or would have been considered with nonfert for the B strains. Even in the Northern Hemisphere, study that's ongoing. And so I think our confidence of being able to clear that bar is high and supported by that data even before we made the improvements in the in the B strain update for the current Phase 3 study. We obviously have a preclinical data. We have a lot of experience with updating our antigens and vaccines now with COVID and others. And so we'll look forward to that data in P303. -- but I think we believe we will do even better than we just did in the P302 study with non-inferiority, and we hope to to see that we'll be achieving something perhaps trending towards superiority. But that's not the goal specifically for the study. In terms of reactogenicity and dose, I'll just say that we are not changing the dose for this 1010P303 study is still 50 micrograms, so it's not a change in dose level. And in general, as we think about reactogenicity across our respiratory pipeline, we have a large number of candidates and vaccines where we've gone to doses substantially higher than 50 micrograms. Even in the flu study, we did that up to 100 micrograms on data we shared before. And we believe that there are populations for whom that works well. And in fact, there are vaccines like our RSV vaccine where 50 micrograms is extremely well tolerated. We're very pleased by that profile to date. And so we actually think it's going to be a vaccine by vaccine case. You can't look at the dose and decide. And as we start going into combinations, we will be optimizing the reactogenicity, the tolerability profile of that vaccine against the benefits in terms of high efficacy that we hope to deliver and often measured by immune responses in those combination studies. So, we currently don't believe that there's a limit on that. But in the specific P303 study, we're continuing down a 50-microgram dose level for 1010. Now on RSV, yes, we are working closely with regulators on filing globally. And that includes all of the markets in which we hope to commercialize that product and launch it next year. And of course, we'll keep you appraised as we move into those regulatory -- that regulatory process in our normal quarterly updates.
Operator:
One moment for our next question. Our next question comes from Luca Issi with RBC. Your line is open.
Luca Issi:
Maybe on IMT, I think you've been highlighting the opportunity adjuvant and neoadjuvant settings. However, I wonder if you could comment on what's the plan in the metastatic settings, is there a place for IT to metastatic settings? And maybe related to it, are you planning to update the metastatic head and neck cancer data set that we have seen at 2021? Any color there much appreciated.
Stephen Hoge:
Great. Thank you for this question. So I think we do think that INT can go both earlier and later in terms of its use. And it's a challenging question about which one do we prioritize in the short term. I think there's a huge opportunity we perceive, we believe, in adjuvant. That is where you hear all of our current activities. That's our focus. That's where we're trying to move into pivotal studies, very, very quickly Phase 3 studies. But if you look to adjuvant, we do have adjuvant experience. You pointed to the head and neck data. We also have from our Phase 1 to management melanoma, there's actually some adjuvant -- sorry, the metastatic melanoma, metastatic non-small cell lung cancer. And I think situations where we think the burden of the tumor, the size of that tumor is a little bit of a barrier to the potential active activity of any immune therapy. In fact, generally, immune therapies have struggled in later-stage disease and where the real power of the technology, its safety tolerability profile, potential benefit probably is upstream. So while we are following closely the metastatic space and we did see some intriguing signs in the metastatic head and neck study that you referenced, we are right now waiting for a little more data to decide whether or not we want to go into those metastatic settings in the short term with our partner, Merck, of course. The other area that I referenced is the earlier stage. And so Stage 3, Stage 2 disease, disease where immune therapies are not traditionally used right now, but we're a well-tolerated approach like INT that we believe does provide a boost of specific T cell responses against the cancer may have a unique benefit. And again, that's a place that we're eager to explore the INT approach with our partner mark in the very near term. We don't have at the present as substantial an effort going into those two areas, but we could pivot very quickly. So for now, what we're focusing on is the pivotal studies in adjuvant, we're watching very closely the evolution of our data in metastatic, and we're trying to think about how we could move whether biomarker enabled or otherwise into earlier-stage diseases. I'm sure we will find ways to explore all of those areas if there's a potential for benefit for INT and then it's just now a matter of working down the opportunities as fast as we can.
Luca Issi:
Fantastic. And maybe if I can follow up. When is the earliest that you can have the COVID plus flu combo potential in the market?
Stephen Hoge:
I'll comment very quickly, but the combination vaccine -- so the first point is we want to need to get the flu approved. I think in the Vaccines Day, we talked about our expectations, our hope are to have the COVID-flu combo approved and launched in 2025.
Operator:
One moment for our next question. Our next question comes from Geoff Meacham with BoA. Your line is open.
Alec Stranahan:
This is Alec on for Geoff Meacham. So given the breadth of your clinical pipeline and the potential opportunities for new vaccines, how does Moderna prioritize assets or indications to go after? Once the data has, let's say, proof of concept for its line disease, what is your clinical strategy in terms of targeting additional bacterial indications? And then just finally, how are you thinking about capital deployment for the INT program?
Stephen Hoge:
I'll take the first question. I'll take all three, I think, but I invite my colleagues to Kevin. So -- so first on how we think about opportunities. It's a great challenge we have. We're obviously seeing a very high success rate as we transition into patient populations or pivotal studies across our pipeline. And the short version of it is we look for places where we think our technology through its modalities is well validated. And so we have a highly differentiated probably success in that next indication and where there is a large unmet need in that indication. If there's a large unmet need medically, there is usually a large unmet need financially in terms of health care systems, and that's ultimately the kind of value we want to deliver into it. So that's how we approach it. That can be infectious diseases. That's how we think about expanding our respiratory franchise. That's how we're thinking about expanding our latent franchises. That's how we've established POC there. And what you're going to be seeing us do in rare diseases, as we are already doing, is that same sort of expansion as we start to see proof-of-concept in the rare metabolic disease space. And then, of course, you asked a question about INT and maybe I'll just jump to the third part of your question, which is how do we think about capital allocation in terms of in INT. There are very few things that have a larger burden of disease, social cost of financial costs of obviously morbidity and mortality than cancer. And we do believe that INT has the potential to be a transformational treatment in that space. And so we got to figure out how to do is how do we responsibly, but aggressively grow that investment across a range of different places where we believe that INT will work. Now we have one significant advantage in that exercise, which is we have a partner in Merck, who has a high degree of conviction around this as well as we talked about, and we are co-investing with them. And so from a capital allocation perspective, we start together looking at what are the indications where there is an opportunity, unmet need in oncology with INT. As I said before, big focus right now is in adjuvant, but of course, we'll be looking at metastatic and earlier stage disease as well. Last question, I think I'll address is the middle one, which is how do we think about line POC and what does it unlock in terms of bacteria. I think we're very keen to address Lyme because of the unmet need associated with Lyme disease, particularly in the northern -- in Europe and the United States. But it is a very interesting one for us to demonstrate a bacterial -- antibacterial vaccination, we already have a substantial discovery pipeline looking at other bacteria that if we can demonstrate proof of concept in line that we will bring forward very quickly. I won't provide updates on those specific targets as that is still preclinical research. But you rest assured, our approach in bacteria will look like our approach in respiratory or latent or INT or rare diseases, which is as we see proof-of-concept. We will double down quickly with other programs that we think have high probability of success.
Operator:
Thank you one moment for our next question. Our next question comes from Simon Baker with Redburn. Your line is open. Simon, your line is open. You can ask your question. Do you want me to remove some from the queue?
Lavina Talukdar:
Yes, you can go to the next question. It will be our last question. Thank you.
Operator:
Thank you. One moment for our next question. Our last question comes from Hartaj Singh with Oppenheimer. Your line is open.
Unidentified Analyst:
This is Erica on for Hartaj today. So in the slides, you have provided the estimated 2027 respiratory product sales range with the high end being almost double or below. I'm curious if you can talk about to what extent does the standalone versus combination share of respiratory vaccines affect this revenue range, and we're just trying to understand this has the degree of like market cannibalization by the combo vaccines? And then secondly, a question on the pediatric RSV program, so the burden of disease for children and caused by RSV is immensely high. Can you talk about your progress in the context of competition in the pediatric RSV program and potential time lines for development? Thank you for the questions.
Arpa Garay:
Sure. Thank you. I'll start with the first question around the respiratory opportunity. We are hearing significant enthusiasm from both consumers as well as our customer base combination vaccines. We believe that it will enhance compliance and adherence to address the broad respiratory burden of disease by putting these two, potentially three vaccines into one. So, as we think about the commercial opportunity, we do think combinations will be the majority of the opportunity as we look forward in 2027. And with that, we do anticipate cannibalization of the monotherapy.
Jamey Mock:
With that efficacy, too, so as we continue to improve our products over time, particularly in fluid and COVID, I think the better the strain matches better the efficacy, I think, will also advantage us in the future.
Stephen Hoge:
And pediatric RSVV question. So, we've actually been working in pediatric RSV as long as we've been in RSV. And so, we have ongoing clinical trials including monotherapy and combination respiratory vaccines across a couple of different diseases that impact that population. We completely agree. It is a huge unmet need and area. In terms of guiding forward what timing will look like, we're conducting clinical research. We'll provide the updates on the data as we see it. We've already actually shared some of the early data from our pediatric programs. There will be both seropositive so kids who have previously been RSV, there is some benefit there. Those children do get reinfected and that will look more like a boosting set of studies. And then there will be seronegative children. Those are the -- those are the places where there might be the highest unmet need, those who have not yet had their first instance of RSV, so very young children onto the age of one. What you'll probably see us do over time is bifurcate those development programs because it en- up being very different take target populations. And more likely than not, the seropositives will move faster and the seronegatives will move slower as is normally the case with pediatric development.
Operator:
Ladies and gentlemen, this does conclude our Q&A session. I'd like to turn the call back over to Stephane Bance for any closing remarks.
Stephane Bancel:
Well, thank you, everybody, for joining us today and for your questions. The next month is going to be exciting, PA data May 18 and new INT data at ASCO in early June. Have a great day. Bye.
Operator:
Ladies and gentlemen, this does conclude today's presentation. You may now disconnect, and have a wonderful day.
Operator:
Good morning. My name is Kevin and welcome to Moderna’s Fourth Quarter 2022 Earnings Call. [Operator Instructions] Please be advised this call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone and thank you for joining us on today’s call to discuss Moderna’s fourth quarter and full year 2022 financial results and business update. You can access the press release issued this morning as well as the slides that we will be reviewing by going to the Investors section of our website. On today’s call are Stephane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Arpa Garay, our Chief Commercial Officer; and Jamey Mock, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance or results to differ materially from those expressed or implied in these forward-looking statements. With that, I will now turn the call over to Stephane.
Stephane Bancel:
Thank you, Lavina. Good morning or good afternoon everyone. Welcome to our Q4 2022 conference call. Today, I will start with a quick business review of 2022. Stephen will then review our clinical programs before Arpa gives an update on our commercial progress and plans. Jamey will then present our financial results and I will come back to share some posts on where we are heading. We are pleased to report today revenues of $19.3 billion for fiscal year 2022, GAAP net income of $8.4 billion, and GAAP diluted earnings per share of $20.12, cash and investment balances of $18.2 billion at the end of the year. We continued our disciplined capital allocation policy, reinvesting first in our company. In 2022, we invested $3.3 billion in R&D, our highest level of R&D investments ever. We invested $1.1 billion in SG&A and $400 million in capital investments. We made investments in Metagenomi for access to new gene editing enzymes and Carisma in oncology. We announced an investment in CytomX and the acquisition of OriCiro in Japan to continue to streamline our manufacturing processes. And just yesterday, we announced the collaboration with LifeEdit. $3.3 billion was returned to shareholders through a buyback of 23 million shares. I am proud of the strong results by our team in 2022 as we made history with a number of outstanding accomplishments for patients. In respiratory vaccines, we developed new products with remarkable speed, getting the mRNA-1273.214 against Omicron BA.1, the strain recommended by WHO and mRNA-1273.222 against Omicron BA.5, the strain asked by the U.S. FDA. We developed 1273.222 in less than 2 months. We are able to protect millions of people from potentially severe disease resulting from new COVID strains. Our RSV vaccine went from Phase 1 start to Phase 3 data in 24 months and met its primary efficacy endpoint in the Phase 3 trial. In oncology, our personalized cancer vaccine was the first demonstration of positive results from an mRNA cancer treatment in the randomized clinical trial. In rare diseases, our propionic acidemia program showed early positive clinical results in a repeat dose chronic disease setting in reducing metabolic decompensation events in patients. And we announced what we hope will become the first effective inhaled mRNA therapy in humans as our partner, Vertex, entered a Phase 1 trial using our technology in the therapy for cystic fibrosis patients who lacks the CFTR protein. Finally, we had our first ESG Day and published our first ESG report, providing additional transparency in how we conduct our business. I want to take a moment this morning to touch on transition the Moderna Executive Committee. As we announced in late 2022, Marcello Damiani decided to retire as Chief Digital Officer after more than 7 years with the company. Marcello joined Moderna before our first clinical trial and we are today a digital-first company, as a big testament of his ability to scale digital resources. I am grateful to Marcello for his contribution during the early years of Moderna. I am excited to have worked already with Brad Miller since early January. Brad brings a wealth of enterprise solution and platform organization experience in several of the top technology companies. This will be instrumental as Moderna scales into a fully-integrated biotechnology company. I want to also share with you that Juan Andres, currently President of Strategic Partnership and Enterprise Expansion has informed me of his decision to retire and will be retiring at the end of May. Juan has played a tremendous role since joining Moderna in 2017 from Novartis, where he led all manufacturing for them. Juan served as Moderna’s Chief Technology, Operations and Quality Officer where he led our manufacturing from an early-stage clinical development company to a commercial company. I believe Juan did a historic job with his team in 2020 and 2021 to scale Moderna for global commercial launch during the pandemic. It is literally unbelievable that he led the team from having made across our entire portfolio, less than 100,000 doses in 2019 to more than 800 million doses in 2021, all during the pandemic. We and hundreds of millions of people across the globe who received the Moderna COVID-19 vaccine owe Juan our gratitude. I believe very few manufacturing leaders would have led such an achievement. Most recently, Juan has focused on building out our organization to support Moderna’s growing pipeline, leading our efforts in producing of personalized cancer vaccine. Jerh, who used to work for Juan at Novartis, has joined us since early fall and has been leading manufacturing since then. I am thankful for Juan who has ensured a very smooth transition, helping Jerh every step of the way. Upon his retirement at the end of May, Juan’s responsibility will transition to Stephen Hoge, President of Moderna, to integrate PCV across all functions, with Jerh leading the manufacturing of PCV for multiple Phase 3s, and of course, for getting commercial ready. On behalf of the entire Moderna team, I want to thank Juan for his continued leadership and wish him and his wife, Marina, the best of a well-deserved retirement. I am deeply thankful to have counted him for so many years as a partner at Moderna, and more importantly, for more than 20 years as a friend and a mentor. We will miss him. The company continues to expand at a rapid pace. We now have 3 commercial COVID-19 vaccine products. We have 4 development programs in Phase 3. We have to expand our commercial portfolio very soon. Overall, we have 48 programs underway with a team of 3,900 team members and now present on the ground in 16 commercial subsidiaries across Americas, Europe and Asia-Pacific. Our $18 billion of cash balance at the end of the year is enabling us to scale across research, clinical development, manufacturing, commercial and G&A. With that, let me ask on to Stephen.
Stephen Hoge:
Thank you, Stephane. Good morning or good afternoon, everyone. Today, I will review our progress against our key clinical programs. I will start with our respiratory vaccines. We have approved our Phase 3 development programs against the big three respiratory viruses, COVID-19, RSV and influenza. I will share some additional data on these in a moment, including some presented this morning on our older adult RSV Phase 3 trial. We are also advancing a portfolio of next-generation programs against these viruses, including mRNA-1283, which is a next-generation COVID-19 booster that is referred to as rather stable. We also have multiple next-generation flu programs. We seek to increase the breadth of coverage against influenza by adding additional antigens that are not present in currently-available flu vaccines. Lastly, our respiratory portfolio includes a large number of combination vaccines to provide protection against multiple respiratory pathogens which has advantages for many stakeholders, including healthcare providers, payers and consumers. These include combinations of COVID, flu and RSV as well as two pediatric vaccines that include additional viruses that are important in children, including hMPV and PIV3. As we prepare for endemic COVID in 2023 and beyond, we wanted to briefly recap the recent VRBPAC committee discussions and recommendations. At the January VRBPAC meeting, the committee voted to harmonize the primary series and booster dose vaccines, which is an important step to simplify future guidance. The FDA also indicated that it expects to convene VRBPAC to determine vaccine strain composition for the ‘23/24 season in the second quarter of this year. We believe that our mRNA platform has demonstrated its ability to deliver variant match vaccines on accelerated time horizons and we believe we are therefore well positioned to deliver whatever composition update the FDA and other public health agencies recommend. And moving to RSV, as you know, we shared the top line results from our Phase 3 RSV study in older adults earlier this year. And today, we shared additional data that was presented this morning at RSVVW. The top line results we have seen are incredibly encouraging and we are grateful to the FDA for breakthrough therapy designation for mRNA-1345, which further emphasizes the significant health impact of RSV in older adults and the high unmet need. In the top line data presented in January, the mRNA-1345 demonstrated 83.7% vaccine efficacy and the primary endpoint of lower respiratory tract disease with two or more symptoms. 1345 was found to be generally well tolerated and there were no safety concerns identified by the Data and Safety Monitoring Board. In the data presented today at RSVVW, we confirmed that 1345 was well tolerated and has an acceptable safety profile. Solicited adverse reactions were mostly Grade 1 or Grade 2 and to-date most solicited adverse reactions were mild to moderate, with the most common adverse reactions being injection site pain, headaches, myalgia and arthralgia. Vaccine efficacy was consistently high across all age groups and in participants with pre-existing comorbidities that are at highest risk. Please refer to the scientific and medical meetings section of the Moderna Investor Relations website to see the full RSVVW presentation. And we are very encouraged by these data and look forward to file a Biologics License Application with the FDA in the first half of 2023 as things proceed. With the option of using a priority review voucher, we might see regulatory action on this filing in late 2023 or early 2024. Now moving to flu. Last week, we shared with you data from our Phase 3 immunogenicity and safety study in the Southern Hemisphere, study P301. In this study, our first-generation vaccine, mRNA-1010 demonstrated superiority on zero conversion rates for influenza A H3 and H1 and superiority on geometric mean titers for H3 and non-inferiority on geometric mean titers for H1. mRNA-1010 did not meet non-inferiority on zero conversion or titers for the two influenza B strains. Our separate Phase 3 efficacy study in the Northern Hemisphere, study P302, has now accrued over 200 confirmed cases of influenza-like illness, almost all of which are influenza A, which is expected – this was expected as the overwhelming majority of influenza burden in older adults is caused by influenza A, including over 95% of hospitalization in the most recent season. Now based on the case accrual in P302, we now expect the independent DSMB will review the first interim analysis of efficacy in that study in the first quarter of this year. Now, let’s take a look at our latent vaccines on Slide 14. Our CMV vaccine is in an ongoing Phase 3 study and we have begun dosing participants in the Phase I/II adolescent dose-ranging study. Our EBV vaccine to prevent infectious mononucleosis is in Phase 1, while our EBV vaccine to prevent long-term sequelae of EBV is in pre-clinical development. We have two HIV Phase 1 trials ongoing and our HSV vaccine is in preclinical. And finally, our VZV program has begun dosing in participants in a Phase 1/2 study, which I will discuss further on the next slide. The VZV study is a Phase 1/2 randomized safety and immunogenicity study evaluating mRNA-1468 against Shingrix. This is a relatively large study, enrolling 500 zero negative older adults in multiple doses and dosing intervals and a 12-month study follow-up. Over 35% of participants will be 70 years and older, which is in line with the largest disease burden of shingles. Now, let’s take a look at our therapeutics portfolio on Slide 16 and I will highlight a few of the programs. We have recently reported strong top line data for our personalized cancer vaccine, which I will talk to in a moment. In immunooncology, we are working to address disease burden beyond PCV with our checkpoint and triplet programs, both of which are in Phase 1 trials in various tumor types. In rare diseases, our Phase 1/2 PA program continues to enroll patients and we are looking forward to selecting a dose of the expansion arm. I will provide a brief update on that in just a moment. Earlier this year, our partner, Vertex, announced initiation of a Phase 1 trial in cystic fibrosis patients, which is our first inhaled pulmonary mRNA therapeutic program. And in cardiovascular, we announced Relaxin has initiated dosing in the Phase 1 study. Both of these study initiations represent important milestones for Moderna as we expand our modalities in therapeutic areas. Now in December, we shared exciting top line data from our Phase 2 personalized cancer vaccine program, testing the combination of PCV and KEYTRUDA against KEYTRUDA alone in the setting of adjuvant melanoma. KEYTRUDA is the standard of care in that setting. In this study, we showed the addition of our personalized cancer vaccine treatment mRNA-4157 to KEYTRUDA reduced the risk of recurrence or death by 44% compared to KEYTRUDA alone. This was the first demonstration of efficacy for an investigational mRNA cancer treatment in a randomized clinical trial and we are pleased to announce that 4157 has received breakthrough therapy designation from the FDA. Along with our partner, Merck, we are excited about these results and expect to launch multiple late-stage confirmatory studies for PCV in 2023, starting with melanoma and then moving to non-small cell lung cancer. We are planning to explore additional indications for 4157, where we believe there is a strong biologic rationale for immune-stimulating approaches. These include early-stage and metastatic settings and will include indications where KEYTRUDA is not yet approved. Finally, we expect to release full data from our Phase 2 study at an oncology meeting this spring and in an upcoming publication. Now moving to PA, since our update with our – at our R&D Day, the PA Paramount study has made good progress. Our fourth cohort is now fully enrolled and we are currently enrolling patients in our fifth cohort which doses at 0.9 milligrams per kilogram every 2 weeks. We are encouraged that to-date we have not observed any dose-limiting toxicities. And we are also encouraged that all patients and families have opted to continue treatment electively in our open-label extension study across all prior dose cohorts. Now, the next step in this trial will be to review available data and determine a dose for expansion. I will now hand the call over to Arpa Garay who will provide an update on our commercial activities. Arpa?
Arpa Garay:
Thank you, Stephen and good day to everyone. I will start with a review of sales on Slide 20. In the fourth quarter, total product sales were $4.9 billion. In the U.S., our sales were $1 billion, sales in Europe approximated $2.2 billion, and sales in the rest of the world were $1.6 billion. We ended the full year strong, with total product sales for 2022 of $18.4 billion. Sales in the U.S. for the full year were $4.4 million, sales in Europe were $6.7 million, and in the rest of the world were $7.3 billion. We are reiterating approximately $5 billion in COVID sales for delivery in 2023 from our currently-signed advanced purchase agreements and deferrals. And we do expect additional sales from key markets such as the U.S., EU and Japan. Slide 21 summarizes the current composition of sales for 2023. We have advanced purchase agreements from Canada, Kuwait, Switzerland, Taiwan and the United Kingdom. We expect these sales to be recognized upon delivery of vaccines in the second half of 2023. Additionally, we expect further sales from deferrals from 2022 contracts. These deferrals are from the countries listed on the slide and are expected to be mainly recognized from deliveries in the first half of 2023. Together, these advanced purchase agreements and deferrals totaled approximately $5 billion in sales for 2023. We do expect additional sales from key markets, including the United States, EU and Japan, as well as Australia and other countries in Asia and Latin America. In the U.S., contracting discussions with commercial customers are ongoing and we will provide visibility into expected U.S. sales at a future date after we complete these discussions. In our discussions with commercial customers in the U.S., it is clear to us that our customers recognize that COVID is still a substantial health burden. Throughout 2022, COVID continued to be a leading cause of hospitalizations and deaths. If you look to the chart on the left hand side, what you will see here is data available through September of 2022 COVID was the third leading cause of death in the United States only after heart disease and cancer. And if you look to this right-hand side, [Technical Difficulty] 4 months for the fall and winter season from October 1, 2022, [Technical Difficulty] 2023, hospitalizations from COVID in the U.S. are nearly 450,000, more than double from flu and nearly 3x higher than RSV in that same 4-month period. There continues to be a clear need to protect against severe COVID infections and our customers recognize that. Given this need, we estimate the U.S. fall 2023 COVID market volume to be approximately 100 million doses. We base this assumption after looking at 2022 vaccination rates and including potential recommendation for two-dose booster series for high risk individuals. Taken together, the doses administered represent roughly 30% of the U.S. population. A few factors that can impact this volume include viral evolution, regulatory recommendations as well as vaccine understanding and uptake by consumers. Moderna’s commercial organization is prepared for the transition to a commercial market in the U.S. Let me now take you through how we have been preparing to go to market. First and foremost, we are committed to access, which I will explain in greater detail in just a moment. To ensure coverage of our vaccine, we are engaged in discussions with private customers as well as public entities such as the VA, CDC and the Department of Defense. We are increasing awareness and educating consumers as well as healthcare providers about the benefits of booster vaccinations in alignment with public health agencies such as CDC and ACIP. We are reaching healthcare providers and consumers through innovative digital outreach programs. We have built the infrastructure needed to fulfill customer orders and shipments and our commercial and medical organizations have been scaling to execute on this plan and we are ready for the transition to a commercial market in the United States. Very importantly, as we enter the commercial phase of the endemic COVID market, we want to emphasize our commitment to vaccines access for everyone in the United States regardless of their ability to pay. For all insured individuals in the United States, consistent with preventative health services requirements, current reimbursement rules will be sustained. As an ACIP-recommended vaccine, Moderna’s COVID vaccine will continue to be available for zero out-of-pocket costs for individuals with insurance. And we are proud to say that for uninsured or underinsured people in the United States, Moderna will be launching a patient assistance program that will provide COVID-19 vaccines at no cost. Let me now summarize our COVID vaccine outlook. In 2023, we expect COVID sales of approximately $5 billion. In addition, we are expecting sales from U.S. commercial market orders, EU, Japan and other countries. We will provide visibility into these sales after we complete ongoing discussions with governments and with customers. We can recognize COVID continues to be a burden to healthcare systems and this continues to be an important point as we discuss the value of booster vaccinations with our customers. In the U.S., we expect commercial market volumes to be approximately 100 million doses in 2023 and Moderna’s commercial organization is prepared for the transition to a commercial endemic market. Last but not least, we are committed to patient access in the United States. I now want to turn to another launch in the respiratory vaccine space that the commercial team is preparing for, our RSV vaccine in 2024. As you heard from both Stephane and Stephen earlier, we are very pleased by our Phase 3 RSV vaccine results. Stephen’s organization will be filing for the approval soon and we expect we maybe approved in late 2023 or early 2024. With the potential approval fast approaching, I am very excited for the RSV vaccine launch and I want to provide additional color into the launch plans. The RSV launch will leverage the existing commercial infrastructure that is already in place for COVID and we will continue to invest to support it ensuring strong execution. Both COVID and RSV markets overlap considerably as we look at our target customers as well as potential target patients and audiences and we will leverage this overlap between the two markets. We will ensure awareness of RSV disease and the associated economic burden of RSV in older adults across key stakeholders such as healthcare providers and payers. Upon approval of our RSV vaccine, we will educate consumers on key attributes of our vaccine. These planned activities will be initiated in 2023 and in full force upon approval. We have the added benefit of an in-place commercial infrastructure built for COVID. Many of these resources can be leveraged for flu as well into the future. I look forward to keeping you updated on our progress throughout this year. And with that, I will turn it over to Jamey.
Jamey Mock:
Thanks, Arpa and hello everyone. This morning, I will cover our 2022 financial performance and provide a framework for our 2023 financial outlook. Moving to our fourth quarter results, starting on Slide 29, total product sales decreased by 30% year-over-year to $4.9 billion. The decrease in 2022 was mainly driven by lower sales volume compared to overall higher demand in the prior year. Cost of sales was 39% of product sales compared to 14% of product sales in 2021. The key driver of the increase in cost of sales as a percent of product sales was a catch-up royalty payment to the National Institute of Health, or NIH, of $400 million, representing 8% of product sales in the fourth quarter. In December 2022, we entered into a non-exclusive patent license agreement with the National Institute of Allergy and Infectious Diseases and institute or center of the NIH to license certain patent rights concerning stabilizing prefusion coronavirus spike proteins and the resulting stabilized proteins for using COVID-19 vaccine products or 2P technology. Pursuant to the agreement, we have agreed to pay low single-digit royalties on future net sales of our COVID-19 vaccines. Our cost of sales also includes a charge of $297 million for inventory write-downs related to excess and obsolete COVID-19 products and expense for unutilized manufacturing capacity, and CMO wind-down costs and related charges of $376 million, and a loss on firm purchase commitments and related cancellation fees of $281 million. These charges other than royalties are driven by costs associated with surplus production capacity, overall lower demand and a shift to our most recent Omicron BA.4/5 targeting COVID-19 bivalent booster. Research and development expenses were $1.2 billion, which increased by 87% versus prior year. The increase in R&D spend continues to be driven by our clinical trial expenses, particularly with our Phase 3 studies for RSV, seasonal flu and CMV. The increase in R&D was also driven by the acquisition of a Priority Review Voucher and an increase in personnel-related costs due to increased headcount. Selling, general and administrative expenses were $375 million, also reflecting an increase of 87% year-over-year. The growth in spending was primarily driven by continued investments in personnel and outside services in support of our marketed products and company build-out. The effective tax rate was 11% compared to 10% last year. After-tax net income decreased by 70% to $1.5 billion. Diluted earnings per share in Q4 decreased by 68% to $3.61. Now turning to our full year 2022 financial results on Slide 30. Total product sales for the full year 2022 were $18.4 billion, an increase of 4% year-over-year. The growth was mainly attributable to customer mix and a higher average selling price in 2022 in certain markets. Cost of sales was 29% of product sales compared to 15% of product sales last year. The increase was driven by higher write-downs for excess and obsolete inventory related to our COVID-19 vaccines, unutilized manufacturing capacity and losses related to future purchase commitments for raw materials. The key drivers for these charges are similar to the drivers in Q4. Costs associated with surplus production capacity, overall lower demand for the year, in particular from low-income countries, and rapid product advance shift from our original vaccine to Omicron targeting COVID-19 bivalent boosters. The previously-mentioned catch-up royalty payment NIH of $400 million is also a driver of the increase year-over-year. The effective tax rate was 13% compared to 8% last year. As a reminder, we had a net operating loss carry-forward of $2.3 billion at the end of 2020, which resulted in a non-recurring benefit to the reported tax rate in 2021. After-tax net income of $8.4 billion decreased 31% versus prior year. The decrease of net income was primarily due to higher cost of sales, higher other operating expenses and a higher effective tax rate. Diluted EPS decreased 29% to $20.12. Now turning to cash and cash deposits on Slide 31. We ended 2022 with cash and investments of $18.2 billion, compared to $17 billion at the end of the third quarter. The increase was driven by our commercial activity. Cash deposits for future product supply reduced from $3.8 billion at the end of the third quarter to $2.6 billion by the end of the year. Now turning to Slide 32. I wanted to give an update on the progress we have made on our capital allocation priorities. Our top investment priority has been and will continue to be reinvesting in our base business across multiple areas. Research and development spending increased 65% year-over-year from $2 billion in 2021 to $3.3 billion in 2022, and we are projecting an additional increase to approximately $4.5 billion in 2023. The clinical data from our PCV, RSV and flu trials were encouraging and further validates the potential of our mRNA technology. We are also investing in our digital capabilities, the commercial build-out of the organization as well as expanding our manufacturing footprint. We plan to significantly accelerate our capital expenditures in 2023 as we expand both our international and U.S. manufacturing footprint. Our second investment priority is to see attractive external investments and collaboration opportunities that will enable and complement our platform. We’ve recently announced several new transactions, and I’m happy to report that we have successfully closed our acquisition of our OriCiro Genomics in the first quarter of 2023. OriCiro is a great example of the companies we are valuating to enable our mRNA platform. It will create substantial value from both the speed and cost viewpoint and impact our pre-clinical, clinical and commercial pipeline for years to come. Our collaboration with LifeEdit, which we announced yesterday, is another example for an attractive external investment opportunity. We believe the combination of Moderna’s mRNA platform with LifeEdit proprietary gene editing technologies, including base editing capabilities, has the opportunity to advance potentially life transformative or curative therapies for some of the most challenging genetic diseases. We are in multiple active discussions regarding additional external collaboration opportunities, and we will be disciplined in our approach. After evaluating internal and external investment opportunities, we then assess additional uses of cash. In 2022, we repurchased 23 million shares for $3.3 billion at an average price of $143 per share. and we have $2.8 billion of share repurchase authorization remaining. Now, let’s turn to our 2023 financial framework on Slide 33. As Arpa mentioned earlier, we currently have COVID vaccine sales of $5 billion contracted for delivery in 2023. Also, we are actively working on preparing for the private market and government contracts in the U.S. and additional contracts for Europe, Japan and other key markets. To help you with your modeling purposes, we expect first half ‘23 sales to be approximately $2 billion. Our total cost of sales includes the cost of goods manufactured, third-party royalties, as well as logistics and warehousing costs. We expect full year 2023 reported cost of sales to be 35% to 40% of sales. This includes royalties in approximately 5% of sales, which are payable to UPenn and CellScript for modified chemistry licenses, and to NIAID and NIH for the 2P license that I mentioned earlier. The increase in cost of sales as a percent of product sales compared to 2022 is primarily driven by presentation mix change as we move from a pandemic to endemic setting, with single-dose application significantly increasing in volume. Longer term, as the endemic market normalizes and we add additional respiratory and other products, we expect our cost of sales as a percent of sales will significantly decrease rates we’re experiencing in 2023. For R&D and SG&A, we expect full year expenses to be approximately $6 billion, with approximately $4.5 billion in R&D. The increase is driven by our maturing development portfolio and the global scale up of our company. We expect a negligible provision for income tax in 2023. And finally, we expect capital expenditures of approximately $1 billion. The increase is primarily due to investments in expanding our manufacturing footprint. This concludes my remarks concerning our financial performance, and I will turn the call back over to Stephane.
Stephane Bancel:
Thank you, Jamey, Arpa and Stephen. Let me now share some thoughts about where we’re heading. I’m really excited to see our mRNA platform and the investments we have made in science over the last 11 years leading to such a promising pipeline. We anticipate a number of important developments. Let me start with our first franchise, respiratory vaccines. In COVID boosters, we are working for the switch to U.S. commercial market, and we anticipate being able to quickly meet the full 2023 market needs for updated vaccines after VRBPAC and the FDA make this transaction in the spring of 2023. We plan to submit RSV vaccine for regulatory approval in the first half of 2023, and as you heard from Arpa, we will be ready to launch the RSV vaccine in late ‘23 or early ‘24. In flu vaccine, for Northern Hemisphere, mRNA-1010 Phase 3 trial, the Data and Safety Monitoring Board is expected to complete its interim efficacy analysis in the first quarter of 2023 of second franchise latent virus vaccine. It’s progressing very well with a broad spectrum of programs. In our large CMV Phase 3 study, we look to complete the enrollment. For EBV, HIV and VZV programs, our next milestone will be Phase 1 data. Turning to Slide 37, let me review the milestone for mRNA therapeutics programs. For personalized cancer treatment, we expect to start our Phase 3 study in partnership with Merck in adjuvant melanoma, and we expect to rapidly expand to additional tumor types, including non-small cell lung cancer. Full Phase 2 data will be presented at an upcoming oncology meeting and published in a top-quality medical journel. In PA, we plan to select those and begin the expansion arm of our Phase 1/2 study. MMA, we will have Phase 1/2 data. The next milestone for heart failure drug by Relaxin will be Phase 1 data in patients. And in Inhaled therapeutics, our partner Vertex expects to complete its single ascending dose study and initiate a multiple ascending dose study. To continue to build the best version of Moderna, we have established seven priorities for 2023. Priority number one, execute the operational and sales plan for COVID booster for fall of ‘23. Priority number two, build an unrivaled seasonal respiratory vaccine franchise. Priority number three, execute a bold campaign of cancer vaccine studies. Priority number four, advance rare metabolic disease programs. Priority number five, drive rapid advancement and growth of our latent vaccine portfolio. Priority number six, deliver the next-generation pipeline and platform. As we said before, this is just the beginning. And Priority number seven, build a culture of perpetual learning and strengthen our processes and digital system as we want to scale the company to another level. On Slide 39, some key dates for 2023 Moderna Investor Days. April 11 will be our Annual Vaccines Day. September 13 will be Annual R&D Day, where we will present development pipeline key updates. And December 7 will be our second ESG Day. Now that we have delivered on the promise of mRNA science with our first product launch, our mission has evolved. Our mission is to deliver the greatest possible impact to people for mRNA medicine. We are passionate about our ability to have a profound impact on humanity. We believe we have a technology to eliminate or greatly reduce human suffering caused by respiratory viruses, latent viruses, many cancers, regenerative diseases and a growing list of other diseases. We believe we can have an impact on disease treatment, with our therapeutic first and then with our gene editing programs. This is just the beginning. With that, the team and I are free to now take your questions. Operator?
Operator:
[Operator Instructions] First question comes from Salveen Richter with Goldman Sachs. Your line is open.
Salveen Richter:
Good morning, thank you for taking my questions. Could you speak to the regulatory strategy for flu, given the miss on the B strains for immunogenicity? And if you are confident into the interim efficacy analysis, given the abominating prevalence of [indiscernible]?
Stephen Hoge:
I’ll take that. Thanks, Salveen for the question. Look, I think the honest answer is we’re still having incomplete information to provide guidance on the regulatory strategy. At this point, we are looking to the efficacy results from the P302 study that I described, which will guide us on that filing strategy. It’s important to note that efficacy, ultimate demonstration of non-inferior efficacy against an approved vaccine was always going to be required for full approval, and that the only thing that you could do with immunogenicity would be an accelerated approval path with an obligation to subsequently demonstrate efficacy. And so right now, we are actually very encouraged that the data that we’ve seen from our immunogenicity and safety study, which is running the Southern Hemisphere, P301, shows superiority on three out of four endpoints for the influenza A strains, which drive the overwhelming majority of disease, and the population of interest here are older adults, and account for over 99% of the cases in our efficacy study. And so that first interim efficacy analysis that we’re conducting now in P302 will evolve over 200 cases and 99% of them are influenza A, and it will be our first chance to really see the performance of the vaccine in terms of prevention of influenza-like illness from flu A. That is the first interim analysis. And so it’s quite possible, as you would expect in any efficacy study, and we’ve all got some experience now with these respiratory efficacy studies, that we may end up – need to go to a second subsequent interim analysis and accrue even more cases to demonstrate either non-inferiority or superiority in that study. And so what we will do is we will wait for the results from the DSMB, the independent DSMB, and gauge from that. Based on those results, obviously, if we do see efficacy, that is the gold standard for proceeding with regulatory filing and full approval. If we do not yet meet that threshold, then we will be looking forward to subsequent interim analyses in that study.
Salveen Richter:
Great. Thank you.
Operator:
[Operator Instructions] Our next question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
Thank you. Just quickly follow Salveen’s question. Do you need to show superiority in order to receive approval regarding the efficacy study? And then quickly on the revenue. Did I hear correctly the existing contract of $5 billion mainly will be in the second half ‘23? If that’s the case, is it fair to say that total COVID revenue in 2023 should be around $7 billion? And then regarding the $2 billion in the first half ‘23, how much will be from the U.S. market, i.e., out of estimated 100 million doses in the U.S., what could be your market share?
Stephen Hoge:
I’ll take the first question. Thank you, Gena for that. So first on superiority. You do not need to demonstrate superiority to get a flu vaccine approved. That’s well precedented. Non-inferior efficacy is the threshold. Our goal, though, over time is absolutely to develop a superior influenza vaccine. And so if we don’t see it with the first-generation product, which is mRNA-1010, I would note that we have four other programs – flu programs in development, different stages of clinical trials, that are looking to do even better than perhaps flu mRNA-1010. And our goal over time would be to demonstrate that we have a superior influenza vaccine. But it is not actually required for approval. Non-inferiority should suffice. I’ll turn it over to Arpa, I think, for the other questions?
Arpa Garay:
Sure. Yes, I can take the second question. In terms of the total sales, we are anticipating about $2 billion of the $5 billion in the first half of the year, and none of that $2 billion is coming from the U.S. market. The remaining advance purchase agreements that we have of $3 billion will be coming in the second half of this year. Now, that $5 million is just the total that we have from advanced purchase agreements as well as deferrals from 2022. We do anticipate additional sales from the U.S., Japan, EU and other markets, and we believe the majority of these sales will be in the second half of 2023.
Gena Wang:
And then your market share regarding the U.S. market?
Arpa Garay:
We continue to believe in the strong, differentiated profile of our products. We do not have any updates on market share projections as we are currently in discussions with customers right now for fall 2023 contracting.
Gena Wang:
Thank you.
Operator:
[Operator Instructions] Next question comes from Matthew Harrison with Morgan Stanley. Your line is open.
Matthew Harrison:
Great. Good morning, thanks for taking the question. I was hoping to ask about the regulatory strategy for PCV, and specifically, how you’re thinking about the potential for filing off the Phase 2 data set, as well as how you’re thinking about the time lines for enrollment in the Phase 3 program and how that may impact the time line for potentially filing off the Phase 2 data set? Thanks.
Stephen Hoge:
Great. Thank you, Matt for the question. So we’re obviously really pleased yesterday to announce that we received FDA breakthrough designation therapy for the PCV program. And what that allows us to do is very rapidly accelerate our conversations with the FDA and other regulators on the path forward for filing 4157. As you noted, the Phase 2b study that we’ve run is a randomized study compared against, really, the standard of care with KEYTRUDA alone, and has already shown a quite significant benefit of 44% reduction of the rate of recurrence and – or death. And that study is ongoing, and so we’re continuing to follow over time and conduct additional interim analyses. And it’s possible that those – in fact, we would hope that those data mature and continue to get stronger and stronger. And so it is entirely possible that in our discussions under breakthrough with the FDA and others that we will come up with a path forward for beginning the filing process based on that Phase 2 and potentially proceeding with accelerated approval. Now as you know, in this country, and I think it’s where your question is coming from, as well globally, if there is a path forward there. We haven’t yet engaged with the FDA on because the breakthrough happened yesterday. But if there is a path forward there, it would require us to rapidly enroll a confirmatory Phase 3 study. And in fact, there is more and more attention on perhaps requiring that those Phase 3 studies be enrolled prior to an accelerated approval. And so for that reason, ourselves and Merck are working really quickly now to try and stand up that confirmatory Phase 3 melanoma study and enroll as fast as possible. Now we’re not ready yet to guide on how quickly that will be, but we are fully aware of the fact that in fact, if there is a path forward for accelerated approval, the enrollment of that Phase 3 may be gaining, and therefore, we want to have it enrolled as quickly as possible. So at this point, we’ve just received the breakthrough designation, we’re engaging with regulators, and we’re going to try and develop that path forward. But it is theoretically possible that there is an accelerated approval path and that we would need to enroll that Phase 3 study based on recent regulatory guidance, more generally to the industry. And working hard to make sure that we can do that as fast as possible here, while continuing to conduct the additional interim analyses and see the maturity of this data continue to proceed, and hopefully, the strength of the benefit provided by the combination to be further validated.
Operator:
Thank you. [Operator Instructions] Our next question comes from Edward Tenthoff with Piper Sandler. Your line is open.
Edward Tenthoff:
Great. Thank you very much. And thanks for all the detail on the call today. My question, I had to go back to flu for a minute and just with respect to the follow-on candidates, including I think the one that’s pentavalent hemagglutinin and then fixed hemagglutinin and then also into the neuraminidase, the candidates at that neuraminidase, what should be our expectation both in times of timing for data here? And how do you ultimately see your seasonal flu product offering kind of evolving? Thanks.
Stephen Hoge:
Thanks for the question, Ed. So let me start with the most advanced program, obviously, is our 1010 program, which we’ve talked about. And we have done an update to that vaccine that we think will increase the B immunogenicity for those populations for whom that matters, and we expect to advance that in clinical study quite quickly. That, in combination with the efficacy data that we were just talking about with P302, probably is the most important information for guiding our next step on the second-generation products. Our multiple – sorry, penta and hexavalent vaccines as well as the 1020, 1030 programs, which include neuraminidase, as you said, are all in various Phase 1 studies. And as we have shown repeatedly hopefully over the last couple of years, we can proceed very quickly in the subsequent Phase 3 in pivotal studies once we select one of those candidates to move forward. But the really important gating information is understanding how is our first generation product performing in terms of efficacy as a first mRNA flu vaccine. And so we are waiting for that information before proceeding forward. But we do expect that at least one, if not multiple, of these second-generation products would move into subsequent pivotal studies, Phase 3 studies. And in those cases, because we would be looking to demonstrate some form of superiority either against a broader range of influenza strains or better protection against influenza-like illness because we have included additional antigens, we would expect those studies to include both immunogenicity and safety and efficacy end points as we move forward. And so we will make a selection on which ones we might move forward based on the ongoing interim analysis of efficacy from our 1010 program. We don’t have another way to update at this point on which we will move forward.
Edward Tenthoff:
Great. Thank you very much.
Operator:
[Operator Instructions] The next question comes from Michael Yee with Jefferies. Your line is open.
Michael Yee:
Hi. Thanks. Good morning. A couple of follow-up on the flu vaccines and also a PCV question for Stephen. I guess could you clarify – do you have a hypothesis around why these strains were not non-inferior and what the ramifications are for that, either for this flu vaccine, but also for the infection study, and what that would mean for combinations? So first of all, just to clarify what’s going on there with the B strain, the ramifications for flu vaccine? And then my second question is on PCV. Obviously, we are excited about the adjuvant data. There is also a competitor reading out in metastatic melanoma this summer, so I wanted to understand how we should compare and contrast that and if you could walk us through how to think about metastatic and what competitors might show? Thank you.
Stephen Hoge:
Thank you, Michael for both questions. So, first on the flu, on the B, we are still looking into the data, and we are going to develop a more complete picture of what we think happened in the influenza B immunogenicity study. I would note a couple of things that are important. The first is these are active comparator studies. And so when you look at non-inferiority on zero conversion or titers, it’s important to note that we are going at the standard-dose influenza vaccine active comparator. And between the Phase 2 study and the Phase 3 Southern Hemisphere study, there were changes in the composition of those active comparators and the comparator use, and so that can drive some difference. The second thing I would note is that they were different populations, and so we went from a Northern Hemisphere to a Southern Hemisphere. And obviously, that can drive some differences in background history of influenza illnesses. But the third and perhaps maybe most relevant is we did expect that the influenza B neutralizing titers were lower. As you remember, we shared with the Phase 2 data about a year ago. We did have lower neutralizing HAI titers for the influenza B strain. And for our older adult influenza vaccine, we thought that was acceptable because at the end of the day, our goal was only to achieve non-inferiority, not to demonstrate superiority. Whereas for the influenza A strains, we really wanted to maximize those neutralizing titers and potential for benefit because influenza A really is what drives the illness in older adults, which is our first generation product. So, we did focus heavily on the influenza A. We were aiming at non-inferiority on the influenza B. And as you said, we did not make non-inferiority on those, and we will continue to pull apart the reason as to why. But we have already identified an update that would allow us to improve immunogenicity against the Bs to the extent that that is important going forward, not just in younger populations but perhaps from an overall regulatory perspective. And so we have made that update, we are actually going to be evaluating that in the clinical study very shortly here. And we expect that we will be able to address that lower immunogenicity that we saw on the Bs quite quickly. But as I said a moment ago, what really matters is efficacy and efficacy against influenza disease. And in this case, we really do see that as influenza A related for our first-generation product. But we will obviously be updating influenza B strain for other subsequent generation of products. And it’s important to note that that’s really important as you get into pediatric populations where influenza B is a burden of disease, particularly in the young. Now, on the question of PCV, we are pretty encouraged by the adjuvant melanoma data. We do have some early metastatic data, as you will note from our prior Phase 1 studies. And in the Phase 2, there was some stage 4 disease as well in that study. We did not conduct it only in first-line metastatic, and so we are actually looking forward to understanding the performance of a competitive product in that space because it may actually identify an opportunity for us to move into earlier stages of treatment. Now, for now, our approach has been to focus on places where checkpoint, including KEYTRUDA, our standard of care and have demonstrated a really strong signal, which is why you see us expanding from adjuvant melanoma into adjuvant non-small cell, and ultimately, will go first into other adjuvant indications where we really think there is the most immediate biologic benefit or biologic rationale for potential benefit that we will be looking to demonstrate. But obviously, as others start to move into the space, if they show benefits in other lines of therapy, we will absolutely want to proceed very quickly in those directions as well. So, we are looking forward to those results, and we will obviously launching them as everyone else will.
Operator:
Thank you. [Operator Instructions] Next question comes from Tyler Van Buren with Cowen. Your line is open.
Tyler Van Buren:
Hey guys. Good morning. Thanks very much for taking the question. For RSV, a few years from now, do you expect it to be a 2-player, 3-player or perhaps a 4-player market when including J&J? And how do you believe that the tolerability profile compared to others based on the full data presented this morning?
Stephen Hoge:
Sure. I can maybe – I will take the first portion of that question. At this point, there are obviously three companies that have wrote out their Phase 3 pivotal efficacy studies and are proceeding right now to filing. I think – I don’t have a specific view on J&J, maybe Arpa can offer perspectives on that, but it is an adenovector virus [ph] program, and otherwise still unclear about their regulatory path forward. Now on the question of reactogenicity and tolerability profile, as we presented today or as our collaborator presented today at RSVVW, we do see, we think a favorable tolerability profile. Grade 3 adverse reactions, whether local or systemic, were all below 2% for any of the individual symptoms. And actually compared relatively favorably with a placebo in that arm, often maybe 1.5x as frequent as what we are seeing in placebo, which we think is a compelling overall reactogenicity profile. We then think the other parts of the benefit of the product are obviously efficacy. We are incredibly pleased. If you look at the most common definition of cases, or RSV, lower respiratory tract disease involving two symptoms, which has been relatively consistent across the different products. We have seen high efficacy 83%, which I really think is among the best. And as we presented today, that efficacy actually holds up beautifully as you look at older populations, those over the age of 70, as well as those with higher comorbidities. Those would drive the majority of the expense associated with caring for patients, older adults with respiratory disease from RSV. So, overall, it’s very difficult, obviously, to do cross-trial comparisons. And ultimately, it will fall for public health officials to make those decisions, but we are really encouraged by both the efficacy and tolerability profile of 1345, and look forward to filing and ultimately to the commercialization of that product. Arpa, would you like to add anything in terms of your perception of the market going forward?
Arpa Garay:
Sure. So, I would say we do anticipate it being at least the 3-player market with Moderna, Pfizer and GSK. There is a possibility of a 4-player market if J&J has a regulatory path forward with their adenovector virus vaccine. Nothing more to add there.
Operator:
Thank you. [Operator Instructions] Our next question comes from Jessica Fye with JPMorgan. Your line is open.
Jessica Fye:
Great. Good morning. Thanks for taking the question. Following up on flu or mRNA-1010, just to be very clear, if you hit on non-inferiority in the efficacy Northern Hemisphere study, do you think that mRNA-1010 would be approvable in spite of missing on non-inferiority on the B strains? And related to that, would you envision the approval only being for older adults? And on RSV, can you comment on the expectation for dosing frequency for your RSV vaccine? And how do you think about the value and potential pricing of that vaccine, maybe benchmarking off of other vaccines for that age group? Thank you.
Stephen Hoge:
Yes. Thanks for those questions. So, let me take the flu stuff first. So again, the full approval of standard is a head-to-head efficacy study. And so if we demonstrate non-inferior efficacy against an approved vaccine in the population which we are studying and which in this case in P302 is 50-plus adults. We do believe that could form the basis of an approval. At the end of the day, immunogenicity results, and in particular, are only surrogates for efficacy, and ultimately, efficacy is the gold standard. That’s what’s required for traditional approval, and that’s why we are running the P302 study. So, it will depend upon how a conversation with regulators around that data package, but it is certainly plausible. And in fact, one might say likely that if we meet efficacy in the efficacy study that that would be sufficient to move forward for a full approval. It doesn’t mean that there may not be questions about demonstrating non-inferior immunogenicity with influenza B strains or other things in subsequent studies, but we do believe there is a possibility there. But at the end of the day, it will be dependent upon data and discussions with regulators, including the FDA. And so we will wait until we have that data and have those conversations, but I think it’s certainly a possibility. Now, as it relates to age for approval, we are currently studying mRNA-1010 only in older adults. And so as I have said, the P301 was in 18-plus, P302 is in 50-plus, and that’s really where we see the broadest recommendations for seasonal influenza vaccine and where we have been most focused initially on building out our respiratory portfolio. We will evaluate our influenza vaccine, in fact, many of our respiratory vaccines in younger populations over time. But we will have to do age de-escalation, dose finding and then bridge down from an immunogenicity perspective, very much like what we did with COVID. And so our initial filings for approval, if they proceed based on data, would be in adults and older adults principally. And then eventually, we would follow-on with pediatric populations. And as I said a moment ago in response to Michael’s question, that may involve using updated B antigens to increase immunogenicity in that population. But again, that’s subsequent studies that we would do in children. Could you remind me of the second question?
Jessica Fye:
For RSV, what are you thinking for dosing frequency and how do you think about value and potential pricing of that vaccine, maybe benchmarking off of other vaccines for that age group?
Stephen Hoge:
Sure. So, I will let Arpa take the second part of that question. First on frequency, it is not yet clear on how frequently people will need an RSV vaccine. It’s a seasonal virus, a seasonal epidemic of disease that shows up. Most of us have been exposed to RSV all over a dozen times over the course of our life. And what really happens from a biology perspective is as we get older, our ability to maintain high neutralizing titers to protect us goes down, and what we have is breakthrough disease and ultimately, a disease leads to substantial cost and morbidity and even some mortality in older adults. We do not – we have not yet had approved vaccines, and so what we don’t yet know is what’s the frequency of vaccination, is it going to be seasonal every year, or is it going to be less than seasonal every couple or few years. But what’s pretty clear, based – from my perspective, based on the epidemiology of RSV infection, is that we do see RSV fairly regularly as adults, and unfortunately, over time, it breaks through more frequently, and so there probably will need to be repeated boosting to protect against RSV. At the end of the day, the initial recommendations will come from ACIP as well as from regulators around that frequency, and we will have to defer to them on how they want to administer and roll out the RSV vaccines, whether they want to follow a flu model, which would be annual to make sure that we get the broadest amount of protection, or that they want to initially roll out RSV vaccines and then follow over time for the durability of that efficacy. At this point, none of us, none of the three products that have read out in Phase 3 have a clear answer on the durability of that efficacy, although we would expect it to wane as it does against natural RSV infection over time in those results. Arpa, do you want to take the next part of the question?
Arpa Garay:
Sure. Yes. I can take the question on pricing. So overall, from a pricing philosophy perspective, Moderna is committed to pricing that reflects the value of our vaccines. In terms of what they deliver to patients, to societies and the healthcare systems, while also ensuring full access for patients regardless of their ability to pay. So, with that broader principle around pricing, we will be looking at the full recommendations that come out of ACIP as we get the filing and the ACIP recommendations to look at what the full value that could be provided back is based on things, as Stephen mentioned, around dosing frequency, and their pricing will be based on both value and access. I am not able to share any additional details on what sort of range that pricing might fall into, but it will be consistent with our overall pricing philosophy.
Operator:
Thank you. [Operator Instructions] Our next question comes from Ellie Merle with UBS. Your line is open.
Ellie Merle:
Hey guys. Thanks so much for taking the question. Just another on flu, just – any more details on the titer level specifically that you saw or when we will get more details on the titer levels? And then how should we think about the importance of having titers above that 40 benchmark versus demonstrating non-inferiority. Like I guess what is the comparator vaccine titers in your Phase 3, did very well and were well above 40? How should we think about the implications then, say, you are at 30 or near 40, what that would mean from a regulatory standpoint as well as a commercial standpoint in interpreting the immunogenicity idea? Thanks.
Stephen Hoge:
All very good questions and I think the short answer is we are looking into that data right now. And we will provide an update I am not exactly sure when we will have that, but we do have the Vaccines Day Investor Meeting coming up in April of the spring and then obviously, we will be looking to publish that data and share as it comes in and is it available. You have highlighted one of the key challenges in active comparator studies in influenza in particular, which is that you can see high titers, but actually because you are looking at a ratio, say for whatever reason, your active comparator does really well against one of the strains, that can impact your ability to non-inferiority statistically. And at the end of the day, the challenge is even more complicated as you look at older adults where, for instance, the influenza B strains are not a big driver of efficacy or disease. And so we will look at all of that as will regulators. I would note that it is well precedented. In fact, many of the currently approved influenza vaccines have, in the past, missed on non-inferiority for – and influenza B strain end point here or there and still have received full approval or accelerated approvals. And the reason for that is, as we have said sort throughout, that at the end of the day, influenza B is not a primary driver of concern and it is known to be among the different strains of influenza in the virus in the vaccines of lower import for disease in older adults. In fact, one of the four strains, there have been active debate about the B/Yamagata strain as to whether or not it’s gone extinct, and even should be removed from quadrivalent vaccines in many of the recent WHO and other debates. And so influenza B is a well-trodden path for many of these vaccines as well as now for mRNA-1010, where there is differential performance, and ultimately, there is precedent for moving forward where you do not technically need non-inferiority on immunity your sales [ph] conversion endpoints and still moving forward because of the lower concern about that disease in older adults. So, we will look at that data. We will develop our strategy. We will obviously engage with regulators with that data, and ultimately determine a path forward. The most important thing for us, though, in the near terms continuing with the efficacy study trying to establish whether or not we have non-inferior or even superior efficacy for mRNA-1010 in its current form against influenza A, which is really where we think payers and public health officials will have the most attention because it is prevention of that disease, not the immunogenicity endpoint, prevention of influenza-like illness, hospitalizations, that is the primary objective of the vaccine and that’s where we are focusing our attention right now.
Ellie Merle:
Great. Thanks for the color.
Operator:
[Operator Instructions] Our next question comes from Joseph Stringer with Needham. Your line is open.
Joseph Stringer:
Hi. Good morning. Thanks for taking my questions. Two from us, the first one on 4157-KEYTRUDA combo program. Just curious, if you could give us a little bit more color on how we should think about the cadence of the additional trial starts? Is it something that will be a more stepwise and measured approach, or should we expect sort of a full force kind of multiple trial starts approach? And then secondly, on rare disease, outside of your MMA, GSD and PA program, what is the next rare disease program that we can expect to enter the clinic? Thank you.
Stephen Hoge:
Thank you for those questions. So, on 4157, I think I will just reiterate our prior guidance, I don’t want to expand it, to say that we are trying to move into those pivotal studies, they are Phase 3 confirmatory studies for melanoma and non-small cell lung cancer, both adjuvant settings, this year. And we – you can look at the history of Merck and their ability to execute those studies, enroll quite quickly and now working together with us, we hope to be able to at least do that well. And we will look to enroll those at least as quickly as other confirmatory Phase 3 studies and similar populations have been run generally. But I don’t think we are going to provide more specific guidance on enrollment time horizon except to say we want to go as fast as possible. I will also clarify, too that from an accelerated approval perspective, if that pathway were to become available based on the current Phase 2 data, which again is subject to future conversations with regulators, that we would want to have started those confirmatory studies. We wouldn’t have needed to complete enrollment, but definitely, we want to be demonstrating that we are moving forward in those confirmatory studies as quickly as possible. And so we have double impetus. We are moving fast in enrolling them in the near-term. Now, in the rare disease space, programs moving out of pre-clinical and clinical. The first I would say is we do have a clinical program for MMA, which you referenced, but that MMA program is another place where we expect to see additional data following on, hopefully, the continued strong performance of the propionic acidemia PA program. And then in the pre-clinical development space, we have programs against OTC and PKU, so Phenylketonuria and a urea cycle disorder OTC, and those are both programs that we would hope to move into clinical testing in short order. We haven’t specifically guided on the timing of that yet.
Joseph Stringer:
Many thanks for taking our questions.
Operator:
Ladies and gentlemen, this does conclude the question-and-answer portion of today’s call. I would like to turn the call back over to Stephane for any closing remarks.
Stephane Bancel:
Thank you very much everybody for joining us and for the many thoughtful questions. We look forward to hosting you for Vaccine Day on April 11. It will be live in Boston, for those of you who can join us, and also of course virtual. Have a great day. Thank you.
Operator:
Ladies and gentlemen, this does conclude today’s presentation. You may now disconnect and have a wonderful day.
Operator:
Good morning. My name is Kevin. And welcome to Moderna's Third Quarter 2022 Earnings Call. [Operator instructions] Please be advised that this call is being recorded. At this time, I'd like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's third quarter 2022 financial results and business update. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Arpa Garay, our Chief Commercial Officer; and Jamey Mock, our Chief Financial Officer. After prepared remarks we will take your questions through 9:15 a.m. this morning. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I will turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon everyone. Welcome to our Q3 2022 Conference Call. Today I will start with a quick business review of a quarter before Stephen reviews or clinical programs. Arpa will then take you through commercial dynamic and Jamey will present financials. I will then come back to close before we take your questions. In the quarter we reported $3.4 billion in revenues. We reported net income of $1 billion and cash investments totaling $17 billion. We now expect deliveries under advanced purchase agreements in the range of $18 billion to $19 billion in 2022, due to delay deliveries from our fill-finish contract manufacturers resulting in $2 billion to $3 billion of revenue deferrals into 2023. In Q3, we had to deal with lots of complexity, launching two products, mRNA-1273.214 and mRNA-1273.214 at the same time, and also moving products from 10 dose vials to 5 dose vials. Q3 was actually our greater delivery amount in terms of vial produced and was up about 20% the average of previous three quarters. Arpa will share more thoughts on 2023 in the moment. In the first quarter, we repurchased over seven million shares. The $3 billion share repurchase program we announced in February, 2022 was completed. Since the start of our first share repurchase program in 2021, through the end of the first quarter, we have repurchased more than 23 million shares. Our first share repurchased program announced in August, 2022 for an additional $3 billion in repurchase is ongoing. Let me now review the pipeline highlights and advances since our last update. I am very pleased with the important progress the Moderna team has made on advancing the pipeline closer to product launches and at the same time equating the breadth of a pipeline. We've continue to progress with COVID booster programs and have received for addition around the world for both COVID mRNA-1273.214 program, which targets Omicron BA.1 and 1273.224 which targets BA.4/BA.5. We have, as you know, two respiratory vaccines in Phase 3 trials as we speak that continue to progress quickly. For flu vaccine our IND in September, we announced the first Phase 3 immunogenicity study was fully enrolled. We expect now data in the first quarter of 2023. As a reminder, we plan to pursue an accelerated approval pathway for seasonal flu vaccine. We also started a Phase 3 efficacy study with our flu vaccine and that trial is enrolling quickly. For Phase 3 RSV vaccine we are on track for data without this winter season. We were pleased to announce that Merck exercised the option to develop and commercialize our personalized cancer vaccine, mRNA-4157, paying Moderna $250 million in Q4. We and Merck with share costs and profit fifty-fifty for this program moving forward. We continue to expect data from our Phase 2 study for PCV in Q4 this year. In rare diseases we are pleased to share at our R&D Day that we saw encouraging early sign of clinical benefits above PA and GSD1a and announce a new development candidate for OTC. On Slide 6, you see our usual snapshot of Moderna in November, 2022 showing the breadth of the pipeline with now 48 programs in development across vaccines and therapeutics. The company continues to grow and we have now more than 3,700 Moderna team members. Last week we were very pleased to announce as a top employer by Science for the eighth consecutive year. We have now 15 commercial subsidiaries globally and a strong balance sheet of $17 billion to fund our Company growth. With this, I will now turn it to Stephen to review the pipeline. Stephen?
Stephen Hoge:
Thank you, Stéphane. Good morning or good afternoon everyone. This morning I'll review our clinical progress. We've launched two vaccine boosters for the current fall winter season to meet different market demands and have received authorizations or approvals worldwide for these vaccines. We previously shared that mRNA-1273.214, which targets Omicron BA.1 induce significantly higher titers than 1273 against the BA1 and BA4/5 sub-lineages in the clinical trial. mRNA-1273.214 is now authorized in United Kingdom, Switzerland, Canada, Australia, European Union, Japan, and other countries. For mRNA-1273.222, which targets the Omicron BA.4 BA.5 variants, the Phase 2/3 study is ongoing and we expect data later this quarter. mRNA-1273.222 is authorized in the United States and now also in the United Kingdom, Switzerland, Australia, Canada, the European Union, Japan, and other countries. Now moving to Slide 9. I'll review our respiratory vaccines pipeline. I will cover the Phase 3 studies in detail in the next slide. Here I want to highlight the progress in the earlier stage studies with our respiratory vaccines. mRNA-1020, mRNA-1030 for seasonal influenza is in a Phase 1/2 study and is now fully enrolled. mRNA-1345, for RSV, is in the pediatric population, is now fully enrolled in our Phase 1 study. Moving now to our combination respiratory pipeline where we have made meaningful progress mRNA-1073, our combination vaccine for COVID and flu is in Phase 1/2, and is fully enrolled. I'm very pleased to announce that our combination COVID, flu and RSV vaccine or mRNA-1230 has also started enrolling in its Phase 1/2 study. We announced a new development candidate, mRNA-1045. This one targeting combination of RSV and influenza and that has started in its Phase 1/2 study. And lastly, in our combination vaccine pipeline, we also have a pediatric vaccine covering hMPV and PIV3. That study is ongoing in a fully enrolled Phase 1B study. Finally, our endemic human coronavirus vaccine is in pre-clinical development along with our pediatric RSV hMPV combination vaccine. Now the review our Phase 3 flu and RSV programs on Slide 10. For flu, our Phase 3 immunogenicity study in the southern hemisphere is fully enrolled with 6,000 participants with a data readout expected in the first quarter of 2023. As we've previously noted, regulators have indicated support for an accelerated approval pathway for our seasonal flu vaccine candidate pending the results from this study. We've also started enrolling our Phase 3 efficacy study in the northern hemisphere and have now enrolled more than 10,000 participants. Timing of this confirmatory Phase 3 efficacy readout will be driven by flu case accruals in the study and could come as early as this winter. Looking to RSV, our pivotal Phase 3 efficacy study in older adults has now enrolled more than 35,000 participants. As we previously mentioned, our primary endpoints in this study, are safety and vaccine efficacy. Timing of the Phase 3 efficacy readout will be driven by RSV case accruals in that study. As we're now in the midst of a very strong RSV season, we continue to expect that the result will be available this winter season. Moving on to our latent and public health vaccine portfolio, our CMV vaccine is ongoing in a Phase 3 study. Our EBV vaccine to prevent infectious mononucleosis is in a Phase 1 study, while our EBV vaccine to prevent longer term sequela, such as cancer and multiple sclerosis is in preclinical. We have two HIV Phase 1 trials ongoing and our HSV and VZV vaccines are ongoing preclinical studies. Finally, our public health vaccine for Zika is ongoing in a Phase 2 trial, and our Nipah vaccine is ongoing in a Phase 1 study. Now let's take a look at our therapeutics pipeline on Slide 12. First, I want to note that AstraZeneca notified us that after a portfolio review, they are returning the rights to the IL-12 program to us. They are concluding the Phase 1 study and we will then evaluate next steps for the program for ourselves. Second, we're excited that our checkpoint vaccine has started dosing its first patients in a Phase 1 study. And finally, our partner Vertex expects to submit an IND for our mRNA cystic fibrosis program by the end of this year. We recently shared updates on our personalized cancer vaccine PA and GSD1A programs at our latest R&D Day. And I will talk to those in more detail in the upcoming slides. Now recall at R&D Day that we shared data from two rare disease programs. The first is in our propionic acidemia program, which is a multi-dose study, and as of September, we had accrued six patient years of experience on the drug and administered well over 100 doses. It is generally been well-tolerated, which is encouraging. We've also seen an encouraging trend in the reduction of biomarkers and been observing a numerical decrease in the frequency of metabolic decompensation events, which is also really encouraging given the severity of these events for proponic acidemia patients. In the GSD1A program, we shared an early set of data from the first two patients in the first cohort. The study is a single-ascending dose study that runs a fasting challenge in a controlled and safe environment in patients with GSD1A, who are unable to normally fast or go without food for long periods of time without becoming hypoglycemic. Both patients in the first cohort have demonstrated that m mRNA-3745 was well tolerated and showed an extension of fast duration and normalization of key biomarkers including glucose, which we think is very encouraging signal of activity. To close, I wanted to remind everyone that we expect our Phase 2 results from our personalized cancer therapeutic by the end of the year. The randomized study comparing PCV plus KEYTRUDA versus KEYTRUDA alone enrolled approximately 150 resected melanoma patients in a high risk of recurrence. The primary endpoint is to prevent recurrence-free survival. As Stéphane mentioned earlier, our partner Merck exercised the option to jointly develop and commercialize mRNA-4157 and we look forward to sharing that data this quarter. With that, I will hand it over to Arpa.
Arpa Garay:
Thank you, Stephen. And good day to everyone. I will start with a review of sales on Slide 16. Sales to the U.S. were $1 billion in the third quarter and were mainly from deliveries of mRNA-1273.222 our bivalent booster targeting Omicron BA.4 BA.5. Sales to Europe of mRNA-1273.214 were also $1 billion and the rest of the world totaled $1.1 billion. Through the first three quarters of the year, U.S. sales were $3.4 billion; sales to Europe were $4.5 billion and sales to the rest of the world accounted for $5.7 billion. In both the three and nine month periods, we saw geographical diversification of sales across these key regions. We recognized there are questions regarding the 2023 market and long-term COVID booster potential and we wanted to walk you through how we are thinking about it. As we transition into an endemic market, there are important factors we considered in our commercial outlook. These include the ongoing medical need for COVID boosters and the potential size of the annual booster market. We use the seasonal flu market to frame the opportunity and I will take you through our thinking on that in the next couple of slides. I will also detail some of the factors underlying the transition to the commercial market in the U.S. in 2023. I will then wrap up the COVID section with an overview of our currently signed contracts and our outlook for additional expected contracts in 2023. Finally, I'll close with a reminder what's coming in 2023 from our respiratory vaccine franchise, a potential launch timings that we shared recently at our R&D Day in September. So starting with the medical need for COVID boosters, the chart on Slide 18 compares hospitalizations and deaths associated with the seasonal flu and COVID over a relevant time period. For flu, we looked back at the past 10 influenza seasons in the U.S. before COVID-related interventions disrupted the typical flu season. And for COVID, we looked at hospitalizations and deaths from October 2021 to September 2022, which covers the period of widespread immunity against SARS-CoV-2 and its variants either through vaccination or infection. We believe this period is a close proxy to what population immunity to COVID could be in an endemic season. As you can see here on the chart, hospitalizations due to COVID during the past 12 months are three times higher than the 2017, 2018 flu season, which represents a year with the highest medical burden within the 10-year period included in the analysis. And the deaths due to COVID are even higher coming in at a rate of seven times higher than the same severe flu season. This comparison shows the higher medical burden of COVID in a period that we believe more closely approximates an endemic season and underscores the need for boosters in the endemic phase. It also helps frame the potential endemic annual COVID market, which I want to take you through on the next slide. To size the endemic COVID booster market we start with the flu market volumes. Volume in the annual flu market is approximately 500 million to 600 million doses around the world. The size of the market is highly dependent on the number of doses as well as global price as shown in the sensitivity table on the left hand side of the slide. The prices shown here are purely illustrative to demonstrate a range of outcomes on global pricing. Ultimately, global price for COVID boosters will reflect sales mix and differential pricing across markets. Our endemic pricing will be focused on the value that the vaccine brings to healthcare systems around the world. As we move to an endemic market in 2023 key factors that will impact our volume in the year include the medical need, ongoing viral evolution, recommendations from public health authorities and consumer motivation to vaccinate. So while we believe that volumes in the global endemic COVID market should approximate to at least 600 million doses over time, we believe it is too early to reliably predict the variables impacting the volume of doses in 2023. Turning to Slide 20, and specifically addressing the transition to the commercial market in the U.S., we anticipate a more fragmented customer base including private payers, health plans, pharmacy chains, individual pharmacies and physician offices. We also anticipate reduced predictability in orders, seasonality of deliveries similar to flu vaccine deliveries, a shift to full distribution costs assumed by Moderna, as well as other major factors in the U.S. such as shifting to a single dose presentation and continued innovation and product differentiation. Moving on to Slide 21, we summarize the contracts that have been signed so far for 2023. We have advanced purchase agreements for the United Kingdom, Canada, Switzerland, Taiwan and Kuwait totaling $2.5 billion. Additionally, we expect deferrals of $2 billion to $3 billion from 2022. These deferrals are from the countries listed on the slide. Together, these advanced purchase agreements and deferrals total $4.5 billion to $5.5 billion in sales for 2023. Finally, and very importantly we have a number of countries where we expect additional 2023 contracts to be signed. We are working to secure 2023 orders in the U.S., EU, Japan, Australia, Asia, Latin America, and COVAX. And we expect to have visibility into these orders as contracting seasons begins later this quarter and continuing into next year. So to summarize the COVID outlook on Slide 22, with the continued higher medical burden of COVID relative to flu, we expect the endemic COVID vaccine market could be as large or larger than flu market volumes over time. The U.S. market is transitioning to a commercial market and we believe we are well-positioned to serve its evolving needs. And finally, we have established a base of confirmed contracts of 4.5 billion to 5.5 billion in 2023, to which we anticipate adding a number of important orders in the months ahead from key markets including the U.S., EU, Japan, and others. Now, before I close, I'm excited to review the formation of our respiratory vaccine franchise that we highlighted during our R&D Day in September. As early as next year we could be expanding into flu and RSV. And as we look at flu and RSV combined with COVID, they lead to more deaths annually than Alzheimer's, stroke or diabetes in developed markets. As Steven highlighted depending on case accruals and vaccine efficacy, we could see data from our flu and RSV Phase 3 trials this winter. We are actively preparing for the commercial launches for these additional respiratory vaccines and we'll leverage a COVID commercial infrastructure that we are currently building. With that I'd like to turn it over to Jamie.
Jamey Mock:
Thank you, Arpa, and hello everyone. It's a pleasure to be here with you today. After two months at Moderna I'm even more excited about our company's future and the role we are playing in bringing in new generation of medicines to patients. For those of you whom I haven't had the pleasure of meeting yet, I look forward to working with you in the month and years ahead. Today I will start by providing additional color on our third quarter results and capital allocation priorities and finish with the view on the key drivers on our remaining 2022 financial performance. Turning now to Slide 25, total product sales in the quarter $3.1 billion decreased 35% year-over-year. The decrease was driven by lower sales volumes due to the timing of market authorizations for our updated COVID-19 booster vaccines, and the related manufacturing ramp-up with our CMO partners. As a reminder, we received the marketing authorization for the U.S. on August 31st, for the European Union on September 2nd and Japan on September 12th. We anticipate that product sales will be higher in the fourth quarter of 2022 than in the third quarter as we continue to deliver against our supply contracts for booster vaccines. Cost of sales was 35% of product sales compared to 15% of product sales last year. This includes a charge of $333 million for inventory writedowns related to excess and obsolete COVID-19 products and expense for unutilized manufacturing capacity of $209 million, and a loss on firm purchase commitments and related cancellation charges of $102 million. These charges are driven by a shift in product demand to our Omicron targeting COVID-19 bivalent boosters and cost associated with surplus production capacity. Research and development expenses were $820 million, an increase of 57% versus the prior year. The increase in R&D spend continues to be driven by our increasing and maturing pipeline including Phase 3 studies for RSV, flu, CMV and COVID boosters. Selling, general and administration expenses of $278 million increased by 65% year-over-year. The growth in spending was driven by continued investments in personnel and outside services in support of the accelerated commercial and overall company build-out. The effective tax rate of 14% compared to 6% last year. As a reminder, we had a net operating loss carry forward of $2.3 billion at the end of 2020, which resulted in a non-recurring benefit to the reported tax rate in 2021. After tax net income decreased by 69% to $1 billion, diluted EPS in Q3 2022 decreased by 67% to $2.53. As a result of our share buyback activities, the diluted weighted average share count reduced by 22 million shares to 412 million shares as of the end of Q3 2022, compared to 434 million shares the prior year. Turning now to the year-to-date financial results on Slide 26. Total product sales for the first nine months of 2022 were $13.6 billion, an increase of 26% year-over-year. The growth was mainly attributable to a favorable customer mix and higher manufacturing capacity to fulfill the customer demand. For the first half of 2022 compared to the first half of 2021, cost of sales was 26% of product sales compared to 16% of product sales last year. The increase was driven by substantial demand reduction from COVAX. As mentioned in our Q2 earnings call, a shift in demand to our Omicron targeting probate COVID-19 bivalent boosters and cost associated with surplus production capacity. After tax net income was $6.9 billion, a decrease of 6% versus prior year. The decrease in net income was primarily due to higher cost of sales, higher other operating expenses and a higher tax rate. Diluted EPS decreased by 3% to $16.46. Turning to cash and cash deposits on Slide 27, we ended Q3 2022 with cash and investments of $17 billion compared to $18.1 billion at the end of the second quarter. The decrease reflects the share buyback in Q3 of $1 billion and a federal tax payment of $0.8 billion. The ending balance of cash deposits for future product supply at the end of the quarter was $3.8 billion. Now turning to Slide 28, our capital allocation priorities remain unchanged. Our top investment priorities has been and will continue to be reinvesting in the base business across multiple areas. R&D spending was $2.1 billion in the year to-date September period, a 55% year-over-year increase. We continue to be excited about our mRNA platform and now have 48 development programs with multiple ongoing studies in now on Phase 3. We increased our year-to-date capital expenditures by 88% year-over-year to $308 million as we expand our manufacturing footprint. We also continue to invest in our commercial and digital capabilities as well as the overall company build out. Our second investment priority is to seek attractive external investment and collaboration opportunities to further expand the reach of Moderna's technologies and capabilities. We are considering attractive opportunities that enable and complement our platform and take a disciplined approach in evaluating potential outside investments. We are in multiple active discussions regarding additional external collaboration opportunities. Then after evaluating internal and external investment opportunities, we then assess additional uses of cash. In the third quarter of 2022 we repurchased 7 million shares for $1 billion. Year-to-date through September 30th we repurchased 20 million shares for $2.9 billion. In October we completed the $3 billion authorization approved in February 2022 and began to utilize the additional $3 billion August authorization. Now it turn to our 2022 updated financial framework on Slide 29. We now expect delivery in 2022 again signed advanced purchase agreements of $18 billion to $19 billion, reflecting deferrals of $2 billion to $3 billion into 2023 due to short-term supply chain supply chain constraints. This total includes expected negative foreign exchange impacts compared to the contract value at signing, which we estimate to be approximately 1.5% of sales for the full year 2022. Assuming current exchange rates remain through the year end. Our total cost of sales includes the cost of goods manufactured, third-party royalties as well as logistics and warehousing costs. We now expect our full year 2022 reported cost of sales to be in the 26 to 28 percentage range, driven by the previously mentioned cost incurred year-to-date, a range on our Q4 volume and potentially further charges due to product updates. For R&D and SG&A we continue to expect full year expenses to be approximately $4 billion driven by our maturing development portfolio and the global scale-up of our company. Based on current tax laws we continue to expect our 2022 effective tax rates to be in the low-to-mid teens as a result of benefits from the foreign derived and tangible income driven by our international business mix and stock based compensation deduction. Finally, regarding capital expenditures, we now expect capital expenditures to be approximately $0.5 billion, slightly below the previous range of $0.6 to $0.8 billion driven by the timing of project completion. This concludes my remarks concerning the financial performance, and I will turn the call back over to Stéphane.
Stéphane Bancel:
Thank you Jamey, Arpa and Steven for this update. Before opening to Q&A, I just want to review some of the key priorities as we close out this year and look into 2023. For the remainder of a year we'll continue to focus on delivering our updated Omicron boosters and drive 2022 sales. We are already working on 2023 sales contract; in Europe, Japan and Europe from afar around the world. Our partner team are also setting up the U.S. team and scaling it so that we can go private in the U.S. market in 2023. We continue to execute on our pipeline. We look forward to be able to present when we have it the Phase 2 data for PCV we expected before the end of the year. The team is doing a great job to cause it to enroll our Phase 3 study for flu, RSV and TMV and I'm excited to see the advancement of our rare disease programs. Looking to next year Arpa and the team and the entire organization including manufacturing, is to bring for multiple vaccine launches in the year including commercial COVID market as well as potential for therapeutic programs to move very quickly in phase three. And finally, we hope that many of you can join us next week for first ESG Day on November 10th. The virtual event will be available from our webcast. With this we'll be happy to take your question. Operator?
Q - Salveen Richter:
Good morning, thanks for taking my questions. On pricing here for the COVID vaccine, Pfizer's guiding to price of 110 to 130 in the private market. Is this in line with what your discussions thus far suggested, and when do you think the private market will emerge next year? And then a second question here; you were talking about some of the cost burden for next year with distribution costs and building out the commercial infrastructure around flu, RSV and trials. Could you just give us some directional color on the OpEx situation as we look – to look forward?
Arpa Garay:
So I can take the pricing question. So first I'm not in a position to comment on competitor pricing, but as we think about our pricing as we evolve from a pandemic setting to an endemic setting, the real focus for us is on ensuring that our vaccines are priced based on the value that they provide to the healthcare system and reflect the cost effectiveness guidelines that are set by public health authorities around the world. So for here in the U.S. that would be ACIP. I think it's important to note that some of the pricing guidance that has been released in the past is really at a growth level and we do anticipate some discounting across different channels. Additionally here in the United States as we evolve into the commercial setting, it's also important to remember that for all ACIP recommended vaccines there is a zero out of pocket cost for consumers. So from a consumer access perspective, we do expect that the pricing will not be a barrier to uptake.
Jamey Mock:
And Salveen, maybe I'll take the cost part of that. So yes, our cost will change in endemic mode. Number one, there are presentation preferences moving more to prefilled syringe and single dose files that I think will be different year-over-year. We have to continue to invest in bivalent vaccines as well as Moderna will now pick-up the distribution costs moving forward, particularly in the United States. And so yes, our cost profile will change and we'll come out and update in terms of what that means at a later date.
Salveen Richter:
Thank you.
Operator:
One moment for our next question. Our next question comes from Matthew Harrison with Morgan Stanley. Your line is open.
Matthew Harrison:
Great. Good morning. Thanks for taking the question. I was hoping to ask on PCV; just given that this is a Phase 2 study that's proof of concept a p-value grayer than 0.05 could be considered a success in this study. So maybe you could just help us think about how you're thinking about success in the study and then just given that it's an open label study, how much data is available to you internally? Thanks.
Stéphane Bancel:
Great. Thanks Matthew for the question. So as you said it's a Phase 2 study, but it's actually a quite sizable one. Its 150 patients and they were randomized two-to-one, so 100 patients have received the combination therapy in 50. The standard of care, which in this case is KEYTRUDA, that's actually quite a sizeable sample and does allow for us to look at efficacy. Now it is a Phase 2 study and so we didn't pre-specify a statistical threshold that we'd want to hit, but we are looking at hazard ratios across that and we will get p-values. I won't comment on a specific p-value or hazard ratio at this time. It's a premature to do so, but obviously we are looking against standard of care and demonstrate a significant benefit over that standard of care. It's just important to note that the study wasn't powered for that at 150 participants. And so we will be looking to that hazard ratio and the p-value as indicative. And then depending upon the strength of that result, if it is in fact, as you said a p-value less than 0.05 and there's a very strong result we will then make our subsequent decisions about how to proceed forward with development. Obviously, the stronger the benefit in terms of hazard ratio and the lower the p-value, the more we'll want to move very quickly towards advancing that program.
Operator:
Thank you. One moment for our next question. Our next question comes from Edward Tenthoff with Piper Sandler. Your line is open.
Edward Tenthoff:
Great. Thank you very much and congrats on all the progress. Appreciate all the detail on sort of the outlook both for the COVID market and as we move forward. Questions back to my favorite topic, the orphan diseases with this proof of concept I assume [ph] from the first programs, is there a desire to be expanding the pipeline similar to what you did with vaccines upon proof of concept with COVID? Thank you very much.
Stéphane Bancel:
Thanks Ed for the question. And so I think the short version is absolutely. In any of our modalities whether they're cancer vaccines or our infectious disease vaccines or now our orphan disease where we believe we've achieved a technological proof of concept. Where we've achieved what we wanted to in patients, we look to rapidly expand the number of diseases and implications that we can bring forward that technology in. And in the case of orphan rare diseases as we shared, we are extremely encouraged by the data across two different diseases using two different medicines that now suggest we have obviously acceptable safety and tolerability profile, which is of primary importance. But more importantly for efficacy we're starting to see really encouraging results in terms of biomarkers or even from the dynamic readout, a potential of benefit for patients. And so given the strength of those two pre prior results, we've actually moved quickly to expand our pipeline. We have a number of other programs that are already publicly disclosed and moving forward in clinical trials. Some including MMA are already in clinical studies. And as I mentioned at our R&D day we're looking to substantially expand that pipeline of programs. And so we announced the OTC program as one instance of that just a month ago but you can expect that we'll be adding substantially to that. Our goal will be to more than double that pipeline in the years ahead as we expand our investments in rare diseases on the back of that de-risk clinical data.
Edward Tenthoff:
Great. Excellent. Thank you, Stéphane.
Operator:
One moment for our next question. Our next question comes from Michael Yee with Jefferies. Your line is open.
Michael Yee:
Hi, good morning. Thanks for the questions. Maybe a question for Stéphane. In RSV we've had two players read out results; I think one is 80% plus or minus 166%, plus or minus, depending on the endpoints. So I wanted to ask you how you view the bar in terms of being competitive and where there is some differentiation opportunity for RSV vaccine that could read out soon. So talk a little bit about that? And then just as a follow-up to PCV, again just to clarify Merck had opted in; can you comment on, I guess the idea of the timing just before the data whether they had sufficient information or just talk a little bit about the implications of that opt in [ph] just before the data? Thank you.
Stéphane Bancel:
Good. Thank you for the question, Michael. So first on RSV, obviously we're incredibly encouraged by the results that are been seen by other vaccines. Given our platform has previously demonstrated its potential in COVID specifically in respiratory vaccines, we think it bodes well for us in terms of that study. There's a bar that's been established in terms of severe disease as you referenced in the 80% range. We are looking at three-symptom or "severe disease". It's hard to compare between the studies. They're not conducted the exact same time and they don't always have exactly the same definition as you know. But we would absolutely hope and expect that the type of efficacy we're going to see against severe disease will be on par with, and I would even hope for better than what's been seen by others. It's certainly been the case with our platform technology compared to others in terms of COVID that we've been able to see those sorts of potential benefits. I'll also note that we looked at titers and we previously shared our titers as well as other companies have from their early clinical Phase 1 and Phase 2 results and we believe that the boosting of anti – neutralizing RSV titers against both RSV-A and RSV-B that we were achieving was on par. You could – you should – you could always argue perhaps better or worse but on par with what others had seen. So we're quite encouraged by that. And I think we're looking forward to the RSV efficacy results over this coming winter. Our bar for this is to be good or better than others have been. In terms of PCV our – in terms of PCV, Merck has opted in as you said, and I know Matthew asked the question as well. So it is an open label study as we previously disclosed and so we have been following events through that study. Those that have received the cancer vaccine in combination of KEYTRUDA versus those that have not. And as we've now passed the one-year mark for follow-up of the last patient to be randomized in that study, we now have at least a year of follow-up and then [indiscernible] many cases, two-plus years of follow-up across those two different cohorts. Those that receive combo and those receive standard care KEYTRUDA. That data was known to us and Merck, but it's important to note that's an ongoing un-cleaned and not primary analysis. And so while it is open label the correct thing to do at this point now is begin the closing process, get all of the scans, review all of the data associated with the clinical outcomes, make sure nothing was missed and in that cleaning process then finalize that database and conduct the primary analysis for the study, which is to evaluate the hazard ratio and statistical significance to that hazard ratio between the two arms. That's actually the process we're undergoing right now. And it's important that before we make a decision on whether that data is positive or negative and the strength of that positivity – how quickly we move forward per Matthew's question into whatever the next major development are that we conduct all of the right diligence on those data sets and that's the work that's ongoing right now. So while Merck made their opt-in decision, which was really more calendar and contract driven based on obviously having access to that open label information, the really important analysis – the primary analysis, the one on which we will base our decisions of what to do next as will regulators and others is the one that we're trying to conduct right now and is not yet completed, but we do expect that result in this quarter.
Michael Yee:
Thanks. Very helpful. Appreciate it.
Operator:
One moment for our next question. Our next question comes from Gena Wang with Barclays. Your line is open.
Gena Wang:
Thank you. Steve, maybe just follow what you comment on Merck, just want to confirm that Merck does see in the open label data for the recurring free survival rate? And then also wanted to confirm that the control arm, the PD1 model that will be in line with historical data that's in the low-70s that would be the rate? And also how would you share the data for the PCV Phase 2 data? Another question is regarding Slide 19, the global COVID market opportunity. Since we expect significant increase in price in the U.S. what is your expectation for the ex-U.S. price change across major market since you're giving like $20 to $40 price range?
Stéphane Bancel:
Gena, thanks for that question. So again, it was not a continuous data set. It was an open label study and so per our agreement with Merck they had the right to know what we know about the program at that point in which they were had to make their decision about whether to opt-in or not. And so of course what we did is we provided them the access to the data of the study at that point in time and as you all have noted they elected to opt-in to that program and we are encouraged by that decision on their part. They obviously do have a tremendous amount of experience and so in terms of the control arm you obviously have the Keynote-054 study, which is their prior registrational studies, they have experience of what to expect in a control arm. And I think you can infer whatever you'd like from their decision to just say that they believe it's worth opting into that program and proceeding to the primary analysis that we're conducting right now. We will obviously be able to compare the control arm, the 50 patients who just received KEYTRUDA as standard of care against the registrational studies that others have, and actually the many years of patient experience that companies like Merck and others have, just to be confident that in fact if we are seeing a difference between and we are seeing a hazard ratio difference, that it's not a difference in terms of that control arm, which will board well because again patients were randomized in this study. Now in terms of the data we'll haven't share, at this point all we'd expect to share this quarter once we've completed the analysis. The primary analysis on efficacy and the study is just the top line data in terms of PCV, which as I mentioned previously is looking at the hazard ratio and then a characterization statistically of that. In subsequent and appropriate for including meetings and otherwise we will look to then share the older data set over time.
Arpa Garay:
And then I can take the pricing question. Your question was primarily around ex-U.S. pricing and our expectations. A couple of factors are coming into play here. The first is the timing of where and how quickly ex-U.S. markets are going to be shifting from more of a central procurement pandemic setting to an endemic setting. So we are looking at how different regions and countries are going to be shifting back towards a more endemic or a commercial approach. When we get to that position, we will be again pricing the vaccine according to the value that it provides in different healthcare systems around the world. And again following established systems around cost effectiveness guidelines based on the country regulations and the public health authority guidance. The overall global average price we do anticipate will be largely driven by the regional mix. So it is hard to predict what that price will end up being, but we'll continue to share more as we see evolving demand as well as evolving pricing.
Gena Wang:
Thank you.
Operator:
One moment for our next question. Our next question comes from Tara Bancroft with Cowen. Your line is open.
Tara Bancroft:
Hi guys, thanks for taking the questions. So is it fair to say that the 2023 signed APAs of the $4.5 billion to $5.5 billion is the floor? And what minimum revenue do you think could be added from the geographies that you're expecting contracts from? And related to this then to what extent does it include sales in the key markets that were mentioned in the press release like for example, does it include the option from the latest U.S. agreement? Thanks.
Arpa Garay:
Thank you for the question. So the $4.5 billion to $5.5 billion is the floor sales that we anticipate in 2023 as we already have signed APAs as well as deferrals from 2022 into 2023. This number does not include options from the U.S. government and we do anticipate additional sales coming from key markets such as the US, EU, Japan, Australia as well as regional sales in Latin America, Asia-Pacific, Middle East and COVAX. So again to answer your question specifically, this is what we anticipate to be the floor, but still unknown in terms of the total opportunity as we evolve particularly in the U.S. market into a commercial setting.
Operator:
Thank you. One moment for our next question. Our next question comes from Jessica Fye J.P. Morgan. Your line is open.
Jessica Fye:
Hey guys, good morning. Thanks for taking my question. Are the short-term supply constraints mentioned is a reason for delivery delays resolved at this point? How does bivalent booster uptake so far this fall compare to your expectations? And as it relates to China's potential orders of Western mRNA vaccines, what's your level of optimism that that could come to fruition within some reasonable time timeline, say the next year or so? Thank you.
Stéphane Bancel:
Sure. So it’s Stéphane. I'm going to take the first question on supply and then I'll turn to Arpa for the commercial PCs. As I shared in my remarks, we actually had to deal with very complex third quarter from a manufacturing standpoint, not launching one product, but two doing this kind of in the work hard times as you are aware, we have FDA informed us at the end of June that they wanted [indiscernible] products for the U.S. and that product was available in pharmacy label at weekend. And the shift from 10 dose per vial per provide, which is what we sold with 1273. And for a first time selling five dose per vial, basically doubling the number of vials needed, the same number of doses. And so we've had quite a number of pain points with fill-finish manufacturers. We are working through a lot of those issues. A lot are solved, a lot are still being, as we speak. There are many lessons to be learned that we are working on to put robust fixes for the end of the year, so that we are in a much better place for the fall of 2023. Arpa?
Arpa Garay:
Sure. So the first question around vaccine uptake we are seeing some variability around the world in terms of vaccination rates with the best data thus far really coming from the U.S. market. As we look at 2022 vaccination uptake versus 2021 specifically for COVID boosters, we're actually tracking a similar pattern. And it's a little bit early to see what November, December, and the rest of the fall and winter season will look like. But the early rates in absolute terms, are fairly low. They are tracking the trends that we expected compared to last year. Around the world, we are seeing some markets with very high uptake of vaccination rates really driven by public health authorities. And in other markets we're still looking at sort of the dynamics that have played out this year with populations have recently gotten their fourth booster in the summertime and regulatory bodies recommending that they wait a few months before they get their fifth booster. So I think more to come as we continue to track around the world. But the early signs of uptake are encouraging. Your last question on China, we continue to look at the opportunities in China. Nothing new to report here as of now, but it is certainly a key market of interest for us commercially.
Jessica Fye:
Thank you.
Operator:
One moment for our next question. Our next question comes from Geoff Meacham with Bank of America. Your line is open.
Geoff Meacham:
Good morning guys. Yes, thanks for the question. Just had a couple. Just one to follow- up on your last comment. You mentioned booster adoption has been mixed depending on geographies. Is this something that Moderna expects to further invest in with regard to value of boosters, additional follow-up and studies? That's the first question. The second one, maybe more for Stéphane, the balance sheet remains pretty strong, you guys have done some buybacks, but we haven't seen sort of a cluster of acquisitions or any sort of real capital allocation yet. Where would M&A sort of fall in your priority list with uses of cash? And does that change as COVID continues to sort of wind down with regard to the revenue base 2023 over 2022 and maybe even more modest going forward. Thank you.
Arpa Garay:
So to the first question on if we are investing in a vaccination rates, as I shared earlier today, the medical need continues to be clear for COVID booster vaccination, especially as we compare it to the hospitalizations and deaths compared to flu. So what we're really doing is partnering with governments and public health agencies to share the data that we have in terms of the ongoing medical need at a country level, as well as the value of COVID booster vaccination in their populations. And it's this partnership with the public health authorities that's really driving increased urgency and action around vaccinations. So that's our approach as we believe public health authorities are in the best position to encourage a vaccination for their populations.
Stéphane Bancel:
Thanks, Arpa. And on the balance sheet side, I mean, our biggest strategy as Jamey noted in his remarks, isn't changed. As you know we are looking, and have already signed deals in terms of technology licenses and also looking at M&A. As we've said before, we stay focused on nucleic acid. We don't really think it's a good strategic use of our capital to buy small molecule asset or large molecule or cell therapy, or else we really want to stay nucleic acid. The BT team is very active as Jamey knows, and this team knows they are doing a lot of work where we're doing a lot of things. As I've said in the past, we will remain disciplined in term of understanding the risk either on the technology side, or on the biology side, understanding value. We're here to create value not to manage press release. But the team is very active. We are looking at diligence on a regular basis. But not everything that we look at comes out to be something we think we can create a lot of value with. But we will continue to look and I would not be surprised if we continue to add partnerships in the monsoon quarter to come.
Geoff Meacham:
Great. Thank you guys.
Operator:
One moment for our next question. Our next question comes from Eliana Merle with UBS. Your line as open.
Eliana Merle:
Hey guys, thanks so much for taking the question. Just another one on RSV. In terms of thinking about mRNA as a vaccine modality in the context of RSV as we move beyond COVID vaccines, I guess how are you thinking about the potential for mRNA and potential advantages relative to other more traditional vaccine modalities in the context of RSV? And I guess what we could learn from some of the upcoming readouts, both, I guess for the opportunity in RSV, but also what it could tell us about mRNA potential and vaccines relative to other modalities more broadly? Thanks.
Stéphane Bancel:
Great. Well, thank you for the question. So, first we have previously demonstrated with our platform in COVID and in fact in some recent publications, even in flu that we're we generally see a really broad based and balanced immune response. So we tend to see very high T-cell and cell mediated immunity. And I would argue some of the highest if not the always the highest antibody neutralizing titers. And both of those are really important as you start talking about older adults and respiratory infections, which is probably why we tended to see higher efficacy with the mRNA platform than other approaches in that high risk population, including in COVID, over the last couple of years. And so those features of the platform, the ability to generate really strong cell mediated immunity, boost T cells, which are important for preventing severe disease, but also pair that with achieving very high titers for a seasonal protection where again, you get high neutralizing titers, which provide a barrier to the amount of infection that you're going to get, which is really, really important for older adults because their cell-mediated responses, their innate immune system just isn't as strong, even with a great vaccine. And it’s that combination that we think defines our advantage from a platform perspective and why we're so excited about developing a portfolio of respiratory vaccines across all the leading killers in that space for older adults. And RSV just fits right into that. As we all know right now, there is a huge unmet need in that space. We are very pleased to have enrolled, fully enrolled that Phase 3 study. And we were actually quite pleased with the titers and cell-mediated immunity that we saw in early development, some of which we've presented publicly. So we're looking forward to that FC readout. In terms of other approaches, let's also just celebrate that recombinant protein, and viral vector and adjuvanted approaches, have all shown really exciting progress in the last year, year and a half across a range of companies. RSV is a huge unmet need. It will take many different approaches to have an impact there. And if anything, I think, the success of others gives us optimism that our approach is going to also be successful, but there are likely going to be many different solutions to this for older populations. We do believe that our platform had an advantage. But given the success of the other vaccines and really, I think, quite encouraging efficacy results, it's important to note that there are many good successful options out there. And it may not always be possible to differentiate between them, but we hope to be in that first class.
Eliana Merle:
Great. Thanks.
Operator:
One moment for our next question. Our next question comes from Joseph Stringer with Needham, your line is open.
Joseph Stringer:
Hi, good morning. Thanks for taking our questions. Just clarification on the deferrals from the 2022 contracts, that $2 billion to $3 billion, are those locked in signed APAs and they are just being deferred to 2023 or is there any optionality built into that? And then the second question is just broader based pipeline. You are expanding pipeline 48 programs, 35 clinical trials. But what I was curious if you could comment on the relative mix of the pipeline respiratory vaccines, immuno-oncology, rare disease, where it stands now and how you sort of see that evolving in that relative mix of the pipeline programs over time?
Stéphane Bancel:
So maybe I'll take the first one quickly. So yes, those are locked in advanced purchase agreements for 2023, and so they are just shifting to the right from 2022.
Stephen Hoge:
Great. And on the broader question of the pipeline, as you mentioned, we've got 48 programs now multiple Phase 3s ongoing with some we hope to be quite eminent readouts. As you look at it right now most of our late-stage pipeline is respiratory vaccines for sure. Right? We have four programs there. And we expect the flu and RSV data to come up quickly. And we will then move rapidly in respiratory into combination vaccines. And so we have three adult combination vaccines in clinical studies. We have two pediatric respiratory combination vaccines that are moving forward and that you should expect to expand into very quickly. We would hope registrational studies over the coming period which will allow us to then build out what we hope will be the best respiratory portfolio both from a monovalent vaccine perspective, but most importantly from combinations against the viruses that matter in the different populations that we really want to protect, particularly older adults and the very young. So respiratory will be a growth area for us for the near term. But we will start to see some diversification. Again, talking about the late stage, we already have a CMV Phase 3 program, which is up and running and enrolling, it has been. And you should expect us in our latent virus vaccines to add additional late-stage programs. We're quite passionate about Epstein–Barr virus, as you all know. And we expect to move that in. And so you will start to see some first diversification in terms of our latent virus portfolio, some that's already happened with CMV. The couple of things that will come quickly as well are we hope that the readout from our personalized cancer vaccine therapeutic vaccine program, and it's a really a therapeutic because we are preventing cancer from recurring, but we're intervening in somebody who has got that cancer already. And because of that, it really has a therapeutic profile to it as an intervention and that that therapeutic readout that we expect to have over the next month or so this quarter will trigger if it's successful moving into pivotal stage studies. And as we all know immuno-oncology is obviously a competitive space. It's a space where there's still substantial need to improve upon the current IO therapy, and there are many different histologies and types of cancer in which it will make sense to develop a program if we show a benefit. So that expansion could happen very quickly and will be the large diversification into oncology and the therapeutic context outside of the vaccine space. And then the last one is rare diseases, which we've spoken about. We have a couple of programs that have already started to show some very encouraging results, both pharmacology and potential clinical readouts. And as soon as we decide on doses for instance, the propionic academia program and get confident that we've got alignment with regulators on the path forward for that program, you should expect us to move into pivotal studies there as well. And then, as I said in answering your prior question, that will not just be a one-off for us once we really believe we've got a modality, we will be bringing forward many programs in succession in parallel there. And so as you look forward on how do we think about the diversification of the pipeline it’s a bigger question, it really is a question of time horizon. Right now it's a lot of respiratory and latent very quickly though we expect to be both expanding that respiratory and expanding or moving to therapeutics, particularly cancer and rare metabolic disease. And that could happen in quite short order based on upcoming clinical readouts.
Joseph Stringer:
Great. Thanks for taking our questions.
Operator:
One moment for our next question. Our next question comes from Mani Foroohar with SVB Securities. Your line is open.
Unidentified Analyst :
Hi, and good morning, this is [indiscernible] for Mani Foroohar. I just have a question related to volume to be expected. So I know that your volume production, you had mentioned that you would exclude 2023 based on because of the variability there, but I was wondering for 2023, how much of COVID volumes or I would say probably [indiscernible] do you anticipate to be driven by the APAs? And also as you move to our northern endemic model for locally vaccine, do you continue to anticipate APAs in the future? Thank you.
Arpa Garay:
Sure. Thank you for that question. As I outlined in terms of the volume expectations for next year outside of the signed APAs, we are looking at a number of variables that could impact the total volume. So in the long term, we do believe we should approach a 500 to 600 million volume comparable to the flu volumes that exist today. In 2023, there will be a range depending on the viral evolution, the ongoing medical need across different regions where we see public health authorities coming out with recommendations as well as just a broader consumer appetite to vaccinate. So the reason for the sensitivity on the volume that I shared earlier is we still have a number of variables as we're continuing to transition into an endemic stage. And so in the short term we don't have a clear picture of that total volume and what that could look like, but we do believe from a medical perspective, over time, we should be approaching at least the same volumes as the flu market.
Unidentified Analyst :
Thank you.
Operator:
And I'm not showing any further questions at this time. I like to turn the call back over to Stéphane Bancel for any closing remarks.
Stéphane Bancel:
Well, thank you everybody for joining the call today and for your thoughtful questions. We look forward to speaking to many hours to come, days to come. And also we’re welcoming you next week or our first ESG Day. Have a great day. Bye.
Operator:
Ladies and gentlemen, that concludes today's presentation. You may now disconnect and have a wonderful day.
Operator:
Good morning. My name is Kevin, and I will be your operator. Welcome to Moderna’s Second Quarter 2022 Earnings Call. At this time, all participants are in a listen-only mode. Following the formal remarks, we will open the call up for questions. [Operator Instructions] Please be advised that this call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone. And thank you for joining us on today’s call to discuss Moderna’s second quarter 2022 financial results and business update. You can access the press release issued this morning, as well as the slides that we will be reviewing by going to the Investors section of our website. On today’s call are Stephane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; Paul Burton, our Chief Medical Officer; and Arpa Garay, our Chief Commercial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor Provisions of the Private Securities Litigation Reform Act of 1995. Please see slide two of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I will turn the call over to Stephane.
Stephane Bancel:
Thank you, Lavina. Good morning, or good afternoon, everyone. Welcome to our Q2 2022 to conference call. Today, I will start with a quick business review, before Paul walks you for some new real world evidence data on Spikevax and then, Stephen, will review our clinical programs. Arpa will take you through commercial dynamics and David will present financials and we will come back to growth, just some thoughts, about where we are heading. In the quarter, we continued strong financial performance. We reported revenue of $4.7 billion and net income of $2.2 billion. We are pleased with the Q2 sales. The decreasing net income is driven by higher cost of sales in the quarter, mostly driven by our COVAX contract, David will cover that in the financials, a strong increase in R&D investment to fund our future, including the four Phase 3 programs and the higher tax rate in Q2 2021. For our share repurchase program, we reduced our share count by $9 million shares in Q2. Since the start of our first share repurchase program in 2021 through yesterday, we have repurchased around $18 million shares. We continue to have signed event purchase agreements for expected delivery in 2022 in the amount of approximately $21 billion. Based on our commercial momentum and our balance sheet, today we also announced that our Board of Directors has approved the new share buyback program for an additional $3 billion. Let me now review the business and pipeline update since our last review. I am pleased with the important progress that Moderna team has made on advancing the pipeline across the product launches and increasing the breadth of the pipeline. On the commercial side, we recently signed a new agreement with the U.S. Government for an initial 66 million doses for bivalent booster vaccine against Omicron. The agreement also include options to purchase an additional 234 million doses, bringing the total sales potentially up 400 million doses. The U.S. Government has already exercised option for an additional 4 million doses since the contract was signed and announced. So the current total is actually 70 million doses contracted for the fall of 2022 with the U.S. Government. In addition, both Canada and the U.K. had exercised options for their current agreements for additional deliveries in 2022. On the regulatory front, I am very happy to talk about authorized for pediatric and recent population in the U.S. We further see the thought with regulatory clinical teams at Moderna and the U.S. FDA now allows the protection of the youngest in the U.S. population as they head back to school. As Canada and Australia both authorized the use of our vaccine for children’s six months to five years old. Our research and clinical team continue the momentum in our pipeline. We are look for running Phase 3 development with COVID bivalent booster, flu, RSV and CMV trials ongoing. Importantly, our Phase 1 study with mRNA-1030, our flu vaccine targeting the traditional flu antigen, as well as been novel NA antigen is now fully enrolled. And our combination flu plus COVID vaccine is also fully enrolled for Phase 1/2. We look forward to these readouts on safety and immunogenicity for approvals. In properties, we now have an open IND with the checkpoint vaccine in oncology and we dose our first patients with our GSD1a rare disease program. Finally, we are very pleased to be on track and look forward to sharing data for our proof-of-concept studies this year for two of our therapeutic programs. The Phase 1/2 rare disease program propionic acidemia or PA and our Phase 2 data with our personalized cancer vaccine in combination to KEYTRUDA versus KEYTRUDA monotherapy. Slide six is our usual snapshot of Moderna in August 2022, showing the breadth of the pipeline with 46 programs in development across vaccine with four therapeutics. The company has strong foundation, we have 3,400 teams and growing, 11 commercial subsidiaries and the strong balance sheet with around $18 billion of cash to fuel our growth. I will now hand it over to Paul.
Paul Burton:
Thank you, Stephane, and good day, everyone. As the COVID-19 pandemic continues and the SARS-CoV-2 virus keeps undergoing significant evolutionary change, the subject top of mind for many people is boosting, when to get boosted and what to get boosted with. In the next few slides, I want to show you how the Moderna mRNA-1273 vaccine is performing clinically in the booster setting and how antibody levels are performing in the real world setting in individuals primed and boosted with mRNA vaccines. Here in this next slide, you see the rates of COVID-19 infection in over 3 million individuals in the Spanish National Registry. The first point these data make is boosting is highly effective, reducing the risk of infection. The second point, the authors make is that mRNA-1273 the Moderna vaccine is more effective than BNT162b2 and that the effect size increases over time. In the second example, I am showing you data from the U.S. VA system in over 450,000 individuals boosted with either mRNA-1273 shown here in green or BNT162b2 shown in red. Again, in terms of infection or hospitalization prevention, the authors conclude that mRNA-1273 is more effective than the alternate mRNA vaccine. Finally, here a data from over 3 million individuals in the NHS England database. Again, showing a clinically meaningful and statistically significant reduction in COVID-19 infection and hospitalization with mRNA-1273 boosting, compared to BNT162b2 during the Omicron periods of infection. I want to turn now to data from an innovative, decentralized, pragmatic real world study. We have recently conducted within the Moderna community collaborative platform with invitation of over 100,000 participants here in the United States. This study is called disCOVEries and it enrolled approximately 850 of these people into a real world assessment of antibody levels following primary vaccination and boosting with either Moderna-Moderna-Moderna that’s the MMM group or Pfizer-Pfizer-Pfizer, that’s the PPP group. Importantly, the study population has an average age of 44, approximately 60% of the participants of female and it’s ethnically very diverse. Participants were enrolled into the study, consented and received a home self-administered micro needle blood collection kit from your buyer. As you can see here, across an array of SARS-CoV-2 variants of concern higher and more durable antibody levels shown here on a logarithmic Y-axis scale were observed in those individuals primed and boosted with the Moderna vaccine in red, relative to the Pfizer vaccine in blue from eight weeks to 32 weeks following their first booster. Many of the comparisons were highly statistically significantly different. It’s true even for antibody levels against the Omicron BA.1 variant of concern, where an approximately 12-week duration advantage is seen in Moderna primed and boosted individuals compared to those receiving the Pfizer vaccine. So taken together, these data demonstrate the strong performance of the Moderna vaccine in a real world clinical boosting setting, which could be explained by higher enduring antibody levels against a wide variety of variance of concern. I will now turn the call over to Dr. Stephen Hoge to provide comments regarding the composition of the full COVID-19 booster vaccines, as well as other pipeline updates. Stephen?
Stephen Hoge:
Thank you, Paul. Good morning, or good afternoon, everyone. I will quickly review some of the recent updates around our Omicron containing COVID booster for the upcoming fall season and then review the rest of our pipeline before handing it to Arpa. We are advancing to bivalent candidates for the fall winter season of 2022 to meet different market demands, mRNA-1273.214 is our Omicron BA.1 containing bivalent vaccine. Clinical data previously presented from our Phase 2/3 study showed that 214 significantly increased neutralizing titers against BA.4, BA.5 compared to the currently authorized booster. Following regulatory discussions, we have submitted application for authorization for 214 in the EU, Australia, U.K., Canada, and Switzerland. We have also started a rolling submission in Japan. mRNA-1273.222 is our bivalent vaccine candidate that combines an Omicron containing vaccine based on the BA.4, BA.5 subvariant and our original mRNA-1273. The FDA has asked us to submit for authorization for 222, based on the demonstrated strength of our bivalent platform against variance of concern including data from our Phase 2/3 studies with 214 and 211 bivalent boosters. FDA has also advised manufacturers to initiate clinical trials with 222 as these data will serve useful as the pandemic continues to evolve. We expect to start these trials in August. Now to review our respiratory vaccine pipeline on slide 16, I just want overall -- Omicron containing COVID boosters. We were happy to announce that our flu vaccine mRNA-1010 launched an immunogenicity study in the Southern Hemisphere in June and we are also preparing to launch an efficacy study for mRNA-1010 this upcoming fall, which will either serve as a post-approval efficacy study or if we are not eligible for study approval we will support our application. Our next-generation flu vaccines, which had eight mRNA’s across HA and NA antigens, mRNA-1020 and mRNA-1030, also completed enrollment in their Phase 1.2 study. Our RSV program is ongoing including a Phase 3 in older adults and a Phase 1 in pediatrics. Importantly, we also started and concluded enrollment for the Phase 1-2 study of our COVID plus flu vaccine this quarter. We look forward to sharing that data when we have it. Our combination COVID flu RSV vaccine also expects to enter the clinic this year. Our endemic, human Coronavirus vaccine is in preclinical along with our pediatric RSV plus hMPV combination vaccine and the pediatric hMPV plus PIV3 vaccine Phase 1b is now fully enrolled. Moving onto our latest public health vaccine portfolio, our CMV vaccine is ongoing in a Phase 3. Our EBV vaccine to prevent infectious mononucleosis is in a Phase 1 study and our EBV vaccine to prevent long-term sequelae from the virus is in preclinical. We have two HIV Phase 1 trials and a Phase 2 Zika vaccine trial that are all ongoing. Our Phase -- our HSV and VVV vaccines are in preclinical. And finally we dosed the first participants in our Phase 1 study of our Nipah vaccine, which is being conducted in partnership with the NIH. And to close on our therapeutics pipeline on slide 18, Stephane already mentioned that we expect data from our proof-of-concept studies in our PCV NPA programs later this year. A few other important updates to highlight on this slide, first, we were happy to dose our first patient in our GSD1a trial and open an IND for our checkpoint cancer vaccine. Second, AstraZeneca notified us that after a portfolio review they are returning the VEGF program and we are currently evaluating the next steps for that asset. Finally, based on a review of early clinical data from our IL-2 program for autoimmune disease and the evolving competitive landscape, we have decided to discontinue further development of our program and we were moving from our pipeline. With that, I will hand it over to Arpa.
Arpa Garay:
Thank you, Stephen. Good morning and good afternoon to everyone. As the newest member of Moderna Executive Committee, I am excited to see the impact that we have had in COVID, as well as the commercial opportunities ahead for us in COVID, as well as future vaccines and therapeutics in the long run, leveraging our mRNA technology. Turning now to slide 20, where you will see the regional sales mix in second quarter and in the first half of 2022. In the quarter, North American sales were $1.6 billion. These sales were driven by 1273 booster deliveries to the United States as we continue to fulfill our first U.S. Government order with the vast majority now delivered through the end of second quarter this year. In the quarter, we also delivered primary series of the -- for the pediatric group five years and younger as the authorization for this group came through in this quarter. Sales to EMEA were $1.5 billion and sales to Asia-Pacific were $1.1 billion. As you can see across the first two quarters of the year, North American sales were $2.7 billion, sales to EMEA were $3.9 billion and sales to Asia-Pacific region were $3 billion. Of note, for both the three months and six month periods, we saw geographic diversification of sales across these key regions. This diversification is a reflection of how quickly we built out and executed within our global commercial organization. Now turning to slide 21, we are also happy to see that as we continue to scale globally around the world that Spikevax’s market share and the booster market defined as a third or fourth booster continues to show substantial market share. As Stephane has already mentioned, we see on slide 22, our new U.S. Government contracts for the fall of 2022. Specifically, the agreement initially was for 66 million doses of our bivalent COVID booster to be delivered in 2022. Earlier this week, the U.S. Government informed us that they exercised an option for 4 million doses for the pediatric age group. This brings the total contract value to approximately $1.8 billion. Our U.S. Government order includes options to purchase an additional 230 million doses, bringing the total to 300 million doses if all options are exercised. As you heard from Stephane earlier, after FDA guidance, the U.S. fall booster product will be the mRNA-1273.222, which consists of 1273 and the Omicron BA.4/5 subvariant. I want to spend some time on the evolution of the pandemic and the resulting impact on the commercial outlook. Early in the pandemic, we anticipated several factors including the virus evolution, population immunity at any given time and seasonal trends would result in a shift to the endemic setting and we have shared this illustrative graph before. These same factors have and will continue to emphasize the commercial outlook. With the emergence and then dominance of multiple variance at different time points, we have been able to develop booster to address these variants, which includes our bivalent boosters against the Omicron variant. We are currently advancing two bivalent booster candidates for the fall of 2022 based on different market needs. The two bivalent booster candidates we are advancing are the mRNA-1273.214 and mRNA-1273.222. Both contain 25 micrograms of the currently authorized booster 1273 and 25 micrograms of an Omicron subvariant either BA.1 or BA.4/5. Deliveries for these boosters will start in September of this year and will be more heavily weighted to the fourth quarter, as we ramp manufacturing and obtain regulatory authorizations around the world. And to close, as we look to 2023, we are prepared for a shift to the commercial market in the U.S. for COVID boosters, where the market will be more fragmented than it was during the pandemic where the U.S. Government with a sole purchaser of vaccines. The commercial organization has already engaged with commercial payers and the channel, both channel distributors, as well as key pharmacies in anticipation of the shift. Internationally, we expect public health authorities to remain key purchasers of vaccines, but we are also identifying markets where there may be a private commercial market as well. All in all, we are well positioned for the transition as we have invested in building our commercial infrastructure both in the U.S. and globally. With that, I will turn it over to David.
David Meline:
Okay. Thank you, Arpa. Today we are providing the analysis of actual 2022 second quarter results along with a view of key drivers of financial performance going forward. Overall, we continue to progress well and I am very pleased with our operational and commercial performance. Turning now to slide 25, starting with an overview of our financial performance in the second quarter. Total product sales for the second quarter of 2022 of $4.5 billion, increased by $334 million or 8% compared to the prior period. The total product sales growth in 2022 was primarily driven by higher average sale price of our COVID-19 vaccine due to changing customer mix. Total revenue was $4.7 billion for the second quarter of 2022, an increase of $395 million compared to Q2 of last year, driven by the increase of sales of our COVID-19 vaccine. Cost of sales was $1.4 billion or 30% of product sales in the second quarter of 2022, compared to 18% of product sales for the same period in 2021. This includes a charge of $499 million for inventory write downs related to excess and obsolete COVID-19 products, a loss on firm purchase commitments of $184 million and an expense for unutilized external manufacturing capacity of $131 million. These charges are driven by substantial reduction of our expected deliveries to COVAX as indicated as a potential variable impacting our Advanced Purchase Agreements in our last call and to a lesser extent by deferral of deliveries to other customers, particularly to the European Union in light of the expected upcoming launch of our updated bivalent vaccines. Research and development expenses were $710 million for the second quarter of 2022 and increased by $289 million or 69% compared to the year ago period. The increase in R&D spend continues to be driven by clinical trial expenses, particularly with our COVID-19 and RSV programs, as well as personnel related costs for expanding and maturing development portfolio. Selling, general and administrative expenses were $211 million for the second quarter of 2022, increased by $90 million or 74% compared to the year ago period. The growth in spending was driven by the commercialization of our COVID-19 vaccine globally with continued investments in personnel and outside services in support of the accelerated company build-out. The effective tax rate for the second quarter of 2022 was 11%, compared to 9% for the same period in 2021. Let me remind you of the fact that we had a net operating loss carry-forward of $2.3 billion at the end of 2020, which resulted in a non-recurring benefit to the reported tax rate last year. After-tax net income in Q2 2022 decreased by $583 million or 21% to $2.2 billion compared to the same period in 2021. The decrease was primarily due to higher cost of sales and other operating expenses in the current period. Diluted EPS in Q2 2022 decreased by $1.22 or 19%, the $5.24 a share, which is compared to the same period in 2021. Turning now to year-to-date financial results compared to the prior year on slide 26. Total product sales for the first six months of 2022 were $10.5 billion, increased by $4.5 billion or 76% compared to the prior year period. The total sales growth in 2022 was mainly attributable to our manufacturing capacity ramp up and to a smaller extent to favorable customer mix resulting in increased average selling price. Total revenue was $10.8 billion for the first six months of 2022, compared to $6.3 billion in the same period in 2021. The increase in total revenue was primarily driven by the increase of sales of our COVID-19 vaccine outside of the U.S. Cost of sales was $2.4 billion or 23% of product sales for the first six months of 2022. This compares to 16% of product sales in the prior year period on a reported basis or 19% adjusted for pre-launch inventory costs, which were expensed in 2020. The increase in cost of sales as a percent of product sales was mainly due to higher write-downs for excess and obsolete inventory, and expenses related to future purchase commitments and unutilized external manufacturing capacity, and to a lesser extent the lack of pre-launch inventory benefit that was realized in the first quarter of 2021. Research and development expenses were $1.3 billion for the first six months of 2022, an increase of $442 million or 54% compared to the prior year. The increase in R&D spend continues to be driven by clinical trial expenses, personnel related costs and outside services for expanding and maturing development portfolio, including the development of COVID-19 bivalent boosters. Selling, general and administrative expenses of $479 million for the first six months of 2022 increased by $281 million or 142% compared to the year ago period. The growth in spending was driven by the commercialization of our COVID-19 vaccine globally and support of the accelerated company build-out, including substantial investments in digital. Additionally, in Q1 2022, there was an initial upfront endowment of $50 million for the newly established Moderna foundation. The effective tax rate for the first six months was 13%, compared to 7% for the same period in 2021. The increase was primarily due to the benefit recorded in 2021 related to the release of the valuation allowance on the majority of our deferred tax assets. After-tax net income increased by $1.9 billion or 46%, the $5.9 billion for the first six months of 2022 compared to the same period in 2021. The increase in net income was driven by the growth of our product sales. Diluted EPS for the first six months from 2022 increased by $4.55 or 49% to $13.85 compared to the same period in 2021. Turning now to cash and cash deposits on slide 27, we ended Q2 2022 with cash and investments of $18.1 billion, compared to $19.3 billion at the end of Q1 of this year. The decrease reflects the share repurchase activities in Q2 of $1.3 billion. The ending balance of cash deposits for future product supply was $4.1 billion, compared to $5.3 billion at the end of the previous quarter. The reduction quarter-over-quarter is driven by product deliveries against customer deposits. Now turning to slide 28, our capital allocation priorities remain unchanged. Our top investment priority has been and will continue to be reinvesting in the base business across multiple areas. As previously stated, R&D spending was $1.3 billion in the first half of 2022, a 54% increase on a year-over-year basis. We remain on track with our full year R&D forecast of $2.5 to $3 billion. Our second investment priority is to seek attractive external investment and collaboration opportunities to further expand the reach of Moderna’s technology and capabilities. We are considering attractive opportunities that enable and complement our platform and take a disciplined approach in evaluating potential outside investments. We are in multiple active discussions regarding additional external collaboration opportunities. After evaluating internal and external investment opportunities, we then assess additional uses of cash. In the second quarter of 2022, we repurchased 9 million shares for $1.3 billion. Since inception of our repurchase activities last year and up until August 2nd, we have purchased 18 million shares or approximately 4% of our outstanding diluted shares for $3 billion of total. As a reminder, we announced a share repurchase program for $3 billion in February of this year and currently have approximately $1 billion of remaining capacity from that authorization. As part of today’s press release, we announced that the Board has authorized an additional share buyback program, the $3 billion with no expiry. Now let’s turn to our 2022 updated financial framework on page 29. We continue to have signed Advanced Purchase Agreements for expected delivery in 2022 in the amount of approximately $21 billion. This includes expected sales from the recently announced new agreement with the U.S Government and an adjustment for doses that remain unallocated by COVAX due to lack of demand we indicated this was a possibility on our last call. Furthermore, this total includes expected negative foreign exchange impacts compared to the contract value at signing, which we estimate to be approximately 1.5% of sales for the full year 2022 assuming current rates remain through year end. We anticipate that for sales in the second half of 2022, sales will be greater in the fourth quarter than the third quarter, driven by the timing of anticipated approval of our updated COVID-19 vaccines and the related manufacturing ramp-up of the new products. Our total cost of sales, includes the cost of goods manufactured, third-party royalties, as well as logistics and warehousing costs. We now expect our full year 2022 reported cost of sales to be in the mid-20% range driven by the previously mentioned cost related to a reduction of doses to COVAX and deferral of doses to other customers. Cost of sales could increase to the high 20% range in the event of further charges due to product updates. For R&D and SG&A, we continue to expect full year expenses to be approximately $4 billion, driven by a maturing development portfolio and the global scale up of the company. Based on current tax laws, we now expect our 2022 effective tax rate to be in the low- to mid-teen range as a result of the benefits from the foreign derived intangible income driven by our international business mix, as well as stock-based compensation deductions. Finally regarding capital expenditures, we continue to plan for capital expenditures in the range of $0.6 billion to $0.8 billion as we further build out our manufacturing in general company infrastructure globally. This concludes my remarks concerning the financial performance and I turn the call back over to Stephane.
Stephane Bancel:
Thank you, David, Arpa, Stephen and Paul for those updates. In 2022, we set out to execute on five key priorities. First to execute on delivering vaccine against $21 billion of Advanced Purchase Agreements, with half of the year behind us and the sign deals for the fall of 2022 and a strong manufacturing team we are on target. We have continued the momentum in our late stage clinical trials with our Phase 3 trial is now approved RSV and CMV and I thank our team for delivering on such aggressive timelines. We are on track to share data from proof-of-concept studies from two therapeutic application of our technology with data from our PA and PCV trial in second half. We have continued to make progress with new development candidates in vaccines and therapeutics, some already been announced and now will come in the second half of the year. And finally, the recent announcement of our inhaled IND with our partner Vertex, we have expanded our mRNA platform to target primary disease. As we grow, we are committed to doing the right things in the right way for patients and our stakeholders. As a company, led by a solid team, we are thinking in five-year, 10-year, 20-year increments. To that end, this year we published our very first ESG report, which highlights the key pillars of our corporate versatility framework, medicine for patients, our employees, the environments, our community, and of course, governance. I am proud of what we are doing initially in pillar and so proud of the ambitious goals we set for ourselves. I’d be the first steps to achieving those ambitious goal is to set them to create and foster our purpose-driven culture and go on this framework to meet those goals. As an example, our goal of a net zero carbon emissions globally by 2030 we put on that amongst the global leaders in promoting long-term sustainable growth for our planet and our organization. We will share more detail on the ESG governance at our ESG Day on November 10th. But first, we look forward to asking you at our Annual R&D Day in September 8th. Moderna mission to deliver on the promise of mRNA science to create a new generation of transformative medicines for patients has always been our Northstar. Despite the challenges facing the financial market with consumer inflation and ensuing increase in interest rates and the cost of capital, Moderna is in a very fortunate position. We have a unique amount of platform enabling the generation and personnel speed of innovative medicines. We have a strong team of 3,400 mission-driven employees. We have $18 billion of cash on the balance sheet and with the strong commercial momentum. We have no intention of slowing down our growth. We are putting our head down and doing the work. I have been as excited about the future of Moderna, now is not the time to slowdown, patients are waiting for innovative medicines. Thank you for listening and now we would be happy to take questions. Operator?
Operator:
[Operator Instructions] Our first question comes from Matthew Harrison with Morgan Stanley. Your line is open.
Matthew Harrison:
Great. Good morning. Thanks for taking the question. Stephane, I was hoping you could just maybe address in a little bit more specificity how you are thinking about opportunities for either acquisitions or collaborations. I think at the topic that comes up a lot among investors, and I think, there is a different scope that a lot of people think about including the things that are quite large versus things which might be more complementary on a technology side to the platform. So if you could just address how you are thinking about that in your priorities that would be great? Thank you.
Stephane Bancel:
Sure. Thanks for the question, Matthew. So, as you know, we run it now for over a year our capital allocation strategy, that’s in a cash flow positive situation. Of course, number one; invest in the business; two, stick to our investment to build the company; and three, share buyback. As you have seen through what we have done over the last quarters, we have been I think totally aligned with this strategy in terms of the investment in internal R&D, as David said, 69% increase in R&D investments Q2 this year versus Q2 last year. As you have seen we have announced last year our third share buyback plan today also being very aggressive there. As you see, we have done a few deals in the last year in terms of external collaborations. We have not acquired any company at this stage. We see this, to your point as, how do we expand the potential of the company to deliver on its mission to make innovative medicine. And on the external front, I think, there’s two filters that are important for us is, finding interesting assets and finding them at a price that we think we can create value for. As we have said, and David confirmed today, our BD teams are extremely busy looking at a lot of things. But as you know, it’s a bit the same as investors looking for companies to invest in. We look at a lot of things before we decide to go for something. We won’t be shy if we find great assets. There’s a lot of asset that are very early. There’s a lot of asset that don’t really see the company’s strategy, but we will keep looking. I think doing the large acquisition -- probably like a large acquisition is not our strategy. What we want to do is to be the best if nucleic acid company in the world. As we have said, would be very happy to go outside them on as long as we see to the nucleic acid space. We will be happy to do acquisition if we find good assets, we will happy to new partnerships and acquisition is not regarding itself. So that’s a bit how we think about outside the investment actually. Thank you.
Operator:
One moment for next question. Our next question comes from Elizabeth Webster with Goldman Sachs. Your line is open.
Elizabeth Webster:
Hey, guys. This is Elizabeth on for Salveen. Congrats on the quarter and thank you for taking our question. Could you walk us through the path to authorization for 1273.222 and just clarify exactly what the path is here and then when could we see data from the trials, you mentioned, that are starting this month? And then our second question is, if you had to think of levers for potential upsides to the $21 billion in APAs, where might those come from and could additional new orders come online? Thank you.
Stephen Hoge:
Thank you for the question. I will take the first one. And I assume Arpa for the second. So first on the 222 is a bivalent vaccine that is based on the BA.4, BA.5 variant. This is most -- I think the question is directed mostly to the FDA guidance and the focus particularly in United States on authorization of vaccine. We continue to work hard to pull together both preclinical and manufacturing data, and all of our underlying bivalent platform data, which includes two different Phase 2/3 studies and I referenced mRNA-1273.211 and mRNA-1273.214, both of which demonstrated superiority of the bivalent platform and the performance against Omicron variant concern. So the totality of that data was Phase 2/3 study was frequently data manufacturing data will form the submission, which is consistent with the published FDA guidance. FDA has also asked us as you referenced to run an additional study really to support future deployment, because we do want to understand the performance of the 222 vaccine if there is further variant evolution for instance in December or January has unfortunately happened every year in this pandemic and we want to have samples that allows us to inform that those sorts of decisions about deployment of vaccine. We expect to enroll that study really in August. But this data wouldn’t be available for a few months, because you would follow till approximately a month to get the boosted samples, collect them and test them in the relevant asset. We do not at this point expect nor the FDA suggested that that data would be required prior to authorization. In fact, we will be authorizing based on the prior clinical data for 211 and 214 the preclinical data manufacturing data. But we will have that -- those samples in hand. Now as to the specific timing of when we would be able to share 222 data, other than later in the fall, I don’t think we have any other specific item today, except to emphasize again that it’s really not required for the authorization based on recent FDA guidance, it really is to support future deployment decisions that are in the later part of the winter.
Arpa Garay:
And I can take the second question on the $21 billion guidance. From an Advanced Purchase Agreement perspective, we do believe the majority of the market demand is captured in this $21 billion. That being said, we continue to work with countries around the world on potential additional orders for our bivalent vaccines as many countries are continuing to assess their public health needs, as well as their booster population recommendations and considering potential expansions to those populations. So we are feeling pretty good about the $21 billion, but we do continue to work with countries to see if and when there is additional demand for either 214 or the 222 bivalent vaccine.
Elizabeth Webster:
Understood. Thanks so much.
Operator:
One moment for our next question. Our next question comes from Michael Yee with Jefferies. Your line is open
Michael Yee:
Hey. Good morning. Thanks for the questions and thanks for the updates. A quick question in relates to the comment around getting the BA.4/5, 222 authorized. Stephen you had mentioned the pathway there, but actually want to think a little bit forward, do you envision that future variant and development vaccines would be able to be authorized quickly via just preclinical data, such as like flu or do you think that that’s where the path is going and that the FDA seems to be going along that route? That’s the first question. And then second question, just ironically you had made comments around monkeypox vaccine and now WHO has made comments on global health emergency, I didn’t see any update there and wondering if that actually had made any progress? Thank you.
Stephen Hoge:
Sure. Thank you for the questions, Michael. I will try to take both. So, first on the approach, I think, you are right, we do believe that the flu model for authorization of strain supplements, strain updates every year will make sense in the future for COVID vaccines and boosters. And in that sense the authorization of 222 if the FDA -- that does have with the FDA and other markets follow really becomes the first instance of that. We are still going to have to file for supplemental BLA instantiating that pulling together that framework. But we are actively working with regulators, not just in the U.S., but globally to try and establish that pathway, because at the end of day, in order for us to take full advantage of the platform and respond every year, it makes sense that we don’t conduct clinical studies before authorization in the future. We also think that the flu model and the performance of the platform globally now in billions of people really does start to demonstrate the potential for us to mature to more of an endemic approach that doesn’t require a clinical study every time. So that is our expectation and hope will obviously engage with regulators over the fall and winter as we complete those filings and trying prepare for in 2023, it might look just like that. And there’s, again, I think, that 222 experienced this fall really might just be that first instance what becomes the new norm. Now on monkeypox, we did initiate a research program. We are tracking that very closely. And obviously, given the recent public health announcements and increasing concern about availability of vaccine supply, we have -- we are -- been beginning to look at what it would take for us to use our platform to provide a monkeypox vaccine, both intervene in the current epidemic, but also to try and address long-term issues of supply in this public health threat. We do not have any update on those discussions and we will firm them up, those will include if appropriate some regulatory consultations. And once we have clarity on whether we are moving to clinical development and what that path would be, we will, of course, provide an update on it. But at this stage, it remains a preclinical program, but one that we are tracking very closely given the recent developments.
Michael Yee:
Got it. Thank you, guys.
Operator:
One moment for our next question. Our next question comes from Gena Wang with Barclays. Your line is open.
Sheldon Fan:
Hi. This is Sheldon on for Gena. Thanks for taking our question and congrats on the good quarter. We have two questions. One is on the current APA for 2022. Could you comment on how many doses for the COVAX contracts are still unallocated and how they were accounted for in the current $21 billion guidance and could you also remind us on the current APA for 2023 and how should we think about the demand next year? And a quick follow-up, another question on mRNA-1010 flu vaccine, the Phase 3 immunogenicity trial, how do you think about the expected data timeline or do you need to enroll also in the Northern Hemisphere? Yeah. Thanks.
Stephen Hoge:
Maybe I will take the last question first and -- or do you want to take.
Arpa Garay:
I will…
Stephen Hoge:
I will take obvious for sure.
Arpa Garay:
Let me -- so just to reiterate, the $21 billion in Advanced Purchase Agreements does not include any additional COVAX doses to be allocated. So that already reflects a lower demand that we are seeing from the COVAX countries and we anticipate that for the remainder of the year we will see very few additional orders and demands from these countries. In terms of 2023 orders, we have already signed deals with five countries that we have previously announced; the United Kingdom, Canada, Australia, Kuwait and Taiwan. We have also signed options with Canada, Switzerland, Taiwan as well. And we are actively having dialog with countries around the world for additional orders in 2023. We will be able to provide additional guidance on what these advanced purchase orders look like later this year or early next year.
Stephen Hoge:
Great. And then on the question of the flu Phase 3 study that’s ongoing. So that is a Southern Hemisphere immunogenicity and safety study. As we said, we are moving -- that study is ongoing, we can characterize where we are. The -- we do not expect at this point that we would need to rotate that study into the Northern Hemisphere. So we will be concluding that safety immunogenicity study enrolling in the Southern Hemisphere. We do expect to run a Phase 3 efficacy study in the Northern Hemisphere in the later part of this year. And so we will be studying Northern Hemisphere comparators this year, but not the current ongoing Phase 3 that is the safety immunogenicity study that we believe could support accelerated approval.
Sheldon Fan:
A quick follow-up, the Southern Hemisphere immunogenicity data, so likely data will be available later in 2022?
Stephen Hoge:
So we haven’t specifically guided on when the data will be available. But generally, as you know, the safety immunogenicity studies after they are enrolled follow a non-inferiority or superiority endpoints at day 29, so one month after this. And so the data will follow shortly once we have completed enrollment. But ultimately we will also want to follow up with the safety data and consult with regulators about their expectations of what sort of follow-up and data they would like to see and so until we have had all of those consultations we will specifically guide on time.
Sheldon Fan:
Thank you so much.
Operator:
One moment for our next question. Our next question comes from Tyler Van Buren with Cowen. Your line is open.
Unidentified Analyst:
Hi. Good morning and this is Tara on for Tyler. So I was hoping you could provide some comment on the 11% share gain that Pfizer was referring to since January 1st and where that might be coming from, like most of it coming from the adeno vaccines, given the share that they have lost, and of course, I’d be interested to know where the Spikevax share changes are coming from as well?
Arpa Garay:
Sure. I can take that question. In terms of the share, the shares that we reported today are looking at the cumulative share for Spikevax across just the third and fourth boosters, which is a difference compared to some of the data that Pfizer had shared and also looked at the major markets, OECD markets that we plan. So as we think about the majority of where our source of business is coming from. We are seeing substantial market share holds and in some cases even some market share gains year-to-date. Again, focusing just on the booster population, which is where the vast majority of our current and future growth is coming from.
Unidentified Analyst:
Yeah. Thanks.
Operator:
One moment for our next question. Our next question comes from Geoff Meacham with Bank of America. Your line is open.
Geoff Meacham:
Hey, guys. Thanks for the question. And on slide 23 you guys gave sort of the illustrative example of kind of the longer term opportunity for COVID. But I wanted to just get, as you invest in 2023 and beyond, not asking for guidance, but how do you think about what would be the tail of the endemic phase in terms of either volume or patients, et cetera? And then a related question, in terms of capital allocation, how do you guys think about investing in businesses. I know, Stephane, the goal here is to further expand the reach of Moderna’s technology in one of your slides, but how do you think about adding capabilities that are outside of that that maybe peripheral, but I wasn’t sure how far you are willing to sort of steer a little bit away from the core mRNA? Thank you.
Stephen Hoge:
Okay. So I think maybe Arpa, you and I will tag team on the first question then comes to Stephane on the M&A one. So first, maybe on the morbidity of disease and that picture, we do believe that endemic human Coronavirus is point of picture here would suggest there’s going to be seasonal disease, there will unfortunately be breakthrough infection, some hospitalization, even some death and that will happen indefinitely, particularly in those that are higher risk. The question, how we define that higher risk is 55 plus, 50 plus or is it 18 plus with high risk factors or is it broadly, will really depend upon the evolution of the virus. It’s hard to anticipate. But we do believe over time this will become a seasonal market, where boosting and elevating neutralizing protection to prevent breakthrough infections will happen annually in a large population and they will benefit from just like they do for flu or they would for endemic human Coronaviruses. Arpa if you want to comment on how that market evolves.
Arpa Garay:
So the only comment I would add to that is, we will continue to work closely with Nipah, as well as governments around the world to see how their recommendations evolve for their populations in terms of the populations that are being covered and how often they are recommending booster. So we will continue to work with them and over time see how the demand shape up.
Stephane Bancel:
Thanks guys. And on the capital allocation, as we have shared in the past, we are very interested by applying with information molecules, so in nucleic acid. And so we are not only looking at mRNA to express a human protein. As you know we are also losing mRNA through gene as you think, as you know, we are already done some partnership there and we look forward to giving us some update in the near future. But then also in the gene therapy that we talked about RNA also being put actually the space of interest. So I think when you applying nucleic acid, I think, as we said in the past, going to small molecule or large molecule is not something that we think will be in the best action on Moderna, again taking at three-year, five-year, 10-year view of things. I think there is a lot of things we can do in terms of nucleic acid and we like the ability to plug and play on to the platform across research, across development, across CMC. So this is very true benefits and synergies that is just getting a product for the sake of getting a product and driving more complexity into the company and definitely the technologies that we see have little income with nucleic acid.
Geoff Meacham:
Okay. Great. Thanks guys.
Operator:
One moment for our next question. Our last question comes from Ellie Merle with UBS. Your line is open.
Ellie Merle:
Hey, guys. Thanks so much for taking the question. Just as you think about the pipeline and capital allocation from here. Can you help us think about just with the buybacks, as well as further business development, how you are thinking about prioritization? And then just a second question on monkeypox, just I know that since you have announced that you have been looking at preclinically and given the kind of public health emergency and broader vaccine needs. Can you comment a bit on your plans from here and any further plans for development? Thanks.
Stephane Bancel:
So let me start on capital allocation. This is Stephane. As David illustrative on one of our slide that we have been using for few quarters now. Our priority number one has been investing in the business. We are really excited about the platform that we have. As you know in vaccine, given the vaccine modality has been derisked, w are willing to invest pretty aggressively. It’s kind of remarkable, let’s call, we have four vaccine in place for you right now. And as we said on the last call, we have shown now three times the ability to grow from starting a clinical study in vaccines to starting a Phase 3 on 12 months timeframe, which I think really speak about the platform of CMC capabilities, and of course, how we really -- we have a very strong development team. So I see that’s going to be interesting, of course, is the human proof-of-concept that we talked about coming later this year in cancer and in rare disease, because if one of those two or both of those two were to work in terms of getting interesting clinical signal, you will most probably see us do what we did with vaccines which expand very quickly. As you know, one of the really good thing about mission RNA and how we built the company with a lot of robotics out of the digitalization is giving to scale very quickly. We always felt a new application like rare disease or cancer before we know even technology is working, we can’t have time to stop piano [ph] with only a few programs. But if let’s say, we have positive results in PA, will you see us developing many more programs, I mean, what we have now in rate disease very quickly? The answer is yes. And we see something in oncology. So investing in the business is priority the number one, priority number two is really expanding the platform through partnership, licensing, M&A and then the excess cash we will return to shareholder. And I think the announcement of share buyback plan today we were and we should begin the production by the Board is a confirmation of the ability to return the capital if we cannot find with using the company.
Ellie Merle:
Thanks, Stephane.
Stephane Bancel:
Stephen, you want to say on the monkeypox?
Stephen Hoge:
Yeah. Yeah. So, I think, conceptually, we were obviously very aware with monkeypox concern, and obviously, very sensitive to recent announcements. And so what we are looking to do is understand if we were to develop that program, how would we move it? The purpose of moving it would be to move very quickly, and Stephane, just covered, our platform is pretty well established and our ability to rapidly scale has been demonstrated. And if we were to go after a monkeypox clinical development program, it would be to very quickly progress towards an approvable standard endpoints in a clinical study. And in that sense, we need to engage with regulators and other consultations to determine what that path would be. It’s not a primary or principal discussion to just advance a program and demonstrate in Phase 1 clinical generally. We would really be doing it to try and help generate a public health counter-measure. And to those conversations, we will start to more engage with them. We obviously also recognize, there are other even larger public health threats right now. COVID remains the larger public health threat and many regulators are totally focused on addressing the updated this to work for this fall and so we are respectful of that. But we will engage in those conversations and once we have clarity on whether or not we will go forward given what we think those endpoints will be we will obviously provide it, but at this point, it’s premature to say more, because we have not clarified those endpoints and therefore have not made our own decision about whether we will move the preclinical program into clinical development at this stage.
Ellie Merle:
Thanks for the color.
Stephane Bancel:
Great. Thank you so much everybody for joining the call today and for your questions. We look forward to speaking to many of you and look forward to welcoming you at the R&D Day early September. Have a great day. Thank you.
Operator:
Ladies and gentlemen, this does conclude today’s presentation. You may now disconnect and have a wonderful day.
Operator:
Good morning. My name is Kevin, and welcome to Moderna’s First Quarter 2022 Earnings Call. At this time, all participants are in a listen-only-mode. Following the formal remarks, we will open the call up for questions. Please be advised that this call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone, and thank you for joining us on today’s call to discuss Moderna’s first quarter 2022 financial results and business updates. You can access the press release issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our website. On today’s call are Stephane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; and Paul Burton, our Chief Medical Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. I will now turn the call over to Stephane.
Stephane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Welcome to our Q1 2022 conference call. Today, I will start by a quick business review of the quarter before Paul walks you through an update on Spikevax real world evidence, and then Stephen reviews our clinical programs. David will then present the key financials. And I will then come back to conclude before we take your questions. I’m happy to share that the team delivered strong financial results this quarter. Revenues of $6.1 billion, GAAP net income of $3.7 billion and GAAP diluted EPS of $8.58. We ended Q1 with a cash balance of $19.3 billion. For our share buyback program, we continued to retire shares in Q1 like we already did in Q4. David will share some numbers on share count in a few minutes. For 2022, we are reiterating $21 billion in signed advanced purchase agreements. We have previously shared market share increases seen in the OECD countries with Spikevax when supply was no longer limited and when real-world publications highlighted differentiating data amongst the market vaccine. I’m happy to share that our market share has increased or still consistent across OECD countries. While a subset of our team is focused on delivering on the $21 billion signed APAs for fiscal year 2022, another subset of our team is focused on preparing the next wave of product launches. With our flu vaccine candidate, mRNA-1010, plan to start a Phase 3 study in Q2 in the Southern Hemisphere, Moderna will have very soon four vaccine candidates in Phase 3
Paul Burton:
Thank you, Stephane, and hello, everyone. While there continues to be many studies posted and published on our mRNA vaccine, I would like to highlight some data today on vaccine boosting. But first, it’s important to understand why booster vaccines are so critical as we transition through the COVID-19 pandemic and the understanding that we need to have about the evolution of the SARS-CoV-2 virus. On slide 9, we can see the change in COVID-19 cases over time with the massive rise we observed due to Omicron in the last weeks of 2021 and the early part of 2022. The Omicron wave was caused by BA.1. And while that was certainly the dominant version of SARS-CoV-2 at that time, remarkably BA.1 is now almost extinct here in the United States and around the world, having been replaced by new subvariants. This continues to demonstrate the remarkable evolutionary capacity of this virus. BA.2 is now the dominant strain in the United States with another subvariant of that BA.2.12.1 increasing rapidly, showing enhanced transmissibility. While BA.2 is dominant today, that increased transmissibility and infectivity of BA.2.12.1 is likely going to ensure it will be the dominant circulating strain very soon as we are seeing in New York and in the northeast of the United States. Other new subvariants, BA.4, BA.5 seen in South Africa, have also been detected in the United States. And we will need to carefully monitor their growth trajectories and pathogenicity. The slowing of booster uptake now means there will be individuals who are under vaccinated and underprotected as we move into late spring and summer when we thought we would have declining case counts, a balance, which could now be impacted by BA.2.12.1, BA.4 or BA.5. The speed and breadth of evolution of this virus that we see so clearly again today underpins our prediction for the global need for a variant-adapted booster campaign this coming fall. The good news is that a booster vaccine protects against both, BA.1 and BA.2. This is seen clearly when we look at data from the United Kingdom Health Security Agency. This graph looks at vaccine effectiveness against symptomatic disease for people who received the Moderna, Pfizer or AstraZeneca vaccine for their primary vaccination on the left. You can see that against both, BA.1, Omicron and BA.2, vaccine effectiveness wanes over time. But, a booster dose of either the Pfizer on Moderna mRNA vaccines increases vaccine effectiveness and protection, as you can see on the right. These two vaccines are combined together in this analysis. But again, even in this setting, there’s gradual waning of vaccine effectiveness over time following boosting. This waning of vaccine effectiveness begs a question, will a second booster dose provide clinical utility and help to restore vaccine effectiveness and provide protection against COVID-19? While there are several examples of studies showing the high clinical effectiveness of additional boosting with the Moderna vaccine, on slide 11, I want to highlight the results of a recent study from Ontario that looked at exactly this question and measured the effectiveness between a first and second booster vaccine dose in a high-risk population, those people living in long-term care facilities. A key finding from this study, which was conducted in over 55,000 individuals, was that indeed a second booster vaccination showed increased vaccine effectiveness against Omicron infections, symptomatic disease, and importantly, against severe outcomes in this high-risk setting. In fact, an increase in vaccine effectiveness was seen at 7 days from the second booster vaccination and continued to increase in effectiveness over time. Turning now to slide 12. I would like to provide a perspective on who might gain particular benefit from annual vaccine boosting. There are many health, age-related and environmental or occupational risk factors that lead to populations being at higher risk for COVID-19. First, age greater than 50 years. We know that hospitalization and mortality rates begin to increase steeply for those with COVID-19 who are over the age of 50. And then, turning to people over the age of 18 year old that have other health risk factors, such as people with kidney disease, cancer, autoimmune disease and HIV patients, other health factors that either result in immunocompromise or place people at higher physiological risk for severe disease, if indeed, they are infected with SARS-CoV-2. Finally, environmental or occupational risk factors, such as health care workers, first responders, those in high-density housing or living conditions such as college students, military personnel, or the incarcerated. We believe it is people in these broad categories who could benefit most from annual boosting for COVID-19. And so, in summary, we’ve seen, as we continue to anticipate, SARS-CoV-2 to keep evolving rapidly with multiple new variants and recombinant variants circulating globally. Real-world evidence demonstrates the effectiveness of a booster shot, a third dose of mRNA-1273 against evolving variants of concern. And an additional booster, a fourth dose of mRNA-1273 shows incremental vaccine effectiveness when compared to a third dose against infection, symptomatic infection and severe disease in a high-risk population. We believe people are at high risk due to health, age and environmental or occupational risk factors. And we believe that when taken together with the viral epidemiology and waning of protection, there is an important need for a variant-adapted booster vaccine and for boosting of populations this coming fall. With that, I’ll now turn it over to Stephen to take you through the progress in our clinical development pipeline. Stephen?
Stephen Hoge:
Thanks, Paul. Good morning and good afternoon, everyone. So, Paul just shared with you the effectiveness of our booster MRNA-1273 against Omicron in the real-world setting. But on slide 15, I’d like to pivot to our strategic rationale for why we think a seasonal booster will be necessary. So, first, we think neutralizing titers will wane similar to the endemic human coronaviruses. And that decline in neutralizing titers will increase their risk of breakthrough, infection and hospitalization for those at higher risk, particularly as Paul just described, older adults or those with medical immunocompromised. The emergence of new variants of concern like the BA.4, .5 subvariants could accelerate the impact of that waning and broaden the risk of breakthrough across the population. So, with that, we do believe a booster will be needed in the fall, and we’re working hard to make improvements to our available boosters. The desired features for a Northern Hemisphere fall-winter booster, we think will be that it improves the durability of protective neutralizing antibodies against Omicron and its subvariants beyond six months, i.e., the full Northern Hemisphere fall or winter infection season. We’d like to retain high and durable protection against Delta and ancestral strains. And we’d like to broaden cross-protective immunity to increase the potential for protection against new emergent variants or subvariants that might happen over the coming months. So, on slide 16, I’d like to summarize our work in developing that improved booster. Our primary focus, as you know, has been on developing a bivalent vaccine, and we have taken three bivalents into clinical trials. The first, mRNA-1273.211 includes 9 of the common mutations and was based on a combination of our prototype wild-type vaccine and Beta. The mRNA-1273.213 bivalent included 11 mutations based on what had emerged from Beta and Delta. And our mRNA-1273.214 booster includes 32 mutations, also now based on the wild-type, prototype vaccine and the combination with the Omicron original variant of concern. Our latest bivalent, mRNA-1273.214, that includes those 32 mutations that have emerged, remains our lead candidate for the fall Northern Hemisphere campaign. The objective of that booster will be to demonstrate superior immunogenicity against variants of concern when it compares to our approved current prototype booster or MRNA-1273 at 50 micrograms. And of course, we want to maintain non-inferiority against ancestral strains in case they reemerge. Now, on slide 17, I’ll summarize the ongoing clinical development work across that portfolio of bivalent boosters. I’ll remind you that mRNA-1273 has been authorized or approved in many markets as a third and even a fourth booster. The data for the first bivalent mRNA-.211 has already demonstrated superiority against all variants of concerns tested, including Omicron and Delta, and I’ll cover that data in just a moment. But, our lead candidate remains, as I said a moment ago, our mRNA-1273.214, which is being evaluated in two separate studies, a Phase 2/3 in the United States and a Phase 3 P305 study in United Kingdom. Again, both of those are being conducted at a booster dose of 50 micrograms. Now, on slide 18, just quickly to update you on the data we have from the .211 first bivalent. Not surprisingly, the safety and reactogenicity profile of the bivalent boosters is consistent with what we saw with mRNA-1273. As you’ll see on the chart, both solicited adverse local reactions and systemic reactions are broadly consistent in both frequency and severity. And the frequency and types of unsolicited adverse events were also comparable between the groups with no serious adverse events in the bivalent vaccine group up to 28 days after the booster dose. Moving to slide 19. We have some of the neutralizing antibody data from that study, evaluating again our mRNA-1273.211 bivalent and comparing that with an approved or authorized mRNA-1273 booster. Looking at neutralizing titers and GMTs, both immediately pre-booster at 1 month or day 29 and at 6 months, on day 181 across the 3 variants of concern which we tested, higher neutralizing titers were seen for day 29 and day 181 across all the variants of concern with the bivalent booster. On slide 20, we represent that as a ratio when comparing the performance of the bivalent booster to mRNA-1273 at 1 month and 6 months, and superiority was met for the ancestral and all variants of concern at different time points, as you’ll note here. The clinical endpoint for superiority was defined as a geometric mean titer ratio, or GMR, where the lower bound excluded -- the lower bound of the 95% confidence interval excluded 1. And as you’ll note, at day 29, a GMR for the ancestral SARS-CoV-2 virus was 1.28, Beta was 1.3, Delta was 1.75, and importantly, Omicron was as high as 2.2. And the 95% confidence interval for Omicron, the lower bound was 1.74, again, demonstrating strong trend towards superiority in this data. Excitingly, at day 181 or 6 months, that superiority was also met for the ancestral virus, Beta and the Omicron variant, which is important because the primary goal we have here is to improve the durability of protection by increasing those titers. So, on slide 21, in conclusion, the safety and reactogenicity profile of the 50-microgram bivalent 211 booster was comparable to the 50-microgram of the authorizer approved 1273 booster. And we believe that the superiority already demonstrated by the bivalent platform in .211 bodes well for our overall strategy. We continue to believe that bivalent boosters will ensure the broadest immunity across the evolutionary uncertainty of SARS-CoV-2 and maintain current protection while expanding the breadth and durability of neutralizing antibodies, including, as I just demonstrated a moment ago with .211, out to 6 months. We anticipate the one-month or day 29 data from our Omicron containing bivalent, mRNA-1273.214 in June of 2022. Now, turning to the rest of our respiratory vaccine pipeline on slide 23. We announced positive Phase 2 data from our flu vaccine, mRNA-1010 at our Vaccines Day event. mRNA-1010 is our -- is part of our speed-to-market approach. We plan to start a Phase 3 immunogenicity study in the second quarter of this year and a Phase 3 efficacy trial later this year with mRNA-1010. As part of our flu vaccine strategy, we are also advancing in parallel vaccine candidates that contain both HA antigens and NA antigens. We started a Phase 1/2 trial of mRNA-1020 and mRNA-1030 last month. Our RSV vaccine, mRNA-1345, a Phase 3 trial in older adults is ongoing, and we are enrolling participants worldwide. We also have an ongoing pediatric RSV trial enrolling as well. In combination, we plan to start a Phase 1 trial for both, our COVID plus flu and our COVID flu RSV vaccines this year. Our Phase 1 trial for the hMPV/PIV3 combination vaccine is also now fully enrolled. And our RSV and hMPV combo and our endemic human coronavirus vaccine combo are in preclinical. Before moving from respiratory vaccines, I want to take a step back and reflect on the incredible progress over the past two years. We have or will have progressed 3 candidates into pivotal Phase 3 studies within one year of an IND being opened. This speed is made possible by our mRNA platform. And we believe our COVID vaccine success has derisked our vaccine pipeline, and we can now move quickly into our RSV and flu pivotal studies. Importantly, in RSV and flu, we are looking at seasonal endpoints or immunogenicity endpoints, which we believe can be -- can allow us to progress faster through Phase 3 towards its readouts and ultimately to commercialization. Now, turning to the rest of our pipeline. We have an ongoing Phase 3 study for our CMV vaccine, mRNA-1647, which is enrolling well and is now enrolling participants globally. Our EBV vaccine to prevent infectious mononucleosis is in Phase 1, and our EBV vaccine to prevent long-term sequelae such as multiple sclerosis is in preclinical studies. Our HIV vaccines are in Phase 1 clinical trials with our partners. And the recently announced HSV and VZV vaccines are in preclinical studies. Within public health vaccines, our Zika vaccine continues to enroll in Phase 2. And we are pleased to update that our Nipah virus vaccine IND was opened, and we look forward to starting that trial soon with our partner. Now, moving to our therapeutic pipeline on slide 26. Within oncology, our personalized cancer vaccine is ongoing in a Phase 1 study and a Phase 2 study. And we expect the first look at the data from our Phase 2 study, which is evaluating personalized cancer vaccine plus KEYTRUDA versus KEYTRUDA alone, in the fourth quarter of this year. We have four other candidates in Phase 1 or preclinical stages across oncology. In cardiovascular and autoimmune, we have two candidates in each area in clinical trials or in the preclinical stage. And within rare diseases, our propionic acidemia program, or PA program for short, which I will give more detail on the next slide, is ongoing in a Phase 1/2 study. Our MMA program is also ongoing in the Phase 1/2 study. And our GSD1a program has an open IND and we look forward to enrolling the first patient or participants in that study. We have three other candidates in preclinical and rare disease as well. Now, before I hand it off to David, on slide 27, I wanted to provide a bit more color on our Phase 1/2 study in propionic acidemia. As a reminder, PA is a rare metabolic disorder that is characterized by a deficiency of propionyl-CoA carboxylase, an enzyme that’s involved in the breakdown of several of the building blocks of proteins called amino acids. As a result of deficiency in that enzyme, harmful intermediate compounds can build up to toxic levels in the body. This can lead to serious health problems, including recurrent episodes of life-threatening metabolic decompensation events. Our therapy for PA encodes for two of those proteins that form the deficient enzyme, PCCA and PCCB, and has 1 mRNA for each in the drug. The Phase 1/2 study is an adaptive trial design, enrolling participants greater than one years of age in the United States, United Kingdom and Canada. Participants received one dose of mRNA-3927 every 2 or every 3 weeks for up to 10 doses in that study. The first cohort is fully enrolled, and we are now enrolling patients in the additional cohorts. Five patients have now completed the initial 10-dose course of the study and became eligible for continuing dosing in the open-label extension. And all five of those patients have elected to participate in the OLE. A total of 75 doses have now been administered across Phase 1/2 and the OLE study. Now, the study is focused on evaluating safety and PK/PD. It is also looking at clinical events, those that are most important, including the metabolic decompensation events I mentioned previously. And of course, we are also evaluating potential biomarkers in the study. We look forward to enrolling more patients and sharing the data this year. And with that, I’d like to turn this over to our financial review. And David?
David Meline:
Okay. Thank you, Stephen. We’re providing today the analysis of actual 2022 first quarter results, along with a view of key drivers of financial performance going forward. Overall, we had a good start to 2022, and I’m very pleased with our operational and commercial performance. Turning now to slide 29, starting with an overview of our sales performance. Total product sales in the first quarter of 2022 were $5.9 billion. This compares to product sales of $1.7 billion in the first quarter of 2021. The total sales growth is driven by the fact that we were in an earlier stage of our manufacturing ramp-up in Q1 of last year with our U.S.-based manufacturing lines roughly three months ahead of our international manufacturing capabilities. This also explains the different geographic sales mix. In the first quarter of 2022, sales outside the U.S. were $5 billion, and sales to the U.S. government were $0.9 billion. The majority of sales in Q1 of last year were to the U.S. government. Turning to slide 30 to go into more detail of our Q1 results. Total revenue was $6.1 billion in the first quarter of 2022 compared to $1.9 billion in Q1 of last year. The increase of total revenue was driven by the sale of our COVID-19 vaccine. Cost of sales was $1 billion or 17% of the Company’s product sales in the first quarter. Cost of sales in percent of sales in Q1 of last year was 11% on a reported basis and 22% adjusted for prelaunch inventory costs, which were expensed in 2020. Compared to the prior year adjusted 22%, we are benefiting this quarter from an increased average selling price, driven by customer mix, favorable impacts from the scale-up of our manufacturing processes, partially offset by higher write-downs for excess and obsolescent inventory and a current period expense related to future purchase commitments. Research and development expenses were $554 million in the first quarter of 2022 compared to $401 million in the same period in 2021. The increasing R&D spend continues to be driven by clinical trial expenses for our expanding and maturing development portfolio. Selling, general and administrative expenses were $268 million for Q1 2022 compared to $77 million for the same period in 2021. The growth in spending was driven by the commercialization of our COVID-19 vaccine globally with continued investments in personnel and outside services in support of the accelerated company build-out. Our Q1 2022 results also include the initial upfront endowment of $50 million for the newly established Moderna foundation. Provision for income taxes was $572 million in the first quarter of 2022 compared to $39 million in the prior year period. Our effective tax rate for the first quarter was 14%. Let me remind you of the fact that we had a net operating loss carryforward of $2.3 billion at the end of 2020, which resulted in a nonrecurring benefit to the reported tax rate last year. We recorded after-tax net income of $3.7 billion in Q1 compared to $1.2 billion in the prior year. Diluted earnings per share in Q1 2022 were $8.58. As a final point on the quarter and beyond, we currently do not have any commercial activities or R&D activities in the Ukraine or Russia. Turning to cash and cash deposits on slide 31. We ended Q1 2022 with cash and investments of $19.3 billion compared to $17.6 billion at the end of 2021. The increase is driven by our commercial activities. The balance of cash deposits for future product supply was $5.3 billion compared to $6 billion at the end of 2021. The reduction quarter-over-quarter is driven by product deliveries against customer deposits. Now, turning to slide 32. Our capital allocation priorities remain unchanged. Our top investment priority has been and will continue to be reinvesting in the base business across multiple areas. For R&D, we continue to forecast to spend in the range of $2.5 billion to $3 billion in order to advance and accelerate our pipeline, both for existing and new programs. Our second investment priority is to seek attractive external investment and collaboration opportunities to further expand the reach of Moderna’s technology and capabilities. We are considering attractive opportunities that enable and complement our platform and take a disciplined approach in evaluating potential outside investments. We’re in multiple active discussions regarding additional external collaboration opportunities. After evaluating internal and external investment opportunities, we then assess additional uses of cash. We completed our initial $1 billion share buyback program in January and announced a new share buyback program of $3 billion in February. Across these two authorizations, we repurchased a total of 3.8 million shares for $0.6 billion during the first quarter. The next slide shows a progression of our share count since we initiated our share repurchase program. As a reminder, we received Board approval for our initial $1 billion share repurchase program in August 2021 and began repurchasing shares in the fourth quarter of 2021. Our quarter-end basic shares outstanding declined from 405 million at the end of September 2021 to 400 million at the end of March 2022. We repurchased 7 million shares, more than offsetting 2 million shares of common stock issued in connection with equity compensation over this period. Our diluted weighted average shares outstanding also declined from 434 million in the third quarter of 2021 to 426 million in the first quarter of 2022, primarily as a result of the share repurchase activity. Now, let’s turn to 2022 financial framework on page 34. We have signed advanced purchase agreements for expected delivery in 2022 in the amount of approximately $21 billion. There’s a potential downside to this number from timing of COVAX deliveries if COVAX is unable to confirm demand aligned to their contracted volume in the 2022 calendar year. There is also an upside to this number from potential additional contracts for the fall booster dose, including for the U.S. market. In 2022, we believe that the SARS-CoV-2 virus will evolve into an endemic phase with a more seasonal sales pattern. As a result, we continue to expect the timing of sales to be larger in the second half of 2022 than in the first half. Our total cost of sales includes the cost of goods manufactured, third-party royalties as well as logistics and warehousing costs. We continue to expect cost of sales as a percent of product sales to increase compared to prior year, driven by both a decrease in our average selling price and a forecast increase in manufacturing cost. The year-on-year decrease in average selling price is due to the higher share of COVAX APAs for delivery this year compared to last year. As the COVID-19 pandemic evolves into an endemic phase, we forecast our manufacturing unit costs to increase. This is driven by a move to smaller dose presentations and the cost for adjusting our production and supply chain infrastructure, including future purchase commitments. We continue to expect our full year 2022 reported cost of sales in the low to mid-20% range, which incorporates all the expected adjustments that we’ve identified for the 2022 calendar year. The cost of sales for Spikevax beyond 2022 will be further impacted by geographic customer mix, pricing effects of the private market as it develops, as well as our initiatives to optimize and improve efficiency. For R&D and SG&A, we continue to expect full year expenses to be approximately $4 billion, driven by our maturing development portfolio and the global scale-up of the company. Based on current tax laws, we continue to expect our 2022 tax rate to be in the mid-teens, as a result of the benefits from the foreign-derived intangible income, driven by our international business mix and stock-based compensation deduction. Finally, regarding capital expenditures, we continue to plan for capital expenditures in the range of $0.6 billion to $0.8 billion as we further build out our manufacturing and general company infrastructure globally. As this is the last earnings call for me as CFO of Moderna, I would like to take the opportunity to welcome Jorge Gomez, who’s well qualified to lead Moderna in the next stage of its development. I would also like to recognize my own team and my colleagues for success in ramping up a global commercial company and contributing to taming the pandemic. I have great confidence in the Company’s ability to fully realize the vast potential of its mRNA technology platform against the many remaining unmet medical needs. This concludes my remarks concerning the financial performance, and I would like to turn the call back over to Stephane.
Stephane Bancel:
Thank you, David, for that review of the quarter and those kind of words. More importantly, I would like to thank you for agreeing to come out of retirement in the spring of 2020 to help us scale Moderna at an unprecedented speed from an early-stage development U.S.-centric company to a commercial, global company. I am very grateful for you agreeing to come out of retirement to help us. Your work to get us to this point was crucial. You built a strong team and robust business processes. Thank you for also agreeing to stand as a consultant as we transition to your successor. And so, I want to wish you and your wife well in the next phase of your life. As many of you know, we announced that Jorge Gomez will join us as Chief Financial Officer, effective next Monday, May 9th. Jorge brings with him experience in the capacity of CFO for global health care organizations, which are Dentsply and Cardinal Health. Jorge has the same kind of high-quality finance training as David as a General Motor finance alumni. He has a broad set of international experiences in addition to having been CFO of publicly-traded companies. I would also like to welcome Arpa Garay who is joining us from Merck at the end of this month as our Chief Commercial Officer. She was a member of Merck Executive Committee where she most recently served as Head of Marketing. Arpa also has a very international background and multiple experiences in both sales and marketing. I look forward to partnering with these two leaders to continue to scale rapidly Moderna. Before moving to Q&A, I would like to review with you our 2022 priorities. Priority number one, to execute on the $21 billion of signed APAs and to prepare for a successful fall 2022 booster season. Priority number two, to execute on our four Phase 3 vaccine programs, an Omicron-containing COVID bivalent booster, flu booster, RSV booster and CMV primary series vaccine, which could lead to three respiratory commercial launches over the next two to three years. Priority number third, to expand beyond infectious disease vaccine into therapeutics and share proof-of-concept readouts for PA, MMA and PCV programs. Priority number four, to bring forward more mRNA candidates into clinical development. And last but not least, priority number five, to continue to expand our mRNA platform, so the possibility of mRNA impact to patients continues to increase over time. I also wanted to remind people of our upcoming events this year, including our Science Day in just two weeks on May 17th. We’ll also host our typical annual R&D Day in September, and first ESG Day in November. While we have had a profound impact on humanity over the last two years, we believe that what is ahead of us to help protect and treat hundreds of millions of people is even more exciting. We’ll continue to execute on our mission, and we thank you for your support. This is just the beginning. Operator, we’ll be happy to take questions now.
Operator:
[Operator Instructions] Our first question comes from Salveen Richter with Goldman Sachs.
Salveen Richter:
Good morning. Thanks for taking my questions. And David, it’s been a pleasure working with you. Two questions for me. One is for COVID, based on what’s known right now, what regimen as you think about dosing in candidate do you have the most confidence on for annual boosting as you look to 2023 and beyond? And then secondly, just given your balance sheet and the -- as you look to kind of entering an endemic phase here, how are you thinking about later stage BD and M&A to bolster the revenue line?
Stephen Hoge:
Thank you, Salveen, it’s Stephen. So first, on the COVID booster and what we perceive as of today, subject to data and obviously collaborating with regulators. We are, as I said, most excited about our bivalent vaccine platform and including Omicron as the now dominant variant and family of subvariants that are infecting people today. Our objective, just like with our bivalent .211 booster data that I presented today, is to provide at least six months of protection from that. And because we think this will be a seasonal vaccine, that will be very similar to what people are used to from a flu perspective. Meaning in the Northern Hemisphere, you get boosted in the October time horizon that will cover you well through the early part of spring when transmission will subside. And so, provided that we are able to provide that duration of protection as we’ve demonstrated already with the .211, we actually think that that means a seasonal annual booster in the fall that would cover people for the year. And as I said, we think it will be the bivalent Omicron containing booster for this year. For 2023 plus, we would expect to continue to update the bivalent platform to reflect the then dominant or then at-risk circulating strains of the variant of concern. So, it maybe Omicron this year, it may be Omicron again next year, or it may be something new.
Stephane Bancel:
Thanks, Stephen. Good morning, Salveen. It’s Stephane. So, as you know, in term of capital allocation, our priority number one is to invest in the business. We believe we have this really unique platform that is very different from typical pharma companies or biotech companies where we have an ability to scale very quickly. As we discussed today, we are actively preparing the pivotal studies and the launch of four programs in Phase 3, as we discussed. As you know, there’s a lot of programs in Phase 2. And I think there’s around 30 vaccines in development today. And Stephen has showed you today that we believe we have built for our platform and all our investment in digital and a great team, we have the ability in around 12 months to go from opening an IND in vaccine to starting the Phase 3. So, seeing where the pipeline is and where it’s going to be 12 months, 24 months from now, that is really exciting. And then, there’s therapeutics. We always get a question around COVID-19 for obvious reasons. But as we’ve said in our remarks, this year will be very exciting around therapeutics. We have two rare genetic disease programs that are well recruiting, as Stephen described, for which we are eager when we have it to share data with you. And assuming this is positive, we will do more in rare genetic disease. As you’ll recall, because you’ve been following the Company for a long time, we started with a few infectious vaccines and as we got more confirmation that the technology was working in human, and more recently, more capital, we had a very unprecedented acceleration of our infectious disease vaccine. Well, we plan to do the same in rare genetic diseases if we get positive clinical data and personalized cancer vaccines. And as you know, we have, as Stephen said, more cancer programs, autoimmune programs and also quite exciting to work with our partner, Vertex, toward getting following application, the CF mRNA candidate into the clinic. In terms of M&A, I can tell you, our teams have never been as busy. They are looking at a lot of opportunities literally across the world, across therapeutic area, but also technologies. And so, we will not be shy to invest to expand the platform, have approved technology or approved product, and that’s really where we are in terms of M&A. So, more to come when things get finalized and signed. And as you know, the third bucket of our priorities in terms of balance sheet is returning shareholders to capital via share buyback. And so this, as David said, we’ll continue to buy back share at attractive prices. And we’ll continue to have a dialogue with the Board as to what should we do after the current plan, if needed. But now, we’re focused on executing the current plan. Thank you.
Operator:
Our next question comes from Matthew Harrison with Morgan Stanley.
Matthew Harrison:
I guess, two for me. So first, David, could you just comment a little bit more on the upside and downside levers to COVID revenues this year? And in particular, what would be the features that would not allow COVAX to accept the deliveries, or what should we be looking out for to understand that? And then, secondly, just on flu, could you maybe give some updated comments on where you are in terms of regulatory discussions? It sounds like it’s not clear yet whether or not you’re going to need a full efficacy study for approval. And so, I’m just wondering where those discussions are and when you think you’ll be ready to give people a clearer picture on what the outlook is for the flu program. Thanks.
David Meline:
Sure. So, on the first one, in terms of the outlook for sales in 2022, I think the most important point is COVAX had an option for additional doses in 2022, which they chose not to exercise. So that’s impacted the outlook. We formally were reporting options that could be taken up. That’s been taken off the table as they didn’t exercise. And what we see is in our plans in the $21 billion, there continue to be some volume for COVAX. They are actually the consolidator of demand from the countries. So, what they need to do is get the confirmed requirements from each country and then that becomes a confirmed order to the Company. And notwithstanding having contracts, there’s still that process they have to go through. So, that’s why we want to flag there’s some level of uncertainty associated with the timing of that demand that we’re reflecting in the APAs. And then, of course, we have, as we’ve said before, a number of negotiations and discussions going on with other countries around the world as we look forward to the fall season and look forward to the variant booster offerings that are making their way through the approval process. So, likewise, that presents some potential for additional sales that are not presently in the APA count for this year.
Stephen Hoge:
Thanks. This is Stephen, Matthew. So, on the question of flu and where we are, we have been consulting with regulatory agencies globally, as you might imagine, around the path forward for our mRNA-1010 program. As there are issued guidance around accelerated approval that are still open, at the end of the day, we still believe that there’s a potential path towards an accelerated approval, at least in some markets with our mRNA-1010 program using a safety and immunogenicity endpoint as has been previously discussed. At the end of the day, that will be a review, Matt, and we’ll have to generate that data and have conversations on the back of that data with regulators as a path forward. But even if we do move forward with accelerated approval, which is the study that we’re starting the Q2 Phase 3 study that we’re talking about starting this quarter -- even if we do move forward on the back of that, we will have an obligation even under accelerated approval to demonstrate efficacy at some point in a follow-on study to move from accelerated to full approval. And so, because we’re certain of that need at some point, we are also planning to start a Phase 3 efficacy study, which will then be conducted perhaps in the Northern Hemisphere winter, this coming fall. And we also announced the plans for that. And what that would do is, we would hope, we find a path forward for both, the accelerated approval but also rapidly being able to move that to a full approval on the back of that efficacy result. Now for some reason, the accelerated approval is not available in some markets or all markets, we would instead go directly to full approval with a very short latency, given the timing of those two studies.
Operator:
Our next question comes from Gena Wang with Barclays.
Gena Wang:
Thank you for taking my questions. Congrats on the strong quarter, and David, we will miss you. I have a few questions regarding the revenues. David, you mentioned that second half this year will be higher than the first half. Since first quarter, we already delivered $5.9 billion revenues. Should we expect less than $4.5 billion for the second quarter this year? And then, my second question is regarding the ex-U.S. $5 billion revenue. What is the breakdown between EU and the rest of the world? And the third question is regarding the prices for U.S., EU and the rest of the world. And where do you see these prices change for the remaining of 2022 and also 2023?
David Meline:
Okay. Good. So, I’ll try to cover them all. And if I miss one, come back to it. So, in terms of -- first of all, maybe I’ll start at the end on pricing. We previously disclosed the ranges of pricing that we had in the contracts last year. And what you see is that pricing has continued as we contracted for 2021. So, really no change on the price across the various customers. It’s really net prices being impacted, as I mentioned, by the mix of customers where we have our COVAX sales at the very lowest price offer whereas the other countries, the developed countries, we have varying higher prices. So, those continue. We haven’t commented on pricing for beyond 2022. But certainly, I think it’s fair to assume that to the extent that the market moves to a private market, typically, you see higher prices in driving markets based on the needs of the market as opposed to when you’re addressing the government acquired product for in this pandemic context. So, that’s one. Secondly, in terms of the 2022 outlook, I think you got it right. So basically, first of all, I think it would be a mistake for us to move into quarter-by-quarter accounting of where we see the demand given the variability of this pandemic. But what we see with the $21 billion of signed APAs, we mentioned last quarter and it’s the same this quarter, that $21 billion we think will be somewhat higher second half sales than first half. And as you correctly pointed out, the math would then point to the second quarter being likely the lowest that we’ll see throughout this year. So, I think, your thinking on that is right. And sorry, the third point was?
Gena Wang:
Third point -- thank you very much. That’s very helpful. The third question is ex-U.S. $5 billion revenue, what is the breakdown between Europe and the rest of the world?
David Meline:
Yes. So, we had -- it was a pretty broad spread. I don’t have top-of-mind the specific quantities by country or region. But it was a pretty a broad spread of demand if you look across the world in the first quarter.
Stephane Bancel:
Yes. And maybe just to add, Gena it’s Stephane, on the pricing. As David said, as we move into endemic, as we’ve been saying, we’ll have to discuss with payers as part of kind of health economics and value of products as it’s done for every pharmaceutical product, especially in the U.S. And as you might be aware, CMS has already communicated that for fiscal year 2023, which starts in October 2022, the reimbursement for COVID-19 vaccine is going to be $60.
Operator:
Our next question comes from Michael Yee with Jefferies.
Michael Yee:
I have to ask the last question to David before he gets to go back and then a question for Stephen on the pipeline. I’m going back to the question around the guidance. Can you maybe help us quantify the exposure to COVAX? There was an article I guess talking about some of that recently in the press. And I think that’s part of what your APA commentary out today is. I’m not sure, are you saying that that’s partly in the $21 billion, but then offset by potential USA orders that could come later this year. So, maybe you could just talk about that dynamic a bit? And then a question for Stephen is, again, definitely excited about propionic acidemia. Can you just rightsize your expectations because it’s five patients but at the lowest dose. So, is that a therapeutic dose, you would expect to see biomarker changes? Maybe just talk a little bit about that. Thank you.
David Meline:
Sure. So, yes, so I’m not sure I have too much to add in terms of the color as to the outlook for the balance of the year. Certainly, again, the $21 billion, we don’t, right now, have included any contracts as a result of additional U.S. business, which we think is quite likely, we certainly believe there’s a recognition of the need for boosters in the U.S. and the dialogue is quite active. That’s also true around the world with our customers in other countries and regions. And then, we wanted to be clear that in the case of COVAX, which is as we’ve said before, the lowest priced business in the portfolio, but we have confirmed contracts there in place. And we wanted to flag that they are, as I said, a consolidator of underlying demand. And they continue to work through in those developing markets what is exactly that demand picture and what is the timing of it and the ability of those countries to absorb the product that they’re receiving. So, we wanted to flag that. Quite frankly, if you ask me, should this be an issue of big concern in terms of the total outlook, I would tell you I would put it as quite modest. But, we’re trying to give some sense as to the range of outcomes in terms of this information.
Stephen Hoge:
Thank you, Michael. So, look, you point to the most important thing I’ll say, which is this remains a small number of patients. It’s a rare disease. And as you said, we’re looking now in Cohort 1 and Cohort 2 at our lowest dose levels, and we’re continuing to enroll. And I would expect, as is appropriate for a Phase 1 study where we are doing dose finding Phase 1/2, that we will continue to enroll and explore a range of doses, including potentially higher doses in a Cohort 3. Now, that said, we will have a body of data building. As we said, we’ve got 5 participants who’ve moved into the open-label extension. One has already -- at least one has already been approaching approximately a year on drug, a total of 75 doses. And so, as you look -- as you look at that body of data, it will start to provide potentially an early signal. And I think in that sense, the things that I will personally be looking at, I think we’ll be focused on. First and foremost, it’s clinical endpoints. It is the clinical endpoints that matter most to these patients, obviously. We are developing the medicine to try and prevent the sequelae disease. And so obviously, that includes things like metabolic decompensation events, hospitalization, other interventions, other progressions, signs of rare disease. And importantly, that’s something you really only measure over time as opposed to a biomarker, which I’ll get to in a second. You really need to see about what that looks like over time. Now, the benefit of where we will be this year is that we will have a reasonably large amount of time for these small number of patients on drug. And so, that’s something that we’ll be focused intensely on. But again, it’s small and that we’ll be looking at. When it comes to biomarkers, we’re looking at a range of biomarkers. It’s important to note there are no validated biomarkers in this disease. It’s not even guaranteed that we discover a validated biomarker in this disease. But obviously, it will be helpful as an evidence of pharmacology and the potential for benefit in clinical endpoints that we do try and measure those. And so, we’re looking at a range of biomarkers. We’ve described many of the ones that are associated with the disease. And that will be other data that we will have. As we pull together these first couple of cohorts and we have a cogent story to tell around them. And the balance of the clinical endpoints and the biomarker data together will help us decide whether we’ve found the correct dose to move forward or whether more work is needed to find more optimal dose for this patient population. I think the encouraging thing is, as we stand today, is that we do know there are patients that have been on drug for quite a long period of time. And that longitudinal experience allows us to look at clinical endpoints and also gives us some good indication, hopefully, where we’re going to be on safety, which is obviously essential for us to move forward.
Operator:
Our next question comes from Tyler Van Buren with Cowen.
Tyler Van Buren:
So, the Ontario study provides some interesting initial evidence that the fourth dose is beneficial. And you mentioned the populations that should get the fourth dose. So, how big in aggregate is this population in total? What percent of the U.S. and global population do these patient groups comprise? And the second question was just a follow-up on prior questions to make sure that I’m clear. Did you say that the average selling price per Spikevax dose in 2022 will be lower than ‘21 due to COVAX orders? And does this not account for a potential increased price from future U.S. orders? And could these offset that decline in ASP, especially if you sell a sizable portion of doses at year-end at that $60 per dose price I believe you just mentioned per the CMS announcement?
Stephane Bancel:
Good…
David Meline:
Yes. Go ahead.
Stephane Bancel:
Go ahead, David. You take the first one, and then I’ll comment on the...
David Meline:
Yes, sure. Yes. So, if I track that, in terms of pricing, that’s correct. We have had and we continue to have based on the customer mix this year versus last year, we had indicated that we’ll have a year-over-year decline in the average price of product being sold when we look at the $21 billion of APAs compared to last year’s actual. And the key contributor to that decline for the year is, in fact, the inclusion of COVAX volume in the confirmed APAs that we previously were discussing. So, that contributes to an average sale price decline. Will that sale price on average in 2022 change from that outlook? Yes, it can to the extent there were change in volume for COVAX, including if it were some of the volume were to defer beyond ‘22 into ‘23, for example, that would improve your average price calculation. And then, likewise, to the extent which we’ve indicated, we have an expectation of additional sales of product in 2022 for the fall season, including potentially for private market, should that develop. That would then presumably have a favorable impact on the average price. So, hopefully that is clear.
Stephane Bancel:
And just to comment on the number of individuals who we think are at high risk. Clearly, it can change as we see the vaccine -- we’ve seen with the epidemiology of the virus, as Stephen commented earlier. But just going back to our Vaccines Day earlier, we think that that high-risk population is somewhere in the range of about 1.7 billion people worldwide.
Tyler Van Buren:
Okay. Congratulations again, David, on your second retirement.
Operator:
Our next question comes from Cory Kasimov with JP Morgan.
Unidentified Analyst:
This is Tiffany on for Cory. So, as the U.S. government hasn’t procured a budget for boosters this fall and if they continue to not place orders, can you walk us through some of the implications of what that privatization potentially means and how the Company is thinking about it? You mentioned potentially higher prices, but anything from market research that might suggest demand change or how you’re thinking about discounts, et cetera?
Stephane Bancel:
Sure. Tiffany, this is Stephane. So, indeed, while we are in discussion with the U.S. government, as you would assume, is appropriate. We’re also working toward assuming that there’s no government order -- or American order for vaccines. We want to make sure we can protect as many Americans as we can on vaccine. And so with our commercial U.S. team, we’re working very diligently, and David and I have spent quite some time in the last weeks and months to make sure that we have all the wholesaler contracts and all the pieces you need to be able to be commercial in the traditional sense of the world in vaccine. And we have quite a number of executives that are vaccine self experienced in the U.S. on our team. And so, while we hope that the U.S. government, like other countries in the world, will decide to place an order, like they’ve done in the past, to allow the maximum number people, including people uninsured to get vaccinated, we’re getting fully ready assuming the day zero order from the U.S. government, just in case if that happens. This is a 100% private market in the fall and the Company will be ready for that.
Unidentified Analyst:
Okay, great. And then, just a second one. So, how should we think about operating expenses moving forward and potentially steering away from COVID as a big driver there? Will it be pipeline dependent or something else? Thank you.
David Meline:
Yes. It’s a good question. We’ve tried to give you the indication of the trends for expenses across the business. What I would expect to continue is a very significant investment in R&D as that pipeline progresses and expands. And I would say, it shouldn’t be surprising if you see -- if you move past 2022 that you’ll see a continued increase in the investments in that area, presuming success, which we’re feeling very good about. And then, in terms of the other considerations in terms of cost, in terms of SG&A and if you look at the business now, we’ve substantially built out the Company, as we’ve talked about, as a global commercial enterprise. So, I think we’re -- while perhaps not precisely there yet, we’re largely there in terms of if you look at the structure of the business globally. So, that I think you might consider to be more of a steady state. So, that would be my initial comments on the thinking on that.
Operator:
Our next question comes from Geoff Meacham with Bank of America.
Alec Stranahan:
This is Alec Stranahan on for Geoff Meacham. So, on BD, how do you think about partnerships versus acquisitions? And how large of a deal would you be willing to consider? And then, if I may, can you provide your thoughts on the RSV competitive landscape? And any color on time lines for the readout? Thank you so much.
Stephane Bancel:
Yes. So, this is Stephane. Partnership versus M&A, I think it’s really a question of risk and then willingness of sellers to sell because it needs be two to tango. And so, as we’ve done from the effort we met at Metagenomi, what we do a partnership in terms of licensing for Metagenomi is because we are very excited about the science and the team that we discovered over time and with diligence. We thought at this stage of the gene editing technology of that company, it was not the best thing for us to do from a risk-adjusted basis to acquire the company. And in terms of M&A, we would be very happy to buy the right company that we really believe will drive value to Moderna on commercial basis. In terms of deal size, we’re looking at a lot of different things. But again, we are staying very-disciplined. We are here to create value, not to do things just for the sake of doing things. As you know, a lot of us own a significant share in the Company, and we are already focused on creating value. So, the BD team, as I said, is most busy they have been in a long time. We’re looking at a lot of things, literally, around the world because the best science is not always in this country. There’s a lot of amazing science in this country, but there’s a lot of other smart people around the planet. And we think with Moderna’s infrastructure and capital actually has a lot of technology that we could potentially scale up where there’s commercial teams that have cool science, but not necessarily the right balance sheet and infrastructure to scale and to maximize patient impact. Stephen, do you want to take RSV?
Stephen Hoge:
Sure. So, on RSV, so first, where we are on the data of our program, 1345, we think the titers, the neutralizing antibody titers, GMTs, we’ve seen, look, really strong relative to competitors. And we also feel quite optimistic and positive about our history of generating strong T-cell responses against respiratory viruses. And the mRNA platform, in particular, has demonstrated, we think, through the COVID pandemic, pretty remarkable performance relative to more traditional approaches. And so, the combination has us optimistic. We ultimately don’t need to go demonstrate in a clinical trial that potential benefit. And that’s a Phase 3 study that we’re now in and going full speed at. That Phase 3 study, like our other respiratory virus efficacy studies, is a case-driven design. And so at the end of the day, we have to enroll people, vaccinate them and then make sure that we accrue enough cases to conduct the interim and final analyses. That is something that we’re trying to make sure we’re enrolling in geographies where we expect and anticipate RSV surges. And there have been recent reports from a competitive landscape perspective of others modifying their studies to increase the number of enrolled subjects in their pivotal studies because perhaps they haven’t yet hit that rate of case accrual. In some ways, we think that provides us an opportunity to close even more ground because now we’re all rapidly working to try and demonstrate the potential of RSV vaccines to help the same older adult population. So, it’s beyond our control to know when exactly we will have those cases accrued. But obviously, we’ll be working hard to make sure that we’re as enriched as possible. Now from an overall competitive perspective, other than the vaccine, the freestanding RSV vaccine in older adults, which as I said, we’re quite optimistic about the GMTs T-cell response to platform, we are also studying combos. Because at the end of the day, these are not the only virus -- it’s not the only virus affecting these populations. And so, I’ll remind you that we have a RSV flu COVID combo, which we are going to start a clinical study on, we believe, this year, which would be a combination of all of the three most common respiratory pathogens that are impacting older adults. And as I said, a moment ago, we’ve fully enrolled our RSV and human metapneumovirus combination, which I’ll remind you, that’s in the pediatric population, another important population for RSV, where both of those viruses can lead to morbidity in young children. And so, we are continuing down our strategy of making sure that we’re not just addressing one pathogen, but that we’re providing the best potential health intervention, so that includes that combination strategy. And over time, we think as we demonstrate the potential of our platform against any individual virus, the part of the real value we will deliver to health care systems and patients is the ability to do those combinations quickly to reflect the epidemiology, the underlying patient population is different in younger kids than it is in older adults.
Lavina Talukdar:
Kevin, we have time for one more question.
Operator:
Okay. Our last question comes from Joseph Stringer with Needham & Company.
Joseph Stringer:
I had one on the rare disease programs. And just curious, you have the PA in MMA and clinical development here, an initial readout in PA. How much is the PA readout? Would that be sort of derisking in terms of bringing additional rare disease programs into the clinic? Has it been more of a some of the hurdles and challenges to expanding that area of the pipeline? Is it then more on an indication-specific basis, or have you been sort of waiting for these initial proof-of-concept readouts to bring more programs into the fold, or has it been more of influenced by sort of the focus on COVID and potentially headwinds related to the COVID pandemic? Any additional color on that would be helpful. Thank you.
Stephen Hoge:
Great. Thank you for the question. And it is going to be a mix of all of the factors you listed. So first, let me talk about PA and what that readout means for us as a platform. So, it is our most advanced rare disease platform with an mRNA LNP targeted for those metabolic diseases. And in that sense, MMA and PA, both, will provide validation that the technology risk associated with that are addressed. And that would cause us, from a derisk perspective, from a strategy perspective to expand the number of diseases we go into quite quickly there that could use that same technology. We have been waiting for that readout. We haven’t sat on our hands though, although that we have a third program, GSD1a with an open IND that as we’ve said before, in a slightly -- is in a different lipid nanoparticle system. So, we’ve continued to look at whether other improvements could help. And we’ll continue to look at those. But obviously, if we have a very strong signal out of PA, we will be on gating other programs that can benefit from the technology that would then be derisked from the PA program. When it comes to just the challenges of conducting these studies, I’ll remind you that these are, unfortunately, very Ill children often in these studies and relatively rare diseases. And so, yes, it has been difficult throughout the pandemic in an experimental context to bring people in. And there have been several times during the last two years that we have actually taken an active decision with investigators and families, not to be enrolling people, because obviously bringing sick children into hospitals was exposing them to risks around SARS-CoV-2. They are obviously also even more recently in the Omicron surge. Lots of disruption that these institutions have faced as staff and others have become ill, and therefore, we can’t connect the studies. We hope that a lot of that is behind us now as we move to an endemic phase for SARS-CoV-2, and that that will -- that we will really see a pickup in our ability to continue to execute the study. So, we’re proud of the progress we’ve made in the last year.
Stephane Bancel:
Well, thank you very much for joining us today. We look forward to meeting a lot of you in person in Boston when we do the R&D Day two weeks from now. Have a great day. Thanks.
Operator:
Ladies and gentlemen, this does conclude today’s presentation. You may now disconnect, and have a wonderful day.
Operator:
Good morning. My name is Kevin and I’ll be your operator today. Welcome to Moderna’s Fourth Quarter and Full-Year 2021 Earnings Call. At this time, all participants are in a listen-only-mode. Following the formal remarks, we will open the call up for questions. Please be advised that this call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Kevin. Good morning, everyone. And thank you for joining us on today’s call to discuss Moderna’s fourth quarter and full-year 2021 financial results and business update. You can access the press release we issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our website. On today’s call are Stéphane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; and Paul Burton, our Chief Medical Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. With that, I will turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Welcome to our Q4 2021 conference call. Today, I will start by a quick business review of fiscal year ‘21 before Paul walks you through some real-world evidence data. Stephen will then review our clinical programs before David presents our financial results. I will then come back to close to share some thoughts about where we are heading. We are pleased to report today $18.5 billion of revenues for fiscal year 2021 and the GAAP net income of $12.2 billion, translating to a GAAP diluted earnings per share of $28.29 billion, (sic) [$28.29] with a cash balance at the end of the year of $17.6 billion. We announced this morning that we have completed our August 2021 $1 billion share buyback plan, which reduced our average share count during the fourth quarter for the first time in the Company history. I am very proud of our team’s ability to advance as well as expand with new programs our development pipeline. In respiratory vaccines, for COVID, as you know, we received full BLA approval from the U.S. FDA for Spikevax. With authorization for 12 to 17 years old in key markets globally, we are running a 50-microgram dose study in the U.S. for adolescents following discussions with the FDA. We are pleased to report that we have also started to receive authorization for children aged 6 to 11 years of age, beginning in Australia last week, with more countries expected in the days and weeks to come. We also dosed the first participants in our Omicron-specific vaccine trial, mRNA-1273.529. And today, we’re announcing that we plan to bring mRNA-1273.214, bivalent vaccine, combining the wild-type vaccine mRNA-1273 and the Omicron vaccine mRNA-1273.529 into one single dose booster into the clinic with a total mass of microgram. We are very excited that now RSV is in Phase 3. This is our third vaccine starting the Phase 3. And for flu, we are waiting the Phase 2 data for mRNA-1010. As soon as we have them, we will pick a dose and will initiate our Phase 3 for flu, and we will start our Phase 1 for COVID plus flu program, mRNA-1273. For latent viruses, CMV is enrolling in Phase 3, EBV and HIV are now in Phase 1, and we were very excited to announce last week that we’re adding two new latent virus programs to our development pipeline, one vaccine against HSV and one vaccine against VZV. More latent viruses’ vaccines are in the works in our labs. In therapeutics, PA and MMA are in the clinic dosing and recruiting more patients. We announced a new checkpoint cancer vaccine, mRNA-4359, and Merck has informed us that they are restoring the rights to the KRAS vaccine, while evaluating future steps for this program. The Company continues to expand at a rapid pace. We now have one approved medicine; two in Phase 3, RSV and CMV; and five in Phase 2, for a total of 44 programs. We are deploying capital to accelerate and expand the pipeline. The Company now has 3,000 team members across the world, with a great culture, and 11 commercial subsidiaries across Americas, Europe and Asia Pacific, while $17 billion cash balance at the end of the year is enabling us to scale across research, development, manufacturing, commercial and G&A. With this, let me now turn to Paul.
Paul Burton:
Thank you, Stéphane, and hello, everyone. It is almost exactly three months ago that we first heard about the [Technical Difficulty] And today, we look back on three months that have seen millions of new cases of COVID-19 infection worldwide due to Omicron and a death toll that turned out to be almost 20% higher than that seen during the Delta wave. It is clear that SARS-CoV-2 is a virus capable of making very large evolutionary leaps in its structure and function. And while we are hopeful that we are about to enter a period of relative stability in the northern hemisphere, we believe firmly that a vaccine booster dose that will be required for the fall of 2022 to provide ongoing protection against this virus. We have seen measures taken by the United Kingdom recently to offer an additional booster dose to those at higher risk. And we believe such measures will become more widespread with governments looking to protect their populations from disease later this year and support their healthcare systems. As always, a huge note of appreciation to the frontline workers, the healthcare workers who have continued to immunize us and protect us during this pandemic. Their tireless work has saved millions of lives worldwide. I’m going to review some data today that underpins why we believe an additional booster shot will be required certainly by the fall of 2022. First, let me share with you some data from a study of almost 440,000 individuals in the United States Veterans Administration database, looking at the effectiveness of mRNA-1273, Spikevax used in the primary series vaccination setting. As we know, the mRNA vaccines are extremely safe and effective. And the data here shows the exceptional effectiveness of mRNA-1273. In this well-powered and prospectively designed comparative effectiveness study, mRNA-1273 significantly reduced the risk of COVID-19 infection and hospitalization compared to BNT162b2. These data are reassuring and continue to underpin the strong global confidence that governments, healthcare systems and individuals have in mRNA COVID-19 vaccines and mRNA-1273. Indeed, an example of that is the very recent announcement by the CDC at British Columbia for the preferential use of the Moderna vaccine in anyone over 12 years of age living with immunocompromised [Technical Difficulty]. A similar pattern of results is seen from real-world effectiveness studies in the United Kingdom. The graphs I’m going to show you here come from the United Kingdom Health Security Agency, looking at the effect of either the Pfizer-BioNTech or Moderna vaccines and their vaccine effectiveness against hospitalization. First, let me show you the results in people who for their primary vaccination received the Pfizer-BioNTech vaccine and were then boosted with either the Pfizer-BioNTech vaccine or the Moderna vaccine. We see retained protection against the Delta variant of concern in the field squares, but we see waning of effect in the field circles against Omicron. The same is true, though more pronounced, on the next part of this graph. And here, you see individuals who received as their primary vaccine the AstraZeneca vaccine. Again, in this very large real-world assessment, we can see that mRNA-1273 provides protection against hospitalization due to both, Delta, but to a lesser extent, Omicron infection. Of note, we do see waning of protection over time against hospitalization due to Omicron infection. And this fits with the profound immune evasion we know to be the case with Omicron. But it is also a driver of our belief, not only will a booster vaccination be required later in 2022, but also a booster will be required that protects against both, the Delta and Omicron variants of concern, as Stéphane announced earlier. This is because Delta, as we know, is associated with strong pathogenicity, and Omicron, as we have seen, due to its transmissibility and infectivity, is also associated with substantial morbidity and strain on healthcare systems through sheer bulk of cases, hence, protection against both Delta and Omicron may well be necessary in the next booster vaccination. On slide 10, we see data directly explaining the clinical phenomenon I just showed you. These data come from one of our previously conducted clinical studies. Sera from individuals immunized and boosted with mRNA-1273 is able to substantially neutralize the ancestral SARS-CoV-2 virus, D614G, shown here in black, but to a lesser extent, Omicron, shown in red. And this is particularly true by six months following booster administration. Again, it is these data, coupled with the real-world effectiveness data that I just showed you, that leads us to believe an additional booster dose will be needed later in 2022, providing protection against both, Delta and Omicron variants of concern. You can see this point made clearly again in slide 11, where we estimate the duration of protection against either the ancestral D614G strain or against Omicron. With lower starting titers, protection against Omicron wanes and forward to levels that would begin to allow breakthrough infection to occur due to Omicron and around 9 months after boosting. As leaders in mRNA vaccines and COVID-19 disease, we will continue to monitor this carefully along with other emerging variants and continue to do whatever we can to provide people with a safe and highly effective vaccine booster for 2022 and years to come. We announced in January that we were able to go from identification of Omicron to testing of a new vaccine in humans in just two months. That is a remarkable testament to the Moderna mRNA platform and its utility. Slide 12 will be familiar to many of you as we first presented this slide at our Vaccines Day in April 2021. The graph is an illustration of the different phases of the pandemic to endemic continuum and its effect as measured by morbidity or disease burden over time. The shape of the graph was adopted from what we knew about previous pandemics at the time and what we expected from the current pandemic, taking into consideration the increased levels of immunity that our immune systems collectively became familiarized with the SARS-CoV-2 virus, either through infection or vaccination. As the graph shows, and with hindsight, it is easy to recognize that the very first wave of the pandemic, when all of the world’s population was naïve to this virus, caused the highest levels of morbidity and mortality. With each subsequent wave in mid-2021 with Delta and in late 2021 and early 2022 with Omicron, the morbidity observed from these waves tended to be less severe, certainly relative to the first wave, as their immune systems became more experienced at fighting the SARS-CoV-2 virus. Beyond 2022, there still remains uncertainty. However, given what we know now of other pandemics, like the 1918 influenza pandemic and more importantly, what we have observed with the SARS-COV-2 pandemic, we do believe that we are transitioning into an endemic phase, marked by a period of stability in case counts, hospitalizations and death, at least in the Northern Hemisphere. We will continue to carefully monitor the situation in the southern hemisphere as winter approaches there on a background of continuing Delta and Omicron infection pressure. And we will work as quickly as possible to generate new specific booster vaccine data. In the endemic phase, the virus will continue to circulate but at rates that are static and more predictable. We continue to expect morbidity from the virus when it is endemic. And we believe, like other respiratory viruses, we will see a seasonal pattern of disease burden emerge. But the good news is, because our immune system is now much more familiar with the virus, either through vaccination, infection or both, we do not expect that the majority of the population will be as susceptible to severe disease in the endemic phase as they were in the pandemic phase. Instead, as with other respiratory viruses, we believe protection from SARS-COV-2 will be critical for populations that are likely to be most susceptible to severe disease and high burden of disease. Providing the broadest protection possible will be a focus for us in the endemic phase, and we believe, as Stéphane mentioned, multivalent vaccines against SARS-COV-2 and its variants will help achieve this. As I alluded to earlier, respiratory viruses cause significant disease annually, and endemic coronaviruses are no exception to this. Slide 13 shows the incidence of community-acquired infections leading to hospitalizations in New York City during the 2018 to 2019 season prior to the SARS-CoV-2 pandemic. On the chart, you can see that other endemic human coronaviruses, in fact, cause the highest level of hospitalizations in individuals between 65 and 79 years of age and those 80 years and above, followed by RSV and influenza. Across OECD countries, the estimated impact from endemic human coronaviruses leads to over 1 million outpatient visits, 350,000 hospitalizations and 20,000 deaths in the over 65-year-old population annually. We believe SARS-CoV-2 will follow a similar pattern of seasonal disease as other respiratory viruses do and will impact vulnerable populations. Protecting populations against SARS-CoV-2 will be critical in the months and years to come. And protecting against combinations of respiratory viruses such as SARS-CoV-2, flu and RSV with a single yearly shot will be central to our strategy. Stephen Hoge will shortly provide an update on our robust ongoing strategy to achieve that. So, in summary, the available real-world data continue to show the remarkable effectiveness of mRNA-1273. Real world data shows that a 50-microgram booster dose of mRNA-1273 provides protection against hospitalization caused by Omicron, but we note waning of antibody titers by six months post boosting. We believe that a full 2022 booster will be needed globally. And as leaders in mRNA vaccines, we believe it is important to develop booster vaccines that will give us an opportunity to protect against Omicron and future variants as we continue our fight to help end this pandemic. I will now hand over to Dr. Stephen Hoge to provide further updates on this topic and the Moderna pipeline. Stephen?
Stephen Hoge:
Thank you, Paul. Good morning and good afternoon, everyone. On slide 16, I’d like to briefly summarize our COVID-19 booster development strategy for the endemic phase. So, as Paul covered, the strategic rationale for a seasonal booster has three parts. First, we think neutralizing titers will wane, similar to the endemic human coronaviruses, as Paul just described. That decline in neutralizing titers will increase the risk of breakthrough hospitalization in those at higher risk, specifically including older adults and the immune compromised. We think the emergence of new variants of concern will also have the risk of accelerating waning and broadening the risk of breakthroughs to other populations. So, the desired features for our Northern Hemisphere fall and winter 2022 booster are described here. First, we’d like to improve the durability of protection for neutralizing antibodies against Omicron and Omicron mutations to at least six months that will provide full protection through the Northern Hemisphere fall-winter infection season. We’d like to retain the high and durable protection we’ve been seeing with a prototype vaccine against Delta and the ancestral strains. And third, we’d like to broaden cross-protective immunity to the extent possible to increase the potential for protection against a new emergent variant of concern, which could emerge perhaps from the Southern Hemisphere this midyear. So on slide 17, I’ll quickly summarize our strategy for developing an updated booster for fall 2022. We are currently evaluating three different booster strategies in adults age 18 plus. The first, as both Paul and Stéphane have mentioned, is a bivalent booster vaccine, made up of the prototype mRNA-1273 and an Omicron-specific mRNA-1273.529. This bivalent has been called 214. We are also evaluating, as we previously announced, an Omicron-specific booster, mRNA-1273.529. And of course, we will continue to evaluate our prototype booster, mRNA-1273, for which there is a large body of real-world evidence, as Paul just described. We’re evaluating these three different approaches across two studies in the United States and the United Kingdom, the Phase 2 study in the United States of approximately 750 participants and a 3,000-participant study in the United Kingdom. Both are looking at both bivalent and Omicron-specific boosters. And both will be looking at both, third and fourth dose of those boosters. In the UK study, we will also be looking at heterologous boosting, including on the background of other mRNA vaccines and non-mRNA vaccines. Now, I’d like to take a moment on slide 18 to provide some scientific insight to why we believe the bivalent vaccine booster for the fall of 2022 offers a potential advantage. Slide 18 includes data on some of our prior bivalent boosters. And this data -- this emerging data suggests to us that there may be an opportunity to improve durability against variants of concern while preserving activity against the ancestral variants. This data is based on one of our prior bivalence, mRNA-1273.211, or 211 for short. And 211, as you may recall, was based on the 1273 vaccine and the Beta variant of concern, which emerged approximately a year ago. When we compare over time how the two different booster strategies, our prototype booster on the left-hand column here, and our bivalent booster on the right-hand column here, do against the two different virus variants that were in the vaccine, you see an emergence of potential improvement in durability. So, first, to orient you to the slide. mRNA-1273 was given at 50 micrograms as a booster to those who had previously received two doses of the mRNA-1273 vaccine. And on the top left panel, you’ll see how the neutralizing titers or pseudovirus neutralizing titers against the ancestral strain of the virus, the D614G virus, in our validated clinical assays. Underneath there, you’ll see how those -- the pseudovirus neutralization titers against the Beta variant of concern in blue. And in the middle column, you’ll contrast that with a bivalent 211 booster, which, in this case, included the Beta variant of concern, again, at the same dose level, 50 micrograms. Top right panel is the ancestral variant of concern, ancestral virus, D614G, and lower right is the Beta variant of concern or the B1.351. Now, as you’ll note, comparing the performance against the ancestral D614G virus, both mRNA-1273 and the bivalent Beta containing booster do a good job boosting neutralizing titers by day 29 after the booster. So, one month post booster, neutralizing titers are approximately 1,800 for 1273 and 2,200 for the bivalent. And importantly, as we test the serum six months after booster, noted as day 181 here, you’ll see that neutralizing titers remain high, approximately 1,000 in both boosters. Now, the situation against the variant of concern, in this case, Beta, is slightly different. Again, both boosters, increased neutralizing titers by day 29 one month after booster to quite reasonable levels, approximately 1,000 in both cases. However, when you follow out six months, there is a difference in the neutralizing titers that emerges. And not surprisingly, the bivalent booster, which includes the Beta variant of concern, starts to see more durable neutralizing titers, 402, as noted here, as opposed to 154 for mRNA-1273. So, in summary, six months after a 211 bivalent booster, the neutralizing titers against the Beta variant of concern appear to be more durable than with just a prototype booster. And the durability or the rate of decline for the Beta neutralizing titers more closely matches that seen for the ancestral virus following the bivalent 211 booster. Including the Beta variant of concern, therefore, appears to be improving the durability of neutralizing titers against that variant of concern. Now on the next slide, slide 19, we have that same data, but now plotted as a function of time to help you visualize it a little more clearly. On the left-hand side, we’re contrasting the mRNA-1273 booster as a black line against a bivalent Beta containing booster in the red line. On the left-hand side, you’re again looking at the ancestral D614G neutralizing titers. And as you can see, following a booster approximately six months after booster dose two, neutralizing titers for both of the boosters, both bivalent and the prototype vaccine, increased significantly. And the data we have out to six months, if you project that forward as we do with the dotted lines, suggest that we will maintain quite high neutralizing titers, perhaps as long as one year. Now, the situation on the right-hand side highlights we think the potential for improving durability with a bivalent vaccine. Again, here, the black line is mRNA-1273 and the red line is the bivalent Beta containing booster at the same dose level. Now, while both boosters increase neutralizing titers to approximately 1,000 within one month of boosting, what starts to emerge six months later is a different degree of durability in those neutralizing titers, with the bivalent vaccine containing the Beta antigen doing slightly better, as you can see with the red line. And if you project that forward, it suggests that the bivalent vaccine neutralizing titers against the Beta variant concern will remain quite high, perhaps as long as a year, whereas with a prototype vaccine, those neutralizing titers appear to be decaying more quickly, back towards baseline levels or pre-booster levels by approximately 8 to 9 months. On slide 20, just to quickly summarize therefore where we are in COVID-19 booster development for fall 2022. We believe that a seasonal booster will be necessary to prevent breakthrough diseases, including hospitalization in vulnerable populations. And we believe that the continued evolution of the virus is going to continue to put pressure on pre-existing immunity, whether that’s naturally derived or vaccine provided. We think the fall 2022 booster should reflect the diversity of circulating mutations that are out there in order -- and seek to achieve greater than six months of neutralizing titer durability to increase the potential for protection throughout the entire fall season, in this case, we think September through February. So, Moderna is developing an Omicron-containing bivalent booster, based on data that we have from prior bivalent candidates that suggests that incorporating the variant of concern of those mutations from that variant of concern has the potential to improve the durability against such variant of concern. Now, moving to slide 21, just quickly catching up on other developments in our COVID-19 vaccine. We have made progress in primary series and booster in adolescent and pediatric populations. So, in adolescents first, we’ve received regulatory approvals for Spikevax in Europe, United Kingdom, Australia, Canada and many other countries. In U.S., we plan to submit an EUA for 100 micrograms of mRNA-1273 in adolescents that are immune compromised or an elevated risk of severe outcomes. And we’re also evaluating the potential of a lower dose, 50 micrograms, as a primary series. And lastly, as has been noted previously, we’re preparing to submit data for 50 micrograms as a booster dose in adolescents and 50 micrograms as the booster dose in adults as well. And that will include data on heterologous boosting. In pediatrics, or the 6- to 11-year old, we received provisional approval for Spikevax in Australia and submitted to multiple other international regulatory agencies and expect authorization shortly. The U.S. submission is pending alignment with the United States FDA on the adolescent application. And we will also continue to evaluate lower doses, including a 25-microgram primary series dose. Finally, in the youngest, 6 months to 5-years old pediatric population, we expect data on our 25-microgram two-dose primary series in the first quarter. And pending that data, we’ll plan to submit to regulators. We are also continuing to evaluate lower doses and the potential of a third dose in that population. Pivoting to the broader respiratory vaccine portfolio on slide 22. Beyond COVID-19, we continue to make progress across all of our respiratory vaccines. As Stéphane mentioned, our flu vaccine is fully enrolled with Phase 2. And pending that data, we will prepare to move forward, which we still anticipate doing in 2022 into a Phase 3 study. We’re also preparing to start a combination flu and COVID vaccine, which is currently in preclinical, but we expect to start the Phase 1 study this year. Our older adult RSV program has started its Phase 3 study portion, and that pivotal study is ongoing enrolling. We have a pediatric RSV study, which we will move forward in Phase 1. We have also two different respiratory combination vaccines. First, the human metapneumovirus parainfluenza virus 3 vaccine, which is in Phase 1b and has now fully enrolled, and an RSV plus hMPV vaccine, which remains in preclinical development, and we hope to start shortly. Moving now to latent and public health vaccines on slide 23. As discussed, CMV continues to enroll in the Phase 3 CMVictory study. We have also moved forward two new latent virus vaccines in the clinical testing, our EBV vaccine to prevent infectious mononucleosis is in Phase 1, and our HIV -- our first HIV vaccine, mRNA-1644, is also in Phase 1. We announced two new development candidates, which I’ll cover briefly in the next couple of slides, against HSV and VZV. And our public health vaccines against Zika and Nipah continue to progress. Briefly on slide 24, I’d like to introduce our -- the first of our two new development candidates in this space. mRNA-1608 is our vaccine against Herpes simplex virus 2. HSV-2 primarily infects the genitals and establishes lifelong latent infections within the sensory neurons. There is a significant burden of disease in developed markets, including approximately 18 million people who are HSV positive, HSV-2 positive in the United States. Globally, that represents about 5% of the population. Primary burden of disease is a reduction in quality of life from recurrent lesions. Our mRNA-1608 vaccine encodes antigens on the surface of the HSV virus and has been able to induce very strong immune responses, as illustrated in the figure to the right. Neutralizing titers in mice following an HSV-2 vaccine with mRNA-1608 are significantly above the levels seen in human sero from those who are seropositive. This gives us reason to believe that we will be able to provide a significant benefit with this vaccine in this population. The second development candidate is on the following slide, slide 25. This is our mRNA-1468 program against herpes zoster or shingles. Herpes zoster is caused by the reactivation of latent varicella-zoster virus, or VZV for short. It’s principally a disease that’s seen as a result of declining immunity in older adults, where protection against VZV decline, leading to reactivation of the virus and painful and very itchy lesions. Herpes zoster occurs in about one out of three adults in their lifetime. And the incidence is increasing as populations age, and particularly increases over the age of 50. On the right-hand side is previously published data in the Journal of Vaccine on our VZV vaccine, in this case, mRNA-1468. Our vaccine in nonhuman primates was compared against the protein and protein plus adjuvant, as they stand in, a proxy for the shingles vaccine, which is already approved for this indication. As you note on the right, in nonhuman primates, the mRNA vaccine against gE resulted in significant and elevated neutralizing titers after two doses, and we believe will provide the basis for a strong potential clinical benefit with mRNA-1468. Moving now to our therapeutic pipeline. We continue to make progress across a range of different programs. On slide 26, I’ll note a few very quickly. First, our PCV program in Phase 1 is ongoing and the Phase 2 is fully enrolled. We expect data in the fourth quarter of 2022. We are also going to provide a bit of an update on the checkpoint vaccine and newly announced development candidate in just a minute. Highlighting in other therapeutic areas, our VEGF program continues to move forward in Phase 2 with AstraZeneca. And in rare diseases, our PA and MMA programs continue to enroll in their Phase 1, with a Phase 1 -- first dose level cohort fully enrolled in PA and continued enrolling of additional cohorts. We also continue to make progress across all of our other preclinical programs in rare diseases, including GSD1a, PKU, CN-1 and the cystic fibrosis program with Vertex. On slide 27, I’d like to briefly cover our latest development candidate, a checkpoint vaccine to promote anti-checkpoint T-cell responses in cancer, otherwise known as mRNA-4359. So briefly, the objective of this program is to stimulate effector T cells that target and kill suppressive immune in cancer cells that express high levels of target checkpoint antigen. We previously identified that there are pre-existing IDO and PD-L1 specific T cells that have been identified in cancer patients and tumors. IDO and PD-L1 specific T cells can kill and remove the immunosuppressive regulatory immune cells and cancer cells that overexpress these antigens. It’s an important counterbalance that helps liberate the immune response against the tumor. Our vaccine can expand IDO and PD-L1 specific T cells in preclinical models. And the vaccine induced direct tumor cell killing can facilitate recognition of tumor-associated antigens by other cytotoxic T cells, leading to more broad tumor killing. Systemic blockade with PD-1 or PD-L1 antibodies may further amplify this effect. We will initially be developing mRNA-4359 against indications, including first-line cutaneous melanoma stage IIIb and first-line non-small cell lung cancer. With that, I’d like to turn it over to David to walk you through the financials.
David Meline:
Okay. Thank you, Stephen. We are providing today the analysis of actual 2021 fourth quarter and full year results along with a view of key drivers of financial performance going forward. 2021 was a transformative year for the Company as it marked the transition from an R&D-focused entity to a commercial stage company. I’m very pleased with our performance, and wanted to thank all of our employees at Moderna for their dedication and response to the many challenges during this unprecedented company scale-up. Turning now to slide 29, starting with an overview of our sales performance. Total product sales in the fourth quarter of 2021 were $6.9 billion, representing 297 million doses delivered to our customers. This compares to sales of $4.8 billion for 208 million doses in Q3 and 199 million doses in Q2. We increased our dose supply in the fourth quarter by 43% compared to Q3 after a relatively stable picture in Q2 and Q3 as we successfully focused on removing bottlenecks in our supply chain network. Sales of our COVID vaccine have shifted in terms of geographic mix over the course of the year, in line with our expectations and the ramp-up of our international manufacturing capabilities. Sales outside the U.S. to the rest of the world were $6.1 billion in the fourth quarter, reflecting 252 million doses. And sales to the U.S. government were $0.7 billion in the fourth quarter, reflecting 45 million doses sold. For the full year, we sold 807 million doses, resulting in product sales of $17.7 billion. We generated sales of $5.4 billion in the U.S. and $12.3 billion with customers in the rest of the world. Approximately 25% of our delivered doses went to low and middle income countries, either through direct sales or facilitated by donations from other customers. Turning to slide 30 to go into more detail of our Q4 results. The transformation of Moderna from an R&D-focused biotech company to a commercial-stage business continues to be apparent when reviewing our financial results. The comparison of the fourth quarter of 2021 to prior year is not very meaningful due to the significant growth, which is why I primarily focus on the quarter-over-quarter comparison relative to Q3 on this slide. Total revenue was $7.2 billion in the fourth quarter of 2021 compared to $5 billion in the third quarter and $0.6 billion in the prior year period. The increase of total revenue was driven by the sale of the Company’s COVID-19 vaccine. Product sales in Q4 2021 were $6.9 billion compared to $4.8 billion in the third quarter, an increase of 44%. Cost of sales was $952 million or 14% of company’s product sales in the fourth quarter compared to $722 million or 15% of product sales in the third quarter. The quarter-over-quarter percentage improvement was driven by favorable manufacturing cost as the average selling price remained relatively stable. Research and development expenses were $648 million in the fourth quarter compared to $521 million in the third quarter and $759 million in the same period in 2020. The higher spend versus prior quarter was primarily driven by increased clinical trial expenses from our expanding and maturing development portfolio. The decrease in spending compared to 2020 was mainly due to the fact that the prior year number includes approximately $200 million of prelaunch inventory costs. Selling, general and administrative expenses were $201 million for Q4 compared to $168 million in the prior quarter and $79 million for the same period in the prior year. The growth in spending was driven by the commercialization of our COVID-19 vaccine globally, with continued investments in personnel and outside services in support of the accelerated company build-out. Provision for income taxes was $542 million in the fourth quarter, following $219 million in the third quarter and an insignificant amount in the prior year. Our effective tax rate for the fourth quarter was 10%. The quarter-over-quarter increase was primarily driven by higher earnings. Let me remind you of the fact that we had a net operating loss carryforward of $2.3 billion at the end of 2020. In 2021, we released the valuation allowance against the related deferred tax assets, which resulted in a nonrecurring full year benefit to our effective tax rate of about 5 percentage points. We recorded net income of $4.9 billion in Q4 compared to $3.3 billion in Q3, an increase of 46%. This compares to a loss of $0.3 billion in Q4 of last year. Diluted earnings per share for Q4 2021 were $11.29. Turning now to full year financial results on slide 31. Total revenue was $18.5 billion for the full year 2021 compared to $0.8 billion in 2020. The significant growth was driven by the sales of 807 million doses of the Company’s COVID-19 vaccine, resulting in product sales of $17.7 billion. Cost of sales was $2.6 billion or 15% of the Company’s product sales in 2021, including third-party royalties of $641 million. A portion of the inventory costs associated with this year’s product sales was expensed as prelaunch inventory in 2020. If inventories sold for the full year was valued at cost, our cost of sales for the period would have been $2.8 billion or 16% of product sales. Research and development expenses were $2 billion in 2021 compared to $1.4 billion in 2020. The growth in spending in 2021 was driven by clinical trial expenses for our expanding pipeline and the related organizational build-out. Selling, general and administrative expenses were $0.6 billion for the full year 2021 compared to $0.2 billion in 2020. The growth in spending in 2021 was mainly due to increases in consulting and outside services, personnel-related costs, marketing expense and distributor fees, primarily attributable to the Company’s COVID-19 vaccine commercialization-related activities and increased headcount. Provision for income taxes was $1.1 billion for the full year 2021 compared to an insignificant amount in 2020. The effective tax rate in 2021 was 8%. It was lower than the U.S. statutory rate, primarily due to a nonrecurring benefit related to the release of the valuation allowance, the ongoing benefit of the foreign derived intangible income deduction as well as benefits related to stock-based compensation. Net income was $12.2 billion for the full year 2021 compared to a net loss of $0.8 billion in 2020. Diluted earnings per share were $28.29 for the full year 2021. Turning to cash and cash deposits on slide 32. We ended 2021 with cash and investments of $17.6 billion compared to $15.3 billion at the end of Q3. The increase is driven by our commercial activity. The balance of cash deposits for future product supply was $6 billion at the end of the year compared to $6.7 billion at the end of Q3. The reduction quarter-over-quarter is driven by commercial deliveries against our commitments. Now, turning to slide 33. Our capital allocation priorities remain unchanged. Our top investment priority has been and will continue to be reinvesting in the base business across multiple areas. For R&D, we have significantly increased our spending in 2021 to approximately $2 billion. And we expect to continue to further increase our spending in this area to advance and accelerate our pipeline, both for existing and new programs. We are also further increasing our investment into our global manufacturing network in digital, automation and AI, as well as scaling up our global commercial operations. Our second investment priority is to seek attractive external investment and collaboration opportunities to further expand the reach of Moderna’s technology and capabilities. We are considering attractive opportunities that enable and complement our platform and take a disciplined approach in evaluating potential outside investments. Our announced collaborations with Metagenomi and Charisma Therapeutics fall into this category. After evaluating internal and external investment opportunities, we then assess additional uses of cash. We announced a $1 billion share buyback program in August of last year, which we completed in January of this year. As part of today’s press release, we announced that the Board has authorized a new share buyback program of $3 billion. Similar to last year, we provide you with a financial framework for 2022, which you will find on page 34. We have signed advanced purchase agreements for expected delivery in 2022 in the amount of approximately $19 billion and signed option agreements for delivery in 2022 of approximately $3 billion on a probabilitized basis. In 2022, we believe that the SARS-CoV-2 virus will evolve to an endemic phase. And as a result, we expect the timing of sales to be larger in the second half of 2022 than the first half. Our total cost of sales includes the cost of goods manufactured, third-party royalties, as well as logistics and warehousing costs. For the full year 2022, we expect a cost of sales ratio in the low to mid-20s percent range. The increase compared to prior year is driven by an expected increase in manufacturing costs as well as a decrease in expected average selling price per dose. The forecast increase of manufacturing costs is primarily driven by higher costs for fill/finish activities due to an expected shift from pandemic pack sizes to smaller dose and vial presentations. The decrease in average selling price is driven by the forecast increased deliveries to low-income countries. For R&D and SG&A expenses, we expect full year expenses to be approximately $4 billion, driven by our maturing development portfolio and the global scale-up of the Company. Based on current tax laws, we expect our 2022 tax rate to be in the mid-teens as a result of the benefits from foreign-derived intangible income, driven by our international business mix and stock-based compensation deductions. Finally, regarding capital expenditures, we are planning for capital expenditures in the range of $0.6 billion to $0.8 billion as we further build out our manufacturing and general company infrastructure globally. This concludes my remarks concerning the financial performance. And I now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, David, Stephen and Paul. Let me now share some thoughts about where we’re heading. I am pleased to see how the team is executing on our product strategy. Priority number 1, pan-respiratory annual booster. RSV is already in Phase 3. We’re waiting for flu data to start the Phase 3 on flu and start the Phase 1 on a COVID plus flud candidate, mRNA-1073. Priority number two, latent viruses vaccines. CMV in Phase 3, now we have 5 candidates, including VZV against shingles, and more coming. Priority number three, therapeutics, will be new checkpoint cancer vaccine, and priority number 4, expanding our unique mRNA platform to create new medicine. As I shared in January at the JPMorgan Healthcare Conference, there was a big change in commercial momentum from Moderna between early 2021 and early 2022. In early 2021, vaccine [ph] looked the same after Phase 3 data, and now we see strong real world evidence that Spikevax has long duration or efficacy. In early 2021, we were supply constrained. And now, we’re getting to a place, we continue to scale manufacturing, thanks to investments made last year and are less supply constrained. We have contracts with many governments around the world. But in early 2021, we had few team members on the ground in most countries. On slide 37, you can see that Spikevax booster market share has increased across key markets. You see that shift in market share data from around the world where we have teams on the ground. Even in Germany, where there is a national mRNA champion, we have moved our share of the booster market from 4% in October 2021 to almost 40% in January 2022. And it is also clear that OECD countries or high-income countries have become de facto in mRNA market. We have continued to increase our signed APAs to now around $19 billion. We also have approximately $3 billion in probabilized options. We continue to have numerous discussions with governments and NGOs around the world. For example, the current U.S. contract has its last shipments coming before the summer of 2022. This means that in the $19 billion plus $2 billion option, there is currently no APAs for the U.S. for the second half for 2022. As David mentioned, when we shifted to endemic, we expect to seasonality in sales. This year, we expect to see continued primary vaccination and boosting the Southern Hemisphere in the first half, and a shift to boosters as a fourth dose booster in the Northern Hemisphere in the second half of the year, similar to flu vaccines. We were pleased to announce last week a big expansion of our commercial network. From our current 11 countries where we have Moderna commercial teams on the ground, we announced an additional 10 countries, in Europe, Poland, Netherlands, Belgium, Sweden, Norway and Denmark; and in Asia, Malaysia, Taiwan, Singapore, Hong Kong. You have seen the impact of our strong real-world evidence data, coupled with teams on the ground, drive greater Moderna market penetration. I believe the same phenomena will happen in those 10 new markets. We have a direct commercial presence in these 21 important markets. We will maximize the impact of Spikevax and also have the teams to launch our pan-respiratory annual booster or latent virus vaccines in the rest of the portfolio. That commercial coverage and capabilities will prove critical in our ability to maximize our impact on patients and the resulting value creation. We now have distributors in Central and Eastern Europe. We have distributors in Asia Pacific, and with this week announcement, in Latin America. We, of course, continue to work with COVAX to provide access to a vaccine to low-income countries. Strategically, we want to create a new business model with governments around our pan-respiratory annual booster. We have started to create a subscription or service model with governments around the world. We have announced memoranda of understanding with the governments of Canada and Australia. We are currently in discussions with several other countries. These are 10-year agreements for the supply of a pan-respiratory annual booster. We have also governments which have signed APAs for 2023, as you can see on the slide. Numerous discussions are ongoing as we speak about securing supply for 2023 for the endemic setting to protect people at risk, 50 years and above, people with comorbidity factors, people who stuck with them at risk and adults who simply do not want to get severe disease. As we shared in the past, this is our strategy for capital allocation. As David said, the priority number one is and remains to invest in the Company. We have this unique mRNA platform. And we have $17 billion to invest to grow our pipeline to launch new medicine and to expand the capabilities of our platform. Priority number two is to expand the platform by in-licensing or M&A as we see new interesting nucleic acid technologies that can complement and strengthen the Moderna platform. Priority number 3 is to return capital to shareholders. After completing August 2021 $1 billion share buyback plan and reducing our share count for the first time, we announced this morning a new $3 billion share buyback. As we grow Moderna, we care deeply about building the right company, which is a responsible company to its community. In 2021, 25% of doses we shipped were to low and middle income countries. We’re investing in Moderna Science Center in Cambridge and launching an AI Academy. We plan to achieve net zero carbon emissions globally by 2030. We’ve announced Moderna Charitable Foundation and the global fellowship program. And we announced plans to invest up to $500 million in manufacturing facility in Africa. We hope to be able to announce some positive developments soon about these exciting projects. We will continue to push the boundary of corporate social responsibilities and share this with you. Before taking your questions, we’d like to remind you of 2022 events. March 24th will be our Annual Vaccine Day; May 17th will be our Annual Science Day, where we’ll present new platform advances; September 8th will be our Annual R&D Day where we’ll present development pipeline key updates. And today, we’re announcing that we’ll be hosting our first ESG Day on November 10th. I would like to close by sharing how excited we are about the future of our company. Because mRNA is an information molecule, we always knew this will be a company with a fewer [ph] drug approach. Well, now we know which future it will be. We are passionate about our ability to have a profound impact on humanity. We believe nobody should be hospitalized because of a respiratory virus. We have a technology to do that. We believe nobody should have medical consequences short term or long term because of a latent virus. We have a technology to do that. We believe we can have a profound impact on disease treatments with our therapeutics first and then our gene-editing programs. This is just the beginning. Operator, we’ll be happy to take your questions.
Operator:
[Operator Instructions] Our first question comes from Salveen Richter with Goldman Sachs.
Salveen Richter:
Good morning. Thanks for taking my question. Outside of the flu data that we’re going to see this year, we’re going to get personalized cancer vaccine data as well as rare disease data. Could you just speak to, in rare disease, what success would look like and what the regulatory path there could be? And then, for the personalized cancer vaccine, whether we would be able to get a sense of proof of concept this year?
Stephen Hoge:
Sure. Thank you for the question, Salveen. So first, in the rare disease space, it’s important to recognize that these are our Phase 1/2 studies, both the PA and MMA study. And I’ll speak to the PA study mostly because, as I said a moment ago, it’s the one that is -- has enrolled its first cohort and is moving forward with enrollment. So, first and foremost, we are going to be looking at safety in these studies, as you’d expect from a Phase 1/2. And so, one of the most important things to establish is, can we continue to dose, unfortunately, very ill people, or children in the case of both of these studies with mRNA, LNPs for up to six months or even longer, if they stay on the open-label extension. And establishing the safety of the platform on chronic dosing repeatedly over 6 to 12 months is an important objective of that part of the study. When it comes to efficacy, again, these are early studies and small in number as usually is the case with rare diseases. And so, I have to be careful about interpreting any of the data from the early clinical reads too concretely. But the things we’ll be looking for, first and foremost, we’ll be looking at the performance of the medicines in terms of preventing clinical outcomes. And so, in the case of propionic acidemia, these will be major metabolic decompensation events or hospitalizations that do happen with some regularity, unfortunately, for those folks who suffer from these rare diseases or disease like PA. And we will also be looking at biomarkers, and so specifically, biomarkers that have correlated with preclinical disease, and perhaps to a lesser extent, with some of the existing transplant-based therapeutic interventions in these diseases. But, it’s important to note that across all of these things in the biomarker space, there are not, unfortunately, validated biomarkers for these diseases because they have not yet been therapeutics approved. And so, it will be a balance between looking at efficacy signals early, to be fair, potential signals of improvements in clinical outcomes, and looking at biomarkers across a relatively small number of individuals. And so, there will be heterogeneous disease, both genetically and in terms of their performance. Our hope is when we get composite of data that’s clear, that we’ll be able to then present that and have discussions with regulators, which is the second part of your question, and have a discussion about what outcomes matter most for a potential pivotal study if we continue forward. But we do believe that the things that will ultimately matter most are clinical outcomes, and so, the prevention of major decompensation events and hospitalizations, and obviously, although the studies will be small in patient number, perhaps even hospitalization and death. So, we’ll be looking for those sorts of signals from our early studies. But it’s important to recognize, as I said a moment ago, these are relatively small number, and we’ll want to be circumspect in how we move forward into those efficacy studies. And those regulatory consultations will be based on those data, and we’ll provide updates when we have it. On the PCV program, as you said, we have completed enrollment in the Phase 2 study of PCV. And we expect that that PCV -- as a reminder, that is a head-to-head comparison of KEYTRUDA alone versus KEYTRUDA plus vaccine. And we’re looking at relapse-free survival in approximately a year. That data, we would expect to come in the fourth quarter of this year. And because it is a randomized head-to-head study, I could provide a clear signal of the potential benefit of personalized cancer vaccines versus KEYTRUDA alone, which is obviously the standard we’re going for. That data would emerge based on when we complete enrollment and announce that completement in the fourth quarter this year. And based on that data, we would consult with regulators and obviously decide how to proceed forward if it’s positive.
Operator:
Our next question comes from Gena Wang with Barclays.
Gena Wang:
I have two questions. So, the first one is regarding the flu data, the Phase 2 data in early 2022. Can you be a little bit more specific on timing? And what kind of data you think could be fileable that without the need of a Phase 3 trial with the efficacy outcome? The second question is regarding the 1273, U.S., age 12 to 17. In the press release, you said FDA has not concluded on benefit risk profile of 100-microgram primary series. What additional data you would need to provide? And will you need to provide 50-microgram data in order to receive approval?
Stephen Hoge:
Great. Thank you, Gena, for both those questions. So first, a clarification on the flu data. We do not believe that the Phase 2 data alone would be fileable. And that’s based on previously published regulatory guidance, not specific guidance to Moderna. But ultimately, we don’t think Phase 2 on flu alone, which is an immunogenicity -- safety immunogenicity study of approximately several hundred, is sufficient for filing. The question is, from a filing perspective, what sorts of Phase 3 studies are necessary and whether or not an accelerated approval is possible based on just safety and immunogenicity or whether we will need to demonstrate efficacy in an independent efficacy study, a Phase 3 efficacy study prior to filing. And those are consultations that have not yet happened with regulators, but will on the back of that Phase 2 data that we expect shortly. It’s important to note that there are precedents for accelerated approvals based just on safety and immunogenicity in a Phase 3 study, which would be a few thousand people. But you always have to then follow up with an efficacy study perhaps post approval. And so, in summary, we expect to have to do an efficacy study in flu at some point. The question is whether or not it would be before or after accelerated approval with a Phase 3 immunogenicity and safety study. That Phase 3 study would follow on the current Phase 2 study, and we don’t think that the Phase 2 study alone is viable. We do intend, as I said previously and as we said before, to try and start those Phase 3 studies, whether they’re efficacy safety immunogenicity study this year. On the question of the 1273 adolescent filing in the U.S., and so the FDA has not provided -- has not completed its review of the 100-microgram adolescent primary series. And we have decided on consultation with them to evaluate a lower dose, a 50-microgram primary series dose in -- of adolescents 12 to 17. It’s important to note that the 12 to 17 adolescent 100-microgram primary series has been approved globally in many other markets, and we believe has been administered quite broadly, perhaps up to over 1 million adolescents globally. And so we’ll continue to collect that real-world data as well as the observational data from our monitoring studies. And as and when appropriate, submit that to the FDA for their continued evaluation of the 100-microgram primary series. We are in the interim -- given the strong real-world efficacy data that we’ve seen for Spikevax on 1273, particularly in immune-compromised population. We are in the interim preparing an EUA filing in the United States for 100 micrograms for immune compromised adolescents or those at high risk of severe outcomes from disease because we think the strong efficacy profile of mRNA-1273 at 100 micrograms provides a clear benefit risk in that population. We do believe that 100 micrograms provides a benefit more broadly, which is why it’s been authorized globally. But we’ll continue to work with the FDA and the U.S. to evaluate other potential dose-sparing strategies and submit that data as we develop it.
Operator:
Our next question comes from Matthew Harrison with Morgan Stanley.
Matthew Harrison:
I have two clarifications and then a question. So, first one, on flu. Should we expect that when you present to us the Phase 2 data, whether or not you’ll have had those regulatory discussions or be able to talk about potential next steps in terms of what scope of Phase 3 studies you would need? And then, second, on PA, can you give us a sense of how many cohorts you think you need to see before you may be able to provide that initial data? And then, third, just on the timing of COVID revenues this year, I believe at JPM, you had talked more about first half weighting and second half weighting. And it seems like that’s switched. And I’ve gotten just a couple of questions that I thought would be helpful to clarify. Is that mainly because you’re now targeting boosters for the fall, or is there something else happening here in terms of the weighting of the revenues? Thanks.
Stephen Hoge:
So, thanks, Matthew. I’ll take the first two questions. So first, on flu, it’s obviously not in our hands alone. We will -- whether we would be able to connect with the FDA and get feedback back. As we all know, the agency globally, but also the agency in the United States, agencies are -- have a lot going on right now. And so, there may be some delays in getting that feedback. So, it is not -- we will probably share the Phase 2 data as we have it. And we will obviously rapidly consult with agencies, but we will not negate on hearing back from agencies before we share that data, and our plans are moving forward. But, if it’s possible to get that response more quickly, obviously, that will be something we’ll share at that time. So I wouldn’t expect it, it’s a short version, of being able to have that agency feedback, but it’s possible, and we’ll keep our fingers crossed that perhaps the agencies can turn that around more quickly. On PA, so the number of dose level cohorts. I think it’s important to say that there are sort of two features here that we’ll be looking at. Obviously, the dose level cohorts, so how many different dose level or dose frequency cohorts that we’re looking at in that study? But also, then the duration of time on that study because the primary objective of the intervention of the PA program particularly is to prevent the major metabolic decompensations. And so, that is also something that requires time to accrue, so that you can understand within an individual patient, whether you’ve changed the rate of those recurring events. And so, it’s a mix of two things. I mean, I think we would probably expect to see two to three dose level cohorts. But also, given the rate of enrollment, that would probably also allow us to accrue approximately a year on drug for many of the early recipients. And that combination, those who’ve been on for a while as well as seeing what dose escalation can achieve perhaps in biomarkers, will be the composite data that we think will allow us to make a determination whether we’ve got the right dose level and whether we’ve got a clear indication and benefit, and obviously, whether or not we’re seeing chronic safety and tolerability, which we will expect. And if we have that data and it feels clear, then we will move to regulatory consultation on the next step to clinical studies, and obviously, update all of you with that information. So, not a concrete number of dose level cohorts, but more a function of both time on drug for the early cohorts and perhaps a second or third dose level cohort of data that will provide us with that clarity. And I’ll turn it over, I think, David for the third question.
David Meline:
Yes. So, in terms of the first half, second half timing, as we’ve looked now at -- as we move into ‘22, what do we see? We see that the emerging market countries of COVAX have indicated that they’re having a lot of challenge to absorb all the product that they’re receiving through donations. And therefore, as we looked at, in particular, the second quarter timing versus the third quarter, we refined that outlook to be, as I described, first half, second half. And then secondly, the second half is certainly, as Stéphane mentioned, we think there’s going to be quite strong booster sales that will cause the second half to be a bit higher than the first half.
Operator:
Your next question comes from Michael Yee with Jefferies.
Michael Yee:
We had a two-part question. One was thinking about your COVID boost strategy and the three different strategies you laid out, including the bivalent, which I think is great. Can you just talk about at what point you decide to pick which strategy that would be, and you’re confident that whatever that would be that that would be ready and able to be approved by the FDA to distribute presumably for the fall 2022 booster season, and if that is actually the product that would be in the guidance for 2022? And the second question relates to USA. I know previously you had talked about USA boosting options and purchases. But I can’t actually remember what or where we are for USA for 2022 or for 2023. And maybe just speak to the color about that market. Thank you.
Stephen Hoge:
Great, Michael. So I’ll take the first question and perhaps turn it over to Stéphane on the second. So, in terms of the strategy, so the most important thing to start with maybe is that 1273 as a booster already exists. And so, that is a booster candidate. Although we’re evaluating a fourth dose, we’d expect to have that safety data relatively quickly in the immunogenicity data, and we’ve seen strong real-world evidence of mRNA-1273. And so that booster candidate is obviously something that’s well known to markets and could be available quite quickly. We’re evaluating the Omicron-specific and the Omicron-containing bivalent or 214 in two studies that are ongoing right now. Now, the challenge with all of that, and I think it’s where your question is going, is that when you’re trying to evaluate durability, a difference in durability, that takes time. And so, while we’re running these studies, both the Omicron and Omicron-containing bivalent, and we expect to have data in the first half of this year, that would be one month post boost, and we could turn around and establish, we believe, the safety and the potential benefit of those boosters. And we would hope that the bivalent would continue to be even more compelling. The challenge is that it’s going to be very hard to achieve six months of durability data before Q3. And that’s just a function of time. If you boost people now, it will take time to get that day 181 or six-month durability data that we’ve already started to show with our bivalent vaccines. And so, the question then becomes, how do we proceed from a filing perspective if we’re aiming at the fall for 2022? And those are consultations that are ongoing with regulators now. We do have the benefit of having tested multiple previous bivalent vaccines. And we do have the benefit of being able to use the six-month follow-up data there to start to evaluate this potential for better durability. And so, you could imagine a world where we proceed with the one-month data on our bivalent vaccine showing safety and non-inferiority perhaps against the existing variants, including perhaps the certain variants of concerns known as Omicron. And that we follow up over time, perhaps in the early part of the fall, with data that would confirm the improvement in durability. And then, we would rely on the fact that the previous bivalent vaccine like mRNA-211 had shown that benefit of durability against previous variants of concern, including some that contain some of the same mutations seen in Omicron. And so, it’s a relatively complicated picture because we all -- we do believe, as we’ve said, that it is time to update the vaccine against the mutations that are currently circulating and to improve the durability against those new variants of concern. But it does run into the challenge of timing of those filings. And so, those consultations with regulators are all going. And we’ll provide updates as we align with them on the path forward. That’s quite different than the public health decision from governments as to which vaccines they may want to prepare to stockpile in advance of a fall booster season, which likely can proceed in parallel even without the regulatory filings as it has in the past as it did during the original first wave of the pandemic. So, with that, maybe I’ll turn it over to Stéphane to talk about the fall in the U.S.
Stéphane Bancel:
Thank you, Stephen. Good morning, Michael. So, let me maybe talk about it in two angles. The first one is, as we said this morning, in these increased APAs, we’ve announced at $19 billion, there is no signed APA for -- between Moderna and the U.S. government. So, the number in there from the U.S. is zero. And because there’s no option either that the government has from a previous contract -- the U.S. government has no option currently. So there is $0 in the $3 billion of options probabilized. What is not clear today is what would the U.S. government decide to do for the fall of 2022. We’ve seen what they’ve done in ‘20 and ‘21, buy vaccines from the manufacturers and give them away for free, for the first, second and third dose. Will it be a private market or will be a mix of both the private and free vaccine available because you could see where we are, if we can sell to private markets, there’s private networks and even companies that might want to procure the vaccines. And that’s a bit that is not clear. So it’s why in a very conservative manner, because what we have communicated so far are signed APAs or options, and because the U.S. government has zero, we did not include any of those numbers.
Lavina Talukdar:
Kevin, we have time for two additional questions. Thank you.
Operator:
Okay. Our next question comes from Cory Kasimov with JP Morgan.
Cory Kasimov:
First one, I just want to follow up on what you were just discussing and just kind of looking beyond this fall though. And as kind of we think about moving into this endemic phase and how you’re thinking is evolving, regarding the outlook for the commercial marketplace and like 2023 plus, do you expect APAs for larger countries, be it the U.S. or Europe or anywhere else, or do you think you’ll be doing more selling into private markets? In other words, is there any reason not to shift away from an APA structure? And then secondly, just on capital allocation. Recognize your priorities remain the same here. And clearly, with 44 programs in development, new subsidiaries opening around the world, you have enough capital to do many things simultaneously. But how should we think about priorities with nearly 20% of your year-end cash being dedicated to this latest buyback? Is that 15% to 20% range of kind of your balance sheet an appropriate number to be thinking about in the future? Thank you.
Stéphane Bancel:
So, let me take the first question, Cory, and I’ll turn to David for the buyback question. So, if you look at the countries outside the U.S., they are in, let’s say, non-pandemic markets, they are mostly direct contract with governments anywhere. So, what is it going to look like in terms of shape and form is not very clear. For example, Europe has purchased together the vaccines for the pandemic, which is not the case, let’s say, for seasonal flu, for example. So, will they continue that model for COVID and move to that model for other vaccines or will they go back to a national system is to be seen. But all countries like Japan, Canada and so on are basically a single buyer market. Some countries as you know small product markets, but it’s mostly kind of government orders. And so, we are in discussions with governments about ‘23. As you saw, we’ve already signed contracts for ‘23 because some countries like the UK and others wanted to secure supply because they believe very deeply that the endemic market will require annual boosters. And so, we just want to get ahead of it. And as you saw what we are doing with Canada and Australia, which I think is a very interesting new model of kind of service-based subscription-like partnership, is we basically are trying to secure, as we said, 10-year agreements. And we’re in discussion with several more countries about setting the similar model in the countries where we build the plant. They reserve a given volume for a year, let’s say, 20 million, 50 million-dose, 100 million-dose, depending on the market size. And then, what we commit to them is to be able to customize the respiratory vaccines with what they believe they want. So, as you saw from Paul’s presentation, there’s a lot of respiratory virus that most people are not aware of, even their names. Look at PIE and there people knew that there were coronaviruses circulating and creating so much hospitalization and they are stable. Well, our vision is to bring all of those components together and discuss with local public health experts on an annual basis, what do they want in their vaccine for Canada or vaccine for Australia. As we discussed on the flu call in the fall, sometimes, the WHO picks a flu strain like H3 or -- and then it’s a different flu strain that winter that is between, let’s say, North America, Europe and/or Asia, like Japan and so on. And so, this ability to also customize and work with local public health authorities, to customize a vaccine they want, we think it’s a really unique feature of this platform because of the speed, because every combined components. And so, that’s what we’re already trying to do here, is to really establish very long-term agreements. I’m not aware of other companies having done this type of thing in pharma. And I think this is something we can do. And the teams are working very actively also. We’ll have to wait a few more months to see more coming, but there are quite a lot of discussions. We just have to wait there, because we cannot do in 10 countries at the same time in discussion because they are very-customized and very-complex partnerships. But that’s exactly where we’re trying to head. David, do you want to talk about buybacks?
David Meline:
Sure, yes. Yes. I mean, I guess, what I’d say about the announcement today, I think what you need to think about is a few things as we did. So, why $3 billion, why now? It starts with a few things. One is, as we said, investing internally and externally and ensuring that we have plenty of firepower to allow us to do that. So clearly, where we enter 2022, we’re in a good position to fund all of the opportunities that we think we have, and they’re very compelling. So that’s one. Two is, looking at what visibility we have of additional cash generation for the company. And again, we feel very good about that certainly as we enter 2022. And then thirdly, we look at the valuation levels. And certainly, we think this is quite attractive at these levels. So, combination of all that adds up to an announcement of $3 billion. To your question, should I be filling in the model for that to continue on an ongoing basis? I think it’s a little bit early for us to comment right now on that question. And I’d encourage you just to wait and let’s see how this evolves. But we feel very good about the prospects for the business. I think the statement today is one of a strong confidence that we can achieve all of the objectives that we’ve set out for the Company.
Operator:
Our last question comes from Tyler Van Buren with Cowen.
Tyler Van Buren:
On a similar topic of conversation, could you just provide your latest thoughts on future U.S. pricing for Spikevax? Do you expect the future contracts to have pricing that has normalized relative to ex-U.S. countries? And maybe just on a second point to clarify, do you believe that a new variant wave emerging by year-end is necessary to record the vast majority of the $3 billion in APA options?
Stéphane Bancel:
So, let me start. So, I will not comment on the U.S. pricing for obvious reasons because of our discussions with the governments. I think as we said before, once this goes into a normal private market, we do expect the pricing to be higher. We think the vaccine does not reflect -- sorry, the price doesn’t reflect the value of a vaccine from a pharmacoeconomic standpoint. And I mean, the options, the $3 billion, so there are just different stage -- some are just waiting funding from governments to move into fully signed APAs. Some are things with COVAX that has a place or as for future needs. So, I don’t think they need to be a new variant for some of it to happen, and plus, the U.S. on top of that. But again, we have never managed through a pandemic and through a transition from pandemic to endemic. So, we want to be cautious and prudent. But I think it doesn’t need a new variant for a material chunk, over $3 billion, to move into signed APAs.
Operator:
Ladies and gentlemen, this concludes the Q&A portion of today’s conference. I’d like to turn the call back to Stéphane for any closing remarks.
Stéphane Bancel:
Well, thank you very much everybody for joining. I look forward to talking to you in the coming days and weeks, and especially to welcome you at our Vaccine Day, which will be exciting, in March. Thank you.
Operator:
Ladies and gentlemen, this does conclude today’s presentation. You may now disconnect, and have a wonderful day.
Operator:
Good morning and welcome to Moderna's Third Quarter Earnings Call. At this time, all participants are in a listen-only-mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I would like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please go ahead, ma'am.
Lavina Talukdar:
Thank you, operator. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's third quarter 2021 financial results and business update. You can access the press release we issued this morning, as well as the slides that we'll be reviewing by going to the Investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; and Paul Burton, our Chief Medical Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of Private Litigations Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our performance and results to differ materially from those expressed or implied in these forward-looking statements. On slide 3, please see the important indication and safety information for our COVID-19 vaccine, which has been authorized for emergency use in the United States and many countries around the world. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Welcome to our Q3 2021 conference call. Today, I will start by quick business review of the quarter before David presents the key financials. Paul will then walk you through some real world evidence update. And Stephen will provide the clinical development update. I will then come back to close to share some thoughts about where we are heading. Let's start on slide 5 with a holistic pipeline update. First, respiratory vaccines. We are pleased to have filed the BLA for COVID-19 vaccines with FDA and have now received a priority review designation. We received our first full approval for Spikevax backs in Canada. We received EUA from the FDA for the 50 microgram booster and mix and match has been authorized. We also received authorization for boosters from our major geographies, EMA, UK and so on. We were informed recently that FDA may take until January 2022 to review the EUA request for 12 to 17 years of age. Turning now to flu. mRNA-1010 is fully enrolled for Phase 1 and we expect human data soon now. For RSV, mRNA-1345, after a very positive data in the Phase 1, with high neutralizing antibody levels after one dose, our teams are finalizing the preparation of a Phase II/III in older adults. This study should start soon. We also announced our development candidate for COVID booster plus flu booster in a single dose, mRNA 1073, which we anticipate will be in a clinic soon. Let me now turn to vaccines against latent viruses. As many of you know, latent viruses are a major cause of health problems. Latent viruses are mainly made of DNA viruses. And once a human is infected by a latent virus, that virus will stay in the body. You are familiar with some latent viruses like HIV and HPV. There are many other that helps humans. As a company, we are committed to developing a portfolio of first-in-class vaccines and therapeutics against these latent viruses. Our first programs are CMV vaccine, EBV vaccine, and EBV therapeutic vaccine. We have also partnered to work on two HIV vaccines. Last week marked a big milestone for Moderna. We started dosing in the clinic the first participant of our Phase III pivotal registration study for CMV vaccines mRNA-1647. This marks the second Phase III study that the company has started and brings us one step closer to getting another important vaccine to millions of people around the world. For EBV vaccine, mRNA-1189 should start dosing for its Phase I very soon. And our new development candidate on the EBV therapeutic vaccine, mRNA-1195 is moving toward the clinic. Let me now turn to therapeutics. Our personalized cancer vaccine is in Phase II [indiscernible] with KEYTRUDA monotherapy is fully enrolled. And we expect data in the back end of 2022. Our rare genetic programs continue to progress well in the clinic. PA has its Phase I dose core fully enrolled. MMA is dosing patients in its Phase I. GSD1a has an open IND and has received orphan drug designation by the FDA. So, as you can see, we have three rare genetic disease programs in the clinic or soon to be in the clinic. So, we look forward to showing clinical data in the coming months. We are very proud to have found a way to get Crigler-Najjar Type 1 or CN-1 to patients through a donation of our IP and the assets to the Institute for Life Changing Medicines. CN-1 disease is an ultra-rare disease with very few patients afflicted around the world. For ultra-rare disease, just the cost of clinical developments will result in a very high price for a few patients around the world that could benefit. This model is not one that we support. So instead of developing a drug and charging a very high price, we decided to give the drug away and innovate for a creative partnership. We provide the drug to the institutes, we pay for clinical trial material, the institute pays for the clinical trial cost. If the drug is approved, we'll provide the medicines to the kids and their families for free. And we will not charge any royalty or milestone to the institutes. We believe this is the right thing to do to continue to contribute to society for ultra-rare disease where our mRNA platform can help. One of the important news items announced today is the introduction of a new modality through our existing six modalities. We're very pleased to announce that Vertex and Moderna are now in GLP tox studies for the cystic fibrosis program, VXc-522. This marks our first inhaled preliminary therapeutics program. I would like to congratulate and thank our scientific team as well as the Vertex team for having been able to develop a novel lipid formulation to allow inhaled pulmonary delivery. This is really exciting for patients. As we communicated previously, in addition to doing therapeutics using mRNA coding for human proteins, like the ones I just described, we want to use our mRNA plus lipid technologies to develop therapeutics using mRNA coding for gene editing enzymes. We are pleased to announce our first partnership in the field. We announced earlier this week that we entered into a licensing collaboration with Metagenomi based in California to use their next generation gene editing enzymes. We believe that that technology, coupled with our mRNA plus lipid technologies, and our manufacturing infrastructure will produce some exciting new medicines to take into the clinic. I am very pleased with the progress of the company. We are increasing depth in vaccines, and at the same time increasing breadth of application [indiscernible] and gene editing. On slide 5, you will find financial highlights. For Q3, we reported revenue of around $5 billion enabled by delivering 208 million doses of our COVID-19 vaccine. Net income of $3.3 billion. Our cash and cash equivalents position was $15 billion at the end of September. Since the end of the quarter, we have been working to get better visibility into those variables that will impact our full year results. And we have much better visibility today than we did a month ago. For fiscal year 2021, we're updating the range of doses we plan to deliver to 700 million to 800 million doses. The key variable of this update volume range are first, longer lead time for exports to countries outside the US and for international shipments in general. In Q3, we shipped more deliveries to new international markets and expect Q4 to also have more international deliveries. As we deliver to these countries for the first time, we expect the process to become more routine and delivery time to shorten. However, some Q4 deliveries may still move to early 2022 as a result. Second, in Q3, we have expanded our fill finish capacity with our manufacturing partners, which had the temporary impact on our shipments. That work is complete now. And we should see a positive impact from this expansion very soon. Third, the bottleneck of production has moved down the manufacturing line to product release. We added additional resources, skilled personnel to the release and of the manufacturing line. And we expect to increase the weekly number of doses raised. We believe resolution of these factors put us in a good position heading into Q4 and 2022. Now if I turn to revenue for 2021. The updated revenue range for 2021 is $15 billion to $18 billion. This range reflects our updated delivery schedule with fewer doses delivered in 2021 as some doses are pushed into 2022. And also, the pricing impact of prioritization delivery to COVAX and African Union that have a lower tier pricing. On slide 7, let me start by summarizing the scale-up operational challenges that the team had to work through as we scaled from making less than 100,000 doses in 2019 to hundreds of millions of doses in 2021. Q1 scale up challenges were around drug substance, making mRNA in lipids. Q2 scale up challenges were on drug products and ensuring we could fill enough vials. In the first half of the year, we made great progress in these challenges, as you can see from the numbers on the right. We now have 901 million doses of drug substance that have been formulated [indiscernible] of mRNA products. And we have 770 million doses of drug products already in vials. Q3 scale-up challenges were on product release as the supply chain became more complex with increased deliveries to many countries around the world. At the beginning of the year, we supplied just a few large countries. As mentioned before, we are working through these challenges. And as November 1, we achieved 566 million doses around the world. With the progress year-to-date, we expect to be able to deliver 700 million to 800 million doses for full year 2021. We were pleased to be named last week for the seventh year in a row as one of the top 20 biotech and pharmaceutical company in the world by Science. We continue to invest in science and the culture of our team. We aim at building the best mRNA science in the world, in the culture of boldness, collaboration, curiosity and relentlessness. On slide 9, you will find our usual summary slide, which describes some of the key high-level attributes of the company as of today. Now let me turn it over to David.
David Meline:
Okay. Thank you, Stéphane. We're providing today the analysis of actual 2021 third quarter results, along with an updated view of key drivers of financial performance going forward. Turning now to slide 11, starting with an overview of our sales performance before commenting more broadly on our financial results. Total product sales in the third quarter were $4.8 billion, representing 208 million doses delivered to our customers. This compares to sales of $4.2 billion and 199 million doses in Q2 and sales of $1.7 billion and 102 million doses in Q1 of this year. Sales to the US government were $1.2 billion in the third quarter, reflecting 73 million doses delivered, compared to $2.1 billion and 126 million doses in Q2 and sales of $1.4 billion and 88 million doses in Q1. Sales to the rest of the world were $3.6 billion in the third quarter, reflecting 136 million doses delivered, compared to sales of $2.1 billion and 73 million doses in Q2 and $0.4 billion of sales and 14 million doses in Q1. This reflects the significant manufacturing ramp up outside the US over the last two quarters. The relatively modest increase of delivered doses in Q3 versus Q2 was driven by the factors that Stéphane just explained. We continue to scale our production network and are working to achieve an increased quarter-over-quarter improvement starting in Q4. Turning to slide 12. The transformation of Moderna from an R&D-focused biotech to a commercial company continues to be very apparent when reviewing our financial results. The comparison of the third quarter of 2021 to prior year is not meaningful due to our significant growth, which is why we will primarily focus on the quarter-over-quarter comparison relative to Q2 on the slide. Total revenue was $5 billion in the third quarter of 2021 compared to $4.4 billion in the second quarter and $0.2 billion in the prior-year period. The increase of total revenue is driven by the sale of the company's COVID-19 vaccine. Product sales in Q3 were $4.8 billion compared to $4.2 billion in the second quarter, an increase of 15%. Cost of sales was $722 million or 15% of the company's product sales in the third quarter compared to $750 million or 18% of product sales in the second quarter. The quarter-over-quarter percentage improvement is driven by higher average selling price as a result of shifts in our customer mix and, to a smaller extent, by favorable manufacturing costs. Research and development expenses were $521 million in the third quarter, up from 421 million in Q2 and $344 million in the same period in 2020. The higher spend versus prior quarter and prior year was driven by increased COVID-19 vaccine clinical development activities, including our announced efforts around booster variant-specific and multivalent vaccine candidates. Headcount increases as well as external costs to support our growing and maturing development pipeline beyond the COVID-19 vaccine activities also contributed to the expense increase. Selling, general and administrative expenses were $168 million for Q3 compared to $121 million in the prior quarter and $48 million for the same period in the prior year. The growth in spending was driven by the commercialization of our COVID-19 vaccine globally with continued investments in personnel and outside services in support of the accelerated company build-out. Provision for income taxes was $219 million in the third quarter, following $283 million in Q2 and an insignificant amount in the prior year. Our effective tax rate for the third quarter was 6%. Let me remind you of the fact that we had a net operating loss carryforward of $2.3 billion at the end of 2020. This year, we started to release the valuation allowance against the related deferred tax assets, which results in an estimated non-recurring full-year benefit to our effective tax rates of about 5 percentage points. Our Q3 effective tax rate was lower than the US statutory rate, primarily due to the just-described benefit related to the release of the valuation allowance, the foreign derived intangible income deduction and an excess tax deduction related to stock-based compensation. This last item drove the quarter-over-quarter reduction due to the increase in our share price in Q3 versus Q2. We recorded net income of $3.3 billion in Q3 compared to $2.8 billion in Q2, an increase of 20%. This compares to a loss of $0.2 billion in Q3 of last year. Diluted earnings per share for Q3 2021 were $7.70. Turning now to year-to-date financial results compared to prior year on slide 13. Total revenue was $11.3 billion for the first nine months of this year compared to $0.2 billion for the same period in 2020. The significant growth was driven by the sales of 510 million doses of the company's COVID-19 vaccine. Grant revenue of $473 million in the first nine months of 2021, an increase of $286 million compared to the prior year, was primarily driven by increases in revenue from BARDA related to the company's COVID-19 vaccine development. Cost of sales was $1.7 billion or 16% of the company's product sales for the year-to-date period as of September 30, including third-party royalties of $400 million. A portion of the inventory costs associated with this year's product sales was expensed as prelaunch inventory costs in 2020. If inventories sold for the first nine months of this year was valued at cost, our cost of sales for the period would have been 17% of the product sales. Research and development expenses were $1.3 billion for the nine months ended September 30 of this year compared to $0.6 billion for the same period in 2020. The growth in spending in 2021 was mainly due to increase in clinical trial expenses and, to a lesser extent, personnel-related costs, manufacturing expenses, and consulting and outside services, primarily driven by increased mRNA-1273 clinical development activities. Selling, general and administrative expenses were $0.4 billion for the first nine months of this year compared to $0.1 billion for the same period in 2020. The growth in spending in 2021 was mainly due to increases in consulting and outside services, personnel-related costs, and marketing expenses primarily attributable to the company's COVID-19 vaccine commercialization-related activities and increased headcount. For the nine months ended September 30, we recorded a provision for income taxes of $541 million compared to insignificant amounts in the same period in 2020. Our effective tax rate for the first nine months of this year was 7%. It was lower than the US statutory rate primarily due to non-recurring benefits related to the release of the valuation allowance, the ongoing benefit of the foreign-derived intangible income deduction, as well as a discrete item for excess tax deductions related to stock-based compensation. Net income was $7.3 billion for the nine months ended September 30 of this year compared to a net loss of $0.5 billion for the same period in 2020. Diluted earnings per share were $17 for the first nine months of 2021. Turning to cash and selected cash flow information on slide 14. We ended Q3 2021 with cash and investments of $15.3 billion compared to $12.2 billion at the end of Q2. The increase is driven by our commercial sales and additional customer deposits received in the third quarter for future purchases of our COVID-19 vaccine. Net cash provided by operating activities was $3.3 billion in Q3 after $4.1 billion in Q2. The quarter-over-quarter reduction is driven by a lower amount of cash deposits for future product supply received in Q3 compared to Q2. On a year-to-date basis, net cash provided by operating activities was $10.3 billion compared to $0.8 billion in the prior year. Cash used for purchases of property and equipment was $164 million for the nine months ended September 30 compared to $44 million for the same period in 2020, reflecting continuous investments in our manufacturing infrastructure. Now turning to slide 15. Let me briefly expand further on our cash and investment position and the net balance of cash deposits for future product supply as this is an important point when modeling future cash flows. The cash and investment balance reported as of September 30 was $15.3 billion, up from $12.2 billion as of June 30. The increase is driven by our commercial activities, including cash receipts from product sales and customer deposits for future product supply. The net balance of cash deposits for future product supply was $6.7 billion at the end of Q3, at a similar level to the balance in Q2 of this year. Turning now to the 2021 updated financial framework on slide 16. For the full-year 2021, we expect a product sales range of $15 billion to 18 billion. We now expect to be able to deliver 700 million to 800 million doses at the 100 microgram dose level to our customers. This compares to the prior outlook of 800 million to 1 billion doses. Our total cost of sales includes the cost of goods manufactured, third-party royalties as well as logistics and warehousing costs. Given the favorability we have observed in the year-to-date period, we now expect total cost of sales as a percent of product sales between 16% to 17% for the full year 2021. This compares to our previous outlook of 18% to 20% of product sales. For 2022, we currently expect the cost of sales ratio to exceed 20%, driven by customer mix and multiple presentation types. On the R&D and SG&A expense side, we continue to plan for an increase on a quarter-over-quarter basis and expect this trend to continue for the remainder of 2021 and beyond, driven by a maturing development portfolio and the scale-up of our commercial activities. Based on further increased visibility of the utilization of our accumulated net operating loss carryforward, expected global sales mix and the mentioned discrete benefits in the first nine months of this year, we now expect our all-in 2021 tax rate to be in the high single-digit range. This compares to our previous forecast of approximately 10%. Since we won't have a material benefit from our valuation allowance in future years, we expect our reported 2022 effective tax rate will increase relative to our 2021 rate. It's also too early to further comment on impacts from a potential tax reform. Finally, regarding capital investments. We are now planning for approximately $0.4 billion of capital investment to fall into the 2021 calendar year compared to our previous range of $450 million to $550 million. We are strongly committed to a further build-out of our manufacturing and general company infrastructure, and hence predict capital investment in 2022 will be meaningfully above 2021 levels. This concludes my remarks concerning the financial performance. And I now turn the call over to Paul Burton.
Paul Burton:
Thank you, David. And good morning, good afternoon, everyone. With more than 150 million people worldwide now having received two doses of our vaccine, we are humbled to be able to see the positive impact it is having on people's lives around the world. We are reminded of the fact that almost 250 million people have been infected with COVID-19 globally and 5 million people have lost their lives. Our profound thanks, as always, go out to those on the front lines, working tirelessly to keep us safe in this ongoing fight. Through numerous independent studies, time and time again, we see consistent findings showing that mRNA-1273 is highly effective in saving lives, reducing hospitalizations and reducing the risk of COVID-19 infection. I will highlight a few of these independently conducted studies this morning. In slide 18, I will begin with some of the data from the United States government. These data come from the CDC and show through September of this year the difference in COVID-19 cases between fully vaccinated and unvaccinated populations. An unvaccinated person has an 11-fold greater risk of dying from COVID-19, underscoring the importance of getting vaccinated plays in our protection and ending this pandemic. The data show that vaccination with mRNA-1273 provides the greatest protection, not only against COVID-19 infection, but also death due to COVID-19. In fact, even during the surge of the Delta virus variant during the summer months this year, Moderna's vaccine had the lowest reported deaths associated with breakthrough infection as the CDC data demonstrate. Approximately 160 million doses of the Moderna vaccine have been administered in the United States. And in the next slide, I want to show you some further government-generated data in a country where the Moderna vaccine has also had extensive use, and that country is Switzerland. You can see here exposure data on the left. Approximately 3.6 million people have been fully vaccinated with the Moderna vaccine and 1.9 million people have been fully vaccinated with the Pfizer/BionTech vaccine. These government-generated data from the Federal Office of Public Health in Switzerland, again, are clear and show that vaccination with the Moderna vaccine is highly effective at reducing infections, hospitalizations and deaths due to COVID-19 infection. For every 1 million people vaccinated, use of mRNA-1273 would be expected to result in 643 fewer COVID-19 breakthrough infections, 78 hospitalizations and 38 fewer deaths. Next, in slide 20, our data from two other recent independent studies that build on the results I just showed you from Switzerland. The first study on the left side of this slide followed 38 million vaccinated people in the United States, Iceland and South Korea. And again, found that those people vaccinated with mRNA-1273 were at 53% reduced risk of a COVID-19 infection and 33% reduced risk of a COVID-19 hospitalization compared to those vaccinated with the alternate mRNA vaccine. The data on the right hand of the slide are from a recent publication by Nordstrom and colleagues in Sweden and Norway, showing that mRNA-1273 delivered 87% vaccine effectiveness during a time of high Delta variant circulation in those countries. The data I've shared so far are all from the general population. Let me turn now to the effectiveness of the Moderna vaccine, in particularly vulnerable populations, the immunocompromised. These data were published just two days ago, again, by the United States CDC VISION network and examine vaccine effectiveness at preventing COVID-19 hospitalization, which is so critically important in individuals with a variety of immunocompromised medical conditions between January and September of this year. As the authors of this study note, with the exception of those individuals with rheumatic disorders, vaccine effectiveness point estimates were generally higher for the Moderna vaccine than for the Pfizer/BioNTech vaccine. And the difference in effectiveness in these hard-to-immunize populations is shown in the column on the right. I want to turn now to the topic of myocarditis and to put it in the context of the clinical benefits of vaccination that I've just described with mRNA-1273. It is important to recognize that the cases of myocarditis reported to occur following mRNA vaccination are rare, generally mild, typically respond to conservative treatment and self-limiting. Health authorities have reviewed many data sets and the US CDC and WHO have concluded that there is a risk of myocarditis and pericarditis after receiving any mRNA vaccine. The WHO's Advisory Committee on vaccine safety notes that myocarditis can occur following SARS-CoV-2 infection or COVID-19 disease and that mRNA vaccines have a clear benefit in preventing hospitalization and death from COVID-19. Analyses from our global safety database show that the events of myocarditis are very rare. Overall, we observe 9.5 cases of myocarditis per million vaccinated individuals compared to an expected rate of 21 cases per million individuals. Now, in common with other reports of mRNA vaccines in individuals aged 18 to 24, we see an increased rate of myocarditis of 40 cases versus 17 expected cases per million individuals. These data from our own safety database are consistent with the findings from safety databases evaluated by the CDC and other global health authorities. And the link to the CDC analysis is provided on the slide below. MRNA-1273 has been authorized for use in adolescents aged 12 to 18 in multiple countries around the world. Moderna's global safety database has information on an estimated 1.5 million of them, providing us an opportunity to look at the rate of myocarditis in those individuals, and those data are shown in this table. We see the small, absolute, well-described risk of increased myocarditis rates in males aged 18 to 24 that I just described to you. But we do not see an increased risk in adolescents, neither in boys nor in girls. These data are reassuring and important as we continue to offer mRNA-1273 to adults, adolescents and as we file for authorization in children. So, in summary, the data I've shared with you today from independent government reports in the United States and in Switzerland, countries with high vaccination rates of Moderna's vaccine, show that mRNA-1273 is associated with lowest breakthrough infections, hospitalizations and deaths due to COVID-19. Other independent studies in millions of individuals continue to show that mRNA-1273 is highly effective in reducing the risk of breakthrough COVID-19 as well as hospitalizations and death. And importantly, this is also true in those vulnerable and hard-to-vaccinate immunocompromised patient populations. Moderna's global safety database of 151 million vaccinated individuals shows a rate of myocarditis in males aged 18 to 24 that is consistent with analyses by CDC and others. Moderna's global safety database does not show an increased risk of myocarditis in 1.5 million individuals below the age of 18 and vaccinated with Spikevax. So, this concludes my quick overview of some recent analyses of mRNA-1273 effectiveness data and review of data from our global safety database. And I will now turn the call over to Stephen.
Stephen Hoge:
Thank you, Paul. And good morning or good afternoon, everyone. Today, I'll review the progress we've made across our vaccines and therapeutics pipeline. Let me start with our COVID-19 vaccine, mRNA-1273, for adult ages 18 and above where there are a number of important regulatory updates. First, we have announced that the FDA granted priority review to Moderna's COVID-19 vaccine BLA. In October, we also received an EUA from the FDA and the European Commission's approval for a booster dose of our COVID-19 vaccine at the 50-microgram dose level for the adult cohort ages 18 and above. Turning to the adolescents and pediatric settings. As a reminder, there are two clinical trials for each of these groups
Stéphane Bancel:
Thank you, Stephen. Slide 39 is from our September 9 R&D Day and summarizes the product franchise we focus on at Moderna. Priority number 1 is our pan-respiratory annual booster franchise. Priority number 2 is our first in-class vaccines for latent viruses. Priority number 3 is our therapeutics based on mRNA-encoded proteins. And priority number 4 is the therapeutic space on mRNA-encoded gene editing enzymes. As we have said for several quarters now, SARS-CoV-2 is here to stay and will evolve from a pandemic to endemic setting. We believe that 2022 will see pandemic in low-income countries throughout the year, but at high income countries, the year would be of two halves, pandemic priming for children and boosting for others with an endemic boosting campaign in the fall of 2022. On slide 42, you can see the revenue drivers that we anticipate will play out in 2022. We believe there are three components that will drive our COVID-19 vaccine revenues for next year. First, signed APAs. We have already signed around $17 billion of advanced purchase agreements, or APAs, for delivery in 2022. Second, APA options. There are up to $3 billion of APAs that are in options. Some options have been converted in Q3 from options to firm order. Third, the fall of 2022 commercial market. We also believe that the fall of 2022, assuming that BLA is granted for boosters, will drive the commercial booster market of up to $2 billion. So, in total, at this point, we believe that 2022 revenues could be between $17 billion and $22 billion. We, of course, continue to have discussions [indiscernible] APAs with governments and international organizations, including COVAX, the Pan American Health Organization or PAHO, and the African Union. For this pan-respiratory franchise, our goal is to evolve the COVID-19 vaccine primary series into fall of 2021 boosters, which is happening as we speak, and then fall of 2022 boosters and then add to a COVID booster a flu booster in a single dose, and then added to COVID and flu an allergy booster in a single dose. Our flu vaccine human data should be out soon. And the team is already preparing the Phase II/III for flu. And RSV is moving fast to Phase II/III. If we you look at the health damage of latent viruses, it is profound. EBV is a major cause of infectious mononucleosis. EBV has been reported to increase risk of multiple sclerosis. EBV is associated with certain cancers and autoimmune diseases and EBV is associated with a higher risk of long COVID. CMV associated will be the leading cause of birth defects. CMV is a major driver of immune dysfunctions with aging. CMV is associated with cardiovascular diseases. CMV is associated with cancer and cognitive impairments. For now, our focus is CMV, EBV and HIV, but we are developing in the labs vaccine against other latent viruses [indiscernible] human health. Our goal is to eliminate these viruses. As we discussed earlier, several cancer and rare genetic programs are in the clinic and will provide clinical results soon. On November 15, AZ will present Phase II data in patients from the VEGF program at the American Health Association. And we are pleased to announce the new modality with inhaled pulmonary therapeutics. Moderna [Technical Difficulty] is our effort to expand the use of our platform to create more innovative drugs to help patients. Our strategy to invest internally under the leadership of Dr. Eric Huang with a dedicated team and to set up licenses agreements with next-generation in gene editing companies. The Metagenomi partnership is a first step in that direction. The strategic path of the company is very clear and exciting. We want to stop people getting hospitalized from respiratory infection. We won't stop until this goal is achieved. We want to stop our fellow human beings from suffering from these latent viruses. We want to bring to market mRNA encoded protein therapeutics in oncology, cardiology, rare genetic disease, autoimmune. We want to bring therapeutics using gene-editing enzymes. We believe Moderna could become the most impactful drug company in the world. As we scale, we realize that we needed to go further in setting the right framework for our team to understand what has made Moderna. So, we worked through Q2 and Q3 to articulate our mindset, how we behave, and make decisions at Moderna. There is more information on our website and we'll be happy to spend time with those of you who want to learn more about them. Let me just summarize them at a high level. At Moderna, we act with urgency. Action today compounds the life saved tomorrow. We pursue options in parallel to make the best choice later. We accept risk as the only path to impact. We're obsessed over learning. We don't have to be the smartest. We have to run the fastest. We pivot fearlessly in the pace of new data. We question convention because proven models don't always fuel the future. We push past possible. We behave like owners. The solutions we are building go beyond any job description. We act with dynamic range, driving strategy and execution at the same time and at every step of the way. We remove viscosity to encourage collective action. We prioritize our platform over any single product. We digitalize everything possible using the power of digital information to maximize our impact on patients. We want to be the most impactful drug company in the world. We care deeply about doing it the right way. It means being a great company to work for, as exemplified by our seventh consecutive year ranked as the best company to work for by Science. But it also means building a company that is responsible in minimizing our impact on the planet. We are proud to have announced earlier this week that we will work to achieve net zero carbon emission for our operations globally by 2030. I want to thank the Moderna team for their commitment to our mission, and their relentless work to build the best version of Moderna over the next 20 years. Before we jump into Q&A, we wanted to share the dates of our annual investor event for 2022; Vaccine Day on March 24; Science Day on May 17; and R&D Day on September 8. Operator, we'll now be happy to take any questions.
Operator:
[Operator Instructions]. Our first question comes from the line of Salveen Richter from Goldman Sachs.
Salveen Richter:
For 2022, can you walk through the supply aspects. Are you still guiding to up to 3 billion doses here? And can you also speak to demand dynamics? Is there upside to the guidance that you've commented on today for future signed APAs. And then secondly, how confident are you that you can fix these supply issues and over what time frame?
Stéphane Bancel:
Let me take those different questions. Let me start with the short term. Yes, we really believe we can fix those short-term supply issues. As I tried to explain in my remarks, those are what I would qualify as teething problems of scaling up so fast. In Q1, it was all about making enough drug substance and we are literally now waiting to have enough drug substance to fill vials. And as the teams scaled very nicely, in Q2, the challenge we had internally was all about filling vials. And the complexity of Q3 has really moved to, I would say, the back end of the supply chain, which is releasing product and shipping products. And the complexity has been around just a number of markets we have to serve. Beginning of the year, it was mostly shipping to CDC in the US and Europe, and that was it. But then as we increased the number of countries to many dozens by now, the complexity is just increasing and it's even further now that we are serving COVAX. We need to go country by country. So, that's just the type of teething problems that we are experiencing right now. We have increased personnel. We have invested in digital to help the teams. So, I already expect this to be resolved like we resolved the drug substance challenges in Q1 and the drug product challenges in Q2. In terms of drug substance, yes, we could see make up to 3 billion doses in terms of material. As you know, that number was really depending on the booster dose. And now that we're on the other side of that decision with 50-microgram dose, we confirm we could make up to 3 billion doses, if there was a need for it. The challenge in 2022 is going to really be around product form because, as the market moves to an endemic market, you're going to need basically vials to be able to go to less number of dose per vial. Again, in a pandemic setting, as you know, we launched with 10 dose per vial, which we think was really adapted. In low-income countries, we even add more into vials now. But as we think about the endemic market and we're trying to serve pandemic and endemic at the same time, we just need to keep adding presentation to be relevant to healthcare workers and to healthcare systems around the world. So drug substance will not be an issue. There's, of course, as I described, potential upside to those numbers. We still have quite a number of APAs being discussed with countries around the world. We don't know what will happen in terms of epidemiology. Is there a new variant coming? Is it a variant where the current vaccines is still helpful or we need, as Stephen described, a new booster. There is, of course, a big question mark here that we're all following epidemiology literally on a daily basis. And then, there's so many for boosting people around the world that have had non-mRNA vaccines. As everybody knows, there is a problem of waning immunity over time. So, time is not our friend as people see the level of antibody going down and as we have new variants like Delta or potentially others. But we should not forget that most people around the planet have not had an mRNA vaccine injected in their body to date. And so, we believe there is an important need to boost people with mRNA vaccines. And as Paul showed the real world evidence including the data from CDC and Switzerland, the more we get data and the more time we have been to see, we see a net differentiation between products. And this doesn't go unnoticed by governments around the world who are working really hard to preventing hospitalization and death. And so, there is upside to that. Again, it's a very unpredictable environment. None of us has worked through a pandemic before thankfully. But you will be assured that we will keep scaling up manufacturing, we'll keep in getting with government, so that we can maximize how we can help people and the revenue should follow from that.
Operator:
Your next question is from Matthew Harrison with Morgan Stanley.
Matthew Harrison:
Two for me this morning. So, first, can we just spend a moment on myocarditis. And I think the overlying question here is, why do you think the regulator is more concerned with your vaccine in younger age groups compared to Pfizer, which has obviously already been approved in younger age groups. And then related to that, how much of an impact is this having on uptake and distribution of your vaccine given that we see Pfizer continue to highlight potential differences and bringing that to government's attention? And then secondly, on flu, can you just comment on how you're going to interpret these results? Obviously, we're just going to get titer results. But I think it's your premise that you can achieve a much higher efficacy flu vaccine compared to traditional flu vaccines. So do you think there is a clear correlation in titers to efficacy? And what level of titers would demonstrate very high efficacy?
Stephen Hoge:
This is Stephen. I'll try and take those questions. So, first, look, I think it's most important to say that what we communicated and desired to be maximally transparent last week was that the FDA, unlike other regulators, has asked for some more time to review emerging recent data. And that might take until January. I think your question is how is that different vis-à-vis what happened with the Pfizer vaccine. I think the most important thing to recognize is that the Pfizer adolescent vaccine was authorized prior to any substantial discussion about myocarditis as a benefit or as a risk. In fact, the signal emerged a few weeks later just before we made our filing. And I think a prudent approach there was to – there was a VRBPAC conducted, there was ongoing discussions. But what we've continued to see over the last four or five months is that for both mRNA vaccines, there's a question of whether there's an increased rate of myocarditis above background in 18 to 24-year-old males, a relatively small population, but an important one. And I think it's in the face of those continuing emerging questions that the FDA has been diligent and appropriately conservative in their approach and making sure they have the time to review those. And they have continued to come out over time. And so, I think, principally, what we're seeing here as the difference is a function of timing, which is that the other vaccine had been authorized prior to this concern and there has been continued emerging data around that. We are very grateful to the FDA for that diligence. I would note that the same information are available to other regulators. And as I have said before, we are authorized for that population internationally. And fortunately, as Paul characterized, we have not seen an increased rate of myocarditis in 12 to 17-year-olds. And we think, over time, the substantial benefits of our vaccine will ultimately win out here. And so, we look forward to continuing to work with the FDA. So, that's my best version of what I think we heard from them last week. The question about how we see that evolving over time. Obviously, internationally, we are participating in the market. In the United States, adolescent vaccinations have substantially tailed off, as we all would note. And so, the extent that there is an ongoing need in the United States for vaccination, it is a diminishing market for sure. And again, this is a primary series vaccine. And so, we do hope that most people in the world will seroconvert and not need a primary series moving forward. The question then of what is that endemic market going forward that is of greatest import. And I've tried to summarize that in our view of the evolution of this virus and what that need will be. It is, we believe, in the future, a booster market. And the booster market, targeted at those populations that are at higher risk of respiratory disease. Those populations tend to be older adults and immunecompromised. Places where we think mRNA-1273 is demonstrating really remarkable efficacy, differentiated perhaps. But publicly reported data is really encouraging on that dimension. And I would note that is not a population that's associated to date with any of the vaccines that have an increased rate of myocarditis. And so, the benefit risk there, we think, even swings more favorably to 1273, but we'll allow data to continue to develop and ultimately drive this behavior. So we are quite encouraged by the performance of the vaccine in a population that we think is going to be most important in the years ahead. The last question was on flu, which is what do we think about that data. Obviously, as you pointed to, there are correlates of protection that have been used previously with HAI titers in influenza vaccine. We will be looking at that, as I'm sure everybody will, as well as other responses in the immune system as we continue to try and identify the optimal dose. I'm not going to put out there a view of what I think our view of success will look like. I would just say that we have very high expectations for our platform. Now we do believe that our performance in older adults, including with the COVID vaccine, but also more recently with the RSV booster study in Phase I demonstrates that our platform does incredibly well in those at highest risk of these respiratory viral diseases. And so, we're optimistic that we will continue to show strong performance, hopefully strongest performance in those populations. But I won't give you a specific titer number today.
Stéphane Bancel:
Maybe just to add to Stephen's point, Matthew, I think we should not forget the company's strategy, which is on combination. We believe that combining flu and COVID boosters in a single dose and then adding allergy is the critical central part of our strategy. We think it has tremendous value for compliance, for protection. It has a tremendous value in terms of convenience to the consumers. I do not believe that most people will want the flu shot and a COVID shot and an allergy shot every fall. And as we heard from the payer and healthcare workers, that's the value of our product. So we believe the combination is really a critical success factor.
Operator:
Next question is from Ted Tenthoff with Piper Sandler.
Edward Tenthoff:
My question has to do with the emerging orphan disease pipeline. And I've been really impressed by the progress that you guys are making there in these important patients. And I think the mRNA technology just suits ideally here. So, what do you see as sort of the plans over the next couple of years. Kind of take us forward a little bit. What could this pipeline look like? And maybe you can just give us a sense of where you think Moderna will be in the orphan disease setting in a couple of years.
Stephen Hoge:
I'll take that too. So, first, I think we're very excited about the programs that are already in the clinical space, either already dosing patients or about to start. And so, I think the most important thing is looking forward to next year, the demonstration, we would hope, of proof of concept in that rare disease modality. As you know, we are dosing quite a large number of folks in propionic acidemia and we've been dosing in methylmalonic acidemia. With the opening of the GSD1a IND, we'd hope to be following a short order there. So, all three of those are potentials for us to demonstrate the real proof that this technology can be used to correct inborne errors of metabolism in these populations. I'll also note that there's a quite a wide range of disease going down to as young as two years of age in some of the organic acidemias and also the older adults in some of the GSD1a program. So, we're going to be demonstrating quite a lot there. Crigler Najjar presents another opportunity for that proof-of-concept as well as the PKU program when that moves forward, but those are still in preclinical, as I said. So the question is what do we do on the back of that proof-of-concept from any one or all of those programs. What you've seen us do in respiratory vaccines and in vaccines generally is probably the best predictor of how we will respond, which is, as you know, there are a very, very large number of metabolic diseases that could be addressed to deliver through mRNA therapy. We could list off large groups, the urea cycle of disorders, other organic acidemias, so many beyond that. And even in moving into more broadly present metabolic diseases. So, what we would do is we would define that systemic intracellular therapeutic modality as a core modality, just like we did with vaccines a couple of years ago. And that would cause us to dramatically expand that pipeline. Now I can assure you we're looking at those programs in research right now, but we have held back on moving them into preclinical development and putting them on our pipeline until we've seen the modalities perform. So that is probably the most important thing for me, looking at the rare and orphan disease space over the course of the coming year, is when do we cross that threshold and then, ultimately, when do we expand dramatically that pipeline of programs.
Edward Tenthoff:
I really appreciate seeing the new pulmonary disease areas.
Operator:
We have a question from Michael Yee from Jefferies.
Michael Yee:
Two questions. One is just trying to clarify guidance. I think there's some confusion around guidance. So I would love to understand some clarification. You lowered 2021 a little bit, I think, by $5 billion, but raised 2022 by $5 billion. Is that a timing shift of deliveries? And how much of that is just option contracts as you think about 2022 because you've talked about APAs as firm commitments. I'm just trying to understand how much are commitments versus acceptances and how to think about those two. And then on flu. Following up on the flu question, I think people are looking at labels and looking at fold increases and seroconversion rates of four to seven times and 50 to 60% seroconversion rates. Are those accurate numbers? Are those numbers we should be looking at and comparing to? Maybe you could just help us qualify that because I think that's what people are trying to do.
Stéphane Bancel:
I'll take the first one and give it through to Stephen. So, I think you already highlighted some of the other drivers. So on 2021, so there's two things I think that is driving. First is, of course, the lower volume. And the second one is price. As you know, we are working very hard with several governments to send products that they have bought for high-income countries like the US to low-income countries this side of Christmas. And so, when you think about just the US, the US we see that publicly. When we announced our African Union partnership, the US decided to delay to Q2 the delivery of the December quarter. That volume is going to African Union at a low tier price. So, you have an impact on the turnover just by doing the math of a lower price on the same volume right there. And some orders that are moving from December to January on the supply volume side of things. So, when you're on to 2022 where you have the increase of volume moved from December to January, that is one. We've signed new APAs since the last numbers. And as I said, some options have been exercised. One of them was a COVAX option that was exercised at the end of Q3. That is now counted as a full APA because of the firm commitment and some prepayments and so on. And so, those are the dynamics that are happening. The team also is starting to spend a lot of time, the commercial team, on focusing on the fall of 2022 because we think that's going to be an important moment. The mix and match, we think, is critical. That is now allowed in most places. In some countries, they've been just mix and match for a long time. But the US market, of course, is important, is allowing mix and match. And as Paul just shared, we believe that as time will go, the data will show that what we believe is that we have a longer duration efficacy vaccine on the market. And today, if you look at data and market research, very few consumer in the US know that. People that are listening to this call know that because you read papers on a daily basis, a few people in the US. We believe there is an opportunity for us between now and, let's say, next summer to make sure that people understand the facts, understand the real-world evidence, so that they can make an informed decision and that includes the healthcare workers, the pharmacies, the doctors, the nurses, that include the consumers directly. And so, this is why, as we're starting to sharpen our pencils, everybody is spending a lot of time working toward the fall of 2022, we believe that's another piece that was not – if you go back to our previous numbers, we only disclosed APAs and options of APAs, the commercial opportunities for fall of 2022, we think, is going to be an important vector to the 2022 sales. Stephen, on flu?
Stephen Hoge:
I'll try and clarify that answer on seroconversion rates. It is a Phase I study. And so we are looking at a range of doses and I have not got access to the data. So, I don't want to presuppose the specific answer yet. As soon as we have the data, we will provide our thorough interpretation of it. However, as you point out, generally seroconversion is defined as a fourfold rise in titers above baseline, and we've done that in some of our other studies. And we will be looking at that from the percentage of people who've achieved that seroconversion. And most important to me, we'll be looking at consistency of that percentage across ages. In particular, the older adults where you often don't achieve the type of the same level of immunogenicity, but we believe with our platform will. There are other endpoints as well in terms of seroprotection defined as absolute titers, again greater than 40 is accepted. We will look how high that goes because that can also be reassuring. But I don't think right now we're ready to guide on a specific target that we will declare success other than looking again at that fourfold rise for measuring seroconversion across the range of agents that we'll be studying in the study.
Operator:
Next question is from Gena Wang from Barclays. Your line is open.
Gena Wang:
I have two regarding the COVID vaccine. And the first question is, given the supply restraints, do you see 100-microgram doses being used as 250-microgram doses. And then also, any hesitance due to safety concerns? We did see a few countries put a cautionary action on Moderna vaccine. And my second question is, in the US, if we do the math, you completed delivery of 300 million doses and then US exercised total doses of 410 million for 2021 and the 90 million doses in first quarter 2022. So, just wondering how much of the remaining 110 million doses in 4Q 2021 will flow through next year.
Stephen Hoge:
I can try and take the first question, although invite Paul as well. So, I think in terms of -- you referenced some of the more recent communications that happened from public health officials, for instance, in the Nordics and elsewhere. I would note that those same communications, literally often the same documents, include reference to the fact that there's very strong efficacy for the Moderna COVID-19 vaccine. And in fact, some of those Nordic country communications include reference to the fact that it looks to be potentially greater in terms of protection as emphasis for why the vaccine not only is still recommended in many of those jurisdictions, but ultimately provide very favorable benefit risk. And so, we continue to believe that whole picture looking at the benefit and quantifying that benefit, which we think is substantial and larger than any risks contemplated here is really important. And we, again, look to those communications that clearly state that. Paul, anything you would add to that?
Paul Burton:
Only, Stephen, that recently, I think last week, the WHO gave updated guidance continuing to endorse the vaccines in mRNA-1273. And I think as they are doing their analysis of these data sets, have said that they need to think about potential biases that occur in general practice, looking at the primary vaccination schedule, the difference in timing. We know the vaccines are used differently sometimes, and that may account for these. But I think clear standing behind the data.
Stephen Hoge:
Just as a closing comment, I would just reference, we have entered in some of these markets into a very interesting phase of the pandemic, which is that we are in the lower risk population and we are providing vaccination to them as appropriate. So, as we do that, we're looking at benefit/risk in increasingly cautious ways, and that's appropriate. But as we look forward to next year and we start thinking about boosting, and particularly, the seasonal market of protecting those that are highest risk, I think that calculus obviously changes pretty dramatically. And so, I think we are in a period of time where we are, again, looking at the lowest risk populations. And I think that is transient period time ultimately because it is high risk populations that are of greatest concern looking forward from 2022.
Stéphane Bancel:
Maybe just to add just a bit of color. I've had a chance to speak with a couple of health ministers across the world in the last week or two. And I think people are very clear that the risk is low, that it's very manageable, as Paul said, and it's only in the male 18 to 24. And those health ministers were clear about going back to the risk profile that Stephen just mentioned. They care about having a winter where they don't have hospitals exploding again. And this is driven by the 50 and above, the 40 and above where they know the vaccine, as Paul mentioned, with the data from the US or Switzerland or many other countries that 1273 vaccine seems to be a vaccine providing the longest protection of efficacy, and that's what they care about. And so, I think that people are getting educated and looking at the data. They're just trying to look at the facts and figure out, in a very practical way, how do they keep the economies running, how they keep people out of hospitals and are very focused on the high population. As you know, in many countries, the boosters are not approved in the younger population, whereas they are being really advertised and promoted in the older population, so that people get boosted and don't get sick and hospitalized this winter. On the US government, Gena, we don't comment on volume. US government is a private contract to the government. But indeed, there are some shift from Q4 to next year, both Q1 and Q2.
Operator:
Next question is from Cory Kasimov with J.P. Morgan.
Cory Kasimov:
Two for me as well. So, first of all, in terms of the 2022 APAs that you outlined, are the $17 billion worth of existing contracts constructed as firm commitments or do countries have – the contracts have optionality embedded in them wherein countries don't necessarily have to take the full amount depending on the evolution of the pandemic. And then, my second question is, given your comments on the call about potentially moving into the endemic phase by the second half of next year, should we think about 2022 revenue as being weighted towards the first half of the year?
Stéphane Bancel:
Thank you, Cory. So let me take this apart. The APAs do not have options. When we say $17 billion of APAs, those are firm orders, signed orders and all our APAs have upfront payments. David did a nice job walking you through the cash upfront we are receiving for those contracts. It's one of the way that we are conducting our business. We want people to be very committed and to have a material upfront of the total value of a deal when they sign. The options components that we characterize as up to $3 billion as of today, those are true options, meaning people are reserving capacity. And sometimes it's for financing rhythm because, again, as I said, we do not do APAs without upfront. And one of the best example that's today public is COVAX. We had to do a quarterly option with COVAX. As I said, it just exercised that Q2 option at the end of Q3. But Q3 2022 and Q4 2022 COVAX are still options and it's mostly because of a funding issue. They don't have the cash. They get cash from their different funders, which are mostly government and foundation, on the kind of a month-to-month basis. And so, we did that to help COVAX and to do the right thing to help the planet. As you know, we don't have to do options. We say, if you want the vaccine when you sign a contract and you pay an upfront, but with COVAX we have to do so. We thought it was the right thing to do. And so, we expect, if they want their Q3 volume, that in Q1, they will exercise the options – sorry, end of Q4, they exercise the options [indiscernible]. On the endemic and [indiscernible] of the sales over the year, I think it will be dangerous to assume at this stage one way or another in terms of first half and second half because of volume and also pricing. As you get the sense, the COVAX, the African Union and [indiscernible] deals we have lower prices and David mentioned that, tiered pricing. And so, there might be a large volume, but the price might be way lower than what the commercial market could drive, which we believe is going to be above what has been the pandemic price again because of volume. And so, we have people stocking ahead of when they need to inject in pharmacies. I would be cautioned that assuming Q1 can be lower than Q2 is a bit too early.
Operator:
Next question is from Geoff Meacham from Bank of America.
Geoff Meacham:
I just had one more on the flu program, maybe for Stephen. It's highly likely that you'll see an increase in antibody titers. I was just thinking for the combo study, though, with 1273, what other factors do you need to address to de-risk that study? And just, I guess, I'm trying to determine how to optimize the regimen by things like dose titration or even selecting the population by more urgent need for the COVID booster side of things.
Stephen Hoge:
What we're going to be looking at in the combinations are actually the things that you just pointed to. And so, in particular, we do believe the – most of the value of respiratory vaccination in the near term boosting will be in older adults, 50-plus or 60-plus depending upon how you want to look at it. Very similar to the RSV population, certainly for the flu population and recommendations globally. And we think COVID moves that direction as well. And so, as we look to the first part of the question, where do we intend to optimize that ratio in immunogenicity, it really is towards those higher risk populations that do need, we think, a respiratory booster every year. When it comes to looking at the – what are the combination of things that we're going to be doing in selecting that ratio in dose, obviously, it's mostly going to be immunogenicity and the tolerability profile. Fortunately, these vaccines have been very safe to date in these older adult populations. None of the concerns that we're having about myocarditis really exists in the 50 plus, 60 plus populations. And so, we feel very confident on the overall safety profile. But the question will be, where are we in optimizing the immunogenicity against endpoints that are pretty well validated now in COVID, and where are we against the immunogenicity against endpoints that are also broadly accepted in influenza. We will be doing this for a range of different – it's a four-strain, seasonal strain vaccine. And so, we'll want to make sure there's no interference between those and that we're able to achieve strong balance immunity across the four flu strains plus the COVID vaccine. I think we have approximate targets based on the immunogenicity we saw from Phase III and the immunogenicity we're seeing in the booster studies. But the open question is are we going to need all of that for the COVID portion of our vaccine going forward. And so, we'll continue to follow over time whether we see more durable protection and, therefore, you might be able to optimize further that dose, if there was a reason to do so. And then, the second part is with the seasonal annual flu component. We just want to understand, particularly in the older adult population where flu vaccines have had less clear efficacy every season, is maybe the way I'll say it, and there is an opportunity for improvement by raising those titers. We're going to want to look to whether or not we need to get those titers above what have been achieved by others. And I'll note that there's a precedent even for high dose flu vaccines in that market that have been differentiated on price and grabbing market share. And so, it could cause us to look in that 60-plus or 50-plus population at really indexing more to those flu antigens and making sure that we exceed or at least achieve that similar high degree of seroconversion there. So, those are the factors that we'll look at around the 1073 program that we've announced. We're looking at a range of ratios. Fortunately, we're going to have experience. We already have experience in flu and COVID from our prior work, and it will be a demonstration, we hope, of the ability to combine vaccines. I will note, as a final comment, we have done combination respiratory vaccines in the past, including in adults with the 1653 program. And so, we do have a basis, we're beginning to think, about how we might combine those antigens going forward. But ultimately, we'll let the 1073 study and the early clinical experiences there be confirmatory around this.
Operator:
We have a question from Joseph Stringer from Needham.
Joseph Stringer:
Two from us. Just curious on the guidance for – the 2 billion guidance for the US fall 2022 booster market. Can you help us understand what assumptions are sort of built into that number? Is that based on sort of your internal expectations around a relative percentage of individuals that would – fully vaccinated individuals that would get a booster or just trying to handicap what the variance could be around that. And then, for CMV, can you give us a sense for the relative timelines for enrollment of that Phase III trial and when we could see the initial data from that?
Stéphane Bancel:
On the fall of 2022, so the commercial team has spent quite a lot of time modeling different assumptions in terms of volume of people who want the booster, market share, and, of course, pricing. I cannot comment further for competitive reason right now, especially on the pricing piece, but we will do that in due time. But indeed, it's typical commercial analysis that the team that has done many times before in other companies have run through the last few months.
Stephen Hoge:
And on the enrollment of CMV, thank you for the question. The answer from our target perspective is as fast as we can because we ultimately think there's just such a huge unmet need here, and we're excited to get the study going. There are some caveats that I should put around that, which is it probably won't be as fast as we did in COVID, which was obviously in a couple of months because we are targeting a population at high risk of disease. And in this case, we're looking for women of child-bearing age who are exposed to seroconversion with CMV. And that is often those that are exposed to children because that's the primary vector for those infections. And so, we want to make sure that we get the right population. And that matters because this is a case-driven study. As we're kind of familiar with based on COVID-19, it's the number of cases that we achieve that will ultimately dictate those interim analyses that we're looking for in the time ahead. And so, it makes sense to try and enrich for the at-risk population. So you might see a slower enrollment curve or it might go really fast. We're going to work as hard as we can to go very fast. But the most important thing is that we need to be enrolling those that are at highest risk because we think that will ultimately generate the cases that accelerates the interim readout. Because it is an interim readout that's driven on the cases, I can't predict when that will happen. What I can do is I can try and enrich for those cases in the way I just described.
Operator:
Next question is from Emmanuel Papadakis from Deutsche Bank.
Lavina Talukdar:
We can go to the next question, operator.
Operator:
And next is from Mani Foroohar with SVB Leerink.
Mani Foroohar:
A couple of quick ones. Can you give us a sense of when we might see, if any, the impact of pricing shift to increase deliveries to low and middle income countries that Stéphane mentioned. Is that something that we'd start to see a little bit of early next year? Is that more of a 4Q phenomenon in terms of the budgetary timing and sort of quarterly contract that Stéphane talked about. Secondarily, I have a few small piggy jack question on the numbers. On slide 15, the $6.7 billion in deposits, that presumably all for APAs for 2021 or 2022? Or does some of that apply to APAs for delivery – for contracts for 2023. Thirdly, your CapEx guidance a little lower than it was last Q. Is that $50 million to $150 million that you took out of that, is that absolute savings? Or is that just going to get pushed into next year as some of the contract revenues were? And then finally, a little more substantive of a question. How much perspective can you give us on you potentially compete for share on contracting in EU, the competitor Pfizer/BioNTech locked in pretty large contracts on volume for the next two years? Is there an opportunity for you guys to start capturing more share [indiscernible] relevant in the EU? And last, and finally, we talked on the last quarter about potentially access to China market where you guys are essentially not existing at this point. And you have mentioned going there via JV. Can you give us an update on status of your ability to access that market, discussion of the JV to compete with BionTech/Fosun? Sorry for the barrage of questions, and thanks for taking the questions.
Stéphane Bancel:
I'll take a few and, David, if you can take the financials, that would be great. So, on Q4, we already actually picked COVAX product. And the African Union will be in December, as we communicated in the press release. And it's going to be a big ramp into next year, obviously. In terms of European share, which was one of your questions. So if you look at it, there's a few things. First, in Europe, if you remember, we had a late and slow launch in Europe because we had no manufacturing capacity in Europe. If you recall, when we had the pandemic, we had Norwood that we converted and we were not allowed to export outside the US market. So Lonza came online a bit later than Norwood because we had nothing in Lonza until they started sometime in the summer to just get machines and get the facility ready. And so indeed, in Europe, we have a lesser share than we have in the US, except, for example, for Switzerland like you saw earlier. And so, as the country gets more and more of an understanding first that mRNA vaccines are superior, the other vaccines other than mRNA are not really used anymore. That's my understanding. They're mostly given through COVAX. I think we have an interesting opportunity because as I shared, as you take – you talk to healthcare leaders, health minister, as you get more and more data that Paul shared, we have a better and better understanding that the two mRNA vaccines are not the same. And so, again, as we look into 2022, we already have a contract with Europe for 2022. And we are discussing also with country by country on the data and using the benefit of the Moderna vaccine versus the other one. And then the other question that's going to stay in Europe, which is today, contracts are done at the European level because this was organized and agreed by Europe because of the pandemic. The big question that's going to be out there is when do countries start to be buying by themselves because they have to make guesses of supply when they set up 2021 and 2022 agreements that might not meet the needs that they have, as we understand better the different profiles of the vaccine. And so, I think it's going to be quite interesting to see the share opportunity that we have in Europe. Also as we build our teams, you remember, we had no infrastructure in Europe, zero, not one Moderna employee in commercial. We have now teams in several countries in Europe, in the UK, in Spain, in France, in Germany, building the Italian team as we speak. So I think that will also help because we get feet on the ground to be able to get the message that – of a real world evidence data. David, do you want to take the capital financial questions?
David Meline:
In terms of the deposits, the $6.7 billion we have in hand, I would just say the preponderance of those deposits are for deliveries either in 2021 and in 2022. There may be some small amounts in 2023, but it's pretty early in terms of the contracting there. Secondly, in terms of CapEx, I think the answer is yes, you can expect, as I said – we're going to see a notable increase in our CapEx next year. But generally, in terms of the reduction this year, it's a combination of things moving into the first quarter of next year and also we're running, in some cases, at or below the estimated investment requirements. So it's a combination of those two.
Mani Foroohar:
On potential China JV, I know you guys have mentioned it in the last call, I don't know if there's been any progress?
Stéphane Bancel:
Yes. So nothing to announce today, but [indiscernible] in China.
Operator:
There are no further questions at this time. I would now like to turn the conference back to Mr. Stéphane Bancel.
Stéphane Bancel:
Well, thank you very much, everybody, for joining us. And if you have any further questions, you know where to reach Lavina and the team. Have a great day. Bye.
Operator:
This concludes today's conference call. Thank you for joining. You may now disconnect.
Operator:
Good morning, and welcome to Moderna's Second Quarter Earnings Call. At this time, all participants are in a listen-only-mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I would like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's second quarter 2021 financial results and business update. You can access the press release issued this morning, as well as the slides that we'll be reviewing by going to the investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; Paul Burton, our Chief Medical Officer; Corinne Le Goff, our Chief Commercial Officer; and Jackie Miller, our Senior Vice President, Therapeutic Head of Infectious Diseases. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the safe harbor provisions of Private Litigations Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors, that could cause our performance and results to differ materially from those expressed or implied in these forward-looking statements. On Slide 3, please see the important indication and information for our COVID-19 vaccine, which has been authorized for emergency use in the United States and in many countries around the world. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. Welcome to our Q2 2021 conference call. Today, I will start by a quick business review of our quarter before Corinne walks you through the commercial update. David will present the key financials. Stephen and Jackie will provide the clinical update, highlighting new human data about the final analysis for COVID-19 vaccine Phase 3 COVE study and human data for COVID-19 booster candidate Phase 2. I will then come back to close to share some thoughts about where we are heading. Let me start with Moderna COVID-19 vaccine or Spikevax. We are pleased to announce today that our final analysis vaccine efficacy or VE in our Phase 3 COVE study is holding very nicely at 93%. We are starting to get authorizations for adolescent indication 12 to 17 years of age, receiving authorization in Japan this week and the positive recommendation from the European Medicines Agency in July. An important step toward our vision of an annual respiratory combination booster is the start of a Phase 1/2 for mRNA-1010, which is equivalent for seasonal flu vaccine. Our teams are already preparing the Phase 2/3 for this program. We were delighted to get fast track designation from the U.S. FDA for our RSV vaccine candidate, mRNA-1345 in adults over 60 years of age. There is no approved RSV vaccine. The burden for RSV infection is very high. In adults, over 65 years of age, according to the CDC, 177,000 of hospitalizations and around 14,000 deaths occur annually in the U.S. due to RSV. Our teams are also preparing a Phase 2/3 for this program. Our Zika vaccine has now moved to Phase 2. mRNA-3927 for propionic acidemia or PA, a rare genetic disease has also started building patients in our Phase 1/2. And we are pleased to announce recently that we started dosing healthy volunteers in our first autoimmune disease program, mRNA-6231, coding for IL-2. So Moderna is now in the clinic in five large therapeutic areas
Corinne Le Goff:
Thank you, Stéphane, and good morning or good afternoon, everyone. I am happy to share the productive quarter, the commercial organization has had in the second quarter that continues into today as we ramp up our efforts to supply the Moderna COVID-19 vaccine or Spikevax to countries around the world. Let me start by recapping the advanced purchase agreements that we have signed for delivery in 2021 on Slide 11. Since the last quarterly call, I am very happy to highlight that we have signed APAs with COVAX for 34 million doses this year, an additional 110 million doses to the USG, bringing the total of doses for the United States to about 400 million doses and 10 million doses to Australia for delivery in 2021. In total, we anticipate up to $20 billion in sales from these agreements as we deliver against them throughout the remainder of the year. We are proud to distribute our vaccine directly or through our network of partners across all continents, and most importantly, to ensure access to our vaccines into all countries regardless of their income level, notably through the COVAX facility structure. I also want to mention that we are doing our utmost to -- in supporting the U.S. government to execute the donations of Moderna doses of vaccines. Slide 12 list outs signed APAs for deliveries in 2022 and even in 2023. The construct of the APAs signed for 2022 and 2023 include both confirmed orders, as well as options to be triggered at the future date. To date, we have already contracted for 22 product sales of $12 billion and an additional $8 billion in options. Some of these APAs are for primary series vaccines and others are for potential boosters. In light of the possibility of needing booster vaccines to increase immune responses against waning immunity and the emergence of variants of concern, Moderna has already started our COVID booster strategy that Stephen will speak to shortly. In the meantime, the commercial team is currently engaged in multiple conversations that are gaining urgency as we watch the Delta variant surge among unvaccinated populations. We are notably actively talking to many countries in South America, in Asia and in the Middle East. All these countries are keenly interested in securing both primary series vaccines or booster vaccines for their citizens next year. In the United States, as you know Moderna has initiated the rolling submission process for beta approval and expects to complete its submission in August. This approval will represent an important milestone in achieving herd immunity as it might help combat vaccine hesitancy. It will also allow the commercial organization to stop preparing for the private U.S. market. Turning to Slide 13, which reviewed the sales breakout for the second quarter 2021. Total product sales in the quarter were $4.2 billion representing 199 million doses delivered in the quarter. Sales in the U.S. were $2.1 billion representing 126 million doses delivered to the U.S. government and sales to the Rest of the World -- to the country that are listed on this slide were also $2.1 billion representing 73 million doses delivered in the quarter. And with that, let me hand it over to David to take you through the financial details.
David Meline:
Okay. Thank you, Corinne. We're providing today the analysis of actual 2021 second quarter results along with an updated view of key drivers of financial performance going forward. As in previous quarters, we are presenting our results primarily on a U.S. GAAP basis. In some cases, we also provide additional detail to provide greater clarity on underlying trends. Turn now please to Slide 15. The transformation of Moderna from an R&D-focused biotech to a commercial company is very apparent when reviewing our financial results. The comparison of the second quarter of 2021 to prior year is not as meaningful due to our dynamic growth, which is why we'll primarily focus on the quarter-over-quarter comparison relative to Q1 on this slide. Total revenue was $4.4 billion in the second quarter of 2021 compared to $1.9 billion in Q1. The increase of total revenue is primarily resulting from the sale of the company's COVID-19 vaccine. Product sales in Q2 were $4.2 billion compared to $1.7 billion in the first quarter, an increase of 142%. Cost of sales were $750 million or 18% of the company's product sales in the second quarter compared to $193 million in the first quarter. Research and development expenses were $421 million in Q2 2021 compared to $401 million in Q1 and $152 million in the same period in 2020. The higher spend versus prior quarter and prior year was driven by increased COVID-19 vaccine clinical development activities including our announced efforts around booster, variant-specific and multivalent vaccine candidates. Selling, general and administrative expenses were $121 million for Q2 compared to $77 million for the prior quarter. The growth in spending was driven by commercialization of our COVID-19 vaccine globally with the biggest increases being in personnel and outside services. Provision for income taxes was $283 million in Q2 after $39 million in Q1 and an insignificant amount in the prior year. Our effective tax rate for Q2 was 9%. Let me remind you of the following. The significant investments to develop the mRNA platform over the last decade resulted in a net operating loss carryforward with a balance of $2.3 billion at the end of 2020. As of December 31, we maintained a full valuation allowance against our deferred tax assets related to these loss carryforwards. As discussed in our last call, we started to release the valuation allowance this year. The majority of the allowance will flow through the P&L over the course of this year through our effective tax rate pro rated based on the cadence of our expected pre-tax quarterly earnings. Over the course of 2021, the resulting non-recurring benefit due to the release of the valuation allowance is about five percentage points on our effective tax rate. Our Q2 effective tax rate was lower than the U.S. statutory rate, primarily due to the benefit related to the release of the valuation allowance, the foreign-derived intangible income deduction, as well as a discrete item for excess tax deductions related to stock-based compensation. We recorded net income of $2.8 billion in Q2, after $1.2 billion in Q1, an increase of 128%. Diluted earnings per share for Q2 were $6.46. Turning now to year-to-date financial results compared to prior year on Slide 16. Total revenue was $6.3 billion for the six months ended June 30, compared to $75 million for the same period in 2020. The significant growth was driven by the sales of 302 million doses of the company's COVID-19 vaccine. Cost of sales was $943 million, or 16% of the company's product sales for the six months ended June 30, including third-party royalties of $232 million. Reported cost of sales was reduced by three percentage points, due to the consumption of previously expensed inventory of approximately $200 million. Research and development expenses were $822 million for the six months ended June 30, compared to $267 million for the same period in 2020. The growth in spending in 2021 was largely driven by increased mRNA-1273 clinical development and headcount. Selling, general and administrative expenses were $198 million for the six months ended June 30, compared to $61 million for the same period in 2020. The growth in spending in 2021 was mainly attributable to the company's COVID-19 vaccine commercialization activities. For the six months ended June 30, we recorded provision for income taxes of $322 million compared to an insignificant amount for the period in 2020. Our effective tax rate for the six months ended June 30 was 7%. It was lower than the U.S. statutory rate, primarily due to the non-recurring benefit related to the release of the valuation allowance the ongoing benefit of the foreign-derived intangible income deduction, as well as a discrete item for excess tax deductions related to stock-based compensation. Net income was $4 billion for the six months ended June 30, compared to a net loss of $241 million for the same period in 2020. Diluted earnings per share were $9.30 for the six months ended June 30, 2021. Turning to cash and selected cash flow information on Slide 17. We ended Q2 with cash and investments of $12.2 billion compared to $8.2 billion at the end of Q1. The increase is driven by our commercial sales and additional customer deposits received in the second quarter for future purchases of our COVID-19 vaccine. Net cash provided by operating activities was $4.1 billion in Q2 after $3 billion in Q1, totaling then $7 billion year-to-date. This compares to net cash used in operating activities of $130 million in the prior year. Cash used for purchase of property and equipment was $65 million for the six months ended June 30, compared to $25 million for the same period in 2020. Similar to last quarter, before providing an updated financial framework for the remainder of 2021, let me point out a few areas that are important to keep in mind when modeling expected 2021 financial performance, starting with cost of sales on Slide 18. Cost of sales includes the cost of goods manufactured, third-party royalties as well as logistics and warehousing costs. As you may recall, we began capitalizing our COVID-19 vaccine inventory cost in December 2020 following emergency use authorization. Prior to the authorization, inventory costs were recorded as research and development expenses in the period incurred. In Q1, a zero cost inventory balance of 140 -- $184 million was sold and benefited our cost of sales. If inventory sold during the first quarter was valued at actual cost, our cost of sales would have been 377 million or 22% of product sales. In Q2, cost of sales of 750 million or 18% of product sales were no longer materially impacted by the zero cost inventory, hence, the relevant comparison is the adjusted ratio in Q1. The reduction of cost of sales as a percent of product sales when comparing to the adjusted cost of sales in Q1 is primarily driven by a customer mix-driven increase in our average selling price during the second quarter. Now turning to our cash and investment position on Slide 19. The cash and investment balance reported as of June 30 was 12.2 billion, up from 8.2 billion as of March 31. The increase is driven by our commercial activities, including net increase in customer deposits for future product supply of COVID-19 vaccine. The net cash balance of customer deposits increased from 5.6 billion at the end of Q1 to 6.8 billion as of June 30. Turning to Slide 20. I wanted to share our capital allocation priorities. We seek to optimize our capital deployment and maximize long-term shareholder returns. Our top investment priority will continue to be reinvesting in the base business across multiple areas. For R&D, we have more than tripled spending in the first half of 2021 relative to the prior year, and we will continue to significantly increase spending in this area to advance and accelerate our pipeline. For manufacturing, we previously disclosed our 2021 capital expenditure plans, which will allow us to increase our production capacity. Additionally, we are investing heavily in digital, automation, and AI, as well as scaling up our global commercial operations which will allow us to maximize the impact of our mRNA platform. Our second investment priority is to seek attractive external investment and collaboration opportunities to further expand the reach of Moderna's technology and capabilities. We are considering attractive strategic opportunities that enable and complement our platform and take a disciplined approach in evaluating potential outside investments. After evaluating internal and external investment opportunities, we then assess additional uses of cash. As part of today's press release, we announced that the board has authorized a $1 billion share repurchase program. This program is authorized for a two year period based on the strength of our financial results for the first half of the year and our confidence in our business outlook we believe it is an appropriate time to initiate this program. Turning now to the 2021 updated financial framework on Slide 21. Signed advanced purchase agreements for expected delivery in 2021 reflect the current full year total of approximately 20 billion in anticipated product sales included the 5.9 billion of product sales already generated in the first half of this year. For the full year, we continue to expect a minimum supply of 800 million doses at the 100-microgram dose level. Our manufacturing team and our partners continue working to supply up to 1 billion doses for 2021. We have signed advanced purchase agreements for expected delivery in 2022 for total product sales of approximately $12 billion and options for an additional $8 billion. Numerous additional negotiations are still ongoing for 2022 APAs. We have also started to sign APAs for 2023. As we previously announced we continue to make investments to increase our supply of vaccine including by working with contract manufacturing organizations. We currently anticipate that our supply could be as high as 3 billion doses for 2022, if our sales are primarily at a 50-microgram dose level. If sales are primarily at the 100-microgram dose level we anticipate that supply will be approximately up to 2 billion doses. The ultimate approach on dosage levels for 2022 and where we might end up in that range is subject to ongoing internal review as well as discussions with regulators and customers and will also be impacted by the mix of primary and booster series. Our total cost of sales includes the cost of goods manufactured, third-party royalties as well as logistics and warehousing costs. For 2021 we now expect the average cost total of sales as a percent of product sales to be between 18% to 20% compared to our previous outlook of approximately 20% of product sales. This reflects the successful ramp-up of this global manufacturing network. With regard to planned R&D and SG&A expenses Q2 expenses of approximately $542 million reflect a quarter-over-quarter increase of 13% in line with the outlook we gave in Q1. We continue to plan for an increase on a quarter-over-quarter basis for the remainder of this year and expect this growth rate to accelerate as the business rapidly expands. Based on further increased visibility of the utilization of our accumulated net operating loss carryforward expected global sales mix and the mentioned discrete benefits in the first half of this year, we now expect our all-in 2021 tax rate to be approximately 10%. This compares to our previous forecast in the low-teen range. This forecast is based on current U.S. tax policy and does not include any future potential discrete benefits related to stock-based compensation. Finally regarding capital investments, we maintained our forecast for a range of $450 million to $550 million including the planned capacity expansion investments as announced on April 29 of this year. This concludes my remarks and I turn the call over to Jackie Miller.
Jackie Miller:
Yes. Good morning good afternoon everyone. My name is Jackie Miller and I lead the therapeutic area for infectious diseases. And it's my pleasure today to give you an update to the ongoing accumulation of data in our Phase III clinical study and also to talk about some of the publications of real-world evidence that have occurred outside of Moderna with the use of our COVID-19 vaccine. So on Slide 24, you'll see the top line updates to our COVE efficacy trial. And these are efficacy data that have now been followed through four to six months after subjects received their second vaccination of either mRNA-1273 or placebo. Recall that at the time of our EUA submission, our primary efficacy analysis demonstrated efficacy to COVID-19 of 94.1%. Now four to six months after second dose, we see a maintenance of that efficacy of 93.2% with a lower limit of the 95% confidence interval of 91%. We continue to maintain efficacy against severe COVID-19 disease with updated vaccine efficacy of 98.2% and currently have 100% of efficacy against deaths caused by COVID-19. So unfortunately, there were three deaths in the placebo group and up till now none in the mRNA-1273 group. We continue to see consistency in our subgroup analyses including analyses by gender, by race and by preexisting medical conditions. Our safety profile continues to be consistent with the Phase 3 data over the longer period of safety follow-up and also continues to be consistent across population subgroups. Next slide please, on Slide 25. You'll see the efficacy data broken out by time interval. And so what you see at the top of the table is the overall efficacy we just discussed. According to the primary endpoint, we start measuring vaccine efficacy at 14 days after dose two. And again that's 93.1%. If you look between 14 days post dose two, to less than two months after dose two, we observed vaccine efficacy of 91.8% and 94% if you look two months after dose two to less than four months after dose two. And finally, greater than four months after dose two, we observed 92.4% efficacy. And the conclusion we take from these data is that our efficacy has remained consistently high and durable throughout the period of follow-up. And we intend to continue to follow these data now that the trial is in its open-label phase. The reports will be different moving forward given that subjects in the placebo group have recently been vaccinated, but we think it's important that we continue to follow as subjects remain further out from their initial vaccination. So if you go to Slide 26, you'll see that these data have been consistent also in studies outside of the Moderna clinical trial. So, we begin to see real-world effectiveness data demonstrate that Moderna maintains effectiveness consistent with what was seen in the COVE study. There are reports from Canada, from the United Kingdom and from Qatar and importantly, these trials confirm that there is vaccine effectiveness, not only against the Wuhan strain, but also against staging variance of concern, including the alpha, beta, gamma and delta variants and this is even after partial vaccination or vaccination with a single dose. So if you move to Slide 27, I'm now going to hand over the presentation to Stephen Hoge, who will review our COVID-19 booster strategy and clinical data as well as a review of our pipeline. Stephen?
Stephen Hoge:
Thank you, Jackie. So moving on to Slide 28. I want to start with an update of our perspective on COVID-19 and how it's impacting our strategy for boosters. So, first our emerging perspective. We believe today that the increased force of infection, that's resulting from the delta variant, fatigue with non-pharmaceutical interventions and the seasonal effects of moving indoors, will eventually lead to an increase in breakthrough infections in vaccinated individuals. In fact there have been reports of that already. While we see durable Phase 3 efficacy through six months, which Jackie just described, we do expect that neutralized hydros will continue to wane and eventually that will impact vaccine efficacy. So given this intersection between a rising force infection and waning immunity, we believe a dose three of a booster will likely be necessary to keep us as safe as possible through the winter season in Northern Hemisphere. So how has that informed our booster strategy? Our primary approach, since early this year, has been to advance that portfolio of booster candidates, against all of the potential emerging variants of concern, and so we have a large number of ongoing clinical studies, and I'll provide some update on some today. Those boosters are being evaluated often at two different dose levels, 50 micrograms and 100 micrograms. And they fall broadly into three categories. First is our prototype vaccine mRNA-1273, for which Jackie just described, the primary efficacy data out of our Phase 3 study. Second, we are looking at various specific booster candidates, beta and now a new delta variant-specific candidate. And third, we are looking at a multivalent platform, combining different variants into a single vaccine, first our mRNA-211 program and now a new mRNA-213 program, which includes the delta antigen. The goal of the multi-valent platform is to continue to try and stay ahead of where the virus is going by combining different antigens against emerging periods of concern. So, I'd like to provide a brief update today on the three pre-existing programs, mRNA-1273, our variant-specific booster candidate against the beta strain 351 and our first multivalent vaccine 211. Moving to Slide 29. I have the comparison of those three candidates from our phase 201 study. So quickly starting on the left-hand side, looking in the validated clinical assays. These are the same assays conducted by our collaborators at NIH that were used for our Phase III study and earlier clinical work. We can look at the wild-type virus neutralization of the three different booster approaches. Array left to right you see our prototype 1273, our beta specific variant concern 1273.351 and the multivalent combination of 1273 and beta which is MRNA-1273.211. As a reminder, all three of these were dosed at 50 micrograms. And what I'm showing you on the left-hand side is the pseudovirus neutralization titers in the validated assays, at day one, immediately prior to boosting, day 29 and in some cases day 15. Now, all of the participants in the study who have previously received the Moderna vaccine two doses, just as Jackie had described. And at approximately six to eight months after they had participated in one of our clinical studies, we offered them a chance for this booster. So you can see, at the baseline, day one, six to eight months after primary series, neutralizing titers are somewhat lower in all three cases. With a booster dose of vaccine, all three boosters, we were able to substantially increase the neutralization against the wild-type virus. As you can see for 1273, a 16.7 fold. Approximately, 11-fold at both day 15 and day 29 for the beta variant concern; and 38 to 46 fold with the 211 multivalent platform. So that's against the infectional virus on the left. How do we do against the variance of concern? Well, at the time when we initiated the study, we were particularly concerned about the B.1.351 variant that now its named beta variant; and the P.1 gamma variant. And what we did is we set up assays to evaluate the performance of the booster vaccine, two weeks after booster in all of those and again compared it against the ancestral virus D614G that we used elsewhere. What you can see, in all three cases, when you compare the boosting that's happening and compare that to the level of titers, neutralizing titers that was seen in each of those groups of people against their two months -- one month post their second dose, you can see strong boosting against all the variants of concern by all three booster strategies. In fact, it's very encouraging to see that 1273, our prototype vaccine was able to increase titers against the 351 strain and the P.1 gamma strain. Now as you look to the left, you'll see the level of boosting might be slightly higher with our multivalent platform, which is directionally higher titers, particularly against the 351 and P.1 strain. But overall the boosting remains strong across all three variances -- all three approaches. And as a result of this data, we've made the determination that against these variants of concern, we believe the prototype vaccine mRNA-1273 is more than sufficient as a booster and that there's no obvious advantage to working with the beta including variant boosters at this time. It's also worth noting that the epidemiology has moved away from the beta variant of concern towards the delta variant of concern that we've spoken about extensively. So how did we do then in that 1273 against the delta variant concern. Moving to Slide 30. I'm presenting data here which has been submitted for a manuscript. It's actually in press as we speak. Looking at the performance of 1273 over six months after primary series against the variance of concern. The third dose booster of 50-micron 1273 pseudovirus neutralization titers here in our research assays was shown. And just to orient quickly to the slide, we're looking against wild-type beta, gamma and delta variants of concern. The data on the first three columns deals with the month one post-dose two. So immediately following the primary vaccination series, for which we have the really strong efficacy data that Jackie just described. And as you can see, we see high titers against the wild-type strain 1,200 in this assay in this group and substantially lower titers against the beta and gamma variants of concern that has previously been reported. Following those subjects forward to months six to eight after their second dose, what you can see is waning both for the wild-type virus although it remains detectable levels of titers but also for the variance of concern particularly beta, gamma and delta. In fact, as you can appreciate in those middle four bars beta, gamma and delta neutralizing titers had fallen below the detectable -- the level of detection of the assay for a decent number of participants in all three cases. Now the great news is, that a 50-microgram dose of mRNA-1273 was able to boost against all four of those viral strengths. And 14 days post dose 3, we show the titers on the far right again all four bars. We saw 23-fold boosting against the wild-type strain to levels that are significantly above the level we've seen just after second dose one month post dose 2. Saw a 32-fold boosting against beta; 43 fold boosting against gamma; and 42-fold boosting against delta. Again all reaching levels that are significantly higher than previously seen. And that is very encouraging and we think confirms our selection of the prototype booster as likely to be protective against the circulating variance of concern particularly delta presently. So advancing that mRNA-1273 program at 50-micron booster into Phase II is something we did in parallel. And on Slide 31, I'm happy to provide an update on the Phase II results from that larger study of the prototype booster. So earlier this year our Phase II study of mRNA-1273 was amended to offer a third dose of -1273 at 50 micrograms to all interested participants six months out from their second dose of the vaccine. A total of 344 participants elected to receive that dose. And the top line results are reported here and we have a manuscript in preparation that we'll be submitting shortly. The results are generally consistent with what I've already shared from the [indiscernible] data. And that means that was that neutralizing antibody titers had wind significantly prior to boosting in six months no surprise. Our third dose 50 micrograms of mRNA-1273 boosted neutralizing titers to levels that are above the Phase III benchmark. And again this now being done in our clinically validated assays, with our colleagues at NIH. After a third dose, similar levels of neutralizing titers were achieved across age groups and importantly a subgroup analysis of older adults over the age of 65, so they were both able to boost significantly and achieve levels that are comparable to younger adults which is really encouraging. And the safety profile following the dose 35 micrograms was similar to that observed previously for dose two of mRNA-1273. In total, we think this is really encouraging data on the potential for a 50-microgram booster of mRNA-1273. Now moving to Slide 32, where does that leave us on our COVID booster strategy or a delayed third dose approximately six to 12 months after vaccination. But we believe a booster dose is likely to be necessary this fall, particularly in the face of the Delta variant. Our clinical data right now we think supports a 50-microgram mRNA-1273 booster and we see no obvious advantage for beta containing variant candidates driven both by the data I presented today and the evolving epidemiology. We're going to wait for 100-microgram data in the coming weeks to confirm the dose selection of 50 micrograms as the booster before filing. So moving off from the COVID booster, I'd like to provide a little bit of an update on the other parts of our pipeline. So moving to Slide 33. We were excited to update today that mRNA-1010 our first seasonal influenza vaccine candidate has entered the clinic and that -1010 is moving quickly towards -- in its ongoing Phase I/II study. I'll remind you that mRNA-1010 is a quadrivalent seasonal flu vaccine that's targeting the WHO recommended strength. And our vision is to eventually combine mRNA-1010 as a seasonal flu vaccine in a pan-respiratory vaccine booster for adult and elderly populations that combine flu a COVID-19 booster and potentially our respiratory syncytial virus vaccine. Looking on Slide 34, across our infectious disease portfolio, beyond -1010, we have a number of other important updates. In our RSV vaccine, our positive interim Phase I data was announced at Vaccine Day earlier this year; and Phase I dosing in pediatric and adult vaccine is ongoing. We recently were pleased to announce that we received FDA Fast Track Designation for adults over the age of 60 for this vaccine highlighting the significant unmet need that we believe is there in this disease. Our hMPV/PIV3 vaccine another multivalent respiratory vaccine is in a Phase Ib trial that's, currently enrolling in toddlers and the first cohort has been completely enrolled. Our CMV vaccine against a significant unmet need is on track to start its pivotal Phase III trial this year with roughly 8,000 participants. And lastly our Zika vaccine, Phase II trial is ongoing. We're currently enrolling patients in the United States and Puerto Rico. Moving to Slide 35. Beyond our expanding infectious disease item portfolio. We are advancing an mRNA therapeutics and now have seven programs in ongoing clinical trials. In oncology, our cancer vaccines programs with Merck include the personalized cancer vaccine and KRAS. And we have two intratumoral programs in ongoing Phase I studies. One, a triple program ourselves; and an IL-12 program in combination with AstraZeneca. In our cardiovascular therapeutic area, we have two programs the VEGF program ongoing with AstraZeneca in Phase II; and a preclinical Relaxin program. In autoimmune, we are excited to announce that we have started dosing our first patient in an IL-2 Phase I study and that continues. And the PD-L1 program remains in preclinical development. And lastly in Rare Diseases, we have been dosing for this year in our propionic acidemia program in Phase I; and MMA GSD1a and PKU programs are in preclinical and we look forward to starting this in the clinic. Briefly on Page 36, you can see our expanding pipeline of clinical programs and the continued advancement across all of our therapeutic areas but most notably of our prophylactic vaccines modality and infectious diseases. With that I'd like to turn the call back to Stéphane Bancel for closing remarks.
Stéphane Bancel:
Thank you, Corinne, David, Jackie and Stephen. Before taking your questions let me share a few thoughts. On this slide, we articulate how we have been speaking for many years now about using our mRNA platform to maximize our impact on patients. On the left, we now have two core modalities. For vaccines in blue we have achieved authorized products; and for systemic secreted and cell-surface therapeutics in pink, we have achieved human proof-of-concept with our chikungunya antibodies demonstrating we could get therapeutic level of antibodies and we could repeat those successfully. In the middle, we have four exploratory modalities, which were in the clinic with six programs to understand if we achieve human proof-of-concept. Four medicines in immuno-oncology; one in cardiology; one in rare genetic disease. More coming in the coming quarters. On the right, you see two modalities which are not yet in the clinic but for which we believe we have achieved an important derisking in non-human primates. Delivery of mRNA to the lung and delivery of mRNA into hematopoietic steam progenitor cells. Next slide core modalities. In vaccine we're focusing on bringing to market what we believe could be a game-changing respiratory vaccine that medicine would combine over time COVID-19 sales, seasonal flu boosters plus RSV booster in one single annual shot. We have a great vaccine against COVID-19. Our RSV clinical data have shown a high level of neutralizing antibodies which has led the FDA to grant of fast track designation and quadrivalent flu vaccine is in the clinic now. The flu market is 5 billion to 6 billion per year. Despite flu vaccine efficacy being progressive in several key mRNA 60% in a good year and down to around 30% in a bad year. We believe we can do better with our mRNA platform, given what we have demonstrated with COVID-19. The Moderna flu vaccine candidate could launch as early as 2023. CMV is now close to entering Phase II. And we continue to believe the market opportunity could be $2 billion to $5 billion per year. EBV should be in the clinic soon. There is no vaccine against CMV infection or EBV infection. So we are working towards moving the company from COVID-19 prime vaccination sales in 2021 to COVID-19 booster in 2021 and 2022 and beyond and then adding through as early as 2023 and then adding early and launching CMV and the ABB. I am excited about this vaccine pipeline and how these vaccines could not prevent death, hospitalization and disease in billions of people over the years. Given the research team are not resting. We are working in our labs on more vaccine candidates and we cannot wait to share with you these new vaccine candidates in the future as we take them into clinical studies. This is where the power of mRNA as an information molecule and the industrialization of Moderna and the digitalization of Moderna and our cash position will enable our teams to bring in potent vaccine to protect people at the scale and at the sales, not seem before in the domestical industry. In autoimmune disease, we aim to continue to scale that therapeutic area. Next slide, exploratory modalities. Six clinical programs in four exploratory modalities. If we get positive clinical signal, we scale these modalities quickly as well. That's where the beauty of platform comes into play. Next slide, modalities in research. Vertical of our long-term mindset and our desire to maximize our impact on patients, we continue to invest in science to explore new modalities like the one in the lung and more recently presented at our Science Day in hematopoietic stem cells. In our lung program, we are both working on using mRNA to express a human protein in a cell that was our first Vertex partnership, but we are excited about our exploration of doing gene editing using mRNA to express a gene editing enzyme. That is our second Vertex partnership. Next slide. I believe that the uniqueness of Moderna is that we're expanding our impact on patients in two dimensions. We are expanding in two dimensions at the same time because we have built and industrialized our mRNA platform. Because mRNA is an information molecule, once we make a medicine work in a modality like the COVID-19 vaccine, then with the same four building blocks of life to make another vaccine. So we scale within a modality really fast along the first dimension of the Y axis. On the second dimension is to keep expanding new applications of mRNA into new cell types on the x-axis. And we are doing both at the same time with different dedicated teams, all powered by our science for digital and our manufacturing infrastructure. David shared earlier of prime work for capital allocation, now that we are generating significant amount of cash each quarter. Our number one priority as a company has always been and we continue to be to invest in the business. We continue to believe that we're in the early days of a new class of medicines, mRNA, and these small molecules more than 100 years ago, a large molecule 50 years ago are any indication for next 10 to 20 to 40 years are going to see a very large number of important medicines. And because mRNA is an information molecule, it would happen really fast. We aim to continue to build the world's leading mRNA company. So we're talking about the investments in acceleration of programs already in the clinic, investments in new development candidates moving from the labs to the clinic, investments in our platform to continue to improve the science and to include new modalities, investment in manufacturing, investment in commercial presence, including our digital commercial presence, investment in digital so we can scale our company faster and better than traditional biotech or pharma companies. Our second priority is to expand our horizons by complementing our platform with external technologies or products. This means we are interested in nucleic acid technologies, gene therapy, gene editing, mRNA. On the mRNA front, if we find new delivery technologies that could expand our current capabilities, we will look at them carefully. We would be interested in technology licenses and or in development candidate licenses and or -- if it makes sense, M&A. You can count us to be disciplined. We know what it takes to go from early research to filing regulatory agencies. And for having done it, we know the risk associated with new technologies and everything that works in the mouse will not necessarily work in a human. From a strategic standpoint, we are not interested in small molecule or large molecule development candidates, even if they could complement our commercial portfolio. We love information molecules too much to be interested in acquiring analog molecules, extra molecule, large recombinant molecules, or cell therapy. The share buyback is our first one. We are very optimistic about the future of Moderna and we are just getting started. Our board of directors will continue to regularly review that is the right thing to do to return capital to shareholders in light of our cash generation, balance against investments in our business, and external investment opportunities. Let me talk now about corporate responsibilities. It is very important for us to make Moderna, a model of corporate responsibility and to build a sustainable business. Like everything we do, we don't just want to do things well. We want to do things in the best possible way. We were pleased to announce recently the establishment of our charitable foundation with an upfront endowment of $50 million with a significant focus on other sale populations. We are humbled to have been awarded the number one spot on Fast Company 2021 for best place to work for innovators, which complements our last six consecutive years of recognition by science as a top employer. Both awards reflect the importance we place on our employees and the culture of the company. Recently, Axios-Harris released its 2021 Corporate Reputation survey and the Moderna brand ranked third among 100 of the most visible U.S. companies, our first mention on the list, and a highest ranking for any biopharmaceutical company for corporate reputation. We have recently worked relentlessly with the U.S. government to facilitate donations of the Moderna COVID-19 vaccine to many low-income countries. And we are committed to minimizing our energy footprint. As I look through the next decade, the same north star is guiding us and since we started. We believe we have a responsibility to maximize our impact to protect healthy people and help patients. Our commitment is stronger than ever. This is just the beginning. We look forward to welcoming you at our annual R&D Day on September 9th, whereas in previous years, we will do a detailed review of our development pipeline. The team and I will happy now to take your questions. Operator?
Operator:
[Operator instructions] Your first question comes from the line of Salveen Richter from Goldman Sachs. Your line is now open.
Salveen Richter:
Good morning. Thank you for taking my questions. Could you walk us through the dynamics for 2022 and beyond in terms of APAs for Spikevax essentially demand in the context supply of the 2 to 3 billion doses for 2022, and then how we should think about the years beyond 2022 and pricing? And then secondly, with regard to external BD, how are you thinking about integrating areas like gene editing and gene therapy with your platform?
David Meline:
Maybe I'll start on the answer on 2022-2023. So, as we said, right now, we're -- we've announced that we're increasing our capacity. We see very strong demand continuing in the context of the pandemic well into 2022. And hence, we've given you the range of 2 to 3 billion doses depending very much on whether our customers are still purchasing for primary series or if they're looking at boosters and then depending on the eventual dosage for booster. So I think it's really going to evolve as to exactly what that looks like in terms of dosage. Going beyond 2022, as we said, we are starting to see now the forward-planning countries that are looking beyond the very near-term. We're starting to then have contract discussions and, in fact, have agreed some contracts into 2023. But I think it's early to really know as to how this is going to evolve in terms of the transition from pandemic to the endemic phase. In terms of pricing, I think, it's helpful to start with where we are in 2021 to have a context for understanding pricing going forward. So really three buckets of pricing in 2021, we have the U.S. government where the first 100 million doses was priced at a little over $15. The subsequent 400 million doses were contracted at $16.50. And that pricing was considering a couple of things. One is the BARDA funding we received to underwrite our Phase 3 trial and also the size of the contract, the 500 million dose contract, which is very large. The second category is the higher-income ex-U.S. countries, where as we've said in the past, we start with a price range of $32 to $37 a dose. And there are some cases where we offer discounts based on volume for high volume. And then, the third category is low and middle-income countries, which have received the lowest tiered pricing, including those sales to COVAX, which are considerably lower than the price to the U.S. government. So if we start with that framework for 2021, what we can say is that, the contracts that we've signed now for 2022, the pricing constructs are very consistent with that framework that we've had in 2021. And so we see a continuation in the context again of the pandemic with the pricing framework. If you look at the average price that you calculate in your model, of course, that's going to depend on the mix across these categories. And, of course, we're expecting to see, significant sales to the middle and low-income countries and that increasing in 2022. So it shouldn't be surprising, if the average you see some declines. And then, finally, I would just comment that as we move into a post-pandemic period, then we would expect as we've said in the past to market forces to impact our price negotiations. So hopefully, that answers your question.
Stephen Hoge:
And Salveen, I'll maybe just pipe in at the end with your first question on gene editing, which is -- how we see intersecting. Look, I think we have been, as you all know, the innovator in mRNA and lipid nanoparticle delivery in therapeutics for a while. And we've watched the space quite interestingly or quite significantly in terms of ways that we could help with delivering gene editing cargoes across a range of different tissues where our lipid nanoparticles systems have been shown to go even in humans. And we think it's the right time for us to start to expand in that direction. If there's a general convergence, I think out there in the gene editing space is that messenger RNA and lipid nanoparticles are perhaps the way to go. And that's something we strongly agree with, having spent the last decade working in the technology. So you'll be looking for us to bring new payloads, new capabilities, new enzymes into our existing technological capabilities, which we think are best-in-class.
Salveen Richter:
Thank you.
Operator:
And your next question comes from the line of Matthew Harrison from Morgan Stanley. Your line is now open.
Matthew Harrison:
Great. Good morning. Thanks for taking the questions. A couple of related questions on boosters, if I may. I guess, first question is, maybe you could just put in context some of the information we're hearing from the FDA or the CDC, especially ACIP on their position on boosters and how you would expect that to evolve over the coming months. Second, could you comment on the potential for a multivalent booster? And how you might be thinking about that in the context of the data you presented today, especially just on using a third dose of the existing shot? And then, third, could you maybe just comment on your views of long-term virus evolution? Obviously, typically, viruses tend to evolve towards more infectious, but lower virulence. And so I'm wondering what your thoughts are on the long-term booster market obviously versus the sort of near-term booster market when infections may still be quite high? Thanks.
Stephen Hoge:
Sure. Thank you, Matthew. So let me try and take the first question, first. So I think we are going to always defer to what's happening with the public health officials in terms of when they think the appropriate time to recommend a booster vaccine is necessary. Where we see the data ourselves, I can't speak to the challenges they face. But what we see is the potential for waning immunity. In fact, if you look at -- back at our Vaccines Day, we had Professor Davenport come in and present work that he's done at University of New South Wales in Australia, showing what he predicted back in March would be the picture for waning immunity from the vaccine. It was recently published in Nature Medicine. I had a chance to open and it's looking remarkably prescient because the prediction he was making about the relative strength of the different vaccines suggested that small differences in efficacy would start to emerge to be larger differences in efficacy at about 200, 250 days as neutralizing antibody titers wane. And that may be what we're starting to see. And if you play that forward if you assume he's been right about those predictions, then that picture continues and continues through a year with continued declining -- neutralizing antibody titers over that time. And eventually, we therefore believe a real increase in breakthrough infections and disease even with vaccinated -- even with mRNA-1273. So, we continue to want to be vigilant because that trend and those predictions we think will come to four. And I think the Delta variant has cost us to also be incredibly humble in the face of the virus' ability to fight back and increase its transmission. I mean I think most of us would have thought SARS-CoV-2 was a pretty good infector earlier this year. Delta has shown us that it can make huge steps forward. And so for all those reasons we think that it's appropriate to be cautious. Our approach is not to is to defer public health and when boosters are going to be necessary, but to bring forward the best option as we see them based on the science that we see in the evolving epidemiology. And that's where I think our conclusion today is given beta, gamma, and particularly, delta given the real-world efficacy that we're seeing out there against delta right after vaccination 1273 and the neutralizing titers that we can see that I presented today against delta with 1273 protype dose that we feel pretty confident that that's actually the right way to approach this round of the fight with the SARS-CoV-2. Now, if I give to the second and third questions you asked, this is not the last round of the fight with SARS-CoV-2. We expect it to have at least a couple more rounds and maybe annually we're just going to continue to fight this virus back. And that's where we think multivalent boosters continue to be an important part of the scientific strategy. As you look forward to say early 2022, delta is what we're fighting right now, but what are we going to be fighting in 2022? What new variant of concern? There will be one. And I look at the evolving picture with Delta and the overall variance of concern and there's a couple of specific things that jump out at me. There are now I think five-point mutations in the various variants of concerns, three of them present in beta and gamma. We've talked about them the 417, 484 and 501 mutations. And now there's two mutations in the receptor binding domain in the delta strain at 452 and 478. And those five look like the ways in which the virus has tried to step away from or neutralizing community with our vaccine immune innovation. If you think about how this might play forward, it seems logical to us that those three mutations present in the beta, gamma line and those two mutations present in the delta might find some way to combine in new and potentially scary ways. And if that came with the increased transmissibility force of infection that delta can achieve, that might be a significant threat. And so we view our multivalent platform as the best place for us to try and anticipate that threat. And logically, for us right now that would be looking at a beta gamma or a beta variant a concern combined with a delta variant concern and evaluating that going forward. And that's the 213 program that we're going to be looking at. But we don't think that for this cycle. We do really believe that 1273, a booster dose will hold up against delta right now. The long-term virus evolution question is a great one. And I would say we just got to be humble. We've not faced a variant a virus quite like this. And again I don't think any of us would have predicted the step change in transmissibility that was seen with delta over the last five, six months. And so I wouldn't rule out that the virus doesn't have that kind of surprises in its future. But if you take a very, very long view five, 10 years view, I would say that we continue to think the model for what SARS-CoV-2 will look like in terms of an endemic market is probably predicted by other respiratory infections the endemic coronaviruses like OC43, which every year have rates of reinfection in adult populations and young kids every year results in hospitalizations and some deaths including in this country. And we therefore, believe there will be a long-term endemic market. The virulence of those viruses as you pointed to is lower and that's good news. Hopefully, it's not as big of a threat as we're seeing right now, but we need to be cautious and humble because SARS-CoV-2 keeps surprising us. And maybe that variance will be something more substantial than we see in the endemic coronavirus. So we're hopeful that it will wait that virus will decline, but we really do believe the virus is here to stay for the long-term. And therefore, there's going to be a need to regularly boost particularly high-risk older populations against SARS-CoV-2 into the future. Hopefully that answers your questions.
Operator:
And your next question comes from the line of Ted Tenthoff from Piper Sandler. Your line is now open.
Ted Tenthoff:
Great. Thank you very much and thank you for all the thorough information just on spike that, but also on the pipeline. I guess my first question has to do with capacity and really trying to understand a little bit more fully what goes into continuing to grow capacity, especially overseas. And I guess the second question would be with respect to the worsen disease pipeline which is, I think you guys normalize events just in terms of application for mRNA. How can we be moving faster there? Again appreciating that you guys have a pandemic you're trying to address. But it seems to me like everything is set to go there. And just curious what we can be doing to maybe accelerate some of those important programs. Thank you so much for taking the question.
Stéphane Bancel:
Yes. Maybe I…
Stephen Hoge:
Go ahead.
Stéphane Bancel:
So Ted, I'll take the capacity and then Stephen will talk about rare disease. So as you recall said, we've announced -- I think it was in February that based on the market feedback of the countries given the high efficacy of our vaccine. So there was a lot of demand, and so if you recall we decided to make very significant manufacturing capacity increase 50% addition in the U.S.. It's all at our U.S. site for drug substance in our wood and then the doubling of the OUS capacity at Lonza and Rovi. Our goal is to, as I said in my remarks, depending on the booster dose, which until we have final say by the regulator and until we see the 100-microgram data for 1273 dose, as Stephen described we won't know for sure. But if you model both the prime series APs that have already been ordered, which will be of course at 100-microgram. If you model what we anticipate in the mix between prime series and booster vaccines for Spikevax, if the booster dose was 50 micrograms, we could have up to $3 billion of supply. And if the booster dose were 100-microgram that could take us up to €2 billion of supply, so it's a bit hard to think about it. But we're trying to do is to not have the challenge we're having this year, which is a happy problem. As I said in my remarks, we are still tracking for 800 million to 1 billion dose this year. But I've also said that we are not taking any more orders for 2021, because we are totally maxed out. And of course, we would want to be in a position where we can answer to any countries wanting more vaccines. And so by doing those investments we really are hoping that next year we can make sure that we can fulfill all the demand we're going to get from the market. And as Corinne mentioned, while we have already signed 12 billion of APAs for next year and with those countries and additional 8 billion on top of the 12 billion of options that those countries have, there are still a lot of discussions ongoing. And so what we want to do is to maximize the penetration of Spikevax around the world. And I believe that the new data of this morning showing that the efficacy -- the final efficacy of the cost study is holding very nicely. I think we'll just be yet another argument for countries to want to vaccinate as many people as we can in that country with [Indiscernible].
Ted Tenthoff:
That's helpful.
Stéphane Bancel:
Stephen, do you want to talk about acidemia?
Stephen Hoge:
Sure. Yeah. I mean -- so I said I think you know we've had a long-standing commitment to these populations and that's as strong today as ever. And we hear and want to do everything we can to accelerate these medicines for them. It's important to say this is fast to the finish is the goal, not the fast at the start. And while we're pleased with the start of the propionic acidemia program and hopefully shortly MMA and others in the clinic, our goal is obviously to rapidly move through those Phase 1 studies that find the right dose and hopefully then rapidly move into pivotal studies. And as you know that can happen very quickly. And so particularly in rare diseases, particularly with some early positive clinical data. And so that's what we're trying to do right now is anticipate that positive data, expand and build out our team in that therapeutic area and begin the more foundational preparations for closing very quickly, if we can get some encouraging positive clinical signs. But we're working every day to try and get the -- to accelerate the time that those are incurring early data as well as I said prepare for that future.
Ted Tenthoff:
Fair enough. Thank you gentlemen.
Stéphane Bancel:
Thank you.
Operator:
Your next question comes from the line of Michael Yee from Jefferies. Your line is now open.
Michael Yee:
Hi. Good morning. Thanks for the questions. Two questions. One on boosting. Do you guys have a good sense of what you think the regulatory view or hurdle is to support boosting? You show that great data on Slide 30 showing the waning of the antibodies. And I think we all see that could be a problem and how the third dose gets you way up. But what do you think that specific data is or put another way the tighter level for correlative protection would be to support boosting for the fall or the winter? That's question one. And then question two, a little bit similar. On flu with that data coming up later this year is it your view that significantly higher levels of antibodies will be to significantly higher efficacy and that would just a larger loss to be supportive of a launch I think you said in 2023? Thank you.
Stephen Hoge:
Sure. So I'll try and take the first question which was -- so and you pointed well, we do not currently have a correlative protection in the world unfortunately for any of the vaccines. And so it's very hard to say objectively, what tighter, what level is this sort of the minimum level, which is why in our minds and subject to the regulators to developing their own perspective. But in our mind, the right way to benchmark this has been, let's look at that really consistent high durable efficacy in Phase 3, let's look at the neutralizing titers, let's support that. And let's do that. Let's get above those titers, because if we can exceed those titers and where we were just after the primary vaccination series, then it should stand to reason that we should be able to provide durable protection at or above the levels that we saw before. Now of course the virus is evolving and that's where you see delta and we have to be humble about that. But the good news is -- it looks like in the real-world data that the vaccine 1273 is holding up against delta even with partial vaccination as Jackie mentioned in her slides and with some of those references. And so we do think that getting at or above those levels, should hold up quite comfortably. How much above those levels? Is it 1.0? Is it 2.0? Is it some other number? I think that's ultimately going to be a sense that we want to have between ourselves of the data that we have, the benefits of the different dose levels 50 microgram, 100 microgram. And again, the data we've already seen in terms of Phase 3 and then a dialogue with regulators about how they see that benefit risk. But as it stands today, we think the 50-microgram data that we presented really looks encouraging and likely meet that standard. But we want to hold off, we look at the 100-microgram data in just a few weeks here and decide no that's the right call. We've got to the levels we need to. And we think we can reset immunity in a vaccinated person with a third dose at or above the levels that have been driving this durable protection to date. I'm sorry I missed the second question. Could you just repeat it?
Michael Yee:
Yeah. Similar I guess in flu that when you have data later this year, I suspect you believe the tighter levels will be extremely high. I think there's some better understanding based regulatory guidance documents that for flu there's some acceptable levels and you could certainly compare it to approved products that high levels there would be supportive of a larger study and a faster market?
Stephen Hoge:
That's right. Yes, thank you for reminding me. So we -- obviously, we haven't provided guidance on when we expect the -- to be able to get through those subsequent studies and ultimately, with that regulatory path is because, we have to engage in that discussion with the FDA and global regulators and those are ongoing. And so, it's really subject to them agreeing. But I would agree with your characterization, it's certainly our hope and view that because it's a well-understood market, with things like the HAI titers that even a new platform like messenger RNA might be able to leverage some of that thinking in terms of immunogenicity and safety, as we think about moving forward to approval. We'll ultimately have to show efficacy and real-world efficacy as well, because that's what's going to be of interest for payers. But again, the regulatory path is going to be subject to discussions with regulators that are ongoing. So, I can't provide more guidance at this time.
Michael Yee:
Thanks.
Operator:
And your next question comes from the line of Gena Wang from Barclays. Your line is now open.
Gena Wang:
Thank you. I have three quick questions. The first one is, if boost turned out to be 50 microgram, should we still expect similar price range among the three buckets during pandemic phase? And then number two is, could you walk through the clinical trial development path for a single shot vaccine against COVID flu and RSV. And the third question is regarding the external investment opportunities. You mentioned that expanding to two new modalities, lung and hematopoietic stem cells, with gene editing with liponanoparticle delivery, currently focusing in the liver. Does that mean you are willing to expand to liver diseases?
Stéphane Bancel:
So let me take the first question, Gena. On the boost, the price is not linked to the mass. So we anticipate the price of boosting to be set up and not related to those. I'll let Stephen talk about the second question. Is that okay?
Jackie Miller:
So I'm happy to take that question, Stéphane. This is Jackie. And it's around the clinical development plan for a booster combination vaccine. And the good news is, there have been multiple combination vaccine developments in the past, maybe not for this kind of a groundbreaking indication. But typically, what we do is license the initial components first, and you know that we are working on our BLA for COVID. We're preparing Phase 2/3 for flu and for RSV. And then, we would look to license the combination vaccine through immunobridging.
David Meline:
Thank you, Jackie.
Stéphane Bancel:
And I would…
David Meline:
Go ahead, Stephen. Stephen, go ahead.
Stephen Hoge:
I was going to take the gene. I think unless you want to step on. One comment to on Jackie's point, we already do have combination vaccines like the HPV, we have three vaccines. So we're we -- hopefully, we do have some experience there technically. On the gene editing question, we do have, as you know, programs that target the liver. But as we've presented at previous science days and even today, we have a platform technology that also we think allows us to get in broadly into the immune system and particularly hematopoietic stem cells. And so what you'll -- where we imagine our strong suit to be is in delivering nucleic acid technologies to those areas. And of course, as we look to expanding in gene editing, you'll see us look to those technologies that we've got the most experience with first and then bringing a range of different payloads to -- into our capabilities.
Gena Wang:
Thank you.
Lavina Talukdar:
This is 25 minutes past the hour. So I would hand it over back to Stéphane Bancel to make closing remarks.
Stéphane Bancel:
Thanks, Lavina. Sorry, everybody, if you're still hearing us with the technical problem. We will work with -- we'll provide -- we'll figure what happened. If you have any follow-up questions, please don't hesitate to contact Lavina, who will make sure to reply to you quickly. Thank you for coming in today, and we look forward to talking to you at the latest for R&D Day on September 9. Thank you, and have a great day. Bye.
Operator:
Good morning. May name is Dee Tamar, and I will be your operator today. Welcome to Moderna's First Quarter Earnings Conference Call. At this time, all participants are in listen-only-mode. Following the formal remarks, we will open the call up for your questions. Please be advised, that the call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Dee Tamar. Good morning, everyone. Thank you for joining us on today's call to discuss Moderna's first quarter 2021 financial results and business update. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. On today's call are Stéphane Bancel, our Chief Executive Officer; David Meline, our Chief Financial Officer; Stephen Hoge, our President; Tal Zaks, our Chief Medical Officer, Corinne Le Goff, our Chief Commercial Officer and Juan Andres, our Chief Technical and Operations Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the Safe Harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see slide two of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. On slide three, please see the important indication and safety information for our COVID-19 vaccine, which has been authorized for emergency use in the United States and many other countries around the world. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone, Thank you for taking the time to join our Q1 2021 conference call. We’ll start by quick business review of the quarter before Corinne walks you through commercial update. David will then walk you through the key financials. Stephen will provide the clinical update, especially new human data about two of our COVID-19 booster candidates, mRNA-1273, the currently authorized vaccine, and mRNA-1273.351, the variant-specific booster to B.1.351 first adopted by in South Africa. I will then come back to close. The Moderna COVID-19 vaccine is now available and protecting people in 37 countries around the world. And with the WHO authorization last Friday night, the number of countries where our vaccine will be available will go up significantly. In the first quarter alone 102 million doses have been shipped and many tens of millions of people have been fully vaccinated or received their first dose. 12 months ago in Q1 2020, Moderna had never run a Phase 3 clinical study, never gotten a product authorized by regulator and never made 100 million doses in a single quarter, not even 10 million, not even 1 million doses. I am very proud of what the Moderna team has achieved, and most importantly, I am very thankful for their impact on the world and the incredible customer sacrifices that our team has made to well protect fellow human beings around the world. This is very humbling and I am fortunate to lead Moderna in this moment. I am also thankful for Moderna scientists, engineers, doctors and team members, who have worked relentlessly over the last 10 years now to be ready for when the virus emerged in late 2019. We invented technology to produce safe, well tolerated mRNA vaccines, which made it possible for us to chase this horse. The Company achieved revenues of $1.9 billion in Q1 2021, of which $1.7 billion were COVID-19 vaccine product sales. The net income for the period was $1.2 billion. This marks the Company’s first GAAP profitable quarter in its history after many years of operating losses. At the end of 2021, we had cash and cash investments of $8.2 billion. David will give you more details in a few minutes. We increased our 2021 supply forecast once again. We now believe that we should be able to supply 800 million doses in 2021, and we are still aiming for 1 billion doses for the year. The total advance purchase agreement signed for delivery in 2021 have been increased to $19.2 billion. We are happy to report this morning an interim update to our TeenCOVE study, the initial interim analysis of our Phase 2/3 TeenCOVE study of mRNA-1273 showed vaccine efficacy against COVID-19 of 96%, and mRNA-1273 was generally well tolerated with no serious safety concerns identified to date. We’re still on track to start this month the filing of our rolling BLAs with FDA for COVID-19 vaccine mRNA-1273. Our type [ph] of COVID, we added another first as a Company. And that is the dosing of our first patient with an mRNA therapeutics candidate against a rare genetic disease, propionic acidemia, for genetic deficiency in the liver we have a candidate mRNA-3927. One of the things that I am most excited about is where we’re going. The Q1 results highlighted both the consequence of last year work and decisions we made. So, as I look to where Moderna is going, I get very excited by the level of our increased investments across the board. Using our strong balance sheet to invest to scale Moderna. Just two numbers of some color. In Q1 2021, our R&D investments were approximately 4 times higher than the R&D investments in Q1 of last year. Not 4% or 40%, 4 times higher. For all of you who have known us for many years, Moderna has been built as a digital enterprise since the early days. But we now have the opportunity to do much more and to build new functions like clinical trial operations, pharmacovigilance, commercial, digitally from the get-go. So looking at the next five to 10 years, we’re investing intensively in digital automation and AI. Our plan for 2021 is to invest 3 times more in digital than in fiscal year 2020. We announced last week that we have decided to invest to increase our 2020 supply to up to 3 billion doses. Let me share with you why we decided to recommend to our Board to invest at that scale? First, let’s talk about the science of SARS-CoV-2 virus. New variants of concern continue to emerge around the world, and we believe that over the next six months at the southern hemisphere and its fall and winter, we could see variants of concern emerge. We expect for a while now that we believe booster shots will be needed as we believe that the virus is not going away. We also believe, from a scientific standpoint, that the highest efficacy booster over time will be provided by multi-variant-specific booster. Second, the market has changed quite a lot versus what we knew six months ago. First, mRNA vaccines have emerged as a best-in-class vaccines, high efficacy, good tolerability profile, having to scale manufacturing and speed to chase the variants in the clinic. Many companies are still in the clinic with their first generation vaccine, while we are in the clinic with variant-specific boosters. More importantly, as we are talking to governors around the world in the West and in the East, in the North and in the South, we’re hearing loud and clear from the market, supply us with more mRNA vaccine for primary series and supply us with more mRNA vaccine in the future for boosters for 2022 and 2023. There is a big shift versus what the market perceived six or nine or 12 months ago when protein vaccine or adeno vaccines were thought to be the answer to the pandemic. We believe it has become an mRNA market for COVID-19 vaccine. Third is on our pipeline. We believe we will bring to market several more products in the next two years, that will add to the market demand for COVID-19 boosters for years through vaccine as we discussed at our Vaccine Day and our goal is for the seasonal flu vaccine combined with the COVID variant booster in a single dose product. We’ll have strong clinical data for RSV vaccine and CMV vaccine. Plus we have seven programs in clinical studies in three therapeutics area and more programs to move from preclinical development to clinical studies in the months to come. So we decided to build capacity to deliver up to 3 billion doses of supply in 2022 to serve both the North and the South. We’re doubling our drug substance supply in Europe and increasing by 50% of drug substance supply in the US. We are, of course, adding filling capacity in the US and Europe at our existing partners, but also adding new ones as we speak, more to come. Another piece of feedback we’re hearing from the market is that, Moderna has the best-in-class mRNA vaccine, shipment of minus 20 Celsius and storage, not minus 70 Celsius. Small cartons of 100 doses. Storage up to six months in standard freezer and four weeks in a regular refrigerated temperatures. The only authorized mRNA vaccine that does not require on-site dilution. We believe this is an even more important feature today, that will be more in the future in 2021 and 2022 and 2023 as we move to a booster market and decentralized in pharmacies and at doctor’s office. We believe we have the best mRNA vaccine authorized and we wanted to improve our product, continue to have the best-in-class products of the mRNA market. We are delighted to announce this morning the start of the dosing of our first patient in our Phase 1/2 study with propionic acidemia disease. The study is called Paramount. This is yet another milestone for Moderna. Not only do we have, I believe, the most innovative infectious disease vaccine in clinical pipeline, but we also have therapeutics candidate in clinical studies in oncology, in cardiology and now in the rare genetic disease. Let me close my remark on this familiar slide. We now have 1,500 employees. We’ve recently incorporated Moderna Japan KK, and we continue to build our commercial network. We will push to Asia-Pacific in 2021. And given our strong balance sheet of $8.2 billion, we’re going to continue to accelerate and invest to allow Moderna to scale and maximize the impact of a broad mRNA platform towards as many people as we can. Let me share our perspective on yesterday afternoon announcement by the United States Trade Ambassador, that the US government will support waiving intellectual property protection for COVID-19 vaccines. We believe this will not help supply more mRNA vaccines to the world any faster in 2021 or in 2022, which is the most critical time of the pandemic. There is no idle mRNA manufacturing capacity in the world. There is no industry of talented individuals who are skilled in the art of making high-quality and high purity GMP-grade mRNA vaccines. There are no companies who have developed manufacturing processes, purification processes, analytical processes that would allow them to quickly run a clinical file [ph] And if approved by regulators around the world, then provide hundreds and millions or billions of supply of mRNA vaccine. We have announced in the Company’s statement issued October 8, 2020 that during the pandemic, Moderna will not enforce COVID-19-related patents. You can find that statement on our website. We believe that the best way to end the pandemic is what we’re currently doing
Corinne Le Goff:
Thank you, Stephane, and good morning or good afternoon, everyone. As all of you already know, Moderna’s COVID-19 vaccine is our first authorized product, and on the back of it, we have turned into a commercial Company very quickly. So today, I’m delighted to give you an update on the commercial progress in the first quarter. I will start with our most recently signed supply agreements, those that occurred in the first quarter and at the beginning of the second quarter this year. I am particularly happy to announce our agreement with COVAX, which will provide access to our vaccine to millions of people in low- and middle-income countries, and is in keeping with our global access principles. In total, our COVAX agreement is for 500 million doses for delivery in the 2021 and 2022 period. Specifically, in 2021, Moderna will begin delivery of 34 million doses in the fourth quarter of 2021. COVAX will have an option for an additional 466 million doses in 2022. We are grateful to all the collaborative efforts of CEPI, Gavi, UNICEF, the World Health Organization, and the Moderna commercial teams in making this important supply agreement a reality. Moving now to the additional supply agreements signed for both 2021 and 2022. We have signed additional supply agreements with Israel for 5.3 million doses in 2022, with an additional option of 17.3 million doses for 2022 and 2023 and with Switzerland for 7 million doses in 2022 and options for an additional 7 million in late 2022 and 2023. We have also signed new deals for 2021 delivery with Botswana, Brunei, and in addition, we have also signed an agreement with Zuellig Pharma, our distribution partner in Southeast Asia, Hong Kong, Macau and Taiwan. In total, we announced advanced purchase agreements totaling 845 million doses to be delivered in 2021 to the countries that are listed here on this slide. And we continue to have discussions with countries, both those we have already contracted with and new countries for supply in 2022 and beyond. In our discussions, as Stephane said, we are hearing consistently from governments that in their view there is no other technology that provides the high efficacy of mRNA vaccines and the speed necessary to adapt to variants, while at the same time, allowing reliable scalability of manufacturing. We are grateful for the trust placed in us from the various governments we have signed agreements with and we look forward to supplying the vaccine to other countries and helping end the pandemic and getting ahead of variants. Let me now turn to product sales. Our first — our product sales for the first quarter of this year were $1.73 billion and were recorded for the delivery of 102 million doses. Product sales in the US were approximately $1.4 billion and sales outside of the US to the EU, Canada, Switzerland, Israel and Singapore were approximately $400 million for the 14 million doses delivered in the first quarter. In the US, we have successfully completed the delivery of the first 100 million doses to the US government within 100 days of emergency use authorization, and we expect to complete the delivery of the second 100 million doses to the United States government before the end of the second quarter. As you know, the US production started earlier and it’s roughly one quarter ahead in production ramp, as such, in the second quarter of 2021 we expect the ex-US ramp to be similar to that of the US ramp in the first quarter. To close, I want to reiterate that as we continue to produce and rollout vaccines into the global market, we are humbled and proud to be part of the solution. I will now turn the call over to David Meline.
David Meline:
Okay. Thank you, Corinne. Today, as with our last earnings call, we are presenting our results primarily on the US GAAP basis. In some cases, we also provide additional detail to provide greater clarity on underlying trends. With this background, we are providing an analysis of actual 2021 first quarter results, along with an updated view of key drivers of financial performance going forward. Turning to Slide 18, total revenue was $1.9 billion in the first quarter of 2021, compared to $8 million in Q1 of last year. Following our first-ever product sales of $200 million in December 2020, we recorded product sales of $1.7 billion for our COVID-19 vaccine in the first quarter of 2021. Grant and collaboration revenue increased to $204 million in Q1, primarily due to increases in grant revenue from BARDA to accelerate development of our COVID-19 vaccine. Cost of sales were $193 million in the first quarter, benefiting substantially from previously expensed pre-commercial inventory costs, which I will discuss in more detail on a later slide. Research and development expenses were $401 million for Q1 2021, compared to $115 million for the same period in 2020. The higher spend was driven by increased COVID-19 vaccine clinical development activities, including our announced efforts around booster, variant-specific and multi-valent vaccine candidates. Headcount increases, as well as pharmacovigilance activities related to our COVID-19 vaccine also contributed to the year-on-year expense increase. Selling, general and administrative expenses were $77 million for Q1 2021, compared to $24 million for the same period in the prior year. The growth in spending was driven by increases in personnel outside services and costs associated with commercialization of our COVID-19 vaccine globally. Our provision for income taxes was $39 million in Q1 2021, reflecting a benefit from utilization of our net operating loss carry-forward, as well as discrete items. I will provide further context on the following slides. We recorded net income of $1.2 billion for Q1 of this year, compared to a net loss of $124 million in the same period of last year. Earnings per share on a diluted basis was $2.84. Please note that our share count on a diluted basis now also includes the effect of outstanding options and RSUs as we began to be profitable. Previously when we were in a net loss position, basic and reported diluted number of shares were the same. Turning to cash and selected cash flow information on Slide 19. We ended Q1 2021 with cash and investments of $8.2 billion, compared to $5.2 billion at the end of Q4 2020. The increase is driven by our commercial sales and additional customer deposits received in the first quarter for future purchases of our COVID-19 vaccine. Net cash provided by operating activities was $2.97 billion in Q1 of this year, compared to net cash used in operating activities of $106 million in Q1 of last year. The reversal from net operating cash outflow to cash inflow was driven by our commercial market entry for the entire quarter. Similar to last quarter, before providing an updated financial framework for the remainder of 2021, let me summarize a few areas from our Q1 results that are important to keep in mind when modeling expected 2021 financial performance. Starting with product sales on Slide 20. We started last year to build two distinct supply chains, one in the US and one outside the US for rest of world markets. Our supply chain scale up in the US was roughly one quarter in advance of our ex-US supply chain, which is reflected in the geographic sales mix in Q1. As we move forward in Q2, the ex-US supply chain is also ramping up toward full capability. Turning to Slide 21, cost of sales includes the cost of goods manufactured, logistics and warehousing costs, as well as third-party royalty costs. We began capitalizing our COVID-19 vaccine inventory costs in December of 2020, following the COVID-19 vaccine emergency use authorization, based upon our expectation that these inventory costs would be recoverable through commercialization of the vaccine. Prior to the authorization of our COVID-19 vaccine, inventory costs were recorded as research and development expenses in the period incurred. We expensed $242 million of pre-launch inventory costs in 2020 and started 2021 with the remaining balance of $187 million of zero-cost inventory. Almost the entire balance or $184 million was sold and benefited our cost of sales in Q1 of this year and hence, will not further impact future quarters in a material way. If inventory sold during the first quarter was valued at actual cost, our cost of sales would have been $377 million, or 22% of our product sales, somewhat favorable to what we expected, driven by favorable yields in our US production facilities. Now, turning to our cash and investment position on Slide 22. The cash and investment balance reported as of March 31 was $8.2 billion, up from $5.2 billion as of December 31, 2020. The increase was primarily driven by the net increase in customer deposits for future product supply of COVID-19 vaccine. The net balance of cash customer deposits increased from $2.8 billion at the end of December 2020 to $5.6 billion at the end of Q1 2021. Lastly, let me comment on tax-related items on Slide 23. The significant investments in our research, development and startup activities to develop the mRNA platform over the last decade have resulted in net operating loss carry-forwards with a balance of $2.3 billion at the end of 2020. As of December 31, 2020, we maintained a full valuation allowance against our deferred tax assets related to these loss carry-forwards. We perform a valuation allowance assessment during each reporting period based on the latest available financial information and outlook. After considering the weight of available evidence, both positive and negative, we concluded that as of March 31, it is more likely than not that the Company will be able to realize the substantial majority of its net deferred tax assets. This analysis included, not only our strong first quarter results, but also our April activity. The majority of the valuation allowance will flow through the P&L over the course of 2021 in our effective tax rate pro-rated based on the cadence of our expected pretax quarterly earnings. We also recorded two discrete benefits in our tax provision in Q1, which lowered our first quarter tax rate. The first benefit related to the valuation allowance release for the portion of deferred tax assets, which we expect to utilize in future years. The second related to the excess tax benefits associated with stock-based compensation. Turning now to the 2021 updated financial framework on Slide 24. Signed advance purchase agreements for expected delivery in 2021 reflect the current full-year total of $19.2 billion in anticipated product sales, including doses that have been delivered and recognized as revenue in Q1. Based on continuous progress to ramp up available supply capacity in our network, we have raised the lower end of our global manufacturing plan for 2021 from 700 million to 800 million doses at the 100 microgram dose level. Our manufacturing team and our partners are still working to supply up to 1 billion doses for 2021. Further, we continue to expect a range of deliveries in Q2 2021 of 200 million to 250 million doses. Our total cost of sales includes the cost of manufacturing, logistics and warehousing, and third-party royalties. For 2021, we continue to model average total cost of sales as a percent of product sales to be approximately 20% for the full-year with some variation quarter-by-quarter, largely driven by average selling price going forward. Now, let me comment on planned R&D and SG&A expenses. Q1 expenses of approximately $0.5 billion were stable compared to the underlying Q4 2020 expense run rate on a like-for-like basis. In Q1, our actual expenses were lower than the internal forecast, primarily driven by the timing of clinical development and commercial activities and related costs. We now expect a notable expense trend increase starting in Q2 on a quarter-over-quarter basis for the remainder of this year. Based on better visibility of the utilization of our accumulated net operating loss carry-forward, expected global sales mix and the mentioned discrete benefits in Q1, we now expect our all-in 2021 tax rate to be in the low-teens. This compares to our previous forecast in the mid-teen range. This forecast is based on current US tax policy in effect and does not include any future potential discrete benefits related to stock-based compensation. We will update this view as our business evolves further. Finally, regarding capital investments. We are raising our forecast for capital investment from our previous range of $350 million to $400 million for 2021 to $450 million to $550 million, including the planned capacity expansion investments as announced on April 29. This concludes my remarks concerning financial performance. And I now turn the call over to Stephen.
Stephen Hoge:
Thank you, David. I’ll begin with an overview of our COVID-19 strategy against variants of concern and the initial data from our Phase 2 booster vaccine study before ending with a summary of the rest of our pipeline. And before I go into the data, a reminder that our booster strategy is evaluating single dose booster vaccinations with three different mRNA vaccines
Stéphane Bancel:
Thank you, Stephen, Corinne and David. Our 2021 advanced purchase agreement signed have now been increased to $19.2 billion. As we look into 2022, we’re investing to build 3 billion doses of supply capacity because we believe the market need could be greater in 2022 than in 2021. First, we already have countries signing APAs for 2022 for additional prime series for children, but also for variant-specific boosters, Israel last week and Switzerland this morning. If you recall, they were some of the first countries who signed APAs in 2020, and again, these countries are ahead of the game for 2022 and 2023. Second, with the COVAX partnership announcement Monday, we anticipate to supply up to 466 million doses in 2022. Third, we are having active discussions with all the governments that have signed 2021 APAs with Moderna. For new APAs for 2022 deliveries, again, prime series but also boosters. Fourth, we are having numerous discussions with governments that do not have 2021 APAs with Moderna because we cannot supply them in 2021 fortunately. But many of these governments are already asking us to enter into 2022 APAs because they want high efficacy mRNA vaccines that are easy to store. This is why we decided to invest for more supply in 2022. We believe from our current deals and current discussions that the market wants more supply from us in 2022 than we can supply in 2021. As we look at the next five to 10 years, we have the most innovative vaccine pipeline in the industry, and we’re investing more in research to increase our impact by bringing to the clinic more innovative vaccines against viruses that hurt humans. We are now in the clinic in three therapeutics areas
Operator:
Thank you. [Operator Instructions] Your first response is from the line of Salveen Richter with Goldman Sachs. Please go ahead.
Salveen Richter:
Good morning. Thanks for taking my questions. I have a couple here. So, firstly, with regard to — if the US supports WTO waiver of COVID-19 vaccine IP, what does that mean for Moderna? I mean, if you could just walk us through that? Secondly, if you could just discuss contract dynamics for the vaccine in 2022 as you look to address variants and kind of you see them move towards an endemic market? And third, it’s nice to see the PA program move forward, it’d be great to kind of understand whether we’ll see data from that program this year? And what else we might see from the ex-COVID pipeline? Thank you.
Stéphane Bancel:
Salveen, good morning. It’s Stephane. Let me start with your first question and then I’ll turn the PA question to Stephen. So, on the IP, what does it mean? I believe it doesn’t change anything for Moderna. As I said, we had said last October that we will not enforce our COVID-19-related patents during the pandemic. And as I’ve said in my remarks, there is no mRNA manufacturing capacity in the world. This is a new technology. You cannot go hire people who know how to make mRNA. Those people don’t exist. And then even if all those things were available, whoever wants to do mRNA vaccines will have to buy the machine, invent the manufacturing process, invest purification processes, analytical processes and then they will have to go run the clinical trial, get the data, get the product approved and scale manufacturing. These doesn’t happen in six or 12 or 18 months. We have been working at this for years. And as you know, there are some smaller mRNA companies that are still in the clinic trying to get the products to a finish line. And so, we saw the news last night and I didn’t lose my [ph] sleep over the news during the night. On 2022 contract, the dynamic is, as I just described it, which is the market that’s tremendously changed, since the pandemic started last year before clinical data many countries, as you know, didn’t want to move, especially because of mRNA being a new technology they moved first on protein contracts, on adenos contract. And then the kind of mRNA contracts came later, more just in case, and then the clinical data came along, and the speed to get through approval. And so — and then you have the — in fact, the proteins are still not authorized anywhere in the West. And then you had the low efficacy of the adenos, the safety questions around the adenos, manufacturing scale-up issues that adenos companies have had. And the big question that scientists advising governments have, which is scale actually really boost adenos — with more adenos products because, by definition, you give again the same virus vector to somebody that we believe over time will get less and less response from it. So as you look at the marketplace, which what governments are doing, and given many governments, I think last year believed the pandemic will be gone quickly. Trust me, every governments we’re talking to believed this is going to stay for a long time. They have got massively educated by their scientists and their clinician. And they believe this virus is not going away. They believe boosting is going to be critical. They believe variant-specific boosting is to going to be the right way to do the science, and as you saw from Stephen’s presentation, this is what the clinical data are showing as well. And so, the dynamic is that current governments that have already contracts with us are calling for more, and we’re in active discussion with all of them to supply most of 2022 and 2023, both prime series and specific variant booster. The beauty about the technology is, we can agree right now in the contract to give them next year but we having to choose what they want based on the clinical data and that’s an incredible competitive advantage and this we can do because, as you know, the manufacturing process is the same for 1273 or 1273.351 or 1273.211 or our new 1273-dot-something [ph] new variant. And we can change up on a very short notice because it’s the same equipment in the same room, with the same people, with same raw materials. And then all those governments, which is almost more exciting to me that never called before or that will be called before, but we couldn’t supply them because we are in a unfortunate position to say, look, we’re very sorry, we have no more supply for you in 2021. This is, of course, a very difficult discussion to have given the suffering happening around the world. But the great news with the investments we’ve announced to get up to 3 billion next year, we now can have those discussions with those governments and Corinne and her team are having a lot of discussions with them. So that’s kind of give you a sense for the dynamics. Stephen, on PA?
Stephen Hoge:
Sure. Salveen, thanks for the question. So, as you know, the program, the Paramount study for propionic acidemia is going to be looking at biomarkers as a part of its dose optimization. And so, it’s possible that we’ll be seeing very early indicators of impact there. But we are going to — if there is no guarantee that the first dose level and the first cohort that we’re looking at will be the correct one. And we’re going to make sure that we develop a cogent and consistent data set before we bring that forward. It is a dose optimization study and we will perhaps be looking at multiple dose levels. So, while I think it’s possible that we would see data this year, it’s dependent up on many things that are well beyond our control. Now, you asked a more general question also about our broader portfolio, and if you look at the programs more generally, VEGF, as we mentioned, is a Phase 2 program that’s been enrolling for a while. It’s possible we could see data from that. Our PCV and KRAS programs, again, is open label programs, we’ll continue to track those closely as they enroll. And then similarly, intratumoral programs that we highlighted, many of them are ongoing and producing data and, of course, when we have a complete and cogent data set, we will bring it forward.
Salveen Richter:
Thank you.
Stéphane Bancel:
Thank you, Salveen.
Operator:
Thank you. Your next response is from Matthew Harrison with Morgan Stanley. Please go ahead.
Matthew Harrison:
Great. Good morning. Thanks for taking the questions. I guess, two from me. One, on the sort of next-generation COVID vaccine, where you think it might be a refrigerator stable. Can you just talk about the regulatory path for that vaccine given that it’s not the full spike? Do you think you might have to run an actual efficacy study or do you think a neutralization titer study with safety might be enough for that? And then the second question, Stephen, if I can just follow-up on PPA [ph] I know in the past, right, one of the struggles has been enrollment. Obviously, it’s great to see that you’ve gotten a patient into the study. Can you just talk about now that you’ve gotten a patient in, what your sort of view is around enrollment? And if you think you’ve gotten through some of those hurdles?
Stephen Hoge:
Sure. Thank you, Matthew, for both questions. So, first, on, I believe, you’re referencing our second-generation vaccine candidate, which is mRNA-1283. It is a shorter construct that we think could have a much longer refrigerant stability profile. As we announced previously, we’ve started enrolling in the Phase 1 in that study, and it’s probably a little bit premature to comment on what we think the regulatory path will look like for that until we get some of that initial data and have conversations, obviously, with regulators. But I would highlight that it’s possible that 1283 may not go into a full primary series vaccination study. It could impact in the future function as a booster. But, again, that’s — it’s probably too early to say we will have to wait until we see that data and ultimately, it will be dependent upon conversations with regulators in the future. As it relates to PA enrollment, yes, as you mentioned, we’ve been working very hard on that over the coming — over the past years and we’re quite pleased to have enrolled the first participant, the first patient in that study. And the team is working hard to enroll additional patients as quickly as possible. I think time will tell whether we’ve actually broken through here and addressed any of the issues that we previously had in terms of enrollment. And so, hopefully, we’ll be able to provide subsequent updates on expanding enrollment in the near term that will demonstrate that we’ve made that progress.
Operator:
Thank you. Your next response is from Ted Tenthoff with Piper Sandler. Please go ahead.
Ted Tenthoff:
Great. Thank you very much, and thank you for all of the detailed updates, including running through the financial balances, so detailed David. Congrats on all the success. Stephen, I wanted to pick up on the booster data that you’ve shown, and maybe can kind of take us a step forward, what is the booster strategy going to look like? Do we actually need to maybe re-dose or re-vaccinate sooner than eight to six months because of where the levels were? And maybe you can just tell us what you see as sort of the potential timing? Thank you. For when we’ll be getting boosters?
Stephen Hoge:
Thank you for the question, Ted. Look, I think we have to start by saying, we don’t know. We do not have data on when to expect waning immunity leading to breakthrough infections. But we do know that there is a raging pandemic that re-infections will happen at some point and the best way to ensure that we do not have renewed outbreaks in well vaccinated countries is to boost and maintain the highest possible levels of neutralizing immunity. We, as Moderna, also believe that that means we want to maintain the broadest neutralizing immunity against the largest number of then circulating variants of concern. If you look at the data that we have posted today, as well as some of our published data and others reports, it does feel like immunity to a primary vaccination series or a previous infection seems to weigh in over the six- to 12-month time horizon, at least as measured by neutralizing titers. Again, we don’t know whether that’s a clinical correlate or not, but it certainly is an indication of that waning immunity. And if you look at the data that we — that I presented earlier, approximately half of the participants in our booster studies, no longer have detectable neutralizing immunity against the variants of concern. They have neutralizing immunity against the ancestral strain that they were vaccinated against. So the logical thing we think to do is to boost their immunity against those variants of concern, if you will vaccinate them against those to both increase those titers right now, but also give them a longer duration of protection, perhaps long enough that we can see our way through the pandemic. That probably looks like boosting on a nine to 12-month after primary series as an annual booster for now, at least while we’re continuing to see the evolution of the virus. Now, the last point was about our strategy, more generally here, and we do believe that the virus is not going to follow one path of evolution that we are going to see many variants of concern that there may be divergent paths. And therefore, the best way to ensure that we can protect against the broadest number of variants of concern will be a multi-valent vaccine. Right now, we’re still waiting to see our multi-valent vaccine data, which is a combination, as you know, of ancestral and 351 and the strain first identified in South Africa. But we think this is just the beginning, and we think we’re going to be unfortunately continuing to fight this pandemic through 2022, at least globally. And therefore, we are committed, as a Company, to make as many updates to the vaccine, to add as many variants as we think are necessary to ensure that when people receive a booster, it provides the broadest immune protection against the widest range of variants.
Ted Tenthoff:
Incredibly helpful. Thank you, guys, for all the work you’re doing.
Stéphane Bancel:
Thank you, Ted.
Operator:
Thank you. Your next response is from Michael Yee with Jefferies. Please go ahead.
Michael Yee:
Hi. Thank you. Good morning. Appreciate the questions. I had two important follow-ups. One was going back to the question about the WTO. Can you just offer some color around the view of loss of raw material supply capacity, etc? In other words, shedding some light on any ability to actually increase global capacity even if there were some form of open patents? So maybe just talk about that because I don’t think that you can just make it. I like it to have easy. Can you maybe just offer some color there? And the second question is also a follow-up on the variant strategy, it sounds like the bivalent strategy might be the best, Stephen. So, at what point would you just pull the trigger on beginning to manufacture that and ramp that all up for 2022? Thank you.
Stéphane Bancel:
Thanks, Michael. It’s Stephane. Let me start on the raw material and the IP. Going back to what I said, if somebody was to start from scratch, because again there is no mRNA player that’s with idle capacity out there. One will not start by focusing on large-scale raw material supply. I mean, one will have to first figure how to make mRNA. And you cannot find that patent, which, as you all know, on the Internet, on the US Patent office website. And so, one will have to figure out what machine do you need? How do you make mRNA? What purification methods you need? What analytical methods you need? And once you have figured out all those things, which, trust me, is going to take you time. It is not easy, and there are companies that are on the market with mRNA vaccine that they are carrying [ph] for decades. And even companies that have been working on it for 10 or 20 years are still in the clinic trying to figure out how to get to the finish line. And so, I really believe that this is not the issue. I already believe the IP topic is mostly critically driven. This is not the issue. It might impact other technologies that had adenos [ph] and protein, at least I could have comment on, but for mRNA I really think this is the wrong question. Stephen, do you want to take the variant?
Stephen Hoge:
Yeah. So thank you, again, Michael, for the question. So, on our multi-valent strategy, we have — at this point, we are still waiting for the clinical data to confirm that. And then we expect to have that shortly, as we’ve mentioned, we previously dosed people with the mRNA-1273.211 variant. The preclinical data that we have published does — are presented and does suggest that that is going to be the winning approach. And as I highlighted or as is highlighted by the mono-valent clinical data we already have, there is a benefit to adding additional antigens and potentially therefore benefit with a multi-valent approach. I think it’s important to recognize that we view this is an ongoing battle. And so, your question about when do we pull the trigger and move forward bivalent manufacturing? We’re already on the path of doing that manufacturing, not because we think that we’re done with mRNA-1273.211, the current bivalent vaccine, but because we think we’re going to go down the path of multi-valent vaccines and continue needing to add things. And so, that platform capability, we are already in the process of building and establishing to support multiple updates to a multi-valent vaccine. And we do think that’s going to be required, because we think the virus is not going to standstill and stop evolving, and we suspect there is going to be trivalent, maybe quadrivalent. It will keep happening in the time ahead. We have completed GMP manufacturing of all of those batches and we’re at sufficient scale we think to be able to quickly move into commercial scale distribution if needed. But at this point, we are still waiting for data to come shortly to confirm that performance in clinic.
Stéphane Bancel:
Yes. Just a point to add to Stephen on the multi-valent strategy, a lot of people don’t have pushed it is, it is not easy to do a multi-valent mRNA GMP product from an analytical QC standpoint, because those mRNAs are the same size. They look mostly similar because you just change a few — I mean, a few nucleic acid and that takes time. And it’s where the platform comes to have so much value. As you all know, we have a CMV vaccine on its way to Phase 3, where we developed and as we find over the years, a very complex product, 6 mRNA in the same variant. And so, if you think about what the multi-valent vaccine for COVID is going to look like. It’s not going to be easier. So for people who have not done multi-valent in GMP setting before, trust me, the regulators, because we’ve had these discussion with regulators around CMV over years. They don’t want to see a lot of analytical method characterization, so that you can prove to them that you know what is in the pipe. And that is yet again another big differentiation with Moderna. Thank you, Mike.
Michael Yee:
Got it. Thank you, guys.
Operator:
Thank you. Your next response is from Gena Wang of Barclays. Please go ahead.
Gena Wang:
Thank you for taking my questions. I also have two, one also related to the IP question. So, wanted to ask differently. Just wondering, Stephane, how many contracted global manufacturing sites you have? And how long in general is the contract? And the second question also regarding the new booster data. This is more for Steve. Actually, to me it was a little surprised that differences between 1273 versus 1273.35 was less on narrow than initially I would expect. It seems like 1273 should be also sufficient to protect from variant strain. So, what could be the explanation? And then you did just lay out the plan, you still will be going after the multi-variant approach. But regarding the explanation there, like do you think that just single shot that would still should be sufficient for the protection?
Stephen Hoge:
Thanks, Gena. I’ll — maybe I’ll take the — that question first and then hand it back to Stephane on your IP question. So, a couple of things I would note. The first is the level of titers as you suggested there is little bit less than two-fold difference between them. And you are — but you are seeing substantially higher titers on the order of 1,400 when you give the variant-specific booster, that’s mRNA-1273.351. Now, I would note that this is happening already at day 15. All right. This is an early time point that we’re looking at this point. We will also be looking at day 29. And in this case, we are effectively, it is a prime with 1273.351. It is the first dose of the strain first identified in South Africa. And so, it’s actually, there’s two ways you could look at it, one is, obviously, that it is both look good. I think the other and the way that I’m still looking at this is, it looks like we can very rapidly direct the immune response to an increased level of neutralizing titers against the variant of concern that was first identified in South Africa, 351 in this case. And if you compare the titers that we’ve achieved even by day 15 between these two variants or between the ancestral strain and the 351 strain, it’s really only the 351 strain that’s getting to the same level that we saw against the ancestral strains in that last comparison, so to levels that are approximately similar amount. Now, that’s not to say that mRNA-1273 as a booster couldn’t — wouldn’t provide a benefit, and I think you’re highlighting that, Gena. There is evidence in this data as well that we can substantially increase neutralizing titers generally across the response with a booster dose of mRNA-1273, our authorized vaccine at 50 micrograms, and that is encouraging. That is good news, because I think it suggest that is also a useful strategy. But if you had to choose between the two, and you were primarily concerned about increasing immunity to a higher level so that it can last longer, particularly in patient populations at high-risk of either waning immunity or incomplete immunity, we think this starts to provide very early event [ph] even at day 15, even after a priming dose that there is going to be an advantage to some strain matching of the antigen. And that’s what has us continue to be excited about the multi-valent strategy. Stephane?
Stéphane Bancel:
Sure. Thanks, Stephen. So, Gena, on the contract manufacturers, I would kind of look at kind of raw material, drug substance and drug products. In all of these, we have multi-year contracts. As soon as we get Board authorization to go to a 3 billion supply for 2022, we right away send a lot of orders and a lot of additional supplies to our suppliers. And not to forget, the drug substance, actually, now this is the place, it is a Moderna site where we actually have the biggest capacity of drug substance even in the 3 billion doses 2022 scenario.
Gena Wang:
Thank you.
Operator:
Thank you. Your next response is from Geoff Meacham with Bank of America. Please go ahead.
Geoff Meacham:
Hi. Good morning, guys. Thanks so much for the question. I just had two on COVID. The first one is, what does your data tell you with real world effectiveness of 1273 today, as of now, with respect to some of the main variants? I’m just trying to reconcile that the need for annual boosters versus minimal breakthroughs thus far and high efficacy. And then the second question is, when the next-gen vaccines for COVID-19 when you have some permutations, what’s the potential to leverage the technology to use different parts of the virus versus just modifying the spike protein or do you think this could add regulatory steps that make it difficult, even if it’s theoretically possible? Thank you.
Stephen Hoge:
Thank you, Jeff. Those are both good questions. So, maybe I’ll take the first one. First, which is our real world evidence the largest amount of it that we’ve seen has been published by groups like the CDC and continues to reinforce that the efficacy we saw in the clinical trial seems to be translating well into real world use with very high efficacy against disease — against COVID-19. I think it’s important to note, though, that this is all happening very acutely, right? We are still only months away into our — months into these vaccination campaigns. And the primary concern that we and others have from a public health perspective is, really not what’s going to happen right after vaccination, but what does this look like in nine months? What does this look like in 18 months? And I think the really difficult situation everybody is in, is you could say, well, let’s wait until it’s a year from now, and we see in a reemergence of spikes of cases we see maybe it’s not as bad, but we see a very big in bad flu season in the winters, tens of thousands, maybe hundreds of thousands of death, that kind of scale. That’s not a situation that most are willing to take a risk on because it obviously could be substantially worse than that. And so, we’re probably not going to have a chance to wait for data for cases to really breakthrough a year from after vaccination in the real world setting and let that start to guide re-vaccination decisions. At that point, it’s almost too late. And so, I think at this at this level, we think for the very near term the correct and sort of conservative decision is to continue to try and maintain the highest level of broadest immunity in the populations that are well vaccinated already. Now, if you look beyond this sort of epidemic phase and pandemic phase that we’re in with this variant evolution into the years beyond that, so three, four, five years from now, hopefully, we’re well past the current pandemic. We still believe there’s going to be SARS-Cov-2 re-infections and, as we shared at the Vaccines Day just a couple of weeks ago, we take that lesson from the previous endemic coronavirus epidemics that have happened, where hundreds of years later, and you still see re-infections mortality, substantial healthcare cost associated with those viruses and we don’t know whether SARS-CoV-2 is worse than them or the same, but we believe that that burden of disease that’s created by the fact that respiratory viruses continually re-infect and when they do, they can really have a devastating effect in high-risk populations, particularly older immunocompromised. We think that’s a real probability in the future, in fact, it would almost be unprecedented for that not to be the case in the coronavirus context. And so, for that reason, we believe there is going to be a need for continual boosters, whether it’s annual or not, and whether the multi-valency continues to add more and more valencies, I don’t think anybody can say it. But it’s certainly a situation we’re preparing for.
Geoff Meacham:
And, Stephane, just on the second question on the different modalities or Stephen…
Stephen Hoge:
Yeah, Geoff. I’m sorry. Yeah. I apologize. And this is your second question, on different modalities and different parts of the entry [ph] So, I think what is pretty clear from all of vaccines, if you look across them, but certainly if you focus on the messenger RNA vaccines is that, the high degree of efficacy we’re seeing in vaccines right now is based on the spike protein immunogenicity. That is the antigens being expressed. Is it theoretically possible that non-spike antigens could have provided the same protection? I think it’s definitely possible, so forward looking possible. But I think you would be remiss to look past the multiple large Phase 3 trials that provide pretty conclusive evidence of the value of going after spike protein trying to prevent COVID-19. So, I think you would probably, if you went down that route, you have to re-demonstrate that efficacy, that may be increasingly difficult in a world where we have so many good choices in terms of vaccines. And so, I’m not exactly sure how we go down that path, yeah, even theoretically.
Geoff Meacham:
Okay. Great. Thank you so much.
Operator:
Thank you. Your next response is from Cory Kasimov with JPMorgan. Please go ahead.
Cory Kasimov:
Hey. Good morning, guys. Thanks for taking the questions. I want to go back to the topic of the future contracts. I know this has kind of been asked in a couple of different ways. But based on discussions and negotiations that are currently taking place, how much confidence do you have that there is going to be demand to fill up to 3 billion doses in anticipated supply that you think you could have next year, especially if people are getting a single annual booster in the future? And then the second question is from really a modeling perspective, are the price points currently being negotiated on future contracts comparable to what you have on the existing ones for 2021 just basically want to see if we should be assuming stable pricing for modeling purposes for 2022? Thank you.
Stéphane Bancel:
Yeah. So, let me take a stab at the question, Cory. And if I miss anything, Corinne, just please add some color. So, as I said in my remarks, from what we’re hearing from the customer, this is becoming an mRNA markets looking forward. And so, there are not so many players in the mRNA market. And if you look at what the future needs across the globe, even the fear to vaccinate adults, all adults are not going to get vaccinated this year on the planet. The math doesn’t work. And then you have adolescence, and then you have children across the world. And then you are boosting. So, when you add all those pieces — the reason we are building up to 3 billion of supply as a mix between the prime series and boosters is because we believe that this is what the world is looking for based on the daily engagements we have with governments around the world. It’s a very different set up than what it was a year ago. A year ago, you get people saying, oh, I’m going to get a cheap adenovirus vaccine because being suppliers at cost. This is not a discussion anymore today. The discussion is, I want some mRNA vaccine for multiple, I want variant booster-specific, I want multi-variant, I want the best thing because I don’t want a second and the third and the fourth year with this thing. I need my country to be back on its feet. Understand, Cory?
Cory Kasimov:
Yeah. No. And on the pricing question for modeling purpose?
Stéphane Bancel:
Yeah. On the pricing, I will give you any color, but again, there is no more discussion at all, but your price is this and there is a small company. The company will price at cost at $3. This discussion is gone.
Cory Kasimov:
Okay. All right, perfect. Thank you, Stephane.
Stéphane Bancel:
Thank you.
Operator:
Okay. Your next response is from Hartaj Singh with Oppenheimer & Company. Please go ahead.
Hartaj Singh:
Great. Thank you. Thanks for the question, and all the color. A question I would have is just on the 50 microgram going forward, the booster against variants. Would you see that potentially becoming your initial kind of prime boost vaccine possibly in the future or you think you’ll stick with the 100 microgram route whether it’s with 1283 [ph] also? And then just on OpEx, just any kind of color, when we think about 2022 and 2023, o David, what cost of goods sold could look like once these quarterly variations kind of flush out? And then what your adjusted operating margins could start looking like also? Thank you for the question.
Stephen Hoge:
Thanks, Hartaj. I’ll take a stab at the first one and then hand it to David for the other. So, on 50 micrograms, obviously, the data we shared today is a small number of subjects. But we previously shared and published our Phase 2 data on a slightly larger number of subjects looking at a 50 microgram primary series, that looked quite good and at least as measured by immunogenicity seem to achieve levels that were consistent with the 100 microgram dose. So, it’s certainly something we’re going to look at as to whether or not we could pursue a 50 microgram primary series. But how we get there, will depend upon data that we don’t yet have. Right? So, we will have to look at whether there are clear correlates of protection that we can use to bridge between those doses and/or we’ll have to look at different populations in which we started those doses. As an example, this has been shared, we are evaluating 50 micrograms as a potential primary series even in pediatric populations, as you can imagine, you don’t need perhaps a higher dose in younger people than you do in older ones. So, there’s a lot of things ahead of us in terms of looking at whether or not a primary series for 50 micrograms is possible. Certainly, for a booster series that is the top dose at which we’re looking. And as we look forward, we will continue to carefully evaluate whether or not we can adapt that as a target dose across all of our applications. But it will depend upon data.
Hartaj Singh:
Great. And then question on the - thanks, David. No. Go ahead, David.
David Meline:
Yeah. So, cost of goods and operating expense trends as in 2022 and beyond, I guess, what I’d say is, it’s a little early to start giving that kind of guidance for 2022. What I would say is that, if you look at our cost of goods and the cost of goods manufacturing thus far, we’ve been quite pleased with what we’re seeing as we’ve ramped up production initially here in the US. We’ve seen, as I said, yields have been better than we had foreseen as we did the initial planning. So that’s obviously very helpful. And we reiterated today we think right now the right planning assumption continues to be 20% cost of goods. As you move beyond 2021, you get into a question of vaccines versus therapeutics, we think cost of goods manufactured will be very competitive for this product, and therefore, margins — and the gross margins will depend very much on price levels, which I think it’s early to comment on. And then in terms of operating expenses, we are building out the Company. We continue to do that. And as I mentioned, we — while our overall operating expenses were quite stable at $0.5 billion in the first quarter, we do see that trending up as we now move through the year. And we’ll continue to invest appropriately to drive the portfolio investment and to build out globally. So, I would say, we’ll continue to do that, and we’ll give you better and more precise guidance here as we move closer to 2022 and beyond.
Hartaj Singh:
Great. Thank you.
Operator:
Thank you. Your next response is from Joseph Stringer with Needham & Company. Please go ahead.
Joseph Stringer:
Hi. Good morning. Thanks for taking our questions. Just another one on manufacturing capacity here. As you potentially move to next-generation COVID vaccines, I was wondering if you could give us a sense maybe even qualitatively in terms of the — given the modularity of the technology. What are potential manufacturing ramp would look like for some of these second-gen vaccine in terms of manufacturing capacity in the ramp relative to what we have seen with 1273? Thank you.
Stéphane Bancel:
Yes. It’s Stephane. So, with 1273 [ph] ramp has been constrained by manufacturing capacity. So, if you look at this year, the only reason we quote-unquote only supply 100 million to those in Q1, which is an extraordinary number is because we are building the capacity. And so, the way to think about it is, as Juan and his team are working hard to add new lines and to increase the capacity, the ramps of a follow-on products will be much faster because today manufacturing is slowing down the ramp. So, I anticipate that, as you think about the multi-variant booster launches, as we think about RSV through CMV launch, we will not be on the backfoot. As you know, as part of our 2020 budget that we did at the end of 2019, we didn’t plan for pandemic. We were supposed to be commercial several years down the road. And so, the team has done a remarkable job to get to this point, but we are — and we’re going to stay for, I would anticipate all of the year supply constraint. Corinne and her team would love to be able to sell more products because, trust me, their phone is turning red hot by calls from around the planet. And we would lover to be able to help protect more people, but we just can’t because we were not planning on the pandemic in 2020. So I anticipate that for variants and for new product launch, we will make sure that we’re not capacity constrained, which is why the 3 billion supply volume that some people might think it maybe too aggressive. As I said in my remarks, the pipeline of the companies are still going to pay with these. And so, this is just behind the multi-valent vaccine. So if you look at a couple of years out, and we’re not building manufacturing for six months. We’re going to be really happy to have that capacity so that as we launch products, we can supply the market every single dose that Corinne and her team can make sure that the market wants.
Joseph Stringer:
Great. Thanks for taking the question.
Stéphane Bancel:
Thank you.
Operator:
Thank you. Your next response is from Mani Foroohar of SVB Leerink. Please go ahead.
Mani Foroohar:
Hey, guys. Thanks for taking the question. One quick one, I’m starting on financials. You gave a little clarity on CapEx investments around expanding capacity for reduction. Should we think of that as level setting CapEx going forward — modest increase going forward? Or should we think of that as primarily a one-time build-out? And then secondarily, you’ve given a little bit of clarity, there’s a lot of clarity around COGS for this quarter versus the rest of the year. Going forward, so we think about the absolute COGS per unit, again pretty linearly related to dose. Or are other attributes, royalties, etc., differences in product use between different vaccines, would that suggest that that’s not the right way to think about it?
David Meline:
Yeah. So CapEx, if I understand the question is guidance for 2022 and beyond on CapEx, and again, unfortunately it’s a bit early to be able to comment. We — if you look now, we’ve increased our guidance for this year based on the development side that occurred over the last couple of months. Is that a steady state going forward into the future? I think for a company of what will be our size and scope and level of vertical integration, I think it’s reasonable to expect that we’ll continue to invest in our own capacity and therefore, you can expect we’ll have ongoing CapEx. Is it precisely in this range or somewhat above or below? I really wouldn’t want to give you that precision as of yet because we’re — it’s not clear yet. But I think an ongoing CapEx trend will be appropriate for the Company. In terms of COGS, I think that’s the right way to think about the cost of manufacturing this product, it will be impacted at some level by the amount of materials in the product. But when you’re talking about micrograms of materials, it’s really not that significant. So, I think you can as the simplest assumption assume, it’s a pretty steady cost of manufacture as we’re seeing right now.
Mani Foroohar:
Great. And a quick follow-up on some of the contracting. I know it’s been asked in various forms. You talked about the OUS manufacturing being about a quarter-ish behind US manufacturing. Right now, based on your disclosures on volume, doses actually delivered, versus what we have from the other mRNA competitor and duopoly right now, market share for you guys is somewhere between 5% and 10% in a lot of these countries and to be delivered doses. How do you think about catch up on manufacturing and deliveries? And how that influences the ongoing contracting for next year, i.e., does your stronger incumbent position in the US suggest you are better positioned for US contracting volume? Or do you see 2021, the results for 2021 contracting have nothing to do with 2022 and every year is a whole new game between you two?
Stéphane Bancel:
Yes, it’s a good question. So, as we said, the plan was always the plan we are executing on. And so, when we spoke to countries and set up those 21 APAs, we manage the quarter — delivery to the countries as we knew capacity will come online. We had not anticipated early part of the year that we will be able to export from the US. And the piece we should not forget is, as you know, it’s reported in the MediaDaily, the US is going to be, I mean, way too many vaccine very, very soon, if not, already. And so, the capacity that we are building in the US and we are adding is also going to supply the world. So, I think once we’d anticipate a very big acceleration of shipments, two countries outside the US as we go into Q2 and even more in Q3 and Q4 as we met our obligation to the US government.
Mani Foroohar:
Great. That’s really helpful. And could you guys comment on where you are — what you’re seeing in the real world in terms of vaccine hesitant/end market demand? There has been a couple of media reports that that’s starting to be more of a limiting factor as opposed to supply, at least in the US currently, certainly, not globally.
Stéphane Bancel:
Correct. In the US and you see it on the daily numbers published by the CDC for, I think, since mid-April roughly the number of vaccination per day in the country is going down, and it is not because of supply, if you look at the shipments coming from the companies, they keep increasing exactly as we have been saying. And so, now there is an oversupply of vaccine. It’s really a demand-driven problem now. So you see some states across the country that are still very active in vaccination. And we are looking, as we said, that we have very high rate of vaccination is going up every day with elder parts of the country, as you know, where they have way too many vaccine. As you have heard that the federal government I think yesterday or before has announced that they were going to stop to shift products from one place to the other based on actual demand and this were leading to vaccine as you going to see. The US doesn’t have a supply of vaccine issue anymore. That was true in January, it is not true anymore.
Mani Foroohar:
Great. Thanks. I’ll get back in the queue and let [ph] others.
Stéphane Bancel:
Thank you.
Operator:
Thank you. Your next response is from Simon Baker with Redburn. Please go ahead.
Simon Baker:
Thank you for taking my questions. Firstly, just going back to the debate around IP waivers. Stephane, as you said that the talk of IP waivers rather than misses the point about limited global manufacturing capacity. So, just to support that, given that you announced, you would unilaterally waive IP enforcement back in October. To your knowledge, has any company in the following seven months sought to exercise that freedom to operate? And also sticking with the vaccine, going back to Slide 28, do you have data on T-cell response over that six- to eight-month period post primary vaccination for 1273? And then just a quick question on the financials. David, I think you mentioned that in the SG&A in Q1 there were some costs related to effectively start-up of supply. I just wondered if you could give us any color on the non-recurring one-off elements of SG&A in Q1 as we think about the evolution across the year. Thanks so much.
Stéphane Bancel:
Thank you so much. So I think the first one on IP and Stephen will take the T-cell and David the financials. So, I’m not aware of any company of size that is going after mRNA. Again, going back to what I described, one would have first to figure out how to make mRNA GMP before asking the regulators to start the clinical study. And that doesn’t happen quickly. We are monitoring the field very closely as we always have, but at this stage, I think that this is really not the point that will impact, of course, 2021 impossible but even 2022. Stephen on T-cells?
Stephen Hoge:
So the data we have from today is an initial analysis on the boosters. We are — as we previously announced, we’ve been looking at the study ourselves and with the NIH, they are running a primary series vaccination study and also looking at other elements. And so, we will perhaps get T-cell data in the mid-term. But at this point, we do not have any T-cell data yet on the boosters. We do have ongoing studies with NIH on — in general on our Phase 1 and our own Phase 2 and if it becomes important in the future, we can obviously look at T-cell responses with waning immunity out six, 12 months in those studies as well. We don’t have a specific plan to do that at this point. But we are pretty encouraged by the historical correlation between our previously reported T-cell data and public T-cell data and the neutralizing titers that are obviously much easier to measure over time across a wider range of subjects in T-cells. And David?
David Meline:
Yeah. So, in terms of the expenses we incurred in the first quarter, including in commercial, yeah, we had some one-time expenses to set-up businesses around the world, but the preponderance of the total operating expenses, including in commercial, I would say, will continue. And therefore, that’s why I gave you some guidance that if you start at that $0.5 billion spend level in the first quarter in the fourth of last year, we’re expecting now to see that trend up notably as we move forward through the year, which we thought it would start sooner, but we now expect will start in the second quarter. So, running a business of this size on a global basis, we think that that spend level is quite reasonable, to be honest.
Simon Baker:
All right. Thank you very much.
Operator:
I’m showing no further questions at this time. I would now like to turn the conference back over to Stephane Bancel.
Stéphane Bancel:
Thank you so much for participating in today’s call and for the great questions. We look forward to seeing you at Science Day on May 27. Stay safe, everybody. Have a nice day. Bye.
Operator:
Ladies and gentlemen, that concludes today's conference call. Thank you for your participation. You have a wonderful day. And you may all disconnect.
Operator:
Good morning, and welcome to Moderna's Fourth Quarter 2020 Conference Call. [Operator Instructions]. At this time, I would like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, everyone, and thank you for joining us on today's call to discuss Moderna's fourth quarter and full year 2020 financial results and business update. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. On today's call are Stéphane Bancel, our CEO; David Meline, our CFO; Stephen Hoge, our President; and Tal Zaks, our Chief Medical Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. On Slide 3, please see the important indication and safety information for our COVID-19 vaccine, which has been authorized for emergency use in the United States and many other countries around the world. With that, I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning, or good afternoon, everyone. I hope all of you and your loved ones are in good health. Thank you for taking the time to join our Q4 2020 call. For the call, we propose to start first by looking back to fiscal year 2020 and then looking to fiscal year 2021. For fiscal year 2020, I will share a few slides to summarize the key elements of the year, before David shares with you the 2020 financials and also how we think about 2021 financial framework. For fiscal year 2021, I will share our business objectives and why we are so excited for 2021 and the inflection point this year will represent in our history. Stephen will start with an update on our approach to variance of concern and then Tal will provide an update on our clinical pipeline. 2020 was a historic year for Moderna. We started in January as an early-stage development company. By the end of July, we had become a late-stage development company. And by December, we had positive results from our Phase III COVE study of our COVID-19 vaccine showing efficacy around 94%. Our COVID-19 vaccine was authorized by the FDA and Canada Health in mid-December and we shipped 80 million doses to the U.S. government and the Canadian governments by year-end. We laid the groundwork for a global organization, adding commercial subsidiaries in 8 countries, 8 countries. By year-end, we have signed $11.7 billion of advanced purchase agreement for our vaccine. A team of a little over 1,000 members did all that. And we also continue to invest in science to improve further our mRNA platform. And we also advance on 20 programs in development across 5 therapeutic areas. And we did it without a large global pharmaceutical partner. The Moderna team executed swiftly and superbly in carrying out the complicated Phase III study while achieving remarkable diversity. The Moderna team secured the authorization of the COVID-19 vaccine by regulators. The Moderna team built out our manufacturing capacity, scaling up across many manufacturing processes, partnered with Lonza, ROVI and Catalent, and shipped close to 80 million doses by year-end. For context, we had made less than 100,000 doses of medicine in 2019. This was roughly a 200x increase in 12 months. And we're on our way to produce as many as 1 billion doses in 2021 and more in 2022. In 2019, we had no authorized products. We had negative cash flows from operations every quarter. And we anticipated needing multiple capital raises over at least 5 years until CMV would produce sufficient revenues for Moderna to break even in terms of cash flows and become a self-sustaining company. In 2020, we had our first product authorized and our first product revenues in the last 2 weeks of Q4. We had 2 quarters of positive cash flows from operations. We ended the year with a strong balance sheet and with cash generation in 2 quarters. Moderna has been changed forever. Moderna has been changed in a profound way. Moderna is now a commercial company with subsidiaries in 8 countries and a direct presence in many more through commercial distributors and partners. mRNA is an information molecule. We have a powerful platform, and this is just the beginning. We have a scalable platform due to our investment in IT, robotics and AI over the last 7 years. We have an amazing team, as you witnessed by what the team accomplished in the 12 short months of 2020. And we have a capital to invest and scale. On Slide 6, you have a summary of the countries in which we received emergency use authorization or conditional approvals and all were - started rolling submission that we did. On Slide 7, you get a sense for the commercial infrastructure that we built. For context, in May of 2020, Moderna did not have a single employee dedicated to commercial activities. We closed the year with 8 commercial subsidiaries, an incredible commercial team and commercial partners. We now have commercial operations in the U.S.; in Canada; in Europe, in the 5 largest markets, Germany; France; Italy; Spain; and the U.K. We established one in Switzerland, where we have both commercial capabilities to serve the Swiss market as well as manufacturing for geographies outside the U.S. with our partner Lonza and the support teams that we need in finance, HR, digital and legal. We have partnered with Medison in Israel and with Takeda in Japan. If you step back and think about it, this commercial network which we started building and will expand in 2021 is an incredible asset for the entire Moderna pipeline. We now have the infrastructure to commercialize all our products. These markets on the slide are some of the largest markets in the world. So by building the commercial network for the launch of our COVID-19 vaccine and deciding not to partner, we have actually financed and built the commercial network for all of our programs. We have become a fully integrated company, and in 12 months, proved that we're capable of running complex Phase III studies, scaling up manufacturing to commercial level, making hundreds of millions of doses per year and building our own commercial network. As I said, the Moderna team did an amazing job in 2020. On Slide 8, you get a glimpse of what Moderna looks like in February 2021. While most people in the world think of Moderna as a COVID-19 vaccine company, this is just the first product that we are launching. We have a platform, and our molecule mRNA is an information molecule. This is just the beginning. We are going to soon start a Phase III study for vaccine against CMV, cytomegalovirus, the number onecause of birth defect. There is no approved CMV vaccine on the market. The pharma industry has strived for around 20 years to make a vaccine against CMV. This is a complex virus, a vaccine of 6 mRNAs per dose, over which are 2 from a complex protein called pentamer. We believe that CMV vaccine will present potential annual peak sales between $2 billion and $5 billion. We have three programs in Phase II of personal cancer vaccine in melanoma or OX40 ligand in ovarian cancer and injected in patients' hearts, post heart attack, to revascularize their heart with new blood vessels. We have had 12 positive Phase I results. In addition to our COVID-19 vaccine, we have 8 infectious vaccine candidates, all for first-in-class vaccine like against CMV or what we believe could be a best-in-class vaccine like with influenza. But we also have a remarkable therapeutic pipeline. We have candidate therapeutics in immuno-oncology with 5 medicines in the clinic, 4 in rare disease program, 2 cardio program and 2 autoimmune disease programs. If you think about it, Moderna is going after the 4 leading cause of death by disease area
David Meline:
Okay. Thank you, Stéphane. Before reviewing the financial section of the slide deck, I want to comment on the approach we've taken sharing this information. This starts with the 3 Cs, these being clear, complete and comprehensible. Today, we are presenting our results primarily on a U.S. GAAP basis. And in individual item level, in some cases, we also provide additional detail to provide greater clarity on underlying trends and to facilitate period-over-period comparisons in line with our 3C goal. Reflecting the continued uncertainties related to the course of the evolving pandemic, along with the many challenges and opportunities we face as a newly globalizing commercial company, we also seek to avoid implying an unrealistic level of precision concerning future financial projections in the face of a range of outcomes. With this background, we are providing today the analysis of actual 2020 results, along with a view of key drivers of the financial framework going forward. We will continue to update and refine this information as business evolves. Turning to Slide 10. I want to briefly remind you about some key accounting changes which we presented on this slide in our Q3 call in October. As a result of the successful transition to a commercial company, following the emergency use authorization by the FDA and Health Canada in December 2020, we began to record product sales. We also began to capitalize inventory based on the expectation these costs would be recoverable through commercialization of our COVID-19 vaccine as opposed to expensing costs directly to R&D in the period incurred. And we also began to capitalize purchases of property and equipment and long-term lease assets as opposed to expensing costs in the period incurred through the lack of alternative use. With this context, let me turn now to Slide 11. Total revenue was $571 million for Q4 2020 compared to $14 million for the same period in 2019. Total revenue was $803 million for the full year 2020 compared to $60 million in the prior year. Following the authorization for emergency use by the FDA and Health Canada in December of our COVID-19 vaccine, we generated our first ever product sales. For 2020, we recognized $200 million of product sales for our COVID-19 vaccine, all in late December. Additionally, grant and collaboration revenue increased to $371 million in Q4 and $603 million for the full year, primarily due to increases in grant revenue from BARDA to accelerate development of our COVID-19 vaccine. Now turning to cost of sales. We began capitalizing our COVID-19 vaccine inventory costs in December, starting after the vaccine was first authorized based upon our expectation that these costs would be recoverable through commercialization of the vaccine. Prior to the authorization of our COVID-19 vaccine, inventory costs were recorded as research and development expenses in the period incurred. We expensed $242 million of prelaunch inventory costs in 2020. Hence, our cost of sales were only $8 million in 2020, comprised primarily of third-party royalties. If inventory sold during 2020 was valued at cost, our cost of sales for 2020 would have been $62 million or 31% of our product sales. Research and development expenses were $759 million for Q4 2020 compared to $118 million for the same period in 2019. Research and development expenses were $1.37 billion for the full year 2020 compared to $496 million in the prior year. The increases for both 3- and 12-month periods in 2020 were mainly due to increased COVID-19 vaccine clinical development activities, headcount increases, pre-launch inventory buildup and expenses associated with the equipment and lease facilities that were deemed to have no alternative use at the acquisition of such equipment. Selling, general and administration expenses were $79 million for Q4 2020 compared to $26 million for the same period in 2019. Expenses were $188 million for the full year 2020 compared to $110 million in the prior year. The increases for both periods were mainly driven by increases in personnel, outside services and start-up costs associated with preparation for commercialization of our COVID-19 vaccine globally. We recorded a net loss of $272 million for Q4 2020 compared to $123 million in the same period in 2019 and $747 million for the full year 2020 compared to $514 million in the prior year. Turning to selected cash flow information on Page 12. We ended Q4 2020 with cash and investments of $5.25 billion compared to $3.97 billion at the end of Q3. The increase is primarily driven by $1.7 billion of customer deposits received in the fourth quarter for supply of our COVID-19 vaccine. On the top half of the page, we present information from our 10-K and 10-Q filings. And on the bottom part, we provide the quarterly trend and the cash deposits received related to supply agreements for our COVID-19 vaccine. Net cash provided by operating activities was $2.03 billion for the 12 months ended December 2020, compared to net cash used of $459 million for the same period in 2019. The reversal from cash used to cash provided by operating activities is driven by total customer deposits. Cash used for purchases of capitalized property and equipment was $67 million for the full year 2020 compared to $32 million in 2019. Before looking forward to 2021, let me summarize a few areas from our 2020 results that are important to keep in mind when modeling 2021 financial performance. Starting with research and development and SG&A expenses shown on the top left quadrant of Slide 13. Prior to the authorization for emergency use by the FDA and Health Canada as a pre-commercial research stage company, Moderna expensed all costs related to the production of inventory as well as costs for property and equipment and lease expenses for current and some future contract manufacturing activities due to lack of alternative use. Adjusting for these items, which in the future would be capitalized and expensed as cost of sales, the underlying R&D and SG&A expense run rate for Q4 2020 was $0.5 billion for the quarter. Turning to the upper right quadrant of Slide 13. Cost of sales includes the cost of goods manufactured, logistics and warehousing costs as well as third-party royalty costs. The reported expense in Q4 of $8 million reflects the fact that we expensed all inventory-related costs until authorization of our COVID-19 vaccine. If valued at cost or actual cost of sales, including initial ramp-up costs, would have been $62 million or 31% of product sales. The cash and investment balance reported as of December 31 was $5.25 billion, $2.8 billion of which related to cash deposits from customers for future supply of our COVID-19 vaccine. Lastly, let me comment on certain tax-related items. The significant investments in our research and development and start-up activities to develop the mRNA platform over the last decade has resulted in a net operating loss carryforward with the balance as of December 31, 2020, of $2.3 billion, up from $1 billion at year-end 2019. As of December 31, we maintain a full valuation allowance against our deferred tax assets related to these loss carryforwards. We will continue to monitor the valuation allowance as we progress through 2021 and expect to utilize our loss carryforwards. With this context, let me now move to considerations for 2021, starting with an overview of advanced purchase agreements for our COVID-19 vaccine on Slide 14. We have disclosed advanced purchase agreements to supply our COVID-19 vaccine to 40 countries through the end of 2021, including the U.S. government for 300 million doses with options for an additional 200 million doses; the European for 310 million, with an option for an additional 150 million doses in 2022; plus 10 other countries, including Japan, Canada and South Korea, with announced doses totaling 186 million. We are thankful for the trust of governments around the world have placed in us to deliver a vaccine for their countries. All these agreements contain provisions for deposits and have contributed to our balance of deposits of $2.8 billion at year-end 2020. Negotiations with other countries are also ongoing, including with COVAX. Turning now to the 2021 financial framework on Slide 15. Already-signed APA agreements for expected delivery in 2021 reflect a total of $18.4 billion in anticipated product sales. Based on continuous progress to ramp up available supply capacity in our network, we have raised the lower end of our global manufacturing plan for 2021 from 600 million to 700 million doses at the 100-microgram dose level. Manufacturing is still working to supply up to 1 billion doses for 2021. Further, we expect a range of related - of released doses in Q1 2021 of 100 million to 125 million doses and 200 million to 250 million doses in the second quarter. Our total cost of sales includes the cost of manufacturing, logistics and warehousing and third-party royalties, as discussed previously. For 2021, we currently model total cost of sales as a percent of product sales to be approximately 20% for the full year, with some variation quarter-by-quarter, largely driven by the average selling price. We also expect Q1 reported cost of sales in percent of product sales to be in line with the full year average, including the benefit from the remaining zero cost prelaunch inventory in Q1. Now let me comment on planned R&D and SG&A expenses. The underlying Q4 2020 expense run rate, adjusted for transitional items as discussed on Slide 13, was approximately $0.5 billion. We currently expect our quarterly reported expenses to increase on a continuous basis through 2021 compared to the Q4 2020 adjusted run rate. In Q1, we expect an increase in the low double-digit percentage range relative to the adjusted Q4 2020 expense run rate of $0.5 billion. We will provide additional updates going forward as our global business rapidly expands. I would also like to comment on how we think about modeling our tax rate. In 2021, Moderna will transition to taxpaying status as we deliver on our COVID-19 vaccine contracts to customers. As a U.S.-based company, we start with the statutory 21% tax rate. This is impacted by global sales mix to the extent that non-U.S. rates are generally lower than in the U.S and by recovery against our prior accumulated losses of over $2 billion. As we start the year, we expect the all-in 2021 tax rate to be in the mid-teen percentage level on an ongoing basis, including in the first quarter. We will update this view as our book of business evolves further. Lastly, regarding capital investments. We currently plan capital investment in the amount of $350 million to $400 million for 2021. Roughly half of the investment is to further expand our COVID-19 vaccine supply capability, including supply capacity up to 1.4 billion doses at the 100-microgram level, with the balance of planned capital investments to expand our technical development, clinical manufacturing as well as our other facility footprint. This concludes my remarks concerning the financial performance, and I turn the call back to Stéphane.
Stéphane Bancel:
Thank you, David. Let me now discuss 2021 on Slide 17. We are still in February, and the Moderna COVID-19 vaccine is now authorized in 37 countries, and our team is continuing to engage regulators in new geographies, like Japan, Taiwan, the Philippines and more. We're also well on our way in the rolling submission process to the WHO, which will be key for low-income countries access via COVAX and UNICEF if we get the partnership done. We are committed to our principles of global access to our new class of medicines, where we recognize that the need for scale up of manufacturing operations have led to supply going to the U.S., Europe and other countries. We are working with COVAX to provide low-cost vaccines to the poorest countries and look forward to supplying our vaccines for COVAX Gavi and becoming a company that will be part of the global health infrastructure for many years to come. UNICEF is the procurement arm of COVAX. On Slide 18. As I shared in my 2020 shareholder letter, which we posted publicly on January 4 and which you can access on our block if you have not read it, I believe that 2021 is going to be the most important inflection year in Moderna's history. You can see the evolution of the company over 2019, 2020 and 2021 on this slide over a few dimensions. But to me, the reason I say that 2021 is the most important inflection year in the company's history, is that we are not the same company. As I've told our employees in a recent town hall, I almost wished I could change the company name to help our team understand the magnitude of the change. We used to believe that mRNA vaccine could be authorized and become commercial. We used to have negative cash flows from operations each quarter since our founding in 2010. We used to work to raise additional capital regularly and thought we needed to do that for another 5 plus years until CMV will get the company's cash flow breakeven. But we have been through an incredible pivot. This is not the same company. Now we know that mRNA vaccine can be authorized and become commercial. Now we have positive cash flows from operations for 2 quarters in a row already. So now we are going to double down on our investment in science, in process development, in manufacturing, in IT, in robotics, in AI to scale our company. I believe that in business, there's a drastic difference between believing in an outcome and having negative cash flow versus knowing an outcome and having positive cash flow. Our appetite and our ability to invest has been transformed. We are not the same company now that we were in the last 10 years. Our thinking is going to be much more expansive. On Slide 19. As David shared a moment ago, we have, as of yesterday, already $18.4 billion of signed advanced purchase agreements for fiscal year 2021 deliveries. We still have multiple discussions ongoing about additional APAs for 2021. So these have not been signed yet, so they are not counted in the $18.4 billion. We'll update you each quarter about the signed APAs for fiscal year 2021 as we report sales recognized with the shipment of our COVID-19 vaccine. We increased our manufacturing base plan for the year to 700 million doses and are working hard to get to 1 billion. We are working with COVAX and its procurement arm, UNICEF, to maximize the availability of our vaccine around the world. Let me now turn to Slide 20 to talk about 2022 manufacturing capacity. We announced last night after the market closed that several factors have led us to decide to add more manufacturing capacity. First, many governments have been telling us that they now see 2 classes of COVID-19 vaccine based on efficacy, and they would like more of our vaccine given its high efficacy. Second, variants. Government and political leaders around the world are concerned about the emergence of several variants. They have been very clear about it, and some of them have started to request options for 2022. With the start of the fall season soon in the southern hemisphere, with a large population of the world and with a large population of immune-compromised patients, like HIV-positive patients, we're hearing daily that governments around the world and the COVID teams worry that boosting people with variants will be an important strategy over the next couple of years to get this virus under control. So we decided to add manufacturing capacity. We have communicated previously that our capacity for 2022, given the ramp in 2021, will be approximately up to 1.2 billion doses, assuming 100-microgram dose. We have decided and have started to buy additional capital equipment, hire more people and order more raw materials towards 200 million doses per year of capacity for 2022. So if you assume a 100-microgram dose, we will have 1.4 billion doses of capacity for fiscal year '22. In other words, we will build the manufacturing capacity to make up to 140 kilograms, yes, kilograms, of formulated mRNA in 2022. Now let's talk about output in number of doses. Two factors determine output
Stephen Hoge:
Thank you, Stéphane, and good morning, everyone. Today, I want to take you through our current thinking on variance and our strategy to address them. I'll then turn it over to Tal to provide an update on our pipeline. Let me start with a quick overview of our strategy for SARS-CoV-2 variance of concern. Last week, we published a letter in The New England Journal of Medicine with data that confirmed the Moderna COVID-19 vaccine, mRNA-1273, provides neutralizing activity against all variants of concern tested to date. Nonetheless, as recent reports have increased transmission and potential reinfections of the new variants have emerged, out of an abundance of caution, we have announced multiple strategies to try to increase protection against those variants. As has been widely reported, the immune response generated by original strains appears to be relatively weaker against the B.1.351 variant. If or when immunity wanes in the future, this might lead to a gap in protection. We plan to close this potential gap with an update to our vaccine based on the new strains. We anticipate different approaches for 2 distinct populations. For those who have been immunized or infected by the original strains, we anticipate boosting with a variant-specific booster vaccine, either alone or in combination with our vaccine against the ancestral strains. For those who are still naive to SARS-CoV-2 because they have not been previously infected or vaccinated, we anticipate updating our vaccine to provide immunity to both the ancestral strains and the new variants of concern. Now on Slide 25, you can see the clinical trials that are ongoing or planned for our COVID-19 vaccines. For mRNA-1273, the TeenCOVE Phase II/III study in adolescents ages 12 to 17 years is ongoing and recently completed enrollment. The KidCOVE study, Phase II, in pediatric populations ages 6 months to 11 years, will begin in the near term. And our Phase I/II study in Japan is ongoing, led by our partner, Takeda. For the variant studies, we plan to test a variant-specific booster candidate, mRNA-1273.351 based on the B.1.351 variant, first identified in the Republic of South Africa at the 50-microgram dose level and lower. In addition, we plan to test a multivalent booster candidate, mRNA-1273.211 that combines mRNA-1273 and mRNA-1273.351 in a single vaccine, again at the 50-microgram dose level and lower. And we're also evaluating a third dose of our authorized Moderna COVID-19 vaccine, mRNA-1273, at a 50-microgram dose level, which is already underway. Finally, we are planning to test the next-generation vaccine, mRNA-1283 that encodes for the receptor binding domain and end terminal domain of the spike protein and is being developed as a potential refrigerator stable mRNA vaccine that could facilitate easier distribution and administration in a wider range of settings, including potentially developing countries. We believe messenger RNA is best positioned to address the potential threat of SARS-CoV-2 variance, given several key characteristics, including high vaccine efficacy, speed and agility to make updates, ease with which we can do combinations and our manufacturing flexibility and scalability. I'm happy to say that we have already manufactured the first GMP batch of our variant booster candidate, mRNA-1273.351, and have shipped clinical trial material to the NIH for clinical testing. Now I'll hand it over to Tal, who will walk you through the rest of our portfolio across vaccines and therapeutics. Tal?
Tal Zaks:
Thank you, Stephen. So let me briefly summarize where we are in the rest of our pipeline. We have 4 other vaccine programs that are in clinical trials. The CMV vaccine is on track to start the pivotal Phase III this year. The Zika vaccine is preparing for a Phase II trial that is also expected to begin in this year. And our hMPV/PIV3 vaccine is currently enrolling in toddlers. Our RSV vaccine is being studied in 2 separate trials, 1 in children and 1 in adults. The pediatric trial is enrolling quickly, and the first 3 cohorts in the age de-escalation study have now been fully enrolled. We announced last month that we were taking our RSV vaccine into the adult population, and I'm happy to share that the first participant has since then been dosed in that trial. Just last month, we announced 3 new development programs in infectious disease vaccines. Our influenza vaccine program will evaluate 3 candidates comprising multiple antigen combinations against the 4 seasonal viruses recommended by the WHO, eventually moving 1 candidate into a Phase III trial. Our HIV vaccine program has 2 approaches. mRNA-1644 is a collaboration with IAVI and the Bill and Melinda Gates foundation. It's a novel approach to an HIV vaccine strategy that's designed to elicit broadly neutralizing HIV-1 antibodies. mRNA-1574 is a collaboration with the NIH, and this includes multiple native-like trimer antigens. And finally, for those unfamiliar with the Nipah virus, this is a zoonotic virus transmitted to humans from animals, that is either transmitted in food or through direct human-to-human transmission. It is included in the WHO R&D blueprint list of epidemic threats needed for urgent R&D action. mRNA-1215 is a collaboration with the NIH to develop a Nipah vaccine. We also have 7 clinical proof-of-concept trials ongoing in our exploratory modalities. The Phase II program in VEGF, partnered with AstraZeneca, is ongoing. The personalized cancer vaccine Phase II, which is in combination with KEYTRUDA compared to KEYTRUDA alone, that is partnered with Merck continues. And the Phase 1 of that program in multiple cohorts is ongoing, including the upsized head and neck cohort that is currently recruiting additional patients. The KRAS Phase I that is partnered with Merck continues. And in the intratumoral oncology, we have a Phase II ongoing with OX40 ligand program in ovarian cancer patients. The Phase I dose escalation for the triplet program is ongoing both as monotherapy and in combination with durvalumab. And IL-12 partnered with AstraZeneca continues in a Phase I trial. Finally, within the systemic intracellular therapeutics, the Phase I/II sites for our PA program are being initiated, and we plan to enter the clinic this year. This final slide has our full pipeline. And with that, let me hand it back to Stéphane.
Stéphane Bancel:
Thank you, Tal. On Slide 32. We have a set of clear priorities for 2021, and everybody at Moderna has had a chance to review them and to discuss them. Priority number one, to maximize the impact of our COVID-19 vaccine. The output in 2021 in terms of number of dose that we can ship to countries, the manufacturing scale-up, preparing for 2022 supply, clinical development of variants and boost, and our full commitment to bring variants of concern to the clinic. Priority number two, accelerate vaccine development and continue to bring more innovation from a research lab to clinical development. Priority number three, generate proof-of-concept in therapeutics, based on the success of our ability to repeat dose in human of therapeutics technology with chikungunya antibodies. But also with intratumoral, we expect key data in cardiology, oncology, rate genetic disease and the entry in the clinic of our first autoimmune programs. Priority number four, we want to continue the expansion of mRNA technology. Our appetite to invest in science and process development has not weakened, just the opposite. We believe we are still at the beginning of the S-curve of this exciting new disruptive technology. On Slide 33, as I shared earlier, this is an important inflection year for the company, now that we know that mRNA vaccine can be approved, that we have positive cash flow, we have the ability to invest and to scale like we've never had in our history. If you look at Slide 34, you see some of the key attributes that energized me. mRNA is an information molecule, Moderna has a unique platform. We have a very strong cash position. We have signed very large amount of APAs. We believe Moderna will be cash flow positive this year. We believe Moderna will be profitable in 2021. The team has done a remarkable job, and I'm so proud of this team. We have a fully integrated manufacturing plant in Massachusetts and the network of partners with Lonza, Catalent, ROVI, [indiscernible]. We have already made 100 million doses of drug substance of the COVID-19 vaccine. We have shipped approximately 60 million doses globally. And as the team reviewed, we have 24 exciting first-in-class or best-in-class development programs that we are pushing towards the clinic - sorry, for approval. As our historic investors know, since I've joined Moderna as employee number two in 2011, we have always focused on how do we grow 10x from where we are. We ask this question to ourselves regularly. I have to admit that in 2020, I did not focus on it as intensely as in the previous years. We were focused on moving mRNA-1273 to Phase I, Phase II, Phase III regulatory authorization and on building manufacturing to deliver up to 1 billion doses for 2021. We did not spend much time asking how do we grow 10x? We were focused on how do we get this important vaccine to the finish line and get up to 1 billion doses to help people around the world. In early December, while our team was focused on preparing for FDA VRBPAC meeting on December 17, I was starting to step back and engaged with the Board and some of our executive team members and asked all of them, "Now that we know mRNA vaccine can get approved and that we are generating cash, how do we grow 10x from here?" The last two months have been really fun to invent that 10x Moderna. The team is highly energized by it and so am I. We have the opportunity to become one of the most impactful biopharmaceutical company over the next 10 to 20 years in the world. The potential to maximize that impact on patients is what motivates us. The team and I look forward to welcoming you to our regular investor events. We will host our Vaccine Day on April 14. We will host our annual Science Day on May 27. And we will host our annual R&D Day on September 9. More than ever, we have a unique opportunity to have a very large impact on so many lives. This is humbling to all of us and also highly motivating to always challenge ourselves to build the best version of Moderna that we can. I would like to thank our Moderna team, our partners, our many suppliers around the world, our clinical investigators, our participants in our clinical studies. I would also like to thank our investors for your trust as we embark in this new version of Moderna. The team and I will now be happy to take your questions. Operator?
Operator:
[Operator Instructions]. Your first question comes from the line of Matthew Harrison with Morgan Stanley.
Matthew Harrison:
I guess, first question, can you discuss the dosing strategy around boosting. And in particular, I think I understand why you're going to start with 50 micrograms, but what you think about the probability of using a lower dose there. And then just secondly, as far as I understand, I think the number of doses that have been signed via APA are higher than the 700 million that you've talked about in terms of manufacturing supply. Can you just talk about what your thoughts are in being able to deliver those additional doses this year?
Tal Zaks:
Go ahead, Stephen.
Stephen Hoge:
I'll take the - I'll try and take the first one and then hand it over to Tal as well. And then I think David will obviously take the second. The - so first on 50 micrograms, I'd remind you of a couple of things. So in this case, this is a third dose of a booster. And so I think we're quite optimistic that a substantially lower dose is necessary because the immune system has already been primed and boosted once. And this is just maturing and updating that immune response to new variant. And I'll note that we've shared our Phase II data, which does show that we see at 50 micrograms, even in a primary series, really good neutralizing antibodies. And so I think we're quite optimistic that 50 micrograms or lower will suffice as the third dose booster.
Tal Zaks:
This is Tal. The only thing I'd add is that, that has scientific precedence. There's been data. I, believe we had already a vaccine that was published a while ago that showed that if you come in months later, you can come in with a much lower dose and that is sufficient to provide a boost. So in the context of wanting to maximize the ability to provide a benefit from whatever given capacity, I think that strategy makes sense. On top of which, as Stephen alluded to, it could be that even 50 micrograms as a priming series could suffice. Recall that our dose at 100 supersedes what you see with natural infection in terms of neutralizing antibodies to begin with.
Stéphane Bancel:
Thanks, Tal, and Stephen. On the second question, Matthew, as you know, we have been cautious about talking about supply for the year, and we have raised the number as we have understood better the learning curve of the process, as we've had the lots made and being able to see the demonstrated output. I think it's important to always appreciate this is a new technology we have never made and nobody in the world has made at that scale so fast. And so it's important for people to understand, we invested and built the manufacturing capacity, the ability to make 1 billion doses. The piece, having worked myself in manufacturing at commercial scale, and of course, you know Juan Andres, who runs manufacturing - used to run manufacturing for Novartis worldwide, there are so many unknown at this stage that as we learn more, we are upgrading our numbers. As you know, last year, early in the process, we said we feel comfortable we'll get at least 500 million doses of the 1 billion of infrastructure capacity. Then we raised it to 600. With what we have seen out of the U.S. supply chain, the - as you know, Europe is around 3 months behind the U.S. because of when we started building it. We did not have a Norwood-like site in Europe, as you know. We feel good about what's happening in the U.S. The team are doing really a remarkable job. And so we feel comfortable today to up what we call our base case. We feel very comfortable that we will deliver 700 million doses. The team is working extremely hard to get to 1 billion doses. They know that every extra dose we can get out of Moderna supply chain will be used and will help protect people. So you have our full commitment to do everything we can to get as close as we can to 1 billion. But at this stage, what we're saying is we feel very comfortable we can deliver 700 million, base plan. And we are still working through the upside, and I will not bet against the Moderna team to be able to do better. But at this stage, 700 million is the base.
Operator:
Your next question comes from the line of Ted Tenthoff with Piper Sandler.
Edward Tenthoff:
And just incredible progress over the course of the year. And David, thanks for all the clarity on the quarterly update. I'm wondering, with respect to the $200 million that was booked in the fourth quarter, what is the number of vaccines that is ascribed to that? And then if I may, will we be getting any data from the triplet oncology IO program this year?
David Meline:
Yes. So the $200 million of revenue was associated with the deliveries that we made to the government in the fourth quarter in the U.S., which was around $17 million.
Tal Zaks:
And Ted, this is Tal. As it relates to the triplet, I hope so. Data in oncology is a function of when we see responses. And so once we see them and we confirm them, then, of course, that becomes material information that we share. So it's hard to predict, but I would focus on it.
Edward Tenthoff:
Brilliant. And Tal, wishing you all the best. Thanks for all the hard work.
Operator:
Our next question comes from the line of Salveen Richter with Goldman Sachs.
Salveen Richter:
And Tal, I want to second that. Good luck with the path ahead, and you will be missed here. So with regard to variance, which of the 3 approaches for COVID on the four do you think will be optimal here? And do you have updated thoughts on the correlate of protection or trial design? And then separately, should we expect your pricing strategy for 2022 to be in line with 2021? You did mention options. So I'm just curious how we should think about that.
Stephen Hoge:
Thank you, Salveen. I'll try and take the first part of that for those questions. So as far as what's optimal, I think we have to run the clinical experiment to know, and that's why you see us taking the 3 approaches. It is entirely possible, maybe even desirable, that 1273 as a third dose is able to boost immunity above a level that would be necessary to provide long-term protection against the new strains. But if you look forward and say at some point in the future, we're going to be perhaps in a regular boosting environment, what would be the ideal vaccine, it seems logical that a vaccine that provides the broadest immunity, so not just against ancestral strains or a boost against the new strains would perhaps be the best mode. And that's where I think the blended approach, so 1273.211 as we announced today might ultimately be the right long-term product. But in the near term, we got to run the clinical experiments to get the answer.
Stéphane Bancel:
Thanks, Stephen. And on the pricing question, Salveen, at this stage, we have not disclosed any information on pricing. The piece that I can share as just color is, as you know, we have a high efficacy vaccine. Governments understand the ability that we have to move fast on variants. And of course, they care deeply about that ability versus other technologies and other companies. And so in due course, as the 2021 year progress, we'll be sharing more color on the pricing strategy for 2022 when for both of the prime series as well as the variant. It might be the same, it might not be the same. So we'll discuss that in due course. Thank you.
Tal Zaks:
And Salveen, this is Tal. Thanks for your kind note. And I wanted to answer your question about the correlate. I think that work continues. It's being primary led by NIH who have access to all our samples as well as the other BARDA funded trials. The current assumption is that we should be able to move even without a correlate, and that is consistent with the recent FDA guidance on the development of a variant vaccine. I think if there does emerge a correlative protection, it will make everybody's life easier, especially those coming up in our footsteps in terms of licensing new vaccines. But I think also, for us, the ability to peg the dose level will be substantially made easier with that. So we continue to work towards that goal. We'll see in the coming months whether when one emerges.
Operator:
Your next question comes from the line of Michael Yee with Jefferies.
Michael Yee:
Again, congrats on all the progress. Two questions. One is a variant timing, manufacturing question and one is a competitor question. I guess based on what you described today, can you just kind of walk through the timing of how to develop and what would be needed to get a variant vaccine approved and when you would be able to flip the switch to start making that? That's just question 1 for variant. And then related to that, maybe for Tal, before we let you go, maybe you could comment about how you think about variant vaccines for mRNA versus adeno, particularly with multiple streams in there? How do you think about comparing and contrasting?
Stephen Hoge:
So maybe I'll take the first part of it and then hand over to Tal for the second. I think the - so first, on how quickly we think this goes. We've already, I think, demonstrated that we can pretty quickly produce a new batch. And if you look back to when we announced that we started manufacturing, it might be even faster than last year. We do think the clinical program for testing this is pretty straightforward based on the recent FDA guidance and other public comments. It's likely to be in the hundreds of people and immunogenicity and safety as important endpoints there. But ultimately, that will be subject to discussions with the FDA. And so we think we could get to clinical data here relatively quickly. That will be able to demonstrate the potential for a booster vaccine to close a potential gap in immunity. As far as production, that will depend a lot on the demand picture and possibly that data. But as Stéphane noted, our manufacturing systems, really, we believe we could almost copy and paste the new information into those manufacturing systems and proceed very quickly to be updating our vaccine or produce a booster. Tal, do you want to take the second part of that?
Tal Zaks:
Yes. That's a softball. Look, the mRNA, the beauty of the mRNA technology is the fact that the immune system doesn't recognize the LMPs per se. It only recognizes the protein that we teach the body to make. And so I believe that our vaccine platform is optimally suited to boost, irrespective of what primary series somebody got, whether it was a protein mRNA or, frankly, an adeno vector. I think the challenge with the adeno vector is for both the initial boost and certainly any work with variants down the road is going to be that the adeno vectors generate significant immunity against the other components of the adenovirus and thus, have the risk of limiting the ability to translate the transgene. And therefore, if you look at the boosting delta, that is how much additional antibody levels do you get from a boost, with adeno vectors, you can get a boost, but the magnitude of that boost is invariably less than the magnitude of the boost you get with an mRNA platform. And that is true just in the first boost. I suspect that if you needed to come with a third dose or a fourth dose or variant-specific vaccines, it would be challenging to get the requisite amount of specific immune recognition towards the new transgene.
Operator:
Your next question comes from the line of Gena Wang with Barclays.
Huidong Wang:
Tal, good luck with your next journey. It's a big shoe to fill, and you will certainly be missed here. I have two questions regarding the variance and also next-generation COVID vaccine 1283. So for 1283, is it shorter sequence that make freezer storage feasible? And also with the sequence outside of RM sequence - RBM multi sequence? And then the second part of the question is the COVE II variant for the NIH study. I wondering if you can give a little bit more color regarding type of subjects you will enroll, whether it's a vaccine-naive or experienced subjects and also the booster timing.
Stephen Hoge:
Thank you for the questions, Gena. I'll take the first part of that and then hand it over to Tal to answer the NIH component. So in terms of the mRNA-1283, it is correct. It is - the fact that it is a shorter mRNA does help with the long-term stability and does facilitate, we think, moving that into a refrigerated vaccine. It's not the only feature. As you'll know, we've had advantages on our mRNA platform based on our long history of working in it, for instance, that we're functioning right now already, as has been noted, in a normal freezer for up to 6 months and actually already doing 30 days with mRNA-1273 in a refrigerator post stalling. And so it's a benefit that we're already well on that path, even with just 1273. I think the second part of your question was, is it just the RBM, and 1283 covers both the RBD and the end terminal domain. And as has been widely reported, those are particularly important for neutralizing activity in the immune system against SARS-CoV-2 virus.
Tal Zaks:
Thanks, Stephen. Gena, thank you for your kind comments. The NIH, I'll defer to my colleagues there to describe their trial in the coming weeks once they launch it. But in essence, the answer is both. They will have arms there that will be testing the new variant vaccine as a boost in those who have been previously immunized on their Phase I trial. And then there will be a component that will test the new vaccine in a primary series.
Operator:
Your next question comes from the line of Cory Kasimov with JPMorgan.
Cory Kasimov:
And all the best to Tal, although it's good to know we'll have you for a couple more quarters. So two questions for me as well. Look, I realize there's not much you can say on pricing beyond 2021. But as we think about your booster strategies that could be less frequent at lower doses as well as the greater supply of vaccines we should have on the market, balanced against the potential waning of the pandemic, are there scenarios where the price per dose could be either materially higher or lower than what we're looking at in 2021? And then my second question is from a modeling standpoint. Should we be assuming anything in the model above and beyond what was talked about today that could be owed to the U.S. government for the initial co-development of your COVID vaccine? Or is that what David was referring to with the royalties that are included as part of your cost of sales? And if so, should we just be assuming a generally stable rate going forward? Are those changed at all?
Stéphane Bancel:
So there's a lot to unpack here. Let me start maybe on pricing. So it's quite interesting because, as you know, this is public information, there are some vaccines that have been made available to governments at $3 per dose. And as you know, we walked you through our pricing strategy back, I think, in August, on the Q2 call. The Moderna vaccine is priced much higher. And I think it comes down to a few things. And the most important one is efficacy. Governments care about saving lives. Governments care about getting the economies back on their feet. And so what we're hearing from governments, literally almost on a daily basis as we engage with our existing government contracts versus new ones that are calling us, is there starts to be very clear differentiations between the different products that are either available already or that are getting very close to authorization. And I wish, if I had a crystal ball last year, that we would have built more manufacturing capacity for '21. But I did not anticipate and I don't think anybody did that there will be such difference of efficacy between vaccine, that the mRNA vaccine will be such high performance. As you know, some countries decided to invest first to procure vaccine in adeno or protein technology because they were better understood. And as we know what happened in 2020 in terms of the clinical data, that has played a big role. So it's quite interesting that - also seeing how people are reacting in the countries. If you follow the media, for example, in Europe, where several vaccines have been authorized. As you know, in U.S., we have as of today, I'm hopeful this will change soon with VRBPAC meeting tomorrow, but only to mRNA vaccine authorized in the U.S. But if you look at what's happening in some countries in Europe, also what's happening in different countries around the world, I think governments, because of essence, advisers and clinical advisers and because of really the aim to reduce the pressure on the health care system, in the hospitals. Also what is quite fascinating, and I'm sure you're all observing it, is that, at least the first time I've seen in my career, being 20 years in this business, that in the media every day, you hear about vaccines on TV, in the newspaper, online. And you have the product efficacy that are all over the news every day. This has never happened to my knowledge for any of our products. And so you have this very interesting phenomenon that's happening too, that you have the clinicians, the nurses, the pharmacists, but the consumers too, that are believing that those products are not all the same. And this, I think will be a very important differentiation as we move into '22, as we move more to a traditional commercial market, where it's not governments buying directly, but the traditional kind of retail channel. And the impact, I think, of the consumers' desire to ask for a product when he or she walks into a pharmacy or their GP's office will be quite significantly different from what we are used to with other products.
David Meline:
Okay. Maybe then I'd comment on the question about cost of sales. So Cory, what I did try to say early on was that we don't have precision in all cases. In the case of cost of sales, I would say there are puts and takes that we're monitoring, but we gave you the 20%, because we feel confident that, that's the right range, right zone that we're in. And yes, as we move through time, there's going to be more information as we ramp up our factory capabilities and answer some of the other questions. But I think you're good for now with that modeling assumption.
Operator:
Your next question comes from the line of Geoff Meacham with Bank of America.
Geoffrey Meacham:
Also I wanted to offer up best wishes to Tal. A question on manufacturing investments, I guess, for Stéphane. So I guess I'll focus on variance with the booster. And clearly, you guys have to fund many assets progressing in the pipeline. But how flexible are your manufacturing investments? I guess the question is, as the cases globally continue to decline, is there a way to scale back some of that capacity should lower volumes become the norm?
Stéphane Bancel:
Those are great questions. So in terms of the flexibility, as I described in my remarks, the only raw material that is different from let's say mRNA-1273 to mRNA-1273.351 is only the plasmid. The enzymes are the same, the lipid is the same, with the same equipment in the same rooms with the same people. So the flexibility of this technology is really incredible for people that have worked with recombinant, like I used to do at Lilly. And as you know, I trained as a biochemical engineer, doing neutrophil and E. coli production in bioreactors. It is literally, you come a week after we use disposable reactors, you get new consumable plastic bags and so on as your reactor. And here you go again, you just need to change - take a different plasmid. Because we use disposable reactors and not stainless steel reactors you don't have to spend a lot of time doing cleaning, validation because we basically dispose of things. And so the flexibility is quite incredible. You just need a new plasmid. And so because - so it's not a very expensive part of the cost of the total cost of the finished product that is filled in a vial, because you have to make the mRNA, you have to formulate it and you have to fill it in a vial, our doing a lot of plasmid at risk for variant is a great optionality for the company with very, very good payment. So that's a piece that I think makes this technology very unique and ready to scale and be able to be flexible to respond to different demand. Is there a world where we potentially might end up with a different combination of mutant boost on the road in different continents? Maybe. I don't know, again, it will be market-driven. But if it is what the market requires, we can accommodate that in a way that is totally atypical to what the industry could do and at the speed, which is, I think will surprise many. Because again, we make these other products in the same reactors. In terms of taking down the manufacturing capacity if we don't need, let's say, in '23 or '24, the same amount of volume because of a mass of boosting, as we say, it's actually interesting because, as you know, we have now good plants where we have quite a lot of capacity. But also, we have some capacity at Lonza, in New Hampshire and also capacity in Switzerland, where they have there several lines of manufacturing. So we have actually quite a lot of flexibility to take capacity down at Lonza, either in the U.S. and/or in Europe at different time points. So that's one point. The other piece we should not forget is the pipeline. And I will just make 2 comments, which I think are very important to understand why actually us building manufacturing to that scale is going to be so enabling to Moderna to keep growing and growing and growing. First is the other vaccines. CMV is ready to move into Phase III, as we said this year. We will give you updates on the flu program, but I will remind everybody that there is an approvable endpoint with the regulator for seasonal flu vaccines. And so we anticipate that the development of the flu program will be pretty quick. And as we discussed on previous calls, one of our kind of optimal target product profile for down the road will be a product that is both seasonal flu, with the current strain of flu, but also a boost for COVID-19 with the relevant strain at that time in the same dose. So we anticipate we should have high efficacy for flu as much as mRNA virus. It's a pretty straightforward virus like the SARS-CoV-2 virus. And so that is where we are going. We have more vaccines coming to the market. And then there's also our therapeutic pipeline. And the piece, I think, that is actually quite good for us that we started with vaccine, the scale-up, is I go back to mass, which is if you think about some of our products, as you know 1273 requires 100-microgram per human per dose. But if you look at the chikungunya antibody program, that use the IV-formulated mRNA that we use also in the rare disease program, for example, what we showed is very good data at 0.1 milligram per kilo. So in a 70-kilo adult, it is 7 milligrams compared to 100 micrograms. So as we move the therapeutic pipeline further into development, but especially to commercial, we're going to need much more mass of mRNA. And so I actually - I'm spending a lot of time thinking, one, how do we keep adding capacity to manage the entire pipeline as it's progressing. We have the flexibility with Lonza, that's why we did it this way with Juan, so we are ready to take it - take chunks down if we need to. But if I was a betting man, I would bet with you that we're going to be more adding than taking capacity down because of the probability of technical success. As I've said all along, I believe we will launch the CMV vaccine, given the data we have, the medical need and the biology we're able to do with both the pentamer and GB. And so I'm more spending time wanting to think about how do we do more.
Operator:
Your next question comes from the line of Hartaj Singh with Oppenheimer & Co.
Hartaj Singh:
Great. And Tal, I wish you the very best in your future endeavors. The question I have is, we're hearing more and more about some constraints in the raw materials that go into making vaccines, plasmids, things as simple as pipettes, et cetera. Can you just give us some color and visibility on where Moderna is with its partners, Lonza and others, in this regard to 2021 and 2022 supply? And then could that be a rate-limiting step as you think about other modalities?
Stéphane Bancel:
Thanks. So as we've said all along, which is why we gave a range and not one number for expected supply in the year, I think one number is a very dangerous strategy. As we said, those type of consumables are, of course, important. We are in a regulated business, and we cannot start to make a product until we have all the components, all the raw material, the entire train team and so on and so forth. So we have a lot of - like in an airplane, you have to check a lot of things before you can start the engine. Well, in GMP manufacturing, you have to do the same to ensure the quality and the safety of the product. And so, well, I think the team has done a very good job last year. And as you recall, we started racing against this virus in very early January. And it's in the late January time frame that we started to say, geez, this could turn into a pandemic. And we started to think bigger. And so, one, the team and I spent quite a lot of time as we were starting to think about, okay, how do we go - if we had to make 1 billion doses in '21, how would we do that? I used to run the supply chain at Lilly globally. And we went right away to start to talk to our suppliers. And one other thing we had to do, and if you recall, we did an important capital raise in May of last year, which was very important for getting us to where we are today, in some suppliers. Because the increase of mass of product we're ordering from them was so gigantic, a thousand times more, even 2,000 times more than the year before. As you can imagine, many of them looked at us a bit funny and were very worried. They were worried that if the drug would fail, they might be in trouble, because they will have to get this type of increase of capacity to sometime, buy new equipment, hire more people, buy their own raw materials to make our raw materials for our product, if that makes sense. And so sometimes what we have to do with the team is actually to pay upfront the entire purchase of product so that they will go and take that cash and start to invest to prepare for scale up. So we took a lot of business risk last year that we thought were necessary to ensure we could deliver this year on the output from the manufacturing supply chain. So I think we're in a good place. Do we have from time to time, one component that give us a sleepless night because it's very tight? It's happening because the ramp-up the industry is doing is unprecedented. But so far, so good, and we are extremely on top of it, including as we do things like increasing capacity that we announced last night to right away, not only buy new machine, but right away, place new orders for raw materials and make sure we keep increasing our safety stock so that we run this more and more as a regular business where you have safety stock on site. So if your supplier has a bump in a road, a pump breaking or something else, that we are able to manage that variability, which is normally manufacturing without impacting our ability to maximize our own output.
Operator:
Our next question comes from the line of Simon Baker with Redburn.
Simon Baker:
Two, if I may, please. Firstly, just going back to cost of goods sold. You've helpfully given us the 20% figure for 2021. But presumably, there will be some degree of variation through quarters. So would it be fair to assume that you will be exiting the year at a figure lower than 20%? And I just wonder if you could give us some color on if that would be meaningfully below 20%. And then moving away from COVID to CMV. You've said that the peak sales potential for the CMV vaccine could be around $2 billion to $5 billion per year. It would seem that HPV is a pretty good road map for the potential in that space and set against that, $2 billion to $5 billion looks quite conservative. So I was just wondering if I'm missing something in that analogy or whether that $2 billion to $5 billion is out of an abundance of caution?
David Meline:
Yes. Maybe I'll take the first one. On the cost of goods, percent of sales, our advice right now is to assume that's pretty stable through the year, including as we exit the year. So I would recommend you model it consistently. There will be some ups and downs. And the ups and downs are more related to the mix of business and the price levels of the product that necessarily changes in the cost of the produced product. And hence, given that, we get out towards the year-end, we don't have complete visibility on precisely what the book of business and mix will look like of sales, hence, some uncertainty around the precise percentage number. But again, I would advise just treating it as a constant for now. And then as we get better information, we'll give that to you as we move through the year.
Stéphane Bancel:
Thank you, David. So let me take the CMV question. So a few bit of colors on the $2 billion to $5 billion annual peak sale estimate that we talked about now a while ago. We talked about that as a company that never had run a Phase III at the time, that had never launched a product or get a product approved at the time, that had 0 commercial employee at the time. So we did the analysis with an outside commercial consulting company and with our strategy team and the team here that many of us have commercial experience. So we tend to be cautious, because as a new company we want to make sure that we build credibility and credibility in life has to be earned. And so I will just make the analogy to manufacturing in the sense, we built 1 billion of capacity, and we are very upfront to the financial community about it last year when we did so. But we said, look, given the unknown on raw material supply, given the unknown on yield, we feel comfortable we should get at least 500 million doses out. And then as we learned more, we moved to 600. And as we learned more, we moved it yesterday to 700 million. So do I believe it is possible as we're going to sell more than 5 billion of CMV? Yes, I believe it's possible because it's an incredible unmet medical need. As the world develops, I think the piece one has to be careful about peak sales is peak sales when peak sales slightly up [indiscernible] peak sales 25 years after launch because those vaccines are like annuity-like products. There's, of course, inflation. There's, of course, developing countries that are becoming richer and richer. So is there upside to the number? I believe there's upside to the number. But that's our number right now as a company.
Operator:
Our next question comes from the line of Mani Foroohar with SVB Leerink.
Mani Foroohar:
Tal, looking forward to chatting with you later on today in our fireside chat, and excited to hear at some point what your next adventure is going to be. A quick and sort of boring financial question. As we - as you guys are pretty clearly going to transition to profitability, cash flow positivity on a - at least in the near term, very durable basis, when would you consider changing your current treatment of your NOL allowances? How would that - how would you communicate that? How would that flow through how you communicate your financials from a modeling perspective? And then secondarily, and maybe scientifically, more interestingly, how do you interpret the data suggesting that at least for the adenoviral approach from J&J, but also from your own data and the other mRNA vaccine, that titers appear to improve over time and cellular immunity improves over time and the gap in efficacy against the so-called South African variance and the ancestral Wuhan variant seems to close. That would suggest to me, the 1273 should have similar efficacy versus the South African and the Wuhan variance. Am I interpreting it the same way you are? Or am I missing something?
David Meline:
Yes. So maybe I'll cover the first one. On the tax rate, as I said, what happens is we're carrying this net operating loss with a full valuation reserve right now against the deferred tax asset that comes with it. So that will, of course, get released as we start generating pretax income. And it's likely based on the current book of business that we would consume during the year the entire loss carryforward. So that's point one. Point two is from an accounting perspective, what happens is you basically project the full year tax rate, including the consumption of that deferred tax asset. And then you start accounting quarter-by-quarter based on that average of the full year rate. So it actually, from an accounting perspective, doesn't matter, the timing of the release of the NOL, the deferred tax asset and the valuation reserve. So that's why I tried to be clear with the mid-teen percentage, including in Q1, because from an accounting perspective, it will - it would be stable unless we have some other changes that caused the full year rate to change as we move forward. Hopefully, that's clear.
Mani Foroohar:
That's clear. I misstated my question. I actually meant versus severe disease mortality in terms of the similarity and efficacy alone. I thought of a longer-term follow-up.
Stephen Hoge:
Sure. Yes. So thank you for that question. So let me take a stab at it. I think we're - as you noted, I mean, we had nearly or essentially 100% of efficacy against severe disease, 94% against moderate or any disease. And I think that gives us great optimism about the current 1273 vaccine that's been authorized is protecting and even if the new strains are slightly more virulent, that we'll be in good shape. I think the question, though, is at some point, you would expect coronavirus immunity to wane. And that's because in all of the human circulating coronaviruses, that's what happens. The durability here has always been a question. Is it 1 year? Is it 3 years? Is it 5 years? I think as you see new strains emerge, like the potentially concerning strains that we've been talking about, particularly in the Republic of South Africa and Brazil, you have to ask the question, is that - what does that look like not just now, but what does that look like 6 months from now, 1 year from now, as we start to move into a mode where we're going to see seasonal epidemics of coronaviruses, which we already see for the other coronaviruses, and you would have to assume will happen with SARS-CoV-2. And it's for that - it's those gaps in immunity that I think we're particularly focused on right now, which is, to what extent do you - to what extent in the future do at-risk populations emerge with increased risk against these new strains? It's not something you can see in the data in the first 30, 60, 90 days post vaccination in the current clinical trials. But it's something that you - we did talk a lot about last year, and I think it's going to continue to be something we have to track closely as we look ahead.
Mani Foroohar:
That's helpful. I guess that would imply also that it might be useful to look at sero collected from the already vaccinated patients, 180, et cetera, further days out and perhaps perform the same experiment described in the New England Journal of Medicine article. Would you consider disclosing that data? Or do you think it just makes more sense to just move to your novel variant approaches? And just would the clinical data speak for itself?
Stephen Hoge:
So fair question. I mean, look, I think we will obviously update data on durability of responses across our Phase I, Phase II, Phase III trials as that comes out. And looking forward, it's prudent. And if we can, we will be providing updates as well against the new strains. I think what we're saying about the variants and strategies against it is - I think none of us want to be in a situation where we wait until there are reinfections and increased morbidity and, God forbid, more mortality in a seasonal epidemic, for instance, this winter in the Northern Hemisphere, this coming winter. And then say, okay, now it's time to be boosting people. And so as we're continuing to face an evolving virus that is changing its stripes and perhaps decreasing the durability of our immunity, we're trying to be very proactive with the 3 strategies we described. And I think we'll continue to follow things like the data that you pointed to, which is how are we doing in terms of immunity and blood of people who've previously been vaccinated or previously infected. We'll obviously closely be following reinfections. And then I think we all have to be following very closely what's happening in the southern hemisphere, because as they move into their winter and a potential seasonal spike in the winter as you normally see in temperate climates, it will help us understand to what extent are we still in a - we have to stay ahead in the concerning phase of this epidemic or pandemic or is it a place where we start to get more and more confident that the vaccines have the virus under control.
Operator:
At this time, I'll turn the call back over to Stéphane Bancel for closing remarks.
Stéphane Bancel:
Well, thank you very much, everybody, for joining us. We look forward to seeing you at our next event on April 14 for Vaccine Day, and I wish everybody to stay safe. Have a great day. Bye.
Operator:
Thank you, ladies and gentlemen, that concludes today's conference call. You may now disconnect.
Operator:
Good morning. And welcome to Moderna’s Conference Call. At this time, all participants are in a listen-only mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I’d now turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you. Good morning, everyone, and welcome to the Moderna’s Third Quarter 2020 Conference Call to discuss Financial Results and Business Update. You should access the press release issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our website. On today’s call are Stéphane Bancel, our Chief Executive Officer; Tal Zaks, our Chief Medical Officer; Stephen Hoge, our President; and David Meline, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to safe harbor provision of the Private Securities Litigation Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to revise the information or provide - provided on this call as a result of new information or future results or developments. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. I hope all of you and your loved ones are in good health and remain safe. Thank you for joining our Q3 business update conference call. I will start with reviewing key highlights. I would like to focus on three topics. First, our COVID-19 vaccine. Second, our product development pipeline. And finally, key financials. As many of you know, we completed enrollment of our Phase 3 COVE study for mRNA-1273. 30,000 participants have been enrolled in the study, 37% of whom are from diverse communities. Tal will give you some details in a few minutes. We are grateful for participants in the study and for our principal investigators. I would like to also thank PPD and NIH for great job and collaboration. It is very important to Moderna team that we have a high bar for quality and transparency to ensure the public has trust in COVID-19 vaccine. We have reported weekly enrollment progress of the COVE study and have reported weekly enrollment numbers from diverse communities. While we are not happy with the representation of these diverse communities, given the high burden of disease in these populations, we decided to slow down the overall COVE study enrollment process in order to recruit more people from diverse communities. As you can imagine, it was not an easy decision to slow down, but it was the right decision. We signed the biopharma pledge to not submit for regulatory approval for mRNA-1273 until we have adequate safety and efficacy data. We were also the first company to file the full and redacted version of our Phase 3 protocol online to ensure clinicians around the world could see in full transparency how the COVE study is being run. We were pleased to set the standard and have others in the industry follow early. We continue to expect our first interim analysis to readout in November. The independent Data Safety Monitoring Board, or DSMB will carry out this interim analysis and then inform Moderna. We should have a post-second dose two month safety follow-up after the second vaccination of a 15,000 median participants in the second half of November. As many of you know, this is what the U.S. FDA has asked as part of the EUA submission in their latest guideline published ahead of October 22 VRBPAC meeting. We are working closely with the U.S. FDA to file Chemistry, Manufacturing, and Control or CMC components as soon as they are available to ensure CMC is not on the critical path to an EUA approval. In addition to the ongoing dialog with U.S. FDA, we have announced rolling submissions for mRNA-1273 in the U.K. and Canada and received confirmation of eligibility for submission of Marketing Authorization Application to the EMA for the EU. We are very pleased to announce this morning that we have signed a partnership in Japan with Takeda for an order of 50 million doses for the Ministry of Health, Labor & Welfare of Japan. In the third quarter, we have received $1.1 billion of cash payment from governments around the world, and those are accounted in our financials as deferred revenues. Let me now turn to our broader pipeline. We announced at the R&D Day in September the positive Phase 2 readouts for mRNA-1647, our CMV vaccine. We are on track to start the pivotal Phase 3 registration study for CMV in 2021. I would refer you to our April Vaccine Day presentation, in which we've communicated that we believe our CMV vaccine has a potential for annual peak sales between $2 billion and $5 billion. There is no approved vaccine against CMV, and it is the number one cause of birth defect in the U.S. and in the developed world. Moderna owns global rights to CMV 1647. Our intratumoral OX40 ligand program that many of you have heard about at our recent R&D Day, is now doing patients in the Phase 2 expansion study. In rare disease, start-up activities under an amended protocol for Phase 1 propionic acidemia or PA study are underway. All of our pipeline programs that were enrolling and dosing patients also continue and are ongoing. Let me finish this slide with a few words about the financials. David will go through the financial numbers in a few minutes, but I wanted to share a few thoughts. As I mentioned earlier, we reported this morning that we booked $1.1 billion of deferred revenues in Q3 for supply agreements for mRNA-1273. This cash has been received by the company. These cash receipts have enabled the company to generate $893 million of cash flow provided by operating activities in the third quarter after investments of approximately $300 million in the business. If you turn to Slide 4, I am pleased to report that this is the first quarter in the company's history in which we reported positive cash flow provided by operating activities coming from product supply agreement. It has been 10 years after inception and after investing billions of dollars in science and product development. Thank you to all our employees, scientists, of course to our investors and to all of you that have believed in Moderna. Let me turn to Slide 5 for my closing remarks. As a snapshot of Moderna, in October 2020, we have accomplished a lot and continue to have a broad pipeline that continues to progress well. We have fully enrolled Phase 3 program with mRNA-1273. We now have four Phase 2 trials with CMV, personalized cancer vaccine, VEGF with AstraZeneca and now OX40 ligand, seven ongoing Phase 1 programs and 12 positive Phase 1 studies. Our vaccine franchise has six programs in development, addressing the major unmet needs. We have five immuno-oncology programs in the clinic, four programs in rare disease and two programs in autoimmune disease. Our foundations have never been stronger with 32,000 participants and patients in our trials. We have now over 1,200 employees. We have an international manufacturing capacity and capabilities with our partner Lonza, ROVI, and Catalent, and we have strategic partnerships with companies like Merck, AstraZeneca and Vertex. At the end of September, we have a strong balance sheet of $4 billion. I'm very proud of what we have accomplished so far and where we stand as a company. The next few weeks and months are going to be quite historic for Moderna. I will now turn it over to Tal to talk about clinical updates. Tal?
Tal Zaks:
Thanks you, Stéphane. And good morning, everybody. Let me give you a quick overview of our pipeline progress and starting with our COVID vaccine. The Phase 2 study looking at safety and immunogenicity in 600 participants is ongoing. Dosing has been completed, but we remain blinded while the final immunological testing is being conducted. Once we have the results, we will share that. As a reminder, the safety data from this trial have of course been shared with regulators prior to the start of our Phase 3 COVE study. That is now fully enrolled, and I will share some details shortly. For our CMV vaccine, mRNA-1647, we shared positive Phase 2 data at our annual R&D Day in September and the data enabled us to select the 100 microgram dose to take forward in our pivotal Phase 3 trial, which is expected to begin in 2021. For our Zika vaccine, we showed the positive Phase 1 results and are preparing for Phase 2. On the pediatric front, I'm happy to announce that our hMPV/PIV3 Phase 1b age de-escalation study has resumed dosing toddlers aged 12 to 36 months following a pause that was related to COVID-19 disruptions. This is the first mRNA vaccine to be given to toddlers. In addition, the first cohorts of adults participants in the Phase 1 study of our RSV vaccine, mRNA-1345, has now been fully enrolled. As a reminder, this is also an age de-escalation study similar to the hMPV/PIV3 study and the plan is ultimately to also those toddlers. In another core modality, the systemic secreted and cell surface therapeutics, we showed the positive Phase 1 data from additional cohorts of the Chikungunya antibody program during our annual R&D Day in September. Importantly, we demonstrated not only potentially therapeutic levels of a secreted systemic protein, but that a two dose regimen with a week between doses was safe and well tolerated and led to the predicted increase in Chikungunya antibody productions. This demonstrates our platform's ability for repeat dosing. I'd further note that as a result of rapid systemic clearance, no significant accumulation of our lipid nanoparticle was seen after administration of the second weekly dose. Moving to Slide 8 and the updates from our pipeline of candidates across the four exploratory modalities, and starting with our personalized cancer vaccine, the PCV. The randomized head-to-head trial of PCV with KEYTRUDA -- versus KEYTRUDA alone in the adjuvant melanoma setting is ongoing. A reminder that our Phase 1 study continues with patients in the various tumor types who were treated in the monotherapy Cohort A and in combination Cohort B are in follow-up and the Phase 1 expansion cohorts of Cohort C is enrolling patients. Cohort D in the adjuvant melanoma is also ongoing. For the intratumoral immuno-oncology, I'm happy to report that we have dosed several patients in the ovarian Phase 2 expansion study. Recall that we initiated this part of the study just last month, and I believe the team is focused on moving this trial forward along with the support of our clinical investigators has been very productive. Our Triplet program in this modality also continues to enroll and dose patients in the Phase 1. In our systemic intracellular therapeutics modality study, start-up activities have resumed for the PA program following disruptions due to COVID-19. In addition, Nature published a preclinical study on this program. For the MMA program, recall that we announced at R&D Day that we would take forward our next generation development candidate, mRNA-3705, which recently received a rare pediatric disease designation from FDA. And finally, regarding our partner-led programs, the Phase 1 KRAS with Merck and the Phase 1 Interleukin-12 and Phase 2a VEGF studies with AstraZeneca are all are ongoing. So Slide 9 is a snapshot of our development pipeline. And please note that eight preclinical programs across three different modalities that we didn't touch upon during the clinical review, bringing the total number of programs in development currently to 21. So let me move into more detail to mRNA-1273, our vaccine against COVID-19. Slide 10 is a broad overview of where we are. mRNA-1273, led us to a robust immune response across species and can protect human and non-human primates from the virus taking hold in both the nose and the lungs. In the clinic of the 100 microgram dose we observed consistently high levels of neutralizing antibody titers across all adult age groups and these titers were higher than those seen on average in convalescent sera. And the Phase 3 COVE trial has completed enrollment, meeting our expectations, and I'll give you more details on the demographics in a moment. Let me briefly review the Phase 1 results that were published in The New England Journal of Medicine. mRNA-1273 has been generally safe and well tolerated as the Phase 1 safety data at 100 microgram across the adult age cohorts demonstrate. As it relates to tolerability, the most common solicited adverse events were headache, fatigue, myalgia, chills and injection site pain, the majority of which were mild-to-moderate in severity and self limited. Of note, local and systemic reactogenicity were more common and more frequently moderate in severity after the second dose. One severe solicited systemic adverse event occurred after the second dose and that was fatigue in the above age 71 age cohort who received the 100 microgram dose. What is important is that these flu-like symptoms are expected. They're transient and generally mild-to-moderate in nature. And I believe they correlate with the underlying potency to stimulate an immune response in high levels of neutralizing antibodies. Importantly, there were no vaccine-related serious adverse events in this trial and no patterns of concern for any clinical labs. And while these numbers are still small, we did not see a difference in safety or reactogenicity profile between younger and older adults. As it relates to the immunogenicity, I would make three points regarding the data. First, we see the same level of neutralizing antibodies in younger and older adults. Second, we see these levels consistently in everybody who receive the vaccine. And finally, these levels are higher than those seen on average in the blood of people who had been ill with COVID-19 and whom we expect by and large to be immune to the second infection. Now if you follow the timeline, you will see that the high levels of antibodies are achieved quickly upon boost. And I believe the speed and quality of this immune response speaks to the T cell supports and puts us in a good place to have durable protection. Slide 13 shows an overview of our Phase 3 COVE study, which is now fully enrolled with 30,000 participants. The full protocol can be found on our website. And noteworthy points from this slide include the one-to-one randomization between 100 microgram of mRNA in the vaccine arm and placebo. Every participant in the study was expected to be at higher risk than average of infection, that was inclusion criteria number one. And a significant proportion of subjects were stratified as being risk -- at-risk of worse outcomes from COVID-19 should they get infected. These are people over 65 years old or those under 65 years old, but with chronic conditions that are risk factors for disease. So how did we do in terms of demographics? We're very proud of the hard work of our clinical team, our collaborators at NIAID and our clinical trial sites that led to the successful recruitment of a diverse and representative study population, which is similar to the census of our country, with 37% of study participants coming from diverse communities, and you can see the breakdown on the left side of this slide. We've enrolled 6,000 Hispanic or Latinx participants and 3,000 black or African American participants. Age distribution is shown in the middle. And of note, about two-thirds of the trial participants are older than 45. And the gender distribution was close to evenly split, as is seen on the right. So what about risk factors for severe COVID-19 disease? The greatest risk factor is age, and a quarter of participants were over the age of 65. In addition, 17% were younger, but still at risk of severe disease by virtue of co-morbid conditions, such that 42% of trial participants are in the high risk strata of having worst outcomes should they get infected. There is another way of looking at it, which looks at the breakdown of the chronic conditions that put people at risk, shown on the right. These include diabetes at 36%, severe obesity at 25%, significant cardiac disease at 19% and 18% with chronic lung disease. I would note that all in all, over 8,000 of the participants in our study are living with these chronic conditions. Now if you do the math, you'll realize that a significant proportion of participants in the COVE study had the independent risk factors of both an older age and co-morbid conditions. So Slide 16 sums it up. Our Phase 3 COVE study is representative of the many diverse populations that make up our nation and by extension many parts of the world, and many people could identify with our study and can find themselves in it. As we're anticipating the results of our COVID-19 vaccine study, let me take a few minutes to review the statistical analysis plan on what happens next. As many of you know, we have two interim analyses at 53 and 106 events and a final analysis triggered at 151 events. At the first interim analysis based on the statistical plan in order to cover the success, we will need to show vaccine efficacy of 74% or greater. From the graphs on the left, you can see that there is a 50% probability of meeting that hurdle, assuming a vaccine efficacy rate of 75%. So there is an element of chance here as well. At the second interim analysis, vaccine efficacy of 57% or greater is required to meet the statistical hurdle. And the probability of meeting this is actually 95% if the actual true vaccine efficacy is 75%. For the final analysis, as per FDA guidelines, at least a 50% efficacy is required. And again, the probability of meeting that primary endpoint in the final analysis is in the high-90s, assuming that our true vaccine efficacy rate is 75% or higher. But we also need to remember is what's not on this graph, all of these assumptions are driven by the imperative to ensure that we have a high degree of confidence. And I'm talking about statistical confidence that once one of these boundary conditions are crossed, not only do we have an initial point estimate about the vaccine efficacy, but we have a 95% confidence interval that the true efficacy, not the point estimate of the sample, exclude 30% or is higher than 30%, and I'm sure we'll be coming back to this crucial point in the future. So in each interim analysis there are three potential outcomes; either the study meets the statistical hurdle, which is 74% at the first interim and greater than 57% in the second, enable us to trigger the full analysis required to evaluate whether to proceed with the regulatory submission, and of course the trial remains blinded and data continue to accrue at this time. The study may not meet the statistical hurdle and then would continue to the next milestone or the study is determined to be futile. As we have committed to and have been transparent throughout the Phase 3 clinical program, today we released the informed consent form of our Phase 3 study on our website and we will continue to be transparent. We'll announce the results and next step once the first interim analysis has occurred. Now the pressure on the results are driven by the fact that from a distribution standpoint, we're ready. We expect that mRNA-1273 be distributed within existing infrastructure. There is nothing new required that hasn't already been used for years with many other vaccines. Specifically the advantages of mRNA-1273 that allows us to do this includes the ability to package and ship boxes in any configuration, housing small or large quantities of vaccine, storage conditions of minus 20 degree celsius for six months, refrigeration temperatures of two to eight for up to a week and room temperature's conditions for up to 12 hours after a fall. No special handling or dilution is required prior to vaccination with mRNA-1273. And by the end of this year, we expect to have approximately 20 million doses ready to ship in the U.S. With that, let me turn it over to David to take you through the financials.
David Meline:
Okay. Thank you, Tal. Turning to Slide 21 in the deck. We ended Q3 2020 with cash and investments of $3.97 billion compared to $3.07 billion at the end of Q2. The increase was primarily driven by $1.1 billion of customer deposits received in the third quarter for a potential supply of mRNA-1273. Net cash provided by operating activities was $763 million for the nine months ended September 2020 compared to net cash used of $360 million for the same period in 2019. The reversal from cash used to cash provided by operating activities is driven by total customer deposits for the nine months ended September 30 of $1.2 billion received for a potential supply of mRNA-1273. Cash used for purchases of property and equipment was $44 million for the nine months ended September 2020 compared to $25 million in 2019. Total revenue was $158 million for Q3 2020 compared to $17 million for the same period in 2019. Total revenue was $233 million for the nine months ended September 2020 compared to $46 million for the same period in 2019. Total revenue increased for both the three month and nine month periods in 2020, primarily due to increases in grant revenue from BARDA to accelerate development of mRNA-1273. Research and development expenses were $344 million for Q3 2020 compared to $120 million for the same period in 2019. Research and development expenses were $612 million for the nine months ended September 2020 compared to $378 million for the same period in 2019. The increases for both three month and nine month periods in 2020 were mainly due to increased mRNA-1273 clinical development activities and headcount and pre-launch inventory buildup. Overall, in both periods, we saw a significant increase in expenses for the prophylactic vaccines modality as a result of our focus on mRNA-1273. General and administrative expenses were $49 million for Q3 2020 compared to $28 million for the same period in 2019. Expenses were $109 million for the nine months ended September 2020 compared to $84 million for the same period in 2019. The increases for both periods were mainly driven by increases in personnel, outside services and set-up costs associated with preparation for commercialization of mRNA-1273 globally. Turning to selected cash flow information on Slide 22. On the top half of the page, we present information from our 10-K and 10-Q filings. And on the bottom part, we provide the quarterly trend and also items to take into consideration when assessing the evolution of this trend, in particular for Q2 in Q3 of this year. Cash provided by operating activities and for purchase of property and equipment was $719 million for the nine month period ended September 30 and $874 million in the third quarter alone. Excluding deposits received for potential supply of mRNA-1273 and the Vertex upfront payment, cash used in operating activities and for purchase of property and equipment was $296 million in the third quarter. This compares to $118 million in the second quarter of this year, excluding customer deposits received in Q2. The increased cash used of around $200 million in Q3 compared to previous quarters is consistent with our expectation as we are making substantial investments in manufacturing and potential global commercialization activities for mRNA-1273 COVID vaccine candidate. Let me now give you an overview about where we stand with regard to commercialization optimization activities. Please turn to Slide 23. On Slide 23, you see listed supply agreements that we have announced publicly today. As a reminder, these include the agreement with the U.S. government for 100 million doses in options for an additional 400 million doses. We just announced a deal with Japan for 50 million doses. Canada has confirmed 20 million doses with an option for an additional $36 million. We have also signed agreements with Switzerland, Israel and Qatar, and there are several other countries that have signed agreements that have not been publicly disclosed. We are thankful for the trust the governments around the world have placed in us to deliver a vaccine for their countries. All of these agreements contain provisions for deposits and have contributed to our 3Q '20 deferred revenue value of $1.2 billion, including $600 million from the U.S. government. We continue to work with the European Union where we are in advanced discussions to supply 80 million to 160 million doses. Negotiations with other countries are also ongoing, including with COVAX on the tiered pricing proposal. As a reminder, pricing for agreements with smaller volume were executed at $32 dose or $64 for two vaccination course with $47 per dose or $74 per course. We remain on track to fulfill these contracts with anticipated supply between 500 million and 1 billion doses in 2021. Turning now to our 2020 financial update on Slide 24. On an overall basis, we now expect net cash provided by operating activities and purchases of property and equipment in 2020 in the range of positive $0.1 billion to $0.3 billion. The change compared to our update in Q2 was primarily driven by the increase in customer deposits for the potential supply of mRNA-1273 as well as upfront payments for recently announced collaboration agreements with Vertex and AZ. Let me provide you more color on the individual components of our financial outlook. With regard to the ongoing investment in our portfolio, excluding our COVID vaccine candidate and associated activities, we remain on track to our prior outlook. We expect net cash used in operating activities and purchases of property and equipment to be approximately $0.4 billion in 2020, reflecting an improvement of $0.1 billion from prior outlook. This change is entirely driven by business development activities and related upfront payments as investment levels remain consistent with prior outlooks. Turning now to the financial impacts of our rapidly advancing mRNA-1273 COVID vaccine. First, expenses that fall under the scope of our BARDA agreement. These are primarily research and development activities to drive the COVID vaccine to licensure and scale-up activities on the technical development and manufacturing side. As we expect a relatively close matching of expenses and reimbursement, we do not expect these activities to materially impact our cash flow, and hence, these are not shown separately on Slide 24. Next, looking at the COVID vaccine-related net investments, primarily for manufacturing a product to be commercialized in the U.S. and internationally. We expect the cash impact of COVID-related investments to be $0.5 billion to $0.65 billion in 2020. This includes approximately $0.2 billion in capital investments with the balance of the expenses related to raw materials and production activity in our network. Additionally, this investment includes initial commercial infrastructure build-out and costs related to our supply agreements. The sum total of net cash used in operating activities for all of Moderna's business is currently expected to total $0.9 billion to $1.05 billion before consideration of customer deposits. Including the customer deposits received by the end of September, which sum to $1.2 billion, we expect the total net contribution from cash provided by operating activities and used for purchase of property and equipment of positive $0.1 billion to $0.3 billion. We expect this number to increase as we continue to receive further deposits. Turning now to Slide 25. As we progress towards approval and commercialization of mRNA-1273, there is heightened interest in several areas of accounting that will increasingly impact our reported results as we move forward. Slide 25 highlights some key areas, which will change with an approval event, for example, an Emergency Use Authorization in the United States. Costs associated with pre-launch inventory are currently fully expensed to R&D expense in the period incurred. This includes cost for acquired raw materials as well as production costs. After an approval of that, we will capitalize our inventory to the extent that the commercialization is determined to be probable, and we expect future economic benefits from sales to be realizable. In the third quarter of 2020, we expensed pre-launch inventory of $52 million, largely raw materials. The costs associated with purchases of property and equipment or leased assets related to our mRNA-1273 program are evaluated for assets with alternative use will be capitalized. Examples of these assets include IT and general production infrastructure. Assets acquired to meet the current production needs of mRNA-1273 and before product approval will be expensed immediately as costs are incurred. This reflects the fact that Moderna does not yet have other platform products approved or commercialized. After a regulatory approval event when PP&E at least assets are no longer required to be assessed for alternative use, such assets will be capitalized. In Q3, we recorded $10 million of PP&E as expense. On product sales, customer deposits for potential supply of mRNA-1273 are recorded as deferred revenue and will be recognized as revenue when control of approved product has been transferred to the customer and customer acceptance has occurred. In Q3, we recorded $1.1 billion of incremental deferred revenues associated with potential future supply of mRNA-1273. No product revenue was recognized in the quarter. Accounting for the BARDA grant follows a reimbursement model where we will recognize revenue as we perform services and closely match expenses as they are incurred. As of 12/31/2019, we had $982 million of federal and state accumulated net operating loss carry forwards. And $471 million of net deferred tax assets, which were fully reserved as we concluded that realization of our net deferred tax assets was not yet more likely than not to be realized. After a regulatory approval event, and as we expect to utilize the NOLs, we will reverse and release the valuation allowance or portion of the allowance, which will result in a tax benefit in our income statement. This concludes the financial update. And I turn now the call back to Stéphane.
Stéphane Bancel:
Thank you, Tal and David. In closing, I would like to step back for a few minutes. Across the world, we are all, rightly so, very focused on the pandemic. The race against the virus, both in therapeutics and vaccines. The Moderna team has been incredibly focused in 2020 on getting mRNA-1273 to the market in time. But an important part of my role is to look into the future, not only at the next few weeks, but the next few years and to look at the big picture, not only to look at Moderna. I believe Moderna entered 2020 in a strong position, with a strong cash position of approximately $1.3 billion, with a diverse clinical portfolio of vaccines and therapeutics across six different modalities. We have always focused on a portfolio approach to reduce technology risk. We have 20 development candidates. We had sustained over nine years since inception large investments in platform science, mRNA and LNP formulation. We have established a very broad and strong IP portfolio. We have sustained large investment in process development. We owned a fully integrated plant that allowed us to go from raw materials to fill vials for all of our clinical needs at scale and with unprecedented speed. As I look at the end of 2021, 14 months from now, I believe that if we launch mRNA-1273, we will exit the COVID pandemic crisis in a unique position. We should have a strong cash balance at the end of 2021. It will be made over $4 billion at hand as of September 30, 2020 plus the cash flow that we should generate in fiscal year 2021. The U.S. government has taken a very thoughtful approach with Operation Warp Speed or OWS. They decided to support free vaccine technologies, including mRNA to diversify risk, to ensure we got stable vaccines to the finish line for the U.S. citizens. They decided to back only two companies per technology. Moderna was one of the two companies that the U.S. government ordered 100 million doses from. That is a very important market access. We are grateful for the trust of the U.S. government placing us, our development process and our technology, and we're very thankful for the help. Moderna retains worldwide rights to give lot and commercialize mRNA-1273. Without a corporate partner, Moderna will realize all the profits from the COVID-19 vaccine. We intend to reinvest the returns from the sales of the vaccine into our pipeline development and hope to bring more medicines to the market. I believe that the long-term strategic implication are large. We should have a unique cash position at the end of 2021. Moderna has been built to scale because mRNA is an information molecule. We invested relentlessly in science at scale that no other company could have thought; in robotics, in digital, in process development, in the large manufacturing plant. We have always been limited in the last five year with cash. I believe that this is about to change in a very material way during fiscal year 2021. The approval of mRNA-1273 for commercialization will provide a unique derisking of the entire Moderna vaccine platform. We use the same chemistry to make each mRNA vaccine. We use the same manufacturing process to make the mRNA. We use the same chemistry for our lipid. The same manufacturing process to formulate the mRNA in our lipid. Think about what this team could do over the next five to 10 years starting from a growing credit balance of $4 billion and the knowledge that our technology leads to approved vaccines. I believe 2021 will be the most important inflection year in Moderna's history. Early on, we recognized that mRNA could be an entire new class of medicine. We always said, since day one, that it made no sense that this would be a one-product company. It will be zero if we fail to make safe and efficacious product or it will be a new class of medicine changing medicine forever. Our mission to deliver on the promise of mRNA science to create a new generation of transformative medicines for patients is when we strike for every single band. In 2020 we didn't wait long. We had many partners; the NIH, BARDA, Lonza, ROVI, Catalent, PPD, the clinicians that conduct clinical trials and our team. I'm very thankful for the clinical trial participants for their trust and the participation in our trials. With that, we will now be happy to take your questions. Thank you. Operator?
Operator:
Thank you. [Operator Instructions] And our first question comes from the line of Salveen Richter with Goldman Sachs. Your line is now open.
Salveen Richter:
[Technical Difficulty] COVID-19 vaccine. So just given the commentary from the recent FDA Adcom on COVID-19, just would like your thoughts here about how you intend to unblind potentially on the first interim or post the full three looks in the study? And then secondly just timelines around a vaccine EUA, just given some of the commentary, and I think Dr. Fauci talking about at January green light for vaccines? Thank you.
Tal Zaks:
Yeah. Hi, this is Tal. Let me try and take that. I think the bottom line here is that we feel I think an obligation to the participants of the COVE study, especially those for whom an EUA will be appropriate that there is a way for them to ultimately benefit from what they themselves have contributed to. And of course, I think we all listened intently to Vertex last week. There is a balance to be had here. The way I think this is going to happen at the end of the day, unfortunately, the cases are accruing. We are in a period of increasing transmission. And so I expect that there will be some time between knowing that the bar has been crossed for efficacy, doing the analysis and discussing with FDA. And part of that conversation is finding the right balance of how long do you continue to collect blinded data and what is the type of data that one can collect post blinding, but still on trial. I mean if you do the math, if you're looking for very rare safety events for example, then an unblinded study actually could give you power to detect that even more than a blinded trial. So a lot of these elements I think are going to go into of the conversation that we're having with FDA. And I'm confident that together with them we will find the right balance as to how to operationalize this. As it relates to the timing, I think we're on track to have the first interim in November. I think I expect unfortunately that we're going to be on track for additional cases occurring in December and beyond. And so the totality of data in the coming months I think will cross that threshold I anticipate for efficacy and the rest will be a dialog with FDA and other regulatory agencies on the right process by which to ensure we demonstrate the safety and efficacy and ultimately make the vaccine available.
Salveen Richter:
Thanks, Tal.
Operator:
Thank you. And our next question comes from the line of Matthew Harrison with Morgan Stanley. Your line is now open.
Matthew Harrison:
[Technical Difficulty] all the information this morning. I guess two related questions from me. Tal, can you comment at all about the attack rate that you're seeing in the study? I think given the Pfizer comments earlier this week, there is a lot of confusion among investors about whether the attack rate in these studies mirrors what we're seeing in the general population or if it's somehow lower than what we're seeing in the general population? And then a related question, something else that I think has come up a lot is this concern about functional unblinding potentially due to people recognizing some of the features of the boost and that may be leading towards certain people taking on different behavior, which may lead to this lower event rate. So I was wondering if you could talk about if you have a concern about functional unblinding? And especially related to the fact that you obviously have the placebo data from the Phase 2 study, do you have a much better idea of placebo-adjusted tolerability versus what we've seen from an open label study? Thanks.
Tal Zaks:
Thanks, Matthew. Let me try and give you a sense of how I see the data. First of all, my sense from the emerging data is that attack rates of our trial participants do mirror what we see in those zip codes where the subjects are coming from. It's not a surprise because if you look at the demographics of what we've been able to achieve in the COVE study, having so many people, minorities older with co-morbid conditions, so it's on par with expectations, I think writ large. And unfortunately with the current attack rates not slowing down, the math that we're doing, it's this paradox of more attack rates out there worse off for our subjects unfortunately, but the data will get there. So that's my sense. Now there is a balance to be had, which I think is -- I've tried to address in the first question that not just us but everybody in the field is struggling with, which is the accumulation of data versus the eventual unblinding of participants and how does that all play into regulatory expectations, and I think we'll continue to have this dialog in the coming weeks. The concern you raised about functional unblinding, yes, I share that. I think we all do, to a certain degree. We chose a dose that we believe is optimal in the sense that people may get some transient flu-like symptoms, but it's worth it for the opportunity to prevent this disease. I don't think this leads to lower event rates, per se. I think my biggest concern is that, if anything, it would bias us against vaccine efficacy rate. If people behave because they think they got something and that modifies their behavior, then if anything, you would expect that the behaviors would be such that the placebo recipients would be less at risk of getting infected and the vaccine recipients would be more at risk of getting infected. So at least from a statistical and robustness of the data, it shouldn't have any adverse effect. If anything, it should hurt us. But I don't expect this ultimately to be significantly changing the event rates. And if I look at the macro picture, as I've said, I think we're going to be on track, unfortunately, for where we anticipate being.
Matthew Harrison:
Thanks. Thanks very much.
Operator:
Thank you. And our next question comes from the line of Ted Tenthoff with Piper Sandler. Your line is now open.
Ted Tenthoff:
Great. Thank you very much, and thank you for all of your hard work in bringing 1273 forward. Amazing to see the company turning cash flow positive. A huge milestone. Let me get a sense for cost of vaccine at the price or at the quantities that you're talking about for next year with Lonza? Thank you.
Stéphane Bancel:
Ted, it's Stéphane. As we've said in the past, we are not disclosing cost of goods for obvious competitive reasons. So I won't be able to answer that question.
Ted Tenthoff:
Fair enough. Looking forward to data coming up. Thanks.
Stéphane Bancel:
Thanks, Ted.
Operator:
Thank you. And our next question comes from the line of Michael Yee. Your line is now open.
Unidentified Analyst:
[Technical Difficulty] what do you think is going on with the event rates? I know that you said you actually believe you're on time, so maybe it's a question more in part based on the competitor. And related to that, do you actually know your actual event rates, and therefore, you do you feel every day that you're seeing the numbers and feel very confident about November? Your competitor missed that timeline, so I think everybody is nervous about that. So maybe just make a comment about what you think is going on in general? And then secondly, because if the NRM is actually pretty high interim, and I think you've actually commented on that. Maybe just make a comment about if we don't hit that what that will mean for folks? And I think it has to do mostly with the alpha spend, so that's why I'm not too concerned. But maybe just make a comment about that as well?
Tal Zaks:
Thank you. Yeah, this is, Tal. Look, I can't comment about our competitors. I don't know their data. So I'll leave that to them. I can repeat what I said about our sense. We do of course see the data coming in. There is a small team at Moderna that's aware of the cases that's following up. That number is of course being kept confidential to minimize speculation here. But I can tell you that overall since we are following the zip codes and the counties from which these participants come, we have pretty sophisticated models of what to expect, and I think we're on track for those expectations. So I think we should be on track for that first interim some time in November, as we have articulated. You raised a valid point about the interim, and thank you for asking that. Hitting that interim is going to be a function of what the actual vaccine efficacy is and an element of luck on the distribution of the first batch of data you see. Not hitting it doesn't mean the vaccine doesn't work. And in fact, it doesn't even mean that the vaccine has a less than 75% efficacy. We could easily not hit and yet come back on the second interim and demonstrate an 80% or 85% efficacy. That's kind of the chance of how the stats work. I think the alpha spend, etc., you're correct. This is a conservative design and maybe it's an opportunity to dispel a notion. The fact that we crossed the boundary at an interim doesn't mean it's less powerful than a final from the peer statistical standpoint. Once you cross that boundary, you have the same statistical conviction that you would have had on the final. Now I think the way I think about the data here is irrespective of the point at which we cross the boundary, the trial will continue blinded for a period of time. Data will continue to accrue. And as long as the trial continues blinded, we will get better and better with more and more data to increase the level of certainty we have on all the endpoints of the trial. So in that regard, for me, this first crossing of the interim is just the basis that allows us to go and have confidence to start doing analysis and proceed down the path of regulatory interactions so that we can ensure that ultimately access is not delayed to people when there is such a high need out there in our communities.
Unidentified Analyst:
Thank you.
Operator:
Thank you. And our next question comes from the line of Gena Wang with Barclays. Your line is now open.
Gena Wang:
[Technical Difficulty] do you also see safety in a blinded format? If so, how is that compared to the Phase 1 profile? And then two questions regarding the interim analysis. So if first interim -- if you missed the first interim, is this due four to eight weeks to hit the second interim? And then for the first interim, what is the futility boundary?
Tal Zaks:
Let me take that. The safety, yes, we see the totality in a blinded fashion. I would remind you that the Independent Data Safety Monitoring Board that's been appointed by the NIH and sees not just our trial, but also the other U.S. OWS-sponsored trials, in parallel sees both the blinded and the unblinded data. Writ large, I can tell you that so far we're not seeing anything unexpected, so the trial continues. And in that regard, I don't expect surprises when we eventually unblind. Your second question around the timing is I think it's a matter of weeks between the first and the second interim, not months. Now I can't be more precise because obviously it's a function of the future transmission rates in areas, and I hope that they go down. But for now this is four weeks, not longer. In terms of futility, I think the boundary is to show that this is going the wrong way. It's specified to the degree that we felt it was appropriate to specify in the protocol. Beyond that, I can say that the reason you have a Data Safety Monitoring Board, you've got on that panel several statisticians and several experienced clinicians. So it is where I expect them to exercise their judgment and experience when they're looking at it closely.
Gena Wang:
Okay. Thank you.
Operator:
Thank you. And our next question comes from the line of Cory Kasimov from J.P. Morgan. Your line is now open.
Unidentified Analyst:
Thanks guys for taking my question. This is Matthew on for Cory. Just on the COVE trial in light of subject mix and commentary that event rates are tracking geographically with infections. I'm wondering what your assumptions are for the proportion of patients with SARS-CoV-2 infection that you expect to ultimately going to become symptomatic?
Tal Zaks:
Yeah. So let me make -- let me answer in maybe a simplistic form. The tracking is first and foremost of symptomatic cases because we're not routinely swabbing people just to check PCR as people self-identify. The asymptomatic infection rate will be determined based on serology that distinguishes between infection and immunization by later comparing antibodies against nuclear capsid versus spike protein. So we are primarily aware of the symptomatic infections, to begin with. And that is the parameter that we're primarily tracking as it relates to the epidemiology. Hope that answered the question?
Unidentified Analyst:
Yeah. I guess maybe just to follow-up. So when you say that rates are tracking the zip codes, what is the data that you are gathering from outside the trial to benchmark it to zip codes?
Tal Zaks:
Reporting of case rates and infection rates together. It's -- we've got a couple of independent expert panels and teams here that compile the totality of the data that emerges from the various surveillance programs and sophisticated models to create those predictions.
Unidentified Analyst:
Great. Thank you.
Tal Zaks:
If you're asking me to describe what's under that hood, it's a complicated black box for me. But I can tell you that the number that it spits out ultimately looks very close to the number that we see on a daily and weekly basis at this stage, and that's what gives me the confidence.
Unidentified Analyst:
Got it. Thanks.
Operator:
Thank you. And our next question comes from the line of Geoff Meacham with Bank of America. Your line is now open.
Unidentified Analyst:
Hey, guys. This is Alec on for Geoff. Thanks for taking our questions. Can you talk a bit more about the tiered pricing proposal with COVAX? What would this look like? And if you can talk about the agreement specifically, what's the logic of a tiered structure versus just a set price for a certain amount of doses seen in the other supply agreements? And on the Japan supply agreement, and I suppose on most of the OUS supply agreements, how do you plan to allocate manufacturing between your plants and launches? And are there any logistical constraints that would limit the vaccine produced in Norwood geographically? And I guess, just lastly. What's being done to ensure consistency of vaccine produced between both your own internal and the external manufacturing sites? Thanks.
Stéphane Bancel:
Hey, Alec. It's Stéphane. So it's a lot of questions. If I forget something, let me know. So on COVAX, what we want to do is to make sure that the vaccine is available around the world. As you know, we believe based on -- I mean on the early development data that the vaccine has a chance to be protective across different age range, and we think it's very important. And so we want the vaccine to be available to help as many people as we can. The tiered pricing is very typical of what Gavi has done in the past. As I'm sure you are aware, COVAX is being run by Gavi and CEPI. And so the idea here is to propose to low-income countries and middle-income countries a price that is lower than the price being paid by developed countries or high-income countries. And so that is basically the idea. And so of course, we're not in a position to disclose those prices today because the discussions are ongoing with COVAX. But we want to make sure as a company that we are able to provide vaccines at a lower cost for low-income and mid-income country. In terms of quality across the board, I would say it's typical of what is done in many biopharmaceutical companies when you have several sites in term of a quality organization, technology transfer validation and so on to ensure that the -- a product that is made at Lonza in Visp or at Lonza in New Hampshire or at Moderna in Massachusetts is of the same quality and the same spec. And then in terms of allocation. So as we said in the past, we setup the supply chain, the U.S. supply chain, Moderna Massachusetts and Lonza New Hampshire to focus initially on the U.S. We setup the supply chain in Lonza Visp to be initially focused outside the U.S. And I think this is going to really hold for just I would say two quarters after launch. After such a time, we are going to setup the supply chain in term of regulatory filing so that products can be moved around. So if we get to a point where we have kind of sold all the products in the U.S. that makes sense to be sold in the U.S., we will be able to ship products from the U.S. to international and vice versa. We want to setup the supply chain so that if at some stage we need more product in the U.S., we can import from Switzerland more products into the U.S. So we're trying to setup a supply chain that allows us to be very flexible and nimble to react to the needs of a marketplace. I hope that was your question, Alec?
Unidentified Analyst:
Yes, very helpful. Thanks.
Stéphane Bancel:
Thank you.
Operator:
Thank you. And our next question comes from the line of Hartaj Singh with Oppenheimer. Your line is now open.
Hartaj Singh:
Great. Thank you. Thanks for all of the data presented. Just a couple of quick questions. One is a non-COVID-related on CMV-1647. You've got a worldwide -- since you have worldwide rights to, how are you thinking in terms of as you grow the organizations, Stephane, on a worldwide basis with 1273? Can you just talk a little bit about how that will help you with 1647 when the time comes to launch that, even prepare for worldwide trial with 1647? And then secondly, is it just wrong for us to assume that part of your initial revenues from 1273, the COVID-19 vaccine, could be sort of one-offs depending on how governments order them versus kind of a more like-for-like basis comparison on a year-for-year basis. Any way to think about that going forward? Thanks for the questions.
Stéphane Bancel:
Thank you, Hartaj. So on the, I would say, non-COVID organization, Stephen Hoge has spent a lot of time since the beginning of the pandemic to refocus on that one. I think we've done to allow us as a company to both deliver 1273 at the kind of historic record time and not slowdown the rest of our pipeline, which of course is very important for Moderna and Moderna's future and for patients. Tal has refocused most of this time on 1273 and taking this product from literally in a computer into Phase 1 into Phase 2 and into competing successfully over the COGS study and all the interaction with regulatory agencies around the world on the one hand. And then Stephen has refocused on the non-COVID portfolio. And so what we have done is, we've been hiring quite a lot. If you look at our headcount numbers, we've been hiring quite a lot and we're going to continue to do so into the next quarters so that we can have the right scale in terms of development, so that we don't slowdown the development pipeline of a great asset like CMV or PCV or all of our asset because we don't have the right teams on the ground. And as we get closer to commercialization, we'll of course hire people dedicated to that. We've already actually higher the Head of Marketing for CMV because it's very important we believe given there is no product on the market for CMV. In terms of disease awareness, a lot of women do not know about CMV and what CMV could do to their pregnancy. And so we think there is a great opportunity while the Phase 3 is running to re-drive up CMV awareness. And I think in the post-COVID world where people are going to be, I believe much more attuned to infectious disease, and given the similarity of outcome between the Zika and CMV and given what's happened in Latin America a few years ago, everybody is highly aware of Zika. And so I think there's a great opportunity for us to actually prepare the market to increase the speed of the uptake of CMV by preparing the market a few years before, which is what we are doing. In terms of our revenues, as we described, I think in the Q2 call, we really believe there is a pandemic phase of the COVID vaccine and then an endemic phase which is going to be setup mostly on boosting. As has been discussed a lot in the medical community, we do not know across the different vaccines how long we're going to be working for. And so we believe most probably especially the elderly people are at high risk who are going to want to boost their immune profile. We do not know yet if it's going to be once a year, once every two year, once every three years, most probably it will depend on the population. We have no way to know that, but to run the clinical experiments, which we are all doing across the industry. If you take a different vaccine will have different duration of protection. And so indeed I think you're thinking about it in the right way, which I think there's going to be for quite a while pandemic contracts that are going to be setup, Blackstone that we have setup to actually start vaccinating people. And then there's going to be a follow-up contract with the currency paid for the boost. So that's kind of how we think about it. Does that makes sense, Hartaj?
Hartaj Singh:
Yeah. No, that helps a lot, Stéphane. Thank you.
Operator:
Thank you. And our next question comes from the line of George Farmer with BMO Capital Markets. Your line is now open.
George Farmer:
Hi. Good morning. Thanks for taking my questions. I was wondering if you could comment on what happens if you don't hit the primary endpoint of this study, of the COVE study? And you do hit meaningfully on secondary endpoints, which could be quite important, I mean, certainly reducing severity of disease could be something regarded as meaningful. Have you thought about that? And my second question is, if you do get an EUA, does the existing supply agreement having come into play, do you start drawing down on your inventory commitments following EUA or do you wait until you get full approval? Thanks very much.
Tal Zaks:
So, George, this is Tal. Interesting point you raised. Typically if you don't hit the primary, you don't get to get -- take credit from secondary is the way the stats are laid out. I think in this case, if there is biological possibility, then of course we'd go and discuss with the agency what the data are looking like. Because I anticipate we will actually have the opportunity to cross it early, we may have those discussions ahead of time while more data is accumulating. And if we fail to hit it, but the trial continues blinded given I think the exigent circumstances, we can go and have a dialog with the agency on what we've seen so far. But I'd say the proximal answer would still be the conservative one, which is if you don't hit the primary, it's very hard to take credit for secondaries. It's going to be up to the regulators. I think the answer to your second question is an easier one. Yes, the supply agreement kicks in and U.S. government I would expect would start to take the supplies once we have an Emergency Use Authorization.
George Farmer:
Great. Thanks very much.
Operator:
Thank you. And our next question comes from the line of Alan Carr with Needham & Company. Your line is now open.
Alan Carr:
Hi. Thanks for taking my questions. Stepping away from COVID, you all mentioned in a months or last month that you'd like to revive that flu program. I'm wondering if you could give us some more details around that? Is it something going to be both as a seasonal flu vaccine or something that we see as a universal vaccine? And if you can give us any updates on timelines around that?
Stephen Hoge:
This is Steve. I'll take that.
Stéphane Bancel:
Yeah. Go ahead, Steve.
Stephen Hoge:
So we announced last month is that we're looking at it strategically and we're going to begin building a business in seasonal influenza. I'll remind you that we do have already clinical data in pandemic influenza from two different programs. And so we're still in the ability to generate a productive immune response to influenza virus as well. We haven't provided clarity yet on when we expect that program to move into the clinic. We will in the future provide those updates as we get closer to that time. But we have suggested both it's an interesting market to try and do better on because there is a substantial still unmet need an influence, and we do think our platform can be differentiated for a couple of reasons. One, obviously, that we can bring many antigens to bear. And the second, the opportunity to do combinations, potentially combinations even with other viruses that might need seasonal boosting. And so we'll provide an update as soon as we declare the development candidates we normally do and move forward towards clinical development. But we're quite excited by that opportunity at this time.
Alan Carr:
Great. Thanks for taking the questions.
Operator:
Thank you. And our next question comes from the line of Umer Raffat with Evercore. Your line is now open.
Umer Raffat:
Hi, guys. Thanks so much for taking my questions. I had a couple if I may. First, maybe starting from an easier one. What's the cycle threshold you're using for the PCR positivity? Is it about 34? Second, if you could confirm if there have been any one-off cases of COVID infection for participants who got 1273 in Phase 1 or Phase 2? And then finally, I know the slides mentioned rapid protection in lung and nose of non-human primates. But in The New England Journal data, we did see at least one primate at the 100 dose with detectable RNA on the nasal swab. So I guess what I'm getting at is, as a base case, despite the requirement of symptoms, is it reasonable to assume that we could actually see one-off cases of nasal swab test positive for COVID? Thank you very much.
Tal Zaks:
Hi, Umer. Let me take those questions. Our cycle threshold, I don't think the number is meaningful because it's not the same number on different assays. It's part of the assay characteristics, so I can't really comment because I don't believe you can compare those numbers across different assays. On the participants in the Phase 2, I think we've had a case or two. But since we're still blinded, I can't really comment on what that means. The question on nasal swabs, yes. Well, first of all on the primate, you're right, there was one primate with transient and relatively low levels in the nose and the rest were sterile. I think that when we talk about sterilizing immunity, I think that concept is confused by the high sensitivity that we have. So what do I mean by that? Well, if I am immune and I've got IgGs and I've got IgAs as our data show we have and I'm walking down the street and somebody sneezes on me a whole bunch of SARS-CoV-2, and I'm on 49th Street, well, if somebody swabs me on 50th Street and sticks a swap up my nose, they're probably going to see some detectable virus from that cloud I just inhaled a block ago. It doesn't mean that the antibodies aren't kicking in and the virus isn't getting clear, that's a question of timing and sensitivity. So it depends then on how you measure and when you measure and what are the triggers for measuring. In our trial, we test people who raised their hand to say they've got a symptom and a reason to be tested. The only asymptomatic testing that occurs is really at day one and day 29 to make sure that we're excluding the right people and not confounding adverse events with actual disease. But short of that, we're not just randomly doing swab tests. So I hope that answers your question as it relates to detection?
Umer Raffat:
Thank you very much.
Operator:
Thank you. And our next question comes from the line of Mani Foroohar with SVB Leerink. Your line is now open.
Mani Foroohar:
Hey, guys. Thanks for taking my questions. I know there has been a lot of focus on the COVID-19, but specifically I wanted to dive in on massive financials a little bit. So did I hear right that for the quarter about $10 million of what would otherwise be PP&E were expensed? And so would moving that back to CapEx give us the base from it would go here? And secondarily on that, how should I think about the tempo of CapEx growing over the next 12 to 24 months presuming a successful EUA and global commercialization? And then I have a follow-up question out of this.
David Meline:
Yeah. So in terms of the capital deployment, if you look at the quarter three, we had $10 million, which I mentioned, that was 1273-related that was expensed. What also we invested was $12 million in the quarter, which was capitalized. So $22 million in total. And I think that's a reasonable run rate. You will see some increase here as we finish the year and we trend up on investing in preparation for launch and the full capacity that we're putting in place. As that continues into next year, I think it's fair to understand that the capital deployment is going to increase somewhat, including the possibility in the fourth quarter of additional expensed capital to the extent it's deployed prior to the approval, which we do expect more to happen. And then if I may, also keep in mind the inventory expensing, which is not strictly capital investment, but the acquisition of raw materials and the cost of production, and of course we're ramping that production in anticipation of an approval. And therefore, it's quite likely that we'll have a fairly significant amount of expensed inventory occurring in Q4 prior to approval. And as you would understand, the impact of that is that as we start commercializing product post-approval, the cost of the product will be essentially zero from a cost of goods perspective until we've sold through that inventory that we've produced prior to approval.
Mani Foroohar:
Okay. That's really helpful. And then hopping over on to the non-COVID pipeline. I may have missed it if it was mentioned. What's the timeline to get the next slug of data from the OX40 ligand program? And when should we think about having a reasonable number of patients with that asset on top of a PD-1 -- on top of an approved PD-1 therapeutic?
Tal Zaks:
Let me try and take that. This is Tal. So it's oncology, and data I think is determined by not just patient accrual, but the events that happen on trial. And so it's really hard for me to predict, which is why we've been very clear on how many patients we have and where are we. I think the next tranche of data I would anticipate will be once we have a sufficient number of these patients with ovarian cancer on trial, and that is already in combination with the PD-L1 blocker. So that should be informative for our response rates once we have sufficient patients with sufficient follow-up.
Mani Foroohar:
Great. That's helpful. And as a final question, bought back to financials. Obviously the stock has been quite volatile over the course of this year, as have many of your peers, given that you continue to build out your infrastructure and headcount anticipation of expanding the pipeline and launching commercially for them to zoom in EUA for COVID-19, how should I think about modeling or how should we think about modeling stock-based compensations relative to the overall R&D expense, SG&A expense? Will the portion go up as you start to have more of a sales force? Will it down as you get scale? Just how should we think about that, either in absolute terms, as a proportion of spend? Just how should we think our modeling it in the next say 12 months?
David Meline:
That's a very good question. And I guess I'd have to get back to you. I guess, I'd start with the point of view that the trend would be stable. But we'll have to get back to you on that.
Mani Foroohar:
All right. Thanks for taking the questions, and congratulations on all the success.
Operator:
Thank you. Our last question comes from the line of Navin Jacob with UBS. Your line is now open.
Unidentified Analyst:
Hi. This is Elana on for Navin. Thank you for taking our question. So from what we've seen in your non-human primate studies, what impact on prevention of infection through the viral colonization and viral transmission do you expect to see from 1273? And then secondly, sort of a broader question is, do you think that your vaccine or any vaccine in general will be able to deliver sterilizing immunity?
Tal Zaks:
Yeah. So that's a good question. Let me tell you how I think about it. Again, as I alluded before, I think the concept of sterilizing immunity is a function of how you actually test for it. We'll be able to demonstrate prevention of infection, at least as measured by serology, which is to say maybe somebody saw a little bit of the virus, but actually the antibodies kicked in and they never established enough to demonstrate antibodies against the other parts of the virus, the nuclear capsid. That's actually what we will be measuring in the Phase 3 as a way to show protection from asymptomatic infection. I'm hopeful that we will be able to demonstrate that. I anticipate that -- I mean, just on first principles, what is the hardest thing for a vaccine to do? It's to sterilize. What's the next hardest? It's to prevent asymptomatic. What's the easiest? It's probably to prevent the most severe disease. And so that's how I think about the order of expectations. But I think what you're really getting at is what is going to be the ability of this vaccine to prevent transmission. And I think because I don't anticipate any easy direct measure of this, we're going to have to do some math once we see the data. I expect that if people get less sick and certainly less severe sick, they will be spreading less because I think there is -- while we have asymptomatic spread, we also have symptomatic spread. And so -- and in fact, I think the higher the symptoms at least earlier in the disease, potentially the more the viral shedding you see. So that's a complicated equation that I think in a nutshell is going to be very hard to answer directly through some concrete measurement in the trial. What we will hope to show is that we prevent asymptomatic infection based on serology. We will clearly hope to show prevention of symptomatic infection, that's the primary endpoint. And based on first principles, which by the way, Phil Krause from FDA alluded to and agreed with last Thursday, one would anticipate that if you prevent symptomatic disease, you should -- it should be even clearer that you're preventing the more severe manifestation.
Operator:
Thank you. And that concludes today's question-and-answer session. I would now like to turn the call back to Stéphane for any closing remarks.
Stéphane Bancel:
Great. Thank you, operator. Well, thank you everybody for your time and your question and your support. Have a wonderful day, and speak soon. Stay safe everybody. Bye.
Operator:
Ladies and gentlemen, this concludes today's conference call. Thank you for participating. You may now disconnect.
Operator:
Good morning. And welcome to Moderna’s Conference Call. At this time, all participants are in a listen-only mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head of Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, everyone, and welcome to the Moderna’s second quarter 2020 conference call to discuss financial results and business update. You should access the press release issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our website. On today’s call are Stéphane Bancel, our Chief Executive Officer; Tal Zaks, our Chief Medical Officer; Stephen Hoge, our President; and David Meline, our Chief Financial Officer. Before we begin, please note that this conference call will include forward-looking statements made pursuant to safe harbor provision of the Private Securities Litigation Reform Act of 1995. Please see slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon, everyone. I hope all of you are in good health and remain safe. Thank you for joining our Q2 business update. We are committed to building mRNA as a new class of medicines. We model our position to remain the leader in this field. As you know, we believe our mRNA medicines have a potential to address the large and unmet medical needs and to treat diseases that are not addressable by recombinant protein or small molecules. Due to the platform nature of mRNA, we believe our mRNA medicines provide a higher probability of technical success to commercial launch and faster timelines to clinical trials and to the market relative to traditional medicines. We also believe that the manufacturing capital intensity of mRNA is materially lower than recombinant protein and the cost of manufacturing at commercial scale will be similar to small molecule injectables. Let me start by summarizing the key achievements of the Company in Q2 on slide 4. mRNA-1273, COVID-19 vaccine candidate, saw the start of its Phase 2. And the team focused on the Phase 3 design and preparation for July start. We signed a strategic long-term partnership with Lonza to a significant manufacturing capacity to make mRNA and formulate it at large industrial scale to complement our Massachusetts manufacturing site capacity in the context of the pandemic. We started the technology transfer to Lonza in Q2. We raised $1.3 billion on May 18th. And as communicated in our S-3, this capital was raised to enable us to scale up at risk our manufacturing capacity in anticipation of a potential approval for mRNA-1273. We started investing the proceeds for additional capital equipment at our Massachusetts plant, as well as the Lonza plants in New Hampshire and in Switzerland, ordering raw materials at large scale and hiring additional personnel to make mRNA-1273. In April, we finalized and announced an award from BARDA for up to $483 million for clinical development costs for mRNA-1273. An additional award was also announced last week for upto $472 million, given the increased size of the Phase 3 to 30,000 participants. The total world is now upto $955 million. In a very difficult environment, during the spring, given the many lockdowns across the U.S. due to COVID-19 outbreak, our clinical team was able to continue to operate most of our clinical trials on schedule. And we remain on track to provide in Q3 the Phase 2 interim data for CMV vaccine candidate, mRNA-1647. The team continues to work relentlessly to start the Phase 3 study for CMV in 2021. We continue to believe that our CMV vaccine candidate annual peak sales could be $2 billion to $5 billion. And like mRNA-1273, COVID vaccine candidate, we own the global commercial rights of mRNA-1647, our CMV vaccine candidate. Q2 marked a new growth phase for Company as we started to build and hire our commercial team. This is a historic moment for those of us who have worked at the Company for many years, since it was a breakthrough research enterprise, working with small animal models. And now, we’re building commercial. In Q2, the team started discussions with several countries around the world about potential supply agreements for mRNA-1273. We saw first deferred revenues book in Q2 on the balance sheet for $75 million of cash receipts from deposits for first potential supply of mRNA-1273. As of the July 27th, we have received approximately $400 million of cash deposit. For today’s call, we will cover three main topics. First, Tal will review and update you on our clinical programs and share some new clinical data on our CMV and Zika vaccine candidates. Then, Stephen and I will share with you our value framework for vaccine COVID-19 candidate and our approach for delivering value during the pandemic. Finally, David will take you through the second quarter financials. Let me now turn the call to Tal.
Tal Zaks:
Thank you, Stéphane, and good morning, everyone. Let me start with our prophylactic vaccines portfolio, a quick overview of the programs is on slide 7. mRNA-1273, our vaccine against COVID-19 has made very significant progress. The interim results of the Phase 1 trial have now been published in The New England Journal of Medicine. The Phase 2 trial was fully enrolled. And importantly, we recently started Phase 3 trial of 30,000 volunteers in the U.S. Our CMV Phase 2 dose confirmation study remains on track for interim data in the third quarter of this year, and the Phase 3 trial is expected to begin in next year. The Phase 1 trial for Zika is fully enrolled. And I’ll share some data from the 100 and 250-microgram dose cohorts shortly, while we’re preparing for Phase 2. Our Phase 1b age de-escalation study with our combination hMPV/PIV3 vaccine remains on pause with enrollment suspended due to COVID-19 disruptions. And finally, the Phase 1 study with mRNA-1172 against RSV led by our partner Merck is ongoing. So, new is additional data from our CMV and Zika Phase 1 programs. For CMV, these include 12 months immunogenicity data from dose cohorts, 30, 90 and 180-microgram and the safety data from the 300-microgram dose cohort. For Zika, we had disclosed data from the 10 and 30-microgram cohorts. And today, I will show you the safety and immunogenicity data for the 100, 250-microgram dose groups. So, starting with CMV. On slide 9, you will see a table of the solicited adverse events after the third and final vaccination in the highest dose we tested, 300-microgram. The safety and reactogenicity data from this dose is despite side by side with the other lower dose cohorts from the trial. At the 300-microgram dose levels, solicited adverse event reaction showed a trend towards increases with the higher dose and who were higher in seropositives. This is consistent with what we’ve seen in the other doses as we moved from 30 to 90 and 180 micrograms. The most commonly reported adverse events were pain at the injection site, headache, fatigue myalgia and chills for seronegative participants with fever and arthralgia as more common adverse events in seropositives. Importantly, at the 300-microgram dose, there continues to be no related serious adverse events reported and no unexposed safety findings. Moving to the 12-month immunogenicity data for the full 30, 90 and 180 micrograms dose cohorts in the Phase 1 CMV trial. We’re happy to report that six months after the last vaccination with mRNA-1647, we have durable neutralizing antibody responses out to the 12 months timeframe. Slide 10 shows the data in the same format we reported previously. At the top part of the table, you see the geometric mean neutralizing antibody titers against epithelial cell infections or the pentamer at the 12-month mark, as well as the fold increases above the seropositive benchmark of 5,917. At the 90 and 180-microgram doses, the fold increase in geometric mean titers above that seropositive benchmark were 3.6 and 3.9-fold higher respectively. And the bottom half of the table figures for the geometric mean antibody titers against fibroblast infection or the gB antigen for the same doses can be seen, and the geometric mean titers against fibroblast cells in the 90 and 180-microgram were 0.7 and 0.9-fold seropositive benchmark level of 1,449. On slide 11 is the graphical representation of the geometric mean antibody titers against epithelial cell infection relative to the seropositive benchmark, geometric mean titter. And the scale is the log scale. You can see the seronegative participants by the solid line and the increase in neutralizing antibody titers for each of the doses after each vaccination. The 90 and 180-microgram doses are shown in the solid blue and orange, and they’re well above the benchmark seropositive level of the 12-month mark. This is six months after the last vaccination. So, the persistence of neutralizing antibodies is encouraging. Seropositives are represented by the dashed lines. These participants entered the trial with neutralizing antibodies as you can see from the start, and they also saw their neutralizing antibody levels further increase with each vaccination, remaining above the geometric mean titer for neutralizing antibody levels to the 12 months mark. CMV is an important unmet need, and we’re encouraged and excited by these 12 months data and are currently running the Phase 2 dose confirmation study where we’re testing a narrow range of doses at the 50, 100 and 150-microgram, 252 seronegative and seropositive results have been enrolled, and we remain on track for an interim readout in the third quarter of this year. We continue to plan for the Phase 3 trial in 2021, pending further regulatory interactions and feedback. The primary endpoint, as we have previously reported, is anticipated to be prevention of primary infection in seronegative women of childbearing age. And we expect to include less than 8,000 participants in the U.S. and Europe. Let me now move to mRNA 1893, our vaccine against the Zika virus where we’ve seen additional data at the 100 and 250-microgram dose levels. Both of these dose levels were generally well-tolerated with a trend towards more observations of local erythema and swelling at the injection site with the higher dose levels and after the second vaccination. Consistent with what we’ve seen across our vaccine platform, there’s a trend of more solicited systemic adverse events noted with 250-microgram dose levels, particularly after the second dose. There were no related serious adverse events. You can see some of the 30-microgram data have been updated from our Vaccine Day Presentation back in April. In terms of the immunogenicity response, both the 100 and 250-microgram dose level induced a strong neutralizing antibody response in both seropositive and seronegative participants. The table on the left hand side focuses on the seronegative participants and the data for the 10 and 30-microgram cohorts that we previously shared. All seronegative participants in the 100-microgram dose cohort seroconverted after the second vaccination, similar to that seen with a lower 30-microgram dose cohort, but now with higher geometric mean titers as depicted graphically on the bottom. We did not see any further increase at the 250-microgram relative to the 100-microgram dose. Now, on this page, you can see the data for participants with preexisting flavivirus in unity or the seropositives where mRNA-1893 was still able to mount to Zika specific neutralizing antibody response. Here, the first vaccination is comparable to a booster response. And the response was similar between all dose levels. Although of course, I would note that these are very small numbers. We are reviewing the data and will make a dose selection decision in preparation for the Phase 2. And this program continues to be supported by BARDA. So, let me quickly review at a high level the data generated to-date with mRNA-1273, our vaccine against COVID-19. The Phase 1 clinical data we published in The New England Journal show that 100% of the participants vaccinated generated neutralizing antibodies and the geometric mean titer level at day 43 were 4.1-fold, higher than those seen in convalescent sera from 3 representative COVID-19 patients. As published recently in The New England Journal of Medicine, in the nonhuman primate, vaccination with the two-dose regimen of mRNA-1273 led to rapid protection against infection in both the lungs and those of the animals. Finally, a preprint from a mouse challenge model has showed consistent protection in the lungs and noses of those animals as well. And this morning, we’re happy to hear that Nature published this data and it is available online today. So, we’ve been able to show protection against viral replication in every species we’ve tested so far, and the levels of neutralizing antibodies that correlated this protection are roughly similar to those we achieved in humans in Phase 1. So, we look forward to the data readouts expected in the coming months, which will include the older and elderly cohorts in Phase 1, the safety and immunogenicity data from the Phase 2, and the potential for ensuring safety and efficacy analysis from the Phase 3 COVE trial. Let me now quickly review our other clinical programs in the Systemic Secreted & Cell Surface Therapeutics. I’m happy to report that the additional two cohorts of the Phase 1 trial for mRNA-1944 have recently completed enrollment. As a reminder, in this program, the mRNA-1944 encodes for IgG antibody against the chikungunya virus. The additional cohorts are testing an IV infusion of mRNA-1944 at the high dose of 0.6 mg/kg with steroid premedication, as well as two doses of 0.3 mg/kg without steroid premedication, given a weaker part. The randomized Phase 2 trial of our personalized cancer vaccine in combination with KEYTRUDA versus KEYTRUDA alone, which is partnered with Merck, is ongoing. And we’ve also made progress with the intratumoral immuno-oncology programs. Our OX40 ligand program has expanded into the Phase 2 dose expansion study in combination with durvalumab in patients with ovarian cancer and is actively recruiting patients. The Phase 1 program with our triplet of OX40 ligand IL-23 and IL-36 gamma is ongoing and data were recently presented at ASCO. You can find a link for the data at the bottom of the slide. Within our systemic intracellular therapeutics modality, both MMA and PA studies remain on pause due to COVID-19 disruptions. Finally, our partner led programs including the mutant KRAS vaccine with Merck and IL-12, and VEGF with AstraZeneca continue. Slide 18 is a snapshot of our development pipeline with mRNA-1273 in Phase 3 and CMV in preparation for Phase 3 start in 2021. We have four trials now in Phase 2 and two preparing for Phase 2 and 10 development candidates in Phase 1 with a further nine in preclinical studies. So, with such a broad development pipeline, we anticipate many readouts and next steps in the near term, and they’re listed on slide 19. These include, as I mentioned, the Phase 1 results from the older and elderly age cohorts, Phase 2 and interim analysis for Phase 3 for mRNA-1273, the Phase 3 results from CMV in the third quarter, and the complete Phase 1 data from the additional cohorts from our antibody against the Chikungunya virus. With that, let me turn the call over to Stephen.
Stephen Hoge:
Thank you, Tal. I’d like to start our discussion on mRNA-1273 value by -- on slide 21, just recapping the scale of the loss that we’ve all been facing. It is remarkable that this pandemic has harmed millions of people already and our hearts go to those who’ve lost loved ones or been made sick themselves. Given the scale of the devastation, it is hard to believe that just seven months ago, none of us have ever heard of SARS-CoV-2 or COVID-19. Globally now, over 18 million people have had confirmed infections with the virus, and almost 700,000 have died. In the U.S., that looks like 4.7 million confirmed cases and 150,000 deaths. And the estimates are that by year-end in the U.S. alone, there could be up to 400,000 deaths in this country. What we’re learning about the virus almost every day is that it may have a number of long-term sequelae beyond the short-term impacts of the disease. So, beyond the pulmonary infection, pneumonia, acute respiratory distress syndrome and pulmonary embolism that we see, there have been increasing reports of other coagulation disorders, cardiac injury and long-term sequelae, as well as disturbing reports of potential long-term neurologic complications and potential inflammatory syndromes in children. Clearly, we’re learning more and more every day about the terrible devastation of this virus. And it’s absolutely imperative that we and others advance the vaccine to try and blunt this pandemic and control these long-term sequelae. So, on slide 22, in trying to assign value to a vaccine during a pandemic such as this, there are a number of approaches, the one of the most established is using incremental cost effective ratios. Using a health economic assessment framework, you usually look just at healthcare costs, really the direct medical costs associated with caring for the disease. Through the ICER analysis, we look at the incremental change in costs divided by the incremental change in health outcome. And the health outcome, the value of that improvement is measured based on willingness to pay thresholds. Generally, $50,000 to $150,000 per quality-adjusted life-years. On slide 22, I’m presenting a recently completed analysis by Quadrant Health Economics, looking at an ICER at a $50,000 quality threshold. This analysis looked just at the short-term benefits of vaccination during a pandemic. And what I mean by that is, just the value created in the first year after rolling out broadly of a vaccine. Scenarios depend on which populations you’re looking at and the value also depends upon the epidemiology of the virus that’s ongoing. But looking at a current trajectory of infection in terms of epidemiology and vaccinating all adults, meaning those 18-plus, the ICER 50,000 value of the vaccine would be approximately $300 per course. If you focused vaccination just initially in the high-risk populations, say those over the age of 65, that value expands substantially, not surprisingly, given the high burden of disease in that population. Obviously, as transmission increases, the potential value of a vaccine is significantly increases beyond that. Now, on slide 23, it’s important to note the limitations of these ICER type analyses. So, while there is obviously a lot of value in a vaccine in directly impacting the healthcare costs, such analysis does not look at the long-term sequelae and long-term disability potential of a disease like COVID-19. It also does nothing to capture the social disruption that’s having a huge impact on all of our lives, nor does it do anything to reflect the economic loss that is rampant across our economies globally. And with that, I’d like to turn it over to Stéphane to talk to our approach.
Stéphane Bancel:
Thank you, Stephen, for this value framing. I think, it is most appropriate for me to start with Moderna’s mission. We set out nearly 10 years ago now with a mission to deliver on the mRNA science to create a new generation of medicines for patients. Our mission has been on compass for almost a decade, and we continue for the long term. On slide 25, I want to share our approach to delivering value during the COVID-19 pandemic. During this pandemic and from the development process of a mRNA-1273, our promise to deliver for patients has never been clearer. We have a responsibility to do everything we can to develop a safe and effective vaccine. We have invested in manufacturing at risk, ahead of approval to ensure supply, if our COVID-19 vaccine candidate is approved. We are working with governments around the world and others to ensure the vaccine is accessible regardless of ability to pay. And we will be responsible on price, well below value during the pandemic. Let me now turn to slide 26 and give you some more specifics. First, as we thought about responsible pricing, we concluded it is important to consider different time horizons. We are currently under a pandemic as defined by the WHO. At Moderna, like many public health experts, we believe that SARS-CoV-2 virus is not going away, and that there will be a need to vaccinate people or give them a boost for many years to come. So, we think about two time horizons, the pandemic period as defined by WHO, and the endemic period. During the pandemic period, we are priced well below value with pre-approval supply agreements mostly to governments. To-date, smaller volume agreements have been executed between $32 and $37 per dose. Larger volume agreements, under discussion, will be at a lower price for higher volumes. In endemic period, pricing considerations will follow traditional dynamics and market forces including vaccine efficacy and the competitive landscape. We will look to price in line with other innovative commercial vaccines. We expect traditional approaches to vaccine purchases and distribution in the endemic phase. With this, let me now turn to David for a review of our financials. David?
David Meline:
Sure. Thank you, Stéphane. Turning to slide number 28, in today’s press release, we reported our second quarter unaudited 2020 financial results. We ended Q2 2020 with cash and investments of $3.1 billion compared to $1.7 billion at the end of Q1. The increase is driven by the capital raise in May of this year. Net cash used in operating activities was $130 million for the first half of 2020 compared to $253 million in 2019. The decrease in 2020 is mainly due to deposits of $75 million received as of June 30th, for potential future supply of mRNA-1273. Net cash used in operating activities for the first half of 2019 also included $22 million of in-licensing payments to Cellscript to sublicense certain patent rights. Cash used for purchases of property and equipment was $25 million for the first half of 2020 compared to $18 million in 2019. Total revenue was $66 million for Q2 2020 compared to $13 million for the same period in 2019. Total revenue was $75 million in the first half of 2020 compared to $29 million for the same period in 2019. Total revenue increased for both, the three and six-month periods in 2020 due to increases in collaboration and grant revenue. The collaboration revenue increases were mainly attributable to an increase in revenue in the second quarter, particularly from AstraZeneca. The increases in grant revenue were primarily due to our BARDA agreement related to the development of our vaccine candidate mRNA-1273. Research and development expenses were $152 million for the second quarter of 2020 compared to $128 million for the same period in 2019. Research and development expenses were $267 million in the first half of 2020 compared to $258 million for the same period in 2019. The increases for both, three and six-month periods in 2020 were mainly due to increased headcount and mRNA-1273 clinical development activities. Overall, in both periods, we saw a significant increase in expenses for prophylactic vaccines modality as a result of our focus on mRNA-1273. While expenses related to the rest of the portfolio were stable. General and administrative expenses were $36 million for Q2 2020 compared to $29 million for the same period in 2019. Expenses were $61 million for the first half of 2020, compared to $56 million for the same period in 2019. The increases for both periods were mainly driven by increases in personnel and legal expenses. Turning to cash flow from net operating activity and purchase of property and equipment on slide 29. Cash used in operating activity and for purchase for property and equipment was $118 million, excluding the $75 million for deposits received as of June 30th for potential supply of mRNA-1273, in line with previous trends. On a reported basis, including the mentioned $75 million deposits, cash used in operating activities and purchase of PP&E were $43 million for the quarter. Turning now to updated guidance for 2020. Our guidance today maintains a metric of net cash used in operating activities and for purchase of property and equipment. We will adopt additional guidance metrics in the future as the business progresses towards commercialization. As always, we’ll be interested in any advice you may have on how we can be clear, concise and comprehensible. On an overall basis, the net cash used in operating activities and purchases of property and equipment in 2020 will increase to $0.65 billion to $0.85 billion from the prior guidance of approximately $0.5 billion for the year. Updated guidance for 2020 reflects ongoing investments in Moderna’s broad mRNA clinical and preclinical pipeline, as well as the significant activities associated with our efforts to advance our mRNA-1273 COVID vaccine toward approval and launch. With regard to the ongoing investment in our base business, we remain on track to prior plans where we continue to expect net cash used in operating activities and purchases of property and equipment to be approximately $0.5 billion in 2020. Turning now to the financial impacts of a rapidly advancing mRNA-1273 COVID vaccine. First, expenses that fall under the scope of our BARDA agreement. These are primarily research and development activities to drive the COVID vaccine to licensure and scale-up activities on the technical development and manufacturing side. As we expect a relatively close matching of expenses and reimbursement, we do not expect these activities to materially impact our cash flow and hence, these are not shown separately on slide 29. Next, looking at the COVID vaccine related non-reimbursable investments, primarily for manufacturing of product to be commercialized in the U.S. and internationally. We expect the net -- the cash impact of COVID-related investments to be between $0.55 billion to $0.75 billion in 2020. This includes approximately $0.2 billion in capital investments with the balance of the expenses related to raw materials and production activity in our network. Additionally, this investment includes initial commercial infrastructure build-up. The sum total of net cash used in operating activities for all of Moderna’s business is expected to total $1.05 billion to $1.25 billion before consideration of customer deposits. Also included in today’s guidance are the deposits we have received to-date for potential future supply of our COVID vaccine. Deposits as of July 31 total $0.4 billion, including $75 million received during the second quarter. We remain in active discussions with a number of potential customers, and we’ll update the status as the market evolves. In summary, on a net total company basis, we update our 2020 guidance for net cash used in operating activities to $0.65 billion to $0.85 billion for 2020. We expect this number to reduce as we receive further deposits. As we progress towards approval and commercialization of mRNA-1273, there’s heightened interest in several areas of accounting that will increasingly impact our reported results as we move forward. Slide 31 highlights several of these items. Costs associated with prelaunch inventory are expensed to R&D in the period incurred. We will capitalize our inventory when regulatory approval and subsequent commercialization is determined to be probable and we expect future economic benefits from sales to be realizable. Customer deposits for potential supply of mRNA-1273 are recorded as deferred revenue and will be recognized as revenue when revenue recognition criteria are met at a future date. Accounting for the BARDA grant follows a reimbursement model, where we will recognize revenue as we perform services and closely match expenses as they are incurred. For purchase and supply agreements, we disclose the aggregated amount of the purchase obligation that is fixed and determinable as per GAAP accounting requirements. This is regardless of whether these obligations have been recorded as actual costs in our financial statements in the current period. With that, I turn the call back to Stéphane for closing remarks.
Stéphane Bancel:
Thank you, David. In closing, let me share with you a snapshot of Moderna today. The Company continues to progress its clinical pipeline and got closer in Q2 to becoming a commercial company. We have a first Phase 3 program with our COVID-19 vaccine, but we’re also preparing the Phase 3 for CMV vaccine. We have three programs in Phase 2, CMV; personalized cancer vaccine, PCV; and VEGF. And we’re also preparing Phase 2 for Zika vaccine and OX40 ligand development candidates immuno-oncology. We have eight programs in Phase 1. We have 12 positive Phase 1 readouts across five different technology modalities. The products in development include seven vaccines for unmet medical needs, which if approved could be first-in-class vaccines. This includes our vaccines against COVID-19, CMV hMPV/PIV3 combo, RSV in older adults, and RSV pediatric population, Zika and EBV. We have five immuno-oncology programs in clinical studies. We have four rare disease programs in both, MMA, PA with open INDs. We have two autoimmune disease candidates, IL-2 and PD-L1 in preclinical studies. The foundation of Moderna has never been stronger. With over 2,000 healthy volunteers and participants enrolled in our study, as of the end of July and that number will grow considerably over the coming weeks as we continue to enroll 30,000-participant Phase 3 COVE study for our COVID-19 vaccine candidate. We have over 1,000 employees. We have a fully integrated GMP site in Massachusetts and a strong, experienced manufacturing partner in Lonza. Our biopharmaceutical partners are leaders in their respective fields and Merck, AZ and Vertex. And finally, we had a strong balance sheet at the end of June. We have the ability to invest $3.1 billion to create value. We have entered a new growth phase for Company, mRNA-1273 is now in Phase 3.We have received approximately $400 million of cash for customer deposits for potential supply of mRNA-1273 as of July 31st. And we continue to discuss with many governments around the world who are interested in getting access to mRNA-1273. The last six months have been extraordinary by many measures for our Company. The next six months could see us of our first product filed for BLA approval. We know that we have a special opportunity, and we’re committed to delivering on the promise of a science to bring forward a new class of medicines for patients. That is why we started this company, that is why we work hard every day. mRNA-1273 is ahead of its peers. We have a large pipeline with more than 20 development candidates. Since our founding, we have believed that it makes no sense that Moderna will be one medicine company. It will be zero if we fail to make mRNA science work or it will be many medicines if the first one gets to market, thanks to an FDA approval. That is because mRNA is an information molecule, which makes Moderna a unique biotech platform company. I would like to end our remarks by thanking the many people who participate in our clinical studies, including patients, healthy volunteers, physicians and nurses. I would like to thank our partners at the NIH and BARDA and our biopharmaceutical partners. I would like to say a special thank you to Moderna team. They have done a remarkable job in Q2 during a very complex time, scaling Moderna quickly while managing lockdown and many personal disruptions. I’m very proud of the team achievement in the past seven months of 2020. With that we are happy to take any questions. Operator?
Operator:
[Operator Instructions] I show our first question comes from the line of Matthew Harrison from Morgan Stanley. Please go ahead.
Matthew Harrison:
Great. Good morning. Thanks for taking my question. I guess, two for me this morning. First, on manufacturing for 1273, can you give us an update on how much inventory you’ve built so far? And where you are in terms of scaling to that 500 million dose per year run rate that you previously highlighted? And then, second, I was hoping you could comment on some media reports we’ve seen over the last day or two about FDA potentially convening a panel for vaccines in October. And I guess, also comments that there’s going to be real time review of vaccines, and what exactly that means, given the timelines potentially for your Phase 3 study?
Tal Zaks:
Matthew, this is Tal. Let me maybe take the second question first, and then I’ll defer to Stéphane to take the first one. Look, I can’t comment on media reports. I think that we have been working very closely with the agency, as I’m sure my colleagues have from other companies to make sure that we’re locked up and give them the greatest -- best visibility we can to our data as it emerges. We continue that dialogue as our Phase 3 is now enrolling. The Phase 3 trial has pretty clear, if you will, interim analyses and this statistical -- robust statistical plan that we have described in the past. But obviously, with the level of unmet need and more and more data emerging to substantiate a potential for benefit as we continue to accumulate safety data on the Phase 3, I’m very happy to see that FDA continues to take a very proactive stance in evaluating for the totality of the clinical data as it emerges. And, as soon as I have any more insights than that, I’ll be more than happy to share it.
Stéphane Bancel:
Thank you, Tal. And Matthew, on the first question around manufacturing, we will not share inventory numbers at this time. What I can tell you is that as we speak, our teams are making commercial products, receiving the potential approval of mRNA-1273. So, we are literally making products and stockpiling at risk. In terms of the capacity, I’ll confirm what we have said before, which is for 2021, the 500 million dose per year continues to be our base plan, meaning the team has a good sense of how to execute over that plan. And the team continues to work really hard over many different approaches, from new process, equipment to debottleneck, to find a path to 1 billion, which we still consider at this date as an upside. So, the base of 500, it’s your base case and the team is still working hard to figure out how we can get closer to a 1 billion capacity for ‘21 output.
Operator:
Our next question comes from the line of Ted Tenthoff from Piper Sandler.
Ted Tenthoff:
A question with regard to assumed infection rate for the Phase 3. Obviously, this is going to be somewhat dependent on sort of where you’re enrolling and things like that. So, what can you tell us about the general assumed infection rate and kind of how many infections you may need to see to be able to power the Phase 3 study of that size?
Tal Zaks:
It’s a great question. The challenge is that I don’t know how to build those assumptions, given the huge wide variability we see in actual infections rate. So, the number of infections you need to see, correlates to the number of cases required to pass the interim. And as we’ve disclosed, it’s roughly a little over 50 -- a little over 100 and 150 some for first and second interim and then the final. That’s sort of number of cases. And I think the advantage of running a 30,000-subject trial with 15,000 of them getting active vaccine and the rest as the placebo is that the larger that cohort, the more likely you are to see those cases early. But, it’s of course, a direct function of infection rates. I think, sort of if you step back, philosophically, we designed with our partners at NIH such a large trial with the initial goal, not knowing at the time what the infection rates were. You kind of just take a cross country average, you figure you’re going to be able to enrich it somewhat and you make it broad enough so that it’s representative of the diverse population. I think, what we’ve seen is an anticipated acceleration of the timeline to cases, not just by us. But I think by many people out there, recognizing that infection rates currently in the U.S. are actually higher, certainly in the areas in which we’re doing this trial than maybe we would have thought three, four months ago. So, I think the initial projections and people smarter than we have sort of made them was that we had expected to see the cases come in towards the end of the year, or by the end of the year. I think with infection rates, currently, there may be an acceleration of this. But, it’s extremely hard to predict. We, like everybody else, follow these on a daily and weekly moving average. And I don’t think, I’ve got a better crystal ball than anybody else in that domain.
Operator:
Our next question comes from Salveen Richter from Goldman Sachs. Please go ahead.
Salveen Richter:
So, with regard to the volume agreements that will be executed at a higher price point for smaller versus larger volume orders, how are you delineating between the smaller versus larger internal volumes? And then, secondly, if two different companies developing vaccines for SARS-CoV-2, both use, let’s just say the PRNT50 assays, are those assays identical? So, could you actually look at these programs head-to-head?
Stéphane Bancel:
Good morning, Salveen. So, I will take the first one and I will turn over to Tal or Stephen for second one. On the volume agreements, we’re disclosing very precise cutoff, because as you can appreciate, there’s a lot of different components that go into getting those agreements to the finish line with potential customers. I think, small is more kind of in 1 millions and large, as you can imagine for bigger countries will be very different ballpark. Stephen, do you want to -- or Tal talk about the PRNT50 assays?
Tal Zaks:
Yes. This is Tal. I’ll take a run at that. It’s a great question. I think, all of us are trying to look at them and understand the assays. There are big differences because those assays are not standardized and you’ve seen a range of PRNT50s, you’ve seen assays against live virus, pseudo virus neutralization. What’s clear is that all of these correlate with each other pretty well. But, there also are differences. And I think if you look across, that’s why everybody is trying to sort of report it out as a ratio to the convalescent plasma. The challenge there is, again, there is no standard panel for what the convalescent plasma is. So, everybody’s using different panels of sera. I think, the ballpark estimates are probably roughly comparative. My sense is sort of between half a log and a log of each other, for sure, maybe even tighter than that. But, it’s hard to drill down and get confidence that you’re really comparing apples to apples. And they’re certainly I don’t think identical. People try to set them up in identical ways, but you’re going to have minor variances. There’s also variability in the preclinical models reported. You can see different -- especially in the NHP, you can see people using different inoculums -- or inoculum levels different of severity of infections in the different models and then reporting out protection or not. So, unfortunately, we’re still relatively in the early days. I think, the good news is that you’ve seen for our data pretty robust titers in every which assay you measure as it relates to convalescent plasma. I’m happy for all of us that I think we’re probably not alone. There are going to be other vaccines that also seem to be getting close to that. And so, ultimately, we’ll all get wiser when we see the Phase 3 results over.
Operator:
Our next question comes from Michael Yee from Jefferies.
Michael Yee:
Two quick ones for me. First, you’re seeing -- or the market is seeing a lot of contracts being deployed both in UK, this morning, also another U.S. contract deployed to J&J. Could you just put into some context for us how we should think about Moderna’s position here and presumption of a diversified stockpile and maybe why or why not there hasn’t been anything to announce just yet? Maybe just make some broad comments about that. And then, the second question, I guess is, since you’ve reported data, there has been obviously a lot of other platforms with data that have come out, both adeno [ph] and protein last night. Maybe just make a comment about how to put that into perspective and the market’s enthusiasm or people’s enthusiasm about those data sets?
Stéphane Bancel:
Let me talk a bit about contracts and then I will hand over to Tal. So, as you know, Michael, to get to a contract, you need two parties to agree on terms. As I said in my remarks, we are discussing with governments around the world. As you know, we have a longstanding relationship with U.S. government with different agencies, BARDA, DARPA and NIH. The contracts that have been signed so far are contracts, so that actually literally around the world, in all the key regions. And so, as we get to the right place, of course, we will make the right announcement as appropriate. Our understanding is as has been said publically that different governments have different strategies. Some of them want to build a portfolio to manage risk, because, of course, at this stage, nobody knows which vaccine will get approved. Nobody knows the different efficacy, which will be most probably very important, especially for the population at risk, the elderly people with comorbidity factor. To them, the difference in efficacy might be very, very important. And so, I think what we’re seeing is the government’s doing what I think we would do or you would do, if you were running a country, trying to take care of the health of a large group of people, which is to build a portfolio, thinking about both risk of success, performance of a product and so covering the country. So, as you see, a lot of those agreements have options built into them, which I think again, makes a lot of sense as we sit. We want to be part of the solution and make sure that we can be helpful. As we’ve said, we are cautiously optimistic about the good clinical data so far of our vaccine. And we want to make sure we can help as many people as we can around the world, as many governments as we can to protect as many people as we can to kind of stop this pandemic. Tal, can I turn over to you for the second part of question from Michael?
Tal Zaks:
Yes. It’s a great question on how the platforms are starting to stack up to each other. I think, our data continues to be as good or better than anything anybody’s recorded, both in terms of what we’re able to show in neutralizing antibodies and in terms of the challenge we do in the non-human primate and the ability to completely eradicate or eliminate the power application of both the lung and the nose. It’s been nice to see Pfizer validating our data in a way with the BioNTech construct. Of course, we’re still very curious to see what they’re taking into Phase 2 and 3, because reportedly they’re taking a different construct than the ones for which they’ve shown data. I think the adeno vectors writ large -- it’s been an interesting story. Initially, it’s pretty clear that there’s less expectation for their ability to boost, given the nature of their platform. And, indeed, the first one -- I don’t think boosted at all. The AstraZeneca Oxford data I think are encouraging and that they can get with a boost apparently to levels of convalescent plasma. Hard to say, very early data, as I think even though they dosed more than 1,000 subjects, they showed data for 10, which they gave a prime boost. So, I’m encouraged by those data and I hope that that’s enough. I suspect that if that ends up being the case, then hopefully for all of us, you will see them sort of be able to reach the minimal bar. But, I’m encouraged by our data and expect that they will translate into -- I’m optimistic that they’ll translate into higher point estimates for efficacy. We saw Novavax data last night, I think any of us who’s been following the field of vaccines, anticipated that a recombinant protein on the right way with a strong adjuvant could be effective. I think, certainly, their neutralizing anybody data are encouraging. And ultimately, I think, the Phase 3 results are going to be required now to really understand fully the point estimate for efficacy as well as a better understanding of the reactogenicity and safety profile of these various approaches. So, in summary, I think the mRNA platforms are clearly there with our data looking great. I think, Novavax did a very nice progress. And I think, adeno vectors, I hope they get there. But, I think they’re going to struggle to boost. And I think, all -- anybody who’s done boost or is able to show a boost is able to show as immunology would anticipate, a very nice potentiation with the booster shot.
Operator:
Our next question comes from the line of Gena Wang from Barclays.
Gena Wang:
I have two questions. So, the first one is regarding the Phase 3 primary endpoint. Just wondering how do you count infection? I think, according to the clinical trials.gov, it’s basically number of participants with the first occurrence of COVID-19 starting 14 days after second dose. Just wondering, is there a time cutoff, so that every patient will have a same time exposure or will you report as event per 1,000 patent years? And the second question is regarding the Phase 1, Phase 2 data update, hopefully, this month or next month. So, will we see more meaningful number convalescent sera from severe patients using PRNT50 or PRNT80?
Tal Zaks:
So, let me take those. I’m not sure we’re going to see a significantly additional data on convalescent plasma. We’re going to look at that when we report out the Phase 2. So, I think it’s premature for me to say one way or another. Regarding your question on Phase 3, I’ll make two points. The primary analysis is just a number of cases. It’s not really -- it’s a slightly different method statistically, but you end up with the same place as I think what you’re referring to as the Pfizer methodology. You will see the number of cases and then the split in those cases, and of course, will describe the timeframe. In terms of counting those cases, you are correct in that the formal counting of cases where you want to see the distribution occurs 14 days after the booster shot, that’s the definition of the primary endpoint. There is, however, a secondary endpoint that will look at the totality of cases that emerge once people start the trial. And of course, I think that could be further supportive evidence, especially its infection rates are so high that indeed we start to see some cases even in the first six weeks. The case definition for us is symptomatic disease. Again, there may be some minor variations between sponsors. I’m not sure everybody’s fully harmonized to the same definition. We’ve been very transparent with the exact definition of our endpoints, so that people can draw their own conclusions and comparisons.
Operator:
Our next question comes from the line of Cory Kasimov from JP Morgan. Please go ahead.
Cory Kasimov:
Hey. Good morning, guys. Thank you for taking my questions. Two for me as well. First one is on pricing. I appreciate all the thought and detail that went into your prepared comments on the value proposition this morning. But, I guess what I’m wondering is if we see other companies out there currently agreeing to contracts with various governments with price points that appear to range from the low-single-digits to nearly $20 per dose. Doesn’t that make it inherently more difficult for you to try to charge something materially higher than that? And then, second question is probably for Tal. I’m wondering, how difficult is it to maintain the integrity of a Phase 3 blinded trial? When you would suspect a large proportion of the mRNA-1273 treated patients are expected to get common vaccination, adverse events like fever and other symptomatic -- other systemic events where presumably that wouldn’t happen with placebo. Does that matter much in the end in your view? Thanks.
Stéphane Bancel:
Good morning, Cory. So, it’s Stéphane. I’ll take the first one on pricing and then I’m sure Tal will take the P3 question. So, I think, like in any discussion, Cory, many factors go into arriving on up to an agreement. I would say, first, the totality of data that is available at the time of the discussions, preclinical model, clinical data. Again, given us, also we have a platform, we have the possibility to share public information that has been shared before over dataset. So, I think data is an important one, because I do not think all products are equal. And as Tal said, we would, of course learn much more with Phase 3 data in the fall. Another consideration of course is volume, as we indicated in our remarks. And then, another factor that is important is time of payment, how is the risk distributed between companies, because we discuss in our remarks, like us have started to make products at risk, not knowing if 1273 in our case will be approved or not, and so on -- and there are many more considerations as well. So, I think when you look at the totality of the data, as we’ve shared, we have signed a number of agreements already for potential supply in the $32 to $34 range. And we also said, as volume will increase, we’ll of course integrate that into the analysis. So, we’ll work with the market. But, we also know what we have in terms of product. Tal, do you want to take the P3 question?
Tal Zaks:
So, Cory, it’s astute observation. And we and our partners at NIH have thought about this one long and hard. I think, at the end of the day, you’re right, especially after the second dose. First dose, probably people still won’t know it. Second dose, people may intuit whether they’ve got the active one or not. I think, the important pieces here is that the endpoints are pretty hard endpoints and that and I don’t think that you’re going to be less likely to report significant symptoms, if you’ve got placebo or not and the measurement of a PCR test as a PCR test. We have put in place measures to ensure that there’s no sort of quirks of the unblinding. For example, we’re going to be testing by PCR everybody, both on this one and when they come in from the second dose just to make sure that if they do see some mild flu like symptoms, they are clearly attributable to vaccine or it is an infection and we’re not biasing by measuring more frequently in those that cut a vaccine. I think beyond the first couple of days, I don’t expect them to be significant symptoms in anybody. So, I don’t think there’s going to be biasing of any testing. In terms of the asymptomatics, we’re going to look retrospectively anyway and call that based on serology and everybody will get tested. So, I think by making sure that all the objective measures are done on both arms at the same time points, I think we should be in pretty good shape here.
Operator:
Our next question comes from Geoff Meacham from Bank of America. Please go ahead.
Geoff Meacham:
Hey, guys. Good morning. And thanks for the question. I just had a few. So, what we’ve learned on the value of 1273 is that T cell responses are important as our antibody titers. And so, when you look at the new data today for Zika or for CMV, or really anything going forward, do you think you’ll place more emphasis on optimizing T cell responses just with respect to the platform? And then, just a follow-up on 1273 pricing. When you look at the Phase 3 data, is there a single element that you would say justifies differential pricing, be it T cell or B cell or safety profile, things like that?
Tal Zaks:
So, let me take, Geoff, the first question and then, I’ll let Stéphane answer the second. I think, a lot of hay has been made out of T cell responses. Look, T cells are near and dear to my heart, being a medical oncologist. They’re required to cure cancer. I don’t -- I’m not quite sure, they’re as important to cure COVID-19 frankly. I think, what they are is a measure of the quality of the response. So, if you will, the fact that we see the right kind of Th1 helper, I think makes everybody feel good about the antibody responses. But at the end of the day, the best measure of the immune response you’re getting is look at the ability to boost after a prime. That tells you that the immune -- even after dose, once you get the boost, you can see the antibody levels come up quickly. They come up to high levels, they come up with good neutralizing activity, which is a best predictor that should you get infected, the immune system will trigger again in a similar manner. And that’s how the immune system works. I think we’ve had plenty of data from other infectious diseases demonstrating that whenever you try to find a correlative protection, and Merck has done that with y Zostavax years ago, and that’s even a disease where you think T cells matter more, the correlative protection comes up time and again being neutralizing antibody, not T cells. T cells are an adjunct sort of measure of the quality of the response when it comes to these kinds of viruses. And I think, the totality of the data that you’ve seen with SARS-CoV-2 in terms of all the animal models, you can passively transfer antibodies and get protection. So, I think that there’s been a lot of variability on the T cell side in terms of people recording assays, and I think people are trying to read into T leg [ph] and assume something about the quality of the response. But to me, that’s a more distal measure of what matters when you’ve got the most obvious proximal measure of what we think is the correlate and the thing that will translate to benefits, which is thriving antibodies. So, it’s a bit of a long winded way of saying that I don’t think it matters as much. And certainly, when you look at optimizing our platform, look, we know our platform is optimized to generate T cell responses. We’ve seen that in the cancer T cell space, where we just do -- all we do is raise CD8 positive T cells by putting in concatenated CD8 positive epitopes. We get some CD4s. But, that is an innate function scientifically of the ability to translate the antigen from within the cell, within mRNA. So, I don’t think, it would be fruitful to try and optimize to T cells. I don’t know that it matters and I wouldn’t know how to do it. And by the way, we already have something that seems to do that based on the fundamental science of what our platform is doing. Let me stop here and transfer to Stéphane.
Stéphane Bancel:
So, on the Phase 3 pricing. So, safety of course is really important, but I would say, it’s important for us, the sponsor, for regulators obviously. And that would be kind of an important consideration to look at the totality of the data, especially for approval of a product. So, assuming a good safety profile, we believe that one important parameter, of course, is efficacy. Because again, I think efficacy has different dimension as efficacy that is important to reach the immunity at the population level. But also, efficacy, if you look at it in the eyes of whoever received the vaccine. Because you can appreciate the vaccine with 50% efficacy or vaccine with 90% efficacy will be very different, what it means to you potentially just statistically, in terms of you getting protection or not. And then, I would say, the subpopulation. I mean, of course, healthy adult is what has been mostly reported so far. It’s a very important population, obviously, that we all look forward to looking at the data in the elderly. As we all know, as Stephen described, again, during the remarks, the elderly are very vulnerable to the virus. And so, understanding antibody titer, understanding efficacy in the elderly, we believe is going to be important. As for diversity, we talk about ethnic groups. It has been widely reported that some ethnic groups are impacted very differently by the virus, and so, getting an understanding of that through the study. As you know, we have paid a lot of attention and we talked about it, it was actually in one of our call about the effort that the team is going through to ensure a good representation across ethnic groups. And then, I would say, comorbidity is also very important, again, because of who is being held the most by this virus, to understand is there a difference or different comorbidity groups for different vaccines. So, as you can see, there’s going to be a lot of pieces of the efficacy picture. Again, some public payers in some countries might be willing or not to pay for different efficacy for different efficacy group. The private market might have a very-different inclination. And in places where it’s out of pocket, you will have actually people having also making very different type of decisions. The other dimension of course is duration of protection, which obviously will take time for all the vaccine manufacturers to really understand at large scale in the clinical setting. Because you heard us say, we believe as many of the health experts believe virus is not going away. We believe there’s going to be an endemic market once the pandemic is declared over by the WHO. Duration would be an important factor. As you can appreciate, if vaccine provides one year production, versus six months, versus two years, versus three years, that will all play into the equation. So, those are some of the factors that we will carefully think about, again, both in the pandemic setting where we said, we will make sure that we price a product at a big discount to value, and in the endemic setting, when we look at all this different product performance, and market forces to determine what we believe is the most optimal price for the product.
Operator:
Our next question comes from Hartaj Singh from Oppenheimer. Please go ahead.
Hartaj Singh:
Just a couple quick questions, off the topic of the COVID-19 vaccine. One is, can you just go over quickly, the CMV Phase 3 program, the initiation, the development? And when you’d expect that to readout and potential approval for that product? And then, secondly, in some of our calls with pulmonologists that treat cystic fibrosis, there seems to be a lot of excitement around mRNA therapy for treating cystic fibrosis patients. The belief right now is that it could only treat a minority of those patients. But, our calls seem to suggest that it could treat a majority of the cystic fibrosis patients. Could you talk a little bit to that and where exactly are you with your Vertex collaboration?
Tal Zaks:
So, let me start with the Phase 3. I’ll let Stéphane talk about the Vertex collaboration. The Phase 3, as we had previously articulated, I don’t think anything has changed. The trial should follow participants for at least a couple of years. It’ll probably take around 18 months to enroll. And then, you wrap up the data. So, that sort of gives you a sense of the overall duration. We’re on track to start next year. I think, what should be obvious to everybody is, once we have the Phase 2 data, it’s going to require some regulatory interactions and the Phase 2 meeting and so forth. We had gotten feedback in the past from FDA about the primary endpoints, which I think reassured us that it was feasible because we’re looking to prevent primary infection and women of childbearing age not directly looking at outcomes in babies, at least not as part of the Phase 3 program. That being said, obviously, it will require much more detailed discussions with them on the Phase 3 design and the dose and alignment with them prior to start of the Phase 3. So, I hope that that gives you some additional color. I can tell you my sense on Vertex. As a scientist, I’m super excited by the potential for mRNA to -- not be limited by any particular mutation, just from the fundamental science of it. But, I’ll let Stéphane speak to the overall status of the collaboration.
Stéphane Bancel:
I’ll let Stephen talk about it. He and his team is doing all the work.
Tal Zaks:
I think Stephen had to jump off our call.
Stéphane Bancel:
Sorry. Thank you. So, on Vertex, so as we communicated, I think it was in the Q1 call, Vertex decided to expand the collaboration with Moderna on CF. The joint teams’ discovery have done a really remarkable job in the last few years in term of delivery. As you can imagine, given as well, making the CFTR mRNA is another complicated, given all the things we have done in the current state of the platform and the technology. It is obviously, the challenge entirely in delivery. We look forward to sharing some data soon. Again, it’s a partnership. So, we need to of course align with Vertex on what we believe is the right timing. But, as Tal said, the teams on both sides, I think Vertex and Moderna are very excited about the progress, about the possibility what it will mean for patients. And as we discussed before, if we can find a technology to deliver safely into the lungs and mRNA, obviously, we will be able to use that for obviously disease of lung. And just to remind everybody, the Vertex collaboration is focused on the CFTR gene. And so, the other applications will belong to Moderna from a commercial standpoint.
Operator:
Our next question comes from Alan Carr from Needham & Company. Please go ahead.
Alan Carr:
Just following on the theme of programs other than COVID-19. You mentioned before that enrollment in a few of these programs is still paused or suspended, because of COVID-19. I’m just kind of wondering, if you’re able to still get prioritized and move forward some of the other programs, such as the rare disease programs, do you feel like you can still keep your eye on the ball in terms of the rest of the non-COVID pipeline at Moderna?
Tal Zaks:
So, this is Tal. Let me maybe take a stab at that. I think, the answer is absolutely, yes. And I think, it’s evidenced by the progress that we continue to show for the rest of our pipeline. We’ve expanded the development team quite significantly to go after the COVID-19 vaccine, so that, I think we’ve got very capable teams that are pushing everything else. The fact that we continue to enroll in oncology, it’s seen a little bit of a slowdown, similar to others. And it really depends on the individual center’s data, so who’s enabled and when to treat patient. I think, on the rare disease, not only is our eye on the ball, I think, we’ve -- as we have alluded to in the past, we’re actually using the time to kind of step back and engage deeper with both, investigators and patients and advocates to make sure that when we do restart those programs, because the kids can finally come back in, those programs are better optimized, so that they can -- we can perform better in terms of recruitment and analysis of data on the study. So, we’re continuing to execute across all the fronts. And I think, you’ll see as hopefully the pandemic recedes or as centers figure out how to, despite PPE and social distancing, to continue or restart clinical activities, then, we should come back on line across the board.
Stéphane Bancel:
Yes. And we just -- to add to Tal’s important comments. When things started to look bad from a global basis in terms of the spread of SARS-CoV-2, we spent quite a long time, I would say in the February timeframe, with the Board and with a team to think about how do we structure the Company, how do we resource the Company, to Tal’s point, so best we can do both. It was very clear that a lot of focus and energy was going to be required to march at a very fast pace, without compromise to safety for 1273. But, at the same time, Moderna has always been a platform company, it has always been about managing risk through very diverse pipeline across different therapeutic areas, across different modalities or technology, for those on the call that are not used to Moderna. And so, it was really important for us, given the inherent risk of developing a product at 1273. Again, we feel much better now, given the data we have seen, but I’m looking back in February. And so, we put a quite a large number of very experienced development professionals in clinical, regulatory, clinical operations, to be able to actually do both. That was one of our big challenges. And I have to say, the team has really done a remarkable job to both, push 1273 as safe as we can, as hard as we can, because that every day those matter, and at the same time, continue to execute on the pipeline. It has been very challenging, given the lockdown or given our focus on safety. Some of the studies were easier to keep rolling, some of the studies, because involving children, we wanted to make sure especially children with very severe disease that we didn’t take the risk to expose them to infection by going to clinical trial sites or their caregiver, their family. And so, we have not laid down an inch on the focus on the pipeline outside of COVID.
Operator:
Our next question comes from George Farmer from BMO Capital Markets. Please go ahead.
George Farmer:
I’m quite struck by the fact that neutralizing antibody titers don’t really seem to emerge until after the second dose, whereas antibodies in general seem to be elicited after the first injection. How do you think about this? I mean, it seems to be across the board, not just with your vaccine, but with these other vaccines. And, importantly, how would such patients be treated in the Phase 3? Should they come down with COVD, say after the first injection because of not having a sufficient titer going into the second injection?
Tal Zaks:
Thanks, George. It’s Tal. Yes, it is interesting. I think, it kind of speaks to basic immunology, which is, if your immune system thinks it clear an infection easy, it doesn’t try too hard. If you come back later and basically show that no, the infection isn’t gone, then it tries harder. And that usually manifests with affinity maturation improvements in the antibody qualities as well as the absolute titers. So, I suspect what you’re seeing is a subtle manifestation of that. Now, the question is though, what happens after just the prime? Yes, you don’t have neutralizing antibodies, but clearly the immune system now reacts differently, once it sees the antigen because you get the boost. And so, it’s an interesting question. Well, what happens if the boost happens not with the vaccine, but with the natural infection? I would suspect, based on sort of first principles that you should see some benefit, you probably -- not the full benefit, you would see if you get infected after the boost, but just by nature of the boosting, you would expect that an infection would also boost in the sense of the immune response would react quicker, more vigorously. And maybe you won’t prevent infection and maybe you won’t even prevent disease. But hopefully, you would prevent a more severe manifestation of disease. Because disease, at least as we understand it in the first phase here is a balance between your immune response and clearing the infection. So, it’ll be very interesting, I think, both for us and for other platforms that use a prime boost to do the secondary analysis of understanding whether we’ve seen any cases in that first month or six weeks and what the distribution of cases are between placebo, and you’re treated. The last piece I’d say that I think also gives me some hope here is the non-human primate where a single dose in the non-human primate can -- or at least the mouse models. The experience with the non-human primate is ongoing. But, in the in the mouse models, we clearly see that even though you don’t see -- you don’t measure the neutralizing antibodies just yet, you are already able to limit viral or abrogate viral replication in the lungs. And so, I think that gives you a sort of immunological readout on the phenomenon describing.
George Farmer:
And then, along those lines, do we have a view on durability yet, following these -- following both, the nonhuman primates and maybe the mice, if they live long enough. Do you have a sense for how durable the vaccine effect could be?
Tal Zaks:
Yes. I do not -- I would make two points on that. First, I don’t think any of us yet understand durability. What is emerging is that if you have mild infection, you’re more likely to have lower levels of antibodies and they’re more likely to wane. That kind of goes with the quality of the immune responses, as I described previously. It’s also -- I mean, let’s not mistake the ability to measure antibodies over time as the Sine qua non of having a memory in an immune response, right? Otherwise, by the time you were my age, you’d be walking around with your blood thick and clotting with antibodies against every infection you’ve ever seen. And yes, I am immune to many things that you are not going to necessarily see high levels of neutralizing antibodies in my blood. It’s more about the memory B cells in the persistence. So, I think that initial level of neutralizing antibodies are what gives us the confidence that we’ve got the right quality of the response and the magnitude is at higher -- higher than what you can see with disease with real infection and convalescent plasma. Now, all that being said, I think it’s obviously reassuring to see the quality of a response by maintaining high neutralizing antibodies. I think, if you look at what the platform is able to induce, it’s pretty evident from the CMV data described earlier this morning that certainly in that instance, with a third boost at six months, look out to a year and you see levels that are still higher than what you had targeted. I think, as it all boils down to the utilization in the context of a pandemic here, if what we have is 12 months of durability, I think that’s a great starting point for us to start protecting the population. And we’ll worry about what happens in year two, three and four when we get to 2022, 2023 on the other side of this pandemic. Over.
Operator:
Our last question comes from Mani Foroohar from SVB Leerink.
Mani Foroohar:
Couple for me, one quick one from an investor. Could you lay out the economics you guys receive on a commercialized vaccine? How to think about percentages paid out to academic partners and the NIH commercial on economic interest et cetera, just breaking it out, trying to understand what unit margin might be? And then secondarily, as we look at pricing, it’s been commented by co-people here, a couple of analysts regarding pricing in the low single digits to upto just below $20 for example, in larger volume contract. It sounds like we should think as somewhere in that range as par for larger contracts for you guys during the pandemic. But, as you talk about the endemic environment and potentially higher pricing, could you lay out how you expect to realize that pricing and setup where demand will likely decline as the severity of COVID-19 impact globally is reduced during an endemic versus a pandemic period, in a setting where perhaps a half dozen or more vaccines being commercialized by large established players with commercial experience in tender markets such as these will be actively competing on price and volume. So, if you could lay out how you expect to substantially increase your realized price in a setting of increased competition, increased supply and reduced demand? That would be really helpful.
Stéphane Bancel:
Thank you. Good morning. This is Stéphane. So, this first one is going to be quick because we are not disclosing unit margin per product. On the endemic market, clearly, as we said in our remarks, the competitive dynamics are going to be important. We continue to believe that when you look at the totality of the data, not only across 1273 but across 1273 today that’s public and around the platform that efficacy is going to be an important parameter. We’ve hired people and we continue to have people in the commercial world that comes from large established vaccine players who have relationship with government, who have relationship with people in the channel. And so, we believe that we should be able to establish good commercial programs based on the performance of the product.
Mani Foroohar:
Great. And as a follow-up, when you -- you’ve talked a couple of things about the CD8 T cell response, T cell biology. Just to clear up, the comment that you -- that across your platform you’ve shown effective T cell response that was directed regarding construct in your cancer portfolio, right, and not at this particular vaccine where we haven’t seen a CD8 T cell response of any measurable level as described in your nonhuman primate or human disclosures thus far. You were talking about the cancer vaccines, right? You weren’t talking about any prophylactic vaccines, so in your CD8 T cell response here?
Tal Zaks:
So, this is Tal. That is correct. Although I have to go back because we did disclose some T cells for CMV. I don’t recall, either they were CD8 or just CD4, frankly, off the top of my head. Yes.
Operator:
I show no further questions in the queue. At this time, I’d like to turn the call over to Stéphane Bancel, CEO, for closing remarks.
Stéphane Bancel:
Thank you very much, everybody, for joining today. Stay safe, and we’ll speak soon.
Operator:
Thank you. Ladies and gentlemen, this concludes today’s conference call. Thank you for participating. You may now disconnect.
Operator:
Good morning, and welcome to Moderna’s First Quarter 2020 Conference Call. At this time, all participants are in a listen-only mode. [Operator Instructions] Please be advised that the call is being recorded. At this time, I would like to turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, everyone. Welcome to Moderna’s conference call to discuss our first quarter 2020 business updates and financial results. You can access the press release issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our website. Speaking on today’s call are Stéphane Bancel, our CEO; Tal Zaks, our CMO; Stephen Hoge, our President; and Lorence Kim, our CFO. Before we begin, please note that this conference call will include forward-looking statements. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. With that, I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina, and good morning or good afternoon, everyone. Thank you for joining the call. I hope you and your families are in good health. As you know, we believe mRNA has a potential to be a new class of medicines with the opportunity to address many unmet medical needs, with medicines with higher probability of technical success, with greater speed of research and clinical development versus traditional medicines and with better manufacturing capital efficiency and lower cost of goods than injectable recombinants. Given the amounts of working with new technology, we have been laser-focused on managing risk
Tal Zaks:
Thank you, Stéphane, and good morning, everyone. I’ll start with a quick reminder on the data generated to date with our vaccines. In over 1,500 healthy volunteers and 7 positive Phase 1 datasets to date, we’ve observed the safety profile that’s consistent with the safety of adjuvanted vaccines. And we’ve time and again demonstrated the ability to elicit an immune response in the form of neutralizing antibodies. I’ll start with a high-level progress on mRNA-1273, our vaccines against SARS CoV-2, and will give more detail shortly. As you heard from Stéphane earlier, we have the FDA clearance to move into our Phase II study, and we plan to start it shortly. This study will run in parallel with the NIH-run Phase I study, which has completed enrollment of the first three dose cohorts. Our CMV Phase II dose confirmation study is fully enrolled, and we still expect data readout to come in the third quarter of this year, despite having had some COVID-19-related disruptions. At our Vaccines Day on April 14, we announced positive interim analysis of our Phase I study for our Zika vaccine. At the two lower doses of 10 microgram and 30 microgram, we achieved seroconversion rates of 94% and 100%, respectively. The two higher dose cohorts of 100 microgram and 260 microgram are now fully enrolled. As a reminder, we paused our hMPV/PIV3 Phase Ib study enrollment as a cautionary measure to protect children and their caregivers due to COVID-19 disruptions. Our RSV program with Merck continues. So this has been covered in detail, but just to quickly pace everybody on the same place, our SARS-CoV-2 vaccine, mRNA-1273, which was a subject of much work and discussion in the first quarter of this year, demonstrates the kind of speed that we believe the platform can provide, from first selection of a sequence by our scientists and our collaborators at NIAID on January 13 to the production of a clinical batch on February 7, 25 days later. That had been released by February 24, and by March 4, was associated with an open IND that NIAID had filed. The strong collaboration between us and NIAID led to this – the trial opening within 63 days, and we’ve spoken about this before. On April 17, we were awarded a contract from the U.S. government agency, BARDA, to accelerate the development. And on April 27, we announced an IND was submitted to the U.S. FDA for the Phase II study. Last Friday, we announced a collaboration with Lonza to manufacture mRNA-1273 at scale, with the goal of producing up to 1 billion doses a year. And of course, today, we announced the FDA clearance to start the Phase II part. In parallel, we have been working on the Phase III protocol, and we are finalizing that with an aim to start the study in the summer of 2020. The design of the Phase I study is on Slide 17. The study started as a 45-subject trial with three dose cohorts, 25, 100 and 250 microgram, with each participant receiving two vaccinations a month apart. Phase III dose cohorts have now been fully enrolled, and the safety and immunogenicity data from them will be shared when available. The NIH is expanding the trial to include two additional age cohorts, a 56-to 70-year-old cohort and a 71-and-above age cohort. Each of these age cohorts will include three dose levels also at 25, 100 and 250 microgram at the same vaccination schedule. In terms of the late phase development for mRNA-1273, as mentioned before, the Phase II study is expected to start shortly. This study will evaluate the safety, reactogenicity and immunogenicity of two vaccinations of mRNA-1273 given one-month apart. Volunteers will receive either placebo, 50 or 250 micrograms at both vaccinations. This study will enroll 600 healthy participants and two cohorts of adults, ages 18 to 55 and 55 year old and above. The study is meant to both increase our safety database as well as confirm the immunogenicity seen in the phase that we expect to see in the Phase I. We are finalizing, as I said, the Phase III protocol, and the study is expected to begin this summer. Last week, Moderna and Lonza announced a strategic collaboration with the goal to enable manufacturing of up to 1 billion doses a year, and this is assuming a dose of 50 micrograms. Technology transfer is expected to begin this June, and we anticipate the first batches of mRNA-1273 to be manufactured at Lonza’s U.S. sites in July of this year. I would be remiss not to mention BARDA’s role in this. The BARDA award is allowing for us to move as quickly as we are with scale-up, both internally and with Lonza. Moving on to CMV. Slide 20 reviews our late-stage development plans for CMV. As previously announced, the Phase II dose confirmation study is fully enrolled, and we remain on track for data readout in the third quarter of 2020. Importantly, greater than 70% of participants have now received their second vaccine dose. A protocol amendment was submitted to expand the time frame for the remaining participants to receive their second dose as well. As a reminder, we plan to select a dose for the Phase III after the first interim analysis, which is the data post the second vaccination. We continue to prepare for the Phase III, which is intended to start in 2021 in the U.S. and Europe. During the first quarter of 2020, we also received constructive feedback from the Type C CMC meeting that we’ve had with the FDA. Moving on to mRNA-1893, our Zika vaccine program. Let me recap on Slide 24 the data that we recently presented at our Vaccines Day, where we reported an interim analysis of the ongoing Phase I trial. This study has demonstrated a fairly benign safety profile, consistent with what we’ve seen before for other vaccines. And at the two lower doses of 10 and 30 micrograms after a two dose vaccination regimen, prime and boost, the seroconversion rates were 94% and 100%, respectively. These data are encouraging, and we are preparing to move forward with this program into a Phase II trial. The exploratory modalities are a critical part of our strategy, and we continue to make up a significant part of what we do in the clinic. And on Slide 26, you see a – if you scan the page, you’ll see many readouts and catalysts from each of the programs, both from our core modalities as well as the exploratory ones. With that, let me now turn the call over to Lorence.
Lorence Kim:
Thank you, Tal. Let me first cover an update on the Vertex agreement. In July 2016, we entered into a strategic collaboration and license agreement with Vertex in that discovery and development of potential mRNA medicines for the treatment of cystic fibrosis or CF by enabling cells in the lungs that people see us to potentially produce functional CFTR proteins. In July of 2019, the initial research term was extended by six months. And based upon promising preclinical data generated, in March of 2020, we were pleased that Vertex elected to extend this collaboration for a further 18 months. Now let me turn to financial results. In today’s press release, we reported our first quarter 2020 financial results. Note that these results are unaudited. We raised approximately $550 million in net proceeds from the February public equity offering, which resulted in us ending Q1 2020 with cash, cash equivalents and investments of $1.72 billion. This compares to $1.26 billion at the end of 2019. Net cash used in operating activities was $106 million for the first quarter of 2020 compared to $144 million in 2019. And just as a reminder, that latter number includes an in-licensing payment of $22 million, which will not recur. Cash used for purchases of property and equipment was $6 million for the first quarter of 2020 compared to $8 million in 2019. Revenue for the first quarter of 2020 was $8 million compared to $16 million in 2019. This decrease of $8 million in revenue was mainly due to cumulative catch-up adjustments resulting from changes in our estimated costs for our future performance obligations, coupled with the timing of amortization of deferred revenue due to the satisfaction of our performance obligations. R&D expenses for the first quarter of 2020 were $115 million compared to $130 million in 2019. The decrease of $15 million in R&D was mainly driven by a decrease in lab supplies and materials and clinical trial and manufacturing costs, partially offset by personnel-related costs. G&A expenses for the first quarter of 2020 were $24 million compared to $27 million in Q1 2019. This decrease of $3 million was primarily attributable to decreases in legal and other consulting and outside services spend. And net loss for Q1 2020 was $124 million compared to $133 million in Q1 2019. I’ll turn now to what we expect for the remainder of 2020. If you look at our cash flow line items, you can see our cash used in operating activities and purchases of property and equipment by quarter are laid out here. In Q1 of 2020, we used $112 million of cash on these two items, which is in line with our expectations. If you go back to Q1 2019, we used $152 million of cash on these two items. And remember again, that number included that licensing payment. Overall, you can see the decline in our quarter-over-quarter cash used for these items through Q4 2019 with a slight uptick in Q1 2020. And so consistent with our initial 2020 guidance, which we issued back in November, we expect our 2020 net cash used in operating activities and purchases of property and equipment to be approximately $500 million. While we have seen parts of our spend slow down as a result of the impact of COVID-19, such as certain clinical trial expenses and laboratory supplies, we are also investing in preparedness for the late-stage development and potential BLA filing for our COVID vaccine. That results in bringing our cash flow guidance back to its original levels. We recognize that much of our COVID vaccine spend is covered by the BARDA award. Note that the award is not cash upfront, but rather reimbursement as expenses are incurred. We do expect to incur significant expenses this year in relation to that BARDA award, but we expect in general a matching of expenses and reimbursements. Let’s go down next on our balance sheet strength and the composition of the $2.4 billion of cash and available funding we have to invest and create value. We ended Q1 2020 with cash, cash equivalents and investments of $1.72 billion. On April 16, 2020, we entered into an agreement with BARDA to accelerate development of our mRNA vaccine candidate against the novel coronavirus for funding of up to $483 million, of which $430 million has been committed. Additionally, we are fortunate to have established strategic alliances with private-and government-sponsored organizations, including the Bill and Melinda Gates Foundation, DARPA and another BARDA award comprising additional available funding of $180 million. Together, this creates multiple years of cash runway, considering the cash guidance that we shared today, and a strong ability to invest for the long run in many aspects of the business. The next slide shows our pipeline and the programs through the various phases of development with a snapshot here. But before I turn it back to Stéphane, let me just reiterate an important point from my announced departure this morning, which is that I expect to seamlessly transition my responsibilities through August. But I’ll make a brief remark now. First of all, I’m so grateful to have been invited to be a part of this company, what an opportunity to contribute to Moderna’s mission of turning mRNA into a new class of medicines. I joined the company six years ago, at a time when this story was nascent and the future was full of unknowns. I’m leaving now as the company has multiple BLAs on the horizon with 23 important new potential medicines in the pipeline and I believe many more to come. We’ve invested heavily in the platform to establish the scientific foundations of this new class of medicines, and the team is growing with unbelievable new talent. I’m personally most proud of the financial foundation we’ve built to enable the company to invest appropriately in the business. It’s been energizing and motivating to partner with Stéphane, the Board, its executive team and the passionate Moderna employee base. For me, I’m going to take the next step in my career, which will be to stay close to innovation in biotech, but not as a company executive. I look forward to sharing more about these plans at an appropriate time down the road with many of you on this call. And with that, I’ll turn it over to Stéphane for closing remarks.
Stéphane Bancel:
Lorence, thanks again for your remarks. But having you as my partner for those six years has been quite a incredible ride. On Slide 34, let me close by giving you a quick update where the company stands today, starting with our pipeline. It’s exciting to see that today, we have two candidates for which we are preparing for Phase III, our CMV vaccine and our SARS-CoV-2 vaccine. We have now six candidates that are either in Phase II or preparing for Phase II. 12 Phase I programs and 11 positive Phase I result. What an acceleration since our IPO in December 2018. Our programs are very exciting. We have seven first-in-class vaccines, where there are no approved vaccines on the market against those viruses. Most of these vaccine candidates have multibillion dollar annual peak sales opportunity. As we shared at our Vaccine Day presentation, we believe our innovative vaccines are going to be a very large business for Moderna with long-term annuity life opportunities at a high EBIT margin. We also have five exciting immuno-oncology programs that are already in the clinic, that are all combined with a commercial checkpoint, four rare disease program and two autoimmune disease programs. The company has never been stronger and look at an formations. We have now dosed more than 1,900 healthy volunteers and patients in our studies. The team is strong and getting stronger every month. We now have more than 900 employees who care deeply about our mission and are proud and energized by our progress and the meaning of our work. Last week, with the Lonza agreement, our manufacturing capabilities has changed league. We not only have a fully integrated GMP site in Massachusetts, who many of you know and have visited, but we’ve added a strategic partnership with Lonza that can enable us to up to 1 billion doses annually for vaccine against SARS-CoV-2, but also other products in our pipeline as we need to. We have great partners with AstraZeneca, Merck and Vertex. I am very proud of the scientific progress that the team of Stephen Hoge and Melisa Moore have done in the work with Vertex over the last few years. And we are very pleased that Vertex decided to expand the relationship with Moderna. But we’re also very thankful for a partnership we’ve had over the years with BARDA, DARPA, CP and the Gates Foundation. And of course, we are very thankful for the latest partnership with BARDA, $483 million, to enable us to do the right clinical study as fast as we go, of course, focusing on safety first for the SARS-CoV-2 vaccine. And of course, we are well capitalized with up to $2.4 billion to invest in the business and continue to build the leading mRNA company in the world. We are very thankful for our investors for their trust and partnership as we build this unique company. We are energized by the opportunity ahead of us to build a new class of medicines. We are currently accelerating our development pipeline and readying the company to potentially file its first BLA for mRNA-1273, which will be, as you can appreciate, a historic moment for the company. We’re investing in the processes to get us there, to get the right foundations for potentially many additional BLAs in the future, starting with the Zika vaccine and CMV vaccines behind the SARS-CoV-2 mRNA-1273. We are already scaling up the organization to address the need to supply up to 2 billion doses for potentially first vaccine mRNA-1273. All those efforts, investment and processes will be very enabling for additional vaccines and therapeutics to come. I have never been as excited and optimistic about the future of Moderna in the last nine years. We are humbled and excited by the opportunity to bring forward a new class of medicines for patients. That has been our North Star since we started the company. I would like to thank the great team of Moderna employees working very hard every day, and literally, many of them 7 days a week now since January to fight the SARS-CoV-2 virus. I would like to thank the many people who participate in our clinical studies, including patients, healthy volunteers, physicians and nurses. I’d like to recognize all our partners that have worked with us to share our vision and helping us to achieve this vision to help patients. With that, we are now happy to take any questions. Operator?
Operator:
[Operator Instructions] Your first question comes from Matthew Harrison of Morgan Stanley. Please ask your question.
Matthew Harrison:
Great. I guess, first, you’ve highlighted the 50-microgram dose on the SARS-CoV-2 vaccine. And I think Dr. Fauci in his interview also talked about good responses in low doses in animals. Can you just talk about your confidence in being able to move forward and elicit the right kind of immune response with the low dose as opposed to the other doses that you’re testing? And then secondly, can you just update us on where the field is and figuring out what neutralizing antibody titers are and if you think they’ll be available by the time you report initial data from the Phase I study? Thanks.
Tal Zaks:
This is Tal. Let me take that question. Two spot-on things that we’re looking at. So first of all, 50 microgram is our current best guess. We – this is short of data and it’s based, as you’ve seen Tony Fauci’s remark, on what we expect the platform could deliver. That being said, the final dose selection will really be a factor of, I think, three elements. The first is the overall sense of a dose response of how much more do you get as you go up in dose. Because in the case of a pandemic, we obviously need to balance this with having enough doses available. And so you don’t want to unnecessarily overshoot. The second element is an understanding of what that dose could mean as you compare to convalescent serum. And so there’s a lot of work being done on assay validation, and I’ll get back to that in a minute, to understand what any given level of antibodies mean. And the last element that I think will enable us to connect the dots is understanding the performance of the vaccine in additional animal models of SARS-CoV-2 and then seeing, commensurate with the expected ability to protect those animals, what levels of titers do we get. And obviously, the higher the species the more reliable that data is. But ultimately, what we care about is being able to connect the dots for human disease. So it’s a – long story short, it’s a best guess estimate for now and based on the emerging data, and we will continue to refine it as more data comes in. Your question about the right kind of immune response. Look, I think the data we’ve seen to date, both across the clinical trials, across the experience that we have in the preclinical models, across the board, as I mentioned, in all the other clinical trials, we’ve routinely reported neutralizing antibodies as the measure of immunological success. And if you think about the kind of scientific-first principles of of scientific-first principles of how an mRNA technology presents an antigen from within the cell and mimics the instruction set that a virus would otherwise give the cell to make an antigen, we get the right kind of immune response, however we want to characterize it, neutralizing antibody in Th1 versus Th2, et cetera. The emerging data that we’re seeing preclinically with mRNA-1273 is all consistent with that. Your final question, on neutralizing antibodies, yes, those assays are being stood up as we speak. They’re being validated. They’re being transferred to commercial vendors. And the NIAID is actively looking at in parallel with the simpler types of binding antibodies we expect to be able to report, both kinds of data when we see the data from that Phase I.
Operator:
Your next question comes from Cory Kasimov of JPMorgan.
Cory Kasimov:
I’ve got two of them for you as well. So I guess, first, can you talk more about what the Phase III COVID vaccine design might look like? And there’s, obviously, nothing traditional about this program. So how should we be thinking about kind of like the endpoints interim analysis and amount of follow-up you think you need for either emergency use authorization or full approval? And then my second question for you is regarding COVID – also regarding COVID-19. There have been some conflicting reports out there on emerging mutations with the virus. I’d be very interested to hear your views on this and what it could potentially mean for the effectiveness of your vaccine.
Tal Zaks:
Thanks, Cory. This is Tal. I’ll take those questions. So look, the Phase III design, let me make a couple of points. Any pivotal trial in order to demonstrate efficacy as well as safety has to be placebo-controlled and large enough so that the people – among the people that you will vaccinate, there will by chance occur cases, right? And so it’s a case-driven design and you set your statistics based on what you expect to see and how many cases you expect to see in the placebo and then how many fewer cases do you expect the vaccine to demonstrate. Now any such trial to be effective depends on three things
Cory Kasimov:
Okay, thanks for the full responses and great CD impressive progress you guys are making.
Operator:
[Operator Instructions] Your next question comes from Ted Tenthoff, Piper. Please ask your question.
Lavina Talukdar:
Ted, you might be on mute. Operator, we’ll come back to Ted. Can you go to the next question please?
Operator:
Your next question comes from Salveen Richter of Goldman Sachs. Your line is open.
Salveen Richter:
Where you’re looking at challenging animal models and then examining the antibody levels in humans? And then a second question around with regard to supply and demand constraints, is the Lonza partnership really just kind of where you – I guess is that – are you going to expand beyond that to kind of handle demand and supply?
Tal Zaks:
This is Tal. Let me try and take your first question. I think you got cut off, but if I understood you correctly, you asked whether we’re running animal channel challenge models and whether we will be able to connect the dots between those and what we see in human vaccinees. And I think the answer is, yes. That work is ongoing. It’s been done in close collaboration with Barney Graham’s team at the VRC of the NIH. And the assay development work that is ongoing is being deployed, so that we are able to connect the dots between the challenge models, convalescent serum and the serum that we eventually expect to see from people who have been vaccinated. So I hope that answers that question, Salveen. Let me turn it over to Juan to talk about or Stephane to talk about the Lonza question.
Juan Andres:
This is Juan Andres, Chief Clinical Operations and Quality Officer in Moderna. So let me take the second question that you have. Lonza brings an incredible track record in supporting and manufacturing products worldwide. Lonza’s capacity, together with the capacity that we have in our site in Massachusetts and the capability, will be a great help for Moderna scaling up and also producing the quantities. We are going to manufacture together with Lonza the formulated bulk, and I expect that we will have more partnership with existing CMOs once we’re finished in distribution, and if needed, with new ones. Thank you.
Salveen Richter:
Thank you.
Operator:
Your next question comes from Ted Tenthoff, Piper. Please ask your question.
Ted Tenthoff:
Great. Can you hear me okay?
Stéphane Bancel:
Yes.
Ted Tenthoff:
Great. Sorry about that before. And thank you for all of the hard work. It’s been just an incredible run during this last couple of weeks and the company has really risen to the challenges. Lorence, it’s been so nice working with you, and I’m wishing you all the best too. So my question actually has to do with CMV. And I know you guys have talked about sort of the challenges just with getting the final data sets and the final process and all those things. So I wanted to see if there’s an update on that and whether or not there’s any changes to the expectation for data in the third quarter. And also, how this general progress and investment in the vaccine platform will really help what you’re doing in CMV. Thanks so much.
Tal Zaks:
Thanks, Ted, for the kind words. This is Tal. Let me try and take maybe both of those questions, and Stephane can add on. On CMV, I believe we remain on track, and it’s – you can do the simple math here. If we’ve already vaccinated over 70% of people with the second dose, and it’s that critical post second dose interim analysis, that should confirm our dose for Phase III. If you recall the data from the Phase I, with much smaller numbers of subjects, we had pretty tight error bars and a pretty good understanding of the dose response curve. Now with the much larger study, even if it ran into some sort of difficulty because of COVID-19, we’re going to be more than powered to understand the immunogenicity. And the size was really driven not just by the need to understand immunogenicity, but also to validate the safety profile that we see here. So I’m confident that with the numbers of people that we managed to get into the second dose, the amendment of the protocol, as mentioned for the remaining less than 30%, that we will stay on track, as we’ve discussed before, to have the data and be able to move on. And our plans for Phase III continue and remain fully on track for CMV. Your question on how it’s preparing the platform. I’ll give you sort of a brief answer from approach of maybe medical and development, and then I’ll let Stephane talk as it relates to the company becoming sort of moving to its commercial life phase. The COVID experience is doing really three things for us. It is accelerating our understanding of the safety and immunogenicity at a much wider level for the platform, sort of the leading edge of data, if you will. And I expect that the ability to run a large placebo-controlled trial and expand the safety database at a much more rapid manner than we had so far in Phase I will be informative for all of us as to the performance of this platform. And here, I sort of speak as a Chief Medical Officer with a keen eye on the safety profile of our platform. So far, we’ve seen nothing unexpected and it’s all been sort of consistent across the application. But of course, a database of 1,500 subjects will benefit greatly when we go into thousands with COVID. The second element has to do with expanding our capabilities, building up a development team that can integrate and build a BLA file, building up our competencies on the regulatory front, the pharmacovigilance front, et cetera. All of that ahead of a large Phase III effort on CMV, I think, is a tremendous benefit for us. And I’ll sort of – I’ll let Stephane speak to all the other elements where this is accelerating the progress of our company.
Stéphane Bancel:
Thanks, Tal. I would just add a few things. And I think Tal started to allude to it, which is the power of our platform, which really create some very powerful network effects, where – if you take an example of something new we shared today, which is we had a positive Type C meeting last quarter around CMV for CMNC support manufacturing. As you can appreciate, this dialogue with the agency around CMV Phase III is going to be very instrumental in helping us on the Phase III for SARS CoV-2 mRNA-1273. Because of the urgency that the agency has, we’ve had an amazing dialogue with the FDA. The responsiveness, seven days a week, very engaged, very willing to find every way to shape a deal process without making any shortcut on safety, obviously. So for this BLA process, that we should be able to go through in the next months, both on the clinical side and also on the manufacturing side. This access to the agency, with this ability to ask question, to get clarification quickly will really help us really build that capability within the team, but also all the digital infrastructure in terms of data gathering, which is critical, both on clinical and on CMC. Then you’re going to be able to use that very quickly on Zika, on CMV. And I think that’s going to be very powerful. The network effect, I think are sometime underappreciated because most companies, as you know, do not have platforms, whereas here, because mRNA being information molecule, there’s really an ability to make Moderna very robust and to take it to the next level. So that’s what we do on SARS CoV-2 BLA-wise can be replicated much faster and much stronger on Zika, CMV and all the other programs. I think it’ll be the same things around commercial. With the arrival of Patrick, we are going to build very rapidly in our commercial infrastructure. We’ll give you updates on that in the coming months. But as you can appreciate, all that work that’s going to happen very quickly on COVID will help us on the other products. But totally as a competitive branding standpoint because, as you appreciate, the Moderna brand has been transformed in the last few months because of the results that the team has been able to accomplish. First of all, the product at center of advancement at the clinical level. So I think the momentum of Moderna is going to be extremely strong and extremely enabled by the SARS CoV-2 verifying process.
Ted Tenthoff:
That’s super helpful, thank you very much.
Operator:
[Operator Instructions] Your next question comes from Yasmeen Rahimi at Roth Capital Partners. Please ask your question.
Yasmeen Rahimi:
Hi, team. Thank you for the continued amazing progress that you’re making day over day. Two quick questions for you. The first question is related to how are you defining age cut-off? In the Phase II, you mentioned there will be a cohort of patients who are 55 and above. Is there maybe a range above which you’re not going to be going after? And then is there going to be a cohort among the Phase II and as you’re thinking about Phase III that are healthy but are at highest risk given that maybe they have obesity or cardiovascular disease? How are we thinking about this patient population that are at the highest risk to incorporate that into the Phase II and Phase III enrollment? And then the second question is in regards to manufacturing. If you could help us understand, what is the single largest unknown when it comes to scaling up mRNA therapeutics?
Tal Zaks:
This is Tal. Let me start by answering the clinical ones, and then I’ll let Juan take the manufacturing question. The – in our Phase II, there is no upper limits. I think above 55. You’ve seen the NIH Phase I sort of parse it out a little bit more finely. I think, for us, we’re going to take all comers above 55 with no upper age limit. In terms of your question on cohorts at higher risk for disease should they get infected, this relates to both the elderly and people with distinct comorbidities. As we build the safety database, obviously, we need to get there, but get there responsibly. I think the Phase II, the initial sort of expansion into larger numbers is people that do not have a high-risk of disease should they get infected. In the Phase III, we will clearly then open it up, and we will do that in a manner that’s responsible and takes the appropriate interim looks to make sure that we expand into that population who needs it the most in a way that’s careful. And that’s an ongoing discussion, obviously, between us, NIAID and FDA how to best achieve that goal. Let me let Juan take your manufacturing question.
Juan Andres:
Okay. Thanks. Thanks for the question. Obviously, one of the unknowns we discussed before which is the assumptions associated with dose. And then in terms of the industrialization of the product, obviously, where we’re working very hard is in bringing the equipment, bringing the raw materials, bringing the people capabilities together as we scale up. I don’t think it is a very single unknown in dose. We have done this before, probably not at the scale at which we are going. Having the partnership with Lonza gives me a tremendous confidence that we are going to be doing this very rapidly. And obviously, speed is of the essence. Bringing these three things together is what it’s all about, and that’s what we are doing.
Stéphane Bancel:
Yes. And maybe Yasmeen, just to add something. We are extremely fortunate to have Juan and this leadership team. As you know, he and the team have all come from large organization. They have managed a very large manufacturing complex organization. They have a lot of experience. They know that every extra million dose we can get out of our system will be helping a lot of people. So I’m very thankful for the team. They are literally working seven days a week, pulling all-nighters to shave every day we can so that we can really maximize the mostly output of the system, and I’m tremendously thankful for them.
Yasmeen Rahimi:
Thank you, Stephane. And so on you were very grateful for all the work that everyone has doing behave and humanity.
Operator:
Your next question comes from Geoff Meacham with Bank of America. Please ask your question.
Alec Stranahan:
This is Alec on for Jeff. And Lorence, we’re sorry to see you go, but I assume you’re moving on to bigger and better things. So my question is on capital allocation in the near term. You reiterated your 2020 expense guidance. But I was hoping you could give a bit more color on the gives and takes within that vis-a-vis OpEx versus CapEx. And how much manufacturing build-out and commercial readiness activities for COVID-19 are reflected within that? And my second question is on the commercialization front. Do you intend to take the vaccine forward yourselves? Or do you think it will take partnering to deliver that at scale? Or would the U.S. government potentially step in as well? Any color you can give here. And if there’s some historical context you could point to, that would be great.
Lorence Kim:
Alec, it’s Lorence. Let me handle the financial question. With respect to the guidance and the components, so as I mentioned around sort of the pre-COVID business, if you will, I’d mentioned that we’re seeing a bit of a slowdown in expenses, OpEx related to lab work and some of the clinical trials, as we’ve noted. And so those expenses will be coming down relative to what we thought when we originally set out guidance. The offset is investments that we are making to be ready for all that’s coming down the road. We’ve mentioned this – the rapid acceleration of the COVID vaccine timelines, and there’s a lot that we need to do at the company to be ready for potentially being commercial in 2021. And so those offset, we will continue to update you all as we – as we scope those investments as we move forward. With respect to CapEx, it is not a huge component here right now of the anticipated budget, mainly because of the leverage we’ve got in the platform as well as the benefit of having a great partner like Lonza on board. And again, we’ll continue to update that guidance should anything change. And then the last thing I would just reiterate is that there is substantial OpEx expected with respect to the COVID vaccine work being funded by BARDA, the clinical development at scale up. But that will be paid for by BARDA reimbursed on a very rapid cycle time. And so that’s why I mentioned that there would be this matching of expense and reimbursement through the course of the year that would substantially offset.
Stéphane Bancel:
Yes. Thanks, Lorence. And Alec, on commercialization, as you can appreciate, as we’ve said before, with the case of CMV, we do not anticipate having the capability and investing to sell the products in our complete countries. For SARS CoV-2, I think it’s important to think about the product in two different time horizon. There is a pandemic phase, in which obviously, we all are. And then we believe, as a company, there’s some opportunity for this product in the pandemic phase because we do not believe this virus is going away. So for the pandemic phase, it’s going to be mostly a partnership with governments. So in that case, you don’t have to necessarily manage complex sets of potential buyers, because we’re going to be, as we discussed, at the global level across the industry, we’re going to be supply constrained for some time, which is why, as we’ve said publicly many times, we are working for everybody with – working on the vaccine. We are hoping that many vaccines are going to be finished. And because if you think about it, there are actually very few companies that have both the manufacturing scale that is required for the task ahead and are already in the clinic, meaning they can have a short to midterm impact with our vaccine. In my opinion, there are just a couple of companies that have those two things. And so if you think about it in a very supply-constrained world in 2021, it’s going to be mostly partnering with governments. So that they will do the allocation in the different geographies. We do not intend, for example, in the U.S. to decide who gets the vaccine. That will not be appropriate. So we intend to continue our partnership with the U.S. government, that we’ve already done with NIAID. And Dr. Tony Fauci’s team for a few years, as you know. And in the clinic more recently, with BARDA, and eventually, I assume the CDC, to be able to supply to the U.S. government the doses, for them to decide the allocation that makes sense for the country.
Alec Stranahan:
Great. Thank you, and congrats on the rapid progress.
Operator:
Your next question comes from Alan Carr of Needham & Company. Please ask your question.
Alan Carr:
Thanks for taking my questions, and congratulations on progress. I got a couple of them, in your increased focus and success with vaccines, are you able to accelerate? What sort of extra emphasis are you putting on these early stage vaccine programs? Can you give us an update on 1345 and 1893, your RSV and EBV programs that are internal? How are those moving along? I know you don’t give high res info and timelines, but to the extent you can. Then the other question is around your COVID-19 program. To what extent is it feasible to have even an interim analysis of your the planned Phase III trial in 2020?
Stéphane Bancel:
So let me start maybe with your first question, on EBV and RSV pediatric. As you know, we do not guide on programs' timelines. So the team is working on advancing those important vaccines as fast as possible. But we have given no timelines, and we will not give timelines. On the retail, as you appreciated over the quarters, to give timelines, when we get closer to kind of late stage development and especially with the SARS CoV-2, given the pandemic and given the suffering around the world, we think it’s important to communicate our best plans. And I hope everybody can appreciate that when we say we aim to start a Phase III early summer, this is the best possible physical plan. Fifty things can derail that. But for the early programs, we are not communicating. Tal, do you want to take the COVID interim data question?
Tal Zaks:
Yes. Yes. Yes. It’s a good question. I believe we should be able to have a sense of the cases and a potential early look by the end of the year. But again, that is a function of how soon can we start, how big the trial is and how good are we at immunizing people who are then at risk for cases occurring, because, as I mentioned, it will end up being a case-driven design to be able to analyze it. And we’ll share the details on the expectations once we lock down the design with our partners and vet it with the agency.
Alan Carr:
So do you expect it to be just a U.S. trial, or would you go global? And I guess another, I guess, follow-up to this is, to what extent I mean a who talk about the possibility of a larger trial with multiple vaccine. Are you contemplating that, too? Or is this – the trial that you’re planning for Phase II just a Moderna versus – a Moderna candidate versus placebo?
Tal Zaks:
So let me take both questions in turn. This first pivotal trial is going to be a partnership with NIAID with the NIH. So it will be, at this stage, either solo or a predominantly U.S. trial. We’re in parallel, looking at opportunities to launch parallel pivotal trials in Europe and globally, because I think, ultimately, the more data we have here, the wiser we will be. I think – can you please remind me your second question?
Alan Carr:
Yes, I think you answered it. I was wondering if you were contemplating a trial – or the government contemplating a trial.
Tal Zaks:
All right. Right, the multi-arm trial, yes. Yes, yes, yes. So there’s been a lot of talk about that, both on the WHO side as well as the NIH. As you can clearly intuit, it’s not front and center in my brain for two reasons. First is, we expect to be the first one out there for a pivotal trial, and so we just have to get on and demonstrate our trial – our vaccine’s potential. But the second one is more fundamental. I think it is important for the field to use more or less the case where it actually makes sense to run many vaccines in a single trial. It’s not like we’re lacking for volunteers who would line up to be immunized and understand the benefit of a vaccine. And frankly, the epidemic is so unpredictable in where it shows up, and to what degree and how it comes down, that there is no expectation of a consistency of attack rate over time that would make that add any scientific value. So for my – this is a personal opinion here. I question the merits of that design from a scientific and a public health need perspective. I think what’s critical here is that for every vaccine candidate, as Stephane alluded to, we’re going to need more than one of them. But it matters less whether there’s a few percent difference on the apparent estimate of the point efficacy, what matters is you know it works and you’re able to scale it up and make it available to those who need it the most.
Alan Carr:
Okay, thanks very much.
Operator:
Your next question comes from Gobind Singh of BMO. Please ask your question.
Gobind Singh:
This is Gobind on for George. Two on 1273. The first one would be, can you help us understand if there’s any profit share agreements in place? Were there any other parties, including the NIH, around the vaccine? And just how do you guys see the commercial landscape evolving with so many other vaccine candidates in development? And then just to follow-up maybe with the NIAID’s comments about their preclinical results that they saw. I understand they’re probably doing their studies separate from you guys. But maybe you can help us understand what kind of preclinical results you’ve seen and when might this data be presented. That would be really helpful. Thanks a lot.
Stéphane Bancel:
Yes. So it’s Stephane. I’m going to start and then the team might think of issues I drop. So it is pretty – as Tal said, there’s preclinical work being done, both in the labs and at NIAID, both with Dr. Tony Fauci’s team, as soon as they just – body of data that makes sense, and is complete and holistic, we intend to publish that work. So as soon as it’s public, you’ll be aware. In term of the profit share, we have not disclosed previously any arrangement. So I will not comment on this one. And what was your other question?
Gobind Singh:
The commercial landscape and how other coronavirus vaccines are in development?
Stéphane Bancel:
Yes. Thank you. So on the commercial landscape, as I briefly mentioned a few minutes ago, as you know, I mean there are 100-plus last time I checked on Wikipedia, vaccine candidates being worked on around the world. The thing I think that are important is manufacturing scale and where are those projects in research of the clinic. As I said a few minutes ago, I believe that the project at this stage in research with a group that doesn’t have the ability to do tens of millions of doses come off ramping up to hundreds of millions per month. He’s not going to be able to have a big dent on this pandemic. And so if you use those two as a screen, which is what we do, I think we end up with a very few number of players, that have again a chance in the 2021 timeframe to have an impact on this. Obviously, like always, in drug development, not every candidate is going to get to a finish line. And I could also anticipate that once a few vaccines are in late stage or commercially approved, a lot of the early project might just stop investing because we are deploying capital and talent for something that might have no commercial end. So I think while there is a lot of people on the stop lane, my sense is very few of them get to the finish line, with manufacturing scale that matters. One or two million doses a year is not going to be very, very helpful at the global scale.
Gobind Singh:
Thank you for your help.
Operator:
Your next question comes from Justin Kim with Oppenheimer. Please ask your question.
Hartaj Singh:
This is – it’s actually Hartaj on for Justin. So one thing first, Lorence, thank you so very much. It was a real pleasure working with you. I look forward to seeing you again sometime in the future. And then secondly, just to Moderna for all the work that you’re doing. I think people – a lot of people really don’t understand just the compression of the timelines that you and the government are engaging. It’s really, really a thing of beauty, knock on wood. So two questions. One is manufacturing and a second on regulatory strategy worldwide. So on manufacturing, if you can just talk a little bit about going from clinical to commercial batches. I know you’ve talked, it’s gone about going from millions, tens of millions to now billion with Lonza. Can you just talk broadly about the timing? When can you go from that clinical? I know you’ve mentioned that Lonza will start manufacturing first batches in July. And how that maps against the BLA that you’ll be starting to file? And then secondly, on regulatory strategy. Japan just approved remdesivir. I guess the EU is going through an approval process, a fast approval process for remdesivir. So how are you thinking of the worldwide approval strategy aside from the United States where I assume you’ll file the BLA towards the end of the year?
Stéphane Bancel:
So Hartaj, let me maybe start quickly on manufacturing. We have not done and have shared precise output per month. What we’ve said is that given we’re ramping up both Norwood, which we said could do up to 100 million doses per year at the 50-microgram dose, and then the Lonza site. As you can appreciate basically every month this year, every month next year, the output per month is going to increase. And so the team is working as hard as they can because they do understand, trust me, that every extra 100,000 vial we get out of the system, we will protect more people. We will slow down the spread of this virus. So it’s not a linear process where you start now at 100 million dose per month, of course not. So it’s just going to be an acceleration process, which is why this dialogue with the government in term of allocation, and we’re going to be hand to mouth for quite some time where as soon as product is made and QC’ed we will go to the government, and then they’ll decide how they allocate it. And we’ll just kind of be on a regular basis. Tal, you want to take the regulatory question maybe?
Tal Zaks:
Yes. Thanks, Stephane, and thanks, Hartaj, for the question. Look, we’re in active dialogue now with the regulators beyond the U.S. I think having the partnership with Lonza is a huge enabler to envision the ability to scale up and eventually supply the vaccine on a global footprint to those who need it the most. That will take shape over the coming weeks and months. The expectation I have is that we will do more than one trial to demonstrate the benefit. That being said, at a certain point, we’ll obviously have data, both potential benefit and ultimately for benefit. By and large, that data should be applicable for filing in other territories. And so we’re actively mapping it out, and our intent is absolutely to eventually be able to make this vaccine available to those who need it the most.
Hartaj Singh:
Great, thanks for all the question.
Operator:
There are no further questions at this time. Presenters, you may continue.
Stéphane Bancel:
So thank you so much, everybody, for participating, and we look forward to talking to you, hosting you at the latest on June 2 for the Moderna Science Day. Stephen and his team will host. Thank you very much. Have a good day and stay safe. Bye-bye.
Operator:
Ladies and gentlemen, this concludes today’s conference call. Thank you for participating. You may now disconnect.
Operator:
Good morning and welcome to Moderna's Fourth Quarter and Full-Year 2019 Conference Call. At this time, all participants are in a listen-only mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I'd like to turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, everyone. On today’s call we will discuss Moderna's fourth quarter and full-year 2019 business update and financial results. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. Speaking on today’s call are Stéphane Bancel, our Chief Executive Officer; Tal Zaks, our Chief Medical Officer; Stephen Hoge, our President; and Lorence Kim, our Chief Financial Officer. Before we begin, I would like to remind everyone that this conference call will include forward-looking statements. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina, and good morning, everyone. As you know, we believe mRNA has a potential to be a new class of medicines with the opportunity to address many unmet medical needs. Given the unknowns of working with a new technology, we have been laser focused on managing risk, technology risk, priority risk, execution risk and financing risks. 2019 was an important inflection year for Moderna. We reported clinically validating data from key programs in two of our modalities, prophylactic vaccines on the left and systemic secreted & cell surface therapeutics on the right. Data that we believe fundamentally change the risk profile of each of these two modalities that we now call core modalities. As a result, our strategy is to double down in these two core modalities with many imported medicines. We have already announced five new development candidates in this core modalities since January 13 at the JPMorgan Conference, three new development candidates in infectious disease prophylactic vaccines and two in the systemic secreted & cell surface therapeutic modalities. Why we double down in core modalities? We are still more than ever interested in understanding the potential of our mRNA technology in our exploratory modalities
Tal Zaks:
Thank you, Stéphane, and good morning, everyone. I'll start with a quick reminder on the data generated to date with our vaccines. In over 1,000 healthy volunteers and 6 positive Phase 1 datasets to date, we've observed the safety profile consistent with the safety of marketed vaccines and the ability to elicit an immune response in the form of neutralizing antibodies. Updates for the ongoing programs are shown on this slide. The CMV Phase 2 dose confirmation study is enrolling well. We completed the second dose cohort and are close to completing the third one. We now expect data readouts to come in the third quarter of this year. Our hMPV/PIV3 combination respiratory vaccine has started the age de-escalation study and Merck is on track with their RSV program in adults. Zika is also going well as 3 of the 4 dose cohorts have completed enrollment. Our three new development candidates announced recently are vaccines against pediatric RSV, mRNA-1345, Epstein-Barr virus or EBV, mRNA-1189 and our vaccine against the novel 2019 coronavirus mRNA-1273. Stephan, will take you through each of these candidates in a minute. CMV is an unmet need as there aren't any approved vaccines and the burden of disease from CMV infection is significant. Of the 25,000 newborns in the U.S. affected each year, 20% will have permanent neurodevelopmental disabilities. And the spectrum of sequelae range from hearing loss, vision loss, microcephaly and even mortality in 10% to 30% of the severely affected infants. We believe our gB and pentamer vaccine has the potential to prevent the infection and help thousands of newborns avoid these sequelae. On Slide 16, I'll review the most recent data from our CMV program. In January of this year, we show the 7-month interim data. The safety has been generally well tolerated and we've not seen any related serious adverse events. Immunogenicity data continues to confirm earlier interim data from the trial and continues to exceed our expectations. Specifically, we continue to see higher neutralizing antibody titers in seronegatives and seropositives in both epithelium and fibroblast assays. If you look at those who have never had CMV, the seronegative group after the third vaccination, they had neutralizing titers against epithelial cells that were more than tenfold higher than the levels seen in seropositives at baseline. We also saw a nice increase in titers against the fibroblasts. Even in seropositives, people who have been infected, our vaccine is able to boost them 20 to 40 fold higher above baseline level after the third vaccination. And we see early evidence of durability out to 12 months. These data are what's behind our optimism and belief that we can take this vaccine all the way through to prevent infections in newborns. As mentioned earlier, our Phase 2 dose confirmation study is enrolling well and is currently enrolling the last dose cohort. We now expect data from the first interim analysis in the third quarter of this year. Preparation for the Phase 3 trial that we anticipate will include less than 8,000 participants, including women of childbearing age, are underway. An early estimate of the cost of this trial is in the range of $200 to $250 million. Now as an opportunity for us, CMV is clearly a blockbuster opportunity. We estimate annual peak sales for a CMV vaccine to be in the range of $2 billion to $5 billion in line with the estimates of others in the field. Assuming an average selling price like GARDASIL, a relevant comparator here, we estimate gross margins of about 90% and EBIT margins of approximately 50%. We're excited about the progress towards this opportunity, supported by the fact that just one of the antigens in our vaccine, the gB when encoded for in a traditional recombinant technology in the past, have already shown a 50% efficacy, which was demonstrated by Sanofi several years ago. We believe that targeting both the pentamer and gB antigens as our CMV vaccine dose, will be additive to the vaccines efficacy. As a reminder, our CMV vaccine mRNA-1647 is wholly owned by us. Let me transition it to Stephan to talk about our new development candidates.
Stephen Hoge:
Thank you, Tal. I want to start by introducing three development candidates in our vaccine -- prophylactic vaccine modality that we've recently announced earlier this month. The first to begin Epstein-Barr virus. And just what to pause for a moment and give you an overview of the disease. So Epstein-Barr virus is a member of the herpes family that includes CMV and it spreads through bodily fluids, mostly in the young children and adolescents. EBV is a major cause of a wide range of diseases, but is the leading cause of infectious mononucleosis in the United States, accounting for almost 1 million cases annually. Infectious mononucleosis can debilitate patients for weeks to months and in rare cases can lead to hospitalization and even splenic rupture. EBV infection is also associated with a range of other disorders, including lymphoproliferative disorders, cancers and autoimmune diseases. For instance, it is associated with a significant increase in risk of multiple sclerosis. There are currently no approved vaccines for EBV. So our vaccine candidate mRNA-1189 is designed to provide broad protection against EBV infection and infectious mononucleosis. EBV has a number of surface proteins on its envelope, including gp350, a trimeric complex of gp42/gH/gL, a dimeric complex and also gB. All of those antigens are important for infecting a range of different cell types, but particularly B-cells and epithelial cells. Now vaccination against only one of those antigens, gp350, which provides only partial B-cell protection, reduce the rate of infection in a prior study by up to -- reduce the rate of infectious mononucleosis in a prior study by up to 78%. But it does not prevent the rate of infection. We believe that the combination of multiple antigens, gp350 plus antigens for gp42/gH/gL and gb will provide an opportunity for broader protection both of B-cells and epithelial cells, very analogous to our approach with a cytomegalovirus vaccine. Now we estimate that the opportunity is quite substantial. Worldwide direct costs of EBV linked infectious mononucleosis reach almost $500 million annually and the indirect costs could exceed $1 billion. But prevention of infection, EBV infection, in addition to the prevention infection mononucleosis could represent a much more significant upside because of the associated increased risk in cancers and multiple sclerosis. These represent a long-term potential, but are not currently a focus of our current clinical development plan. Pivoting now to our second recently announced program, which is for respiratory syncytial virus in the pediatric population. I want to pause and provide a little bit of an overview of the disease again RSV is the leading cause of unaddressed severe lower respiratory tract disease and hospitalization in infants and young children worldwide. It's a major cause of hospitalization in this country, accounting for up to 86,000 hospitalizations a year and over 2 million medically attended RSV infections in children under the age of five. Globally, the burden of disease is even more substantial, with over 30 million episodes of acute lower respiratory tract infection annually. We estimate that the direct cost associated with pediatric RSV disease in the children under the age of five exceed 2 billion annually. So our target population for this vaccine is the young under the age of five, children for respiratory syncytial virus. There is currently no approved RSV vaccine in that population. Our candidate mRNA-1345, encodes for a stabilized prefusion F glycoprotein, analogous to our other efforts in RSV vaccines. mRNA-1345 will use the same proprietory lipid nanoparticle as our hMPV/PIV3 vaccine, mRNA-1653 as well as the CMV vaccine that Tal just described. And we believe that neutralizing antibodies elicited by 1345 will lead to a reduction of medically attended RSV disease in the very young. And while that's exciting, we actually intend to combine 1345 with mRNA-1653 to create a combination pediatric vaccine, respiratory vaccine, which will address over 3 million medically attended lower respiratory tract and upper respiratory tract infections annually in the U.S. alone. That combination of mRNA-1345 and 1653 would represent a significant opportunity to address unmet need. The current plan is to develop 1345 and 1653 independently in the near-term through their initial clinical studies. But we would combine them prior to registrational studies and ultimately advance a joint product. Now the third vaccine that we announced earlier this month is our mRNA vaccine against the SARS-CoV-2 virus, recently named the novel coronavirus that's associated with Cov-19 disease. mRNA-1273 is an mRNA vaccine that encodes for a prefusion stabilized form of the Spike protein of that novel coronavirus that had been selected by Moderna in collaboration with the National Institute of Allergy and Infectious Diseases, the vaccine -- and the Vaccine Research Center, which are both part of the NIH. The first clinical batch for our Phase 1, including finishing and filling of vials was completed on February 7th and earlier this week that was shipped to the NIH for the Phase 1 study. NIAID will conduct that Phase 1 study under their own IND in the near-term. Now pivoting to our second core modality, earlier this year, we did announce two additional programs in the auto immune therapeutic area in our systemic secreted & cell surface therapeutics. And as we described them at JP Morgan, I won't go into great detail, but to briefly recap them here. IL-2, our IL-2 program mRNA-6231 encodes for a long acting, tolerizing IL-2. As you can see in the lower right hand corner on this page, we've demonstrated a non-human primate that a single subcutaneous injection of mRNA-6231 can lead to a substantial increase in T reg cells without increasing activated cells. That provides an opportunity to reestablish immune balance across it that might be relevant for a wide range of autoimmune diseases. There are a number of different recombinant IL-2 base therapeutics that have shown potential and we will be advancing this program into the clinic in the near-term. Second program we announced earlier this year with PD-L1, mRNA-6981. It's an mRNA encoded PD-L1 to send of tolerizing signal to immune cells. In this case we are expressing PD-L1 on the myeloid antigen presenting cell. And the purpose of that is to drive a tolerogenic phenotype, a reestablishment of immune homeostasis in effector cells, including T cells and B cells. mRNA-6981 will be IV infusion using the same LNP as our mRNA encoded antibody, mRNA-1944 that had previously been described. And in preclinical disease models across a wide range of autoimmune conditions, we've demonstrated ability to modify the disease. As you can see in the lower right hand corner, one example, which is , which is collagen induced arthritis. The first indication in which we're going to be bringing the PD-L1 program forward is an autoimmune hepatitis, a disease we see a compelling unmet need. Now with that, I'll turn it back over to Tal to talk about our other work in the exploratory modalities.
Tal Zaks:
Thank you, Stephen. Let me just briefly review these modalities that we consider exploratory, but obviously make up a significant part of what we do in clinical research and give you a sense where we are in the prosecution of these programs. Starting with the cancer vaccines, our personalized cancer vaccine mRNA-4157 is in a randomized Phase 2 trial for the treatment of adjuvant melanoma. The combination of PCV with KEYTRUDA against KEYTRUDA alone and its recruiting well. KRAS, vaccine mRNA-5671 is an ongoing Phase 1 study and this one is led by Merck and it has a monotherapy as well as a combination with KEYTRUDA arm. Our intratumoral immuno-oncology therapeutics, we have three programs in this modality. All of them are in combination with PD-1 inhibitors. Dosing of patients is ongoing and I look forward to the future to being able to demonstrate whether the ability to influence the immune system in this matter will be helpful to these patients. On the regenerative therapeutics modality, AstraZeneca is conducting a Phase 2a in patients with our mRNA that encodes for vascular endothelial growth factor. And that's in patients that are undergoing CABG, coronary artery bypass grafting. That study continues to enroll. In the systemic intracellular therapeutics, we have two INDs open now in both MMA and PA methylmalonic acidemia and propionic acidemia. We are pleased to announce that the first patient in MMA has enrolled in this trial. They’re in the observation period right now. This trial is now open at seven institutions in the United States and we're continuing to look for additional patients to enroll, while we follow this first patient closely. With that, let me turn it over to Laurence to describe the rest of our pipeline where we go from here.
Lorence Kim:
Thank you, Tal. So on Slide 29, you see a graphic that represents our whole development pipeline as it stands today. First and foremost, we're focused on the investment and execution around this development pipeline. There are three significant things from us today around the CMV vaccine, where the Phase 3 preparation is very much underway. You’ve had the Phase 2 enrollment for that. CMV vaccine is ahead of schedule and importantly in Phase 1 of MMA has enrolled its first subject. So we're really focused on executing across the entire breadth of the pipeline. And then the other key thing you heard today is that the preclinical programs continue to grow. And you will heard that we announced in the first two months of the year five new development candidates. This is indicative of the productivity that we expect out of the research platform. If you will flip through the next slide, it's a robust list of clinical data, next steps across the pipeline. If you scan the page, you'll see a lot of readouts coming in the near-term. We've got it for the CMV read out the Phase 2 data with a 3-month interim analysis in the third quarter of this year and a Phase 3 start in 2021. The rest of the vaccines you'll see a number of additional Phase 1 readouts that we would expect to occur, as well as advancement of these new -- newly nominated devolvement candidates toward the clinic. If you look at the next category of systemic secreted therapeutics or antibody against Chikungunya will continue to progress through further development of a dose cohort and will continue to advance preclinical work towards IND filings. And you just heard from Tal the rest of our programs will be progressively moving forward here towards clinical data. On Slide 31, in today’s press release, we report our fourth quarter and full year 2019 financial results. Please note these results are unaudited as of this call. We'll be filing audited financials shortly with the 10-K. We ended 2019 with cash, cash equivalents and investments of $1.26 billion. This compares to $1.69 billion at the end of 2018. Net cash used in operating activities was $459 million for 2019 compared to $331 million in 2018. And cash used for purchases of property and equipment was $32 million for 2019, a significant drop versus $106 million in 2018. We placed our Norwood Moderna Technology Center Manufacturing Facility into service in mid 2018. Revenues for Q4 2019 was $14 million compared to $35 million for Q4 2018. And for the full-year, revenue was $60 million compared to $135 million in 2018. I recall that in January 2019 we adopted the mandated revenue recognition standard ASC 606 using a modified retrospective transition method applied to those contracts, which weren't completed as of January 1, 2019. And so the decreases in revenue are largely attributable to the adoption of this new revenue standard together with the completion of the initial 4-year research period under the 2016 work agreement. Total revenue under the previous revenue recognition standard would have been $15 million for Q4 2019 and $95 million for the full-year 2019. R&D expenses for Q4 2019 were $119 million compared to $150 million for Q4 2018 and for the full-year 2019 R&D dollars were $496 million compared to $454 million in 2018. The decrease in Q4 was mainly due to a decrease in our in-licensing payments to Cellscript and its affiliates and a reduction of our lab supplies and materials. The increase for the full year 2019 was mainly driven by an increase in personnel related costs, including stock based comp, driven by an increase in the number of employees as well as higher clinical trial and manufacturing costs. G&A expenses for Q4 were $26 million compared to $38 million in Q4 2018 and for the full-year G&A expenses were $110 million compared to $94 million in 2018. The decrease in Q4 was primarily driven by a decrease in stock based comp, mainly attributable to certain performance based equity awards with vesting or commencement contingent on the IPO in 2018, and the increase for the full-year 2019 was mainly due to the additional costs of operating as a publicly traded company, including the increases in insurance, consulting and outside services and facility costs. On the next slide, we show the progression of selected cash flow line items, namely our net cash used in operating activities and our purchases of property and equipment. The table shows you our GAAP results by period with a total operating cash flow plus PP&E. I'd point you primarily to the bar chart, which shows the quarter-by-quarter progression of this metric and you'll see that our cash is -- shows a steady reduction through 2019. I would note that Q1 contain the impact of the last of the three licensing milestone payments we owe Cellscript, which is $22 million. I don't expect this downward trend to continue to decline quarterly in 2020, but for the full year you can see how we ended up overall flat versus 2019 when we thought about expectations even with the advancement of our pipeline through the clinic. And so the result will reiterate our guidance for 2020, which is that net cash used in operating activities and purchases of PP&E will total between $490 million and $510 million. That approximate $500 million in cash investment into our business is put in the context in the next slide versus the cash that's available to us for investment. Here you see our year-end cash balance of $1.26 billion, and on top of that is the net proceeds of approximately $550 million from our equity offering, which includes the exercise of the underwriters option to purchase additional shares that we anticipate to close later today. And then as we've mentioned before, we have approximately $185 million in potential future grants available to us as well. And so these amounts sum up to $2 billion in available cash, which we expect to invest in our business, and that represents a significant cash runway of multiple years. I will now hand it back to Stéphane to close.
Stéphane Bancel:
Thank you, Lorence, Tal and Stephen. On Slide 35, you can see a bit on Moderna's priorities. Our company has never been stronger. Our pipeline, preparing for CMV Phase 3, for medicines in preparing for Phase 2, the event Phase 1 trial is ongoing and 10 positive clinical readouts. Our programs in development. Seven vaccines where there are no approved vaccines on the market. Most of these vaccine candidates have multibillion dollar annual peak sales opportunities. As I shared in our 2019 shareholder letter, we believe our innovative vaccines are going to be very large business Moderna. With long-term annuity like opportunity and a high EBIT margin. Five immuno-oncology drugs in the clinic, five readiness program with our first Phase 1 started by MMA, two autoimmune diseases program. The foundation of Moderna have never been stronger. Our clinical experience is now more than 1,700 healthy volunteers and patients. The team is strong, with more than 800 employees who care deeply about our mission and are proud of and energized by the progress. I would like to thank my entire team. I would like to extend a special thank you to those who made the coronavirus vaccine from sequence to shipping to NIH for Phase 1 dosing in only 42 days. And we’re proud to be included with those many companies working on the possible response to this continuing global health emergency. Now would -- these are fully digital, fully integrated facility that enables the execution of a pipeline, all the way from raw materials to finished vials ready to ship to the clinic. We have great partners with AZ, Merck, Vertex Defense or DARPA, BARDA, CEPI and the Gates Foundation. As we said on November quarterly call, we are working on expanding that network of partners as we speak. With the financing of our grand [ph] capital and our cash balance, we are in a fortunate position to invest up to $2 billion towards building the leading mRNA company. We are thankful to our investors for their trust and partnership as we build this unique company. For 2020, our priorities are very clear. Priority number one, execute on our development pipeline with a special focus on CMV Phase 2 restart. Priority number two is creating a new development candidate in the into two core modalities. We already have five in the last two months only. And priority number three is to develop new development candidate in new modalities. Stay tuned here. As a reminder, these are the events we are hosting for analysts and investors in 2020. First, Manufacturing and Digital Day is next week on Wednesday at our Norwood facility in Massachusetts. A webcast would be available on our website. Juan Andres and his team will share many new insights, including how the team deliver coronavirus vaccine in 42 days from sequence to shipping. Marcio Damiani and his team will share the progress since opening Norwood in July 2018 on the digital front, including use cases of artificial intelligence and machine learning. We hope to as many of you for first Vaccines Day in New York City on April 14 for a deep dive into mRNA vaccine modality. We'll discuss some of our things clinical data, business model, how we think about value creation, capital allocation and probability of success of mRNA vaccines. In June, we look forward to hosting our first Science Day to be able to share with you the many progress the team has made on mRNA science and delivery in Science Day 2019. In September, we'd also like to welcome you for our fourth R&D Day, where as usual in New York, we'll review in detail clinical data. As I said in my introduction, Moderna is entering a new phase of its development as a company. Our goals are clear. One, file multiple BLAs and launch multiple medicines, which we own commercially. And two, continue to explore modalities in the clinic and invest in science. We have two core modalities and are now laser focused on filing multiple BLA and then scaling model to maximize the impact on patients. We are continuing to realize our vision. We have got 12 innovative medicines currently in the clinic. We are only getting started. mRNA is an information molecule. Because of that we believe that the probability of technical success for our medicines from the lab to approval will be much early higher than traditional medicines. Speed. Our track record speaks louder than words. Coronavirus from sequence to shipping, clinical grade product, 42 days. Evidence of greater capital efficiency relative to traditional recombinant technology is becoming apparent. We can do new DCs like EBV and pediatric RSV without additional capital invested. I’ve never been more optimistic about Moderna's future and potential since joining as employee number 2 in 2011. I believe mRNA is going to be a new class of medicines and I believe Moderna is the leading company in that field. With $2 billion to invest, the great team of science or IP and the manufacturing site at Norwood, we will work to accelerate our leadership in the months and quarters to come. We are excited by the opportunity to bring forward a new class of medicines for patients. I would like to thank the great team of Moderna employees working hard every day and sometime every week to make these vision a reality. I would like to thank the many people who participate in our clinical studies, including patients, healthy volunteers and physicians. I would also like to recognize all our commercial partners -- all commercial partners who work with us and share our vision to deliver transformative medicines for patients. With that, we are now happy to take any question.
Operator:
Thank you. [Operator Instructions] Our first question is from Matthew Harrison from Morgan Stanley. Go ahead with your question please.
Matthew Harrison:
Hey, good morning. Thanks for taking the questions. I guess, two from me. So, one, on coronavirus and I think this is just so people understand. Could you just broadly comment. It sounded like NIH needs the file on IND and then they would obviously start the clinical study. What is your involvement at this point? And are you taking any steps related to ramping manufacturing or any other steps related to this? And then secondly, maybe just on CMV -- well, actually on MMA. Can you just talk about how enrollment is going for additional patients? What sort of led you to be able to get that first patient in? And if you see, promise in terms of being able to rapidly enroll additional patients. Thanks.
Tal Zaks:
hanks, Mike. This is Tal. Let me take both of these questions. As it relates to coronavirus, our part here was to manufacture and ship it. I think the trial now will be run by the NIH and I defer to them to provide updates when they will. You asked about scale-up. I think we're looking at everything that it would take and how to actually get it done. But we'll update everybody once we have a clear picture of that. Obviously, this is a rapidly changing environment. On MMA enrollment, so right now, we've got one patient enrolled. We do not yet have the second and third, but we're actively working with sites to find them. The age barrier, I think, continues to be a difficult one. We only have to find the first three. So -- and then we can go down in age. So I am confident that we will eventually, but I continue to have a dialogue with the agency on trying to reduce that need, so that enrollment can be unhooked from that. And we can get into the age population where we believe the greatest unmet needed potential for benefit is. So I'll update everybody as soon as we have progress in that field.
Matthew Harrison:
Thank you.
Operator:
Thank you. Our next question is from Ted Tenthoff of Piper Sandler. Your line is open.
Ted Tenthoff:
Great. Thank you very much. And just following up on Matt's question first. What -- maybe you can just remind us again so that everybody understands it because we've been giving a lot of questions on this. What is the process for licensure of a bio threat or pandemic vaccine such as coronavirus? And then with respect to CMV, again, great progress just across the board here. Ultimately, what do you see as sort of the vaccination paradigm or timing of vaccinations for women of childbearing age? Thanks so much.
Tal Zaks:
Thanks, Ted. It's Tal. Let me take that. I think what does it take to get licensure is an evolving field. I mean, the pathway to licensure are well understood and they encompass everything from finding surrogates of protection to demonstrating efficacy. What is it going to take here? I don't think anybody knows. I think all options are currently open, but it's a rapidly evolving field. And you can imagine that there actually isn't not yet a surrogate of protection because this is a very new virus and now people are still working to develop those assays and models. I would give NIH a lot of credit for being at the forefront of that effort and work closely collaborating with them on these efforts. As it relates to the vaccination paradigm for CMV, I think the starting point here is clearly going to be in women of childbearing age. That's where you would anticipate the greatest benefit. And I think the next phase is going to be -- to get into a [indiscernible] population of adolescents, because obviously you want to start protection as early as possible, given that's what the disease we're trying to prevent here is ultimately going to be infants born to women. So we're going to try and get down to that age group as we develop this vaccine.
Ted Tenthoff:
Okay, great. Thank you very much.
Operator:
Thank you. Our next question is from salving Salveen Richter of Goldman Sachs. Go ahead, please.
Ross Weinreb:
Thanks for taking the question. It's Ross on for Salveen. Just a few here from me. So just quickly on coronavirus, what's the potential monitory opportunity here? Do you guys have any details around agreements with the NIH about, funding to you guys? So just thinking about the monetary opportunity there to Moderna. And then on the Chikungunya antibody program, how much follow-up time exists since like the initial patient was first dosed and then since he was -- since he receive the second dose? And are you guys seeing any signs that they can meet immune response? And then I have a follow-up.
Stéphane Bancel:
It's Stéphane. I will talk about first of all on corona, only focus as a team is public health. People are sick all over the planet. People are dying. But it's our only focus is to get a vaccine as fast as we can, safely, partnering with the right people to get it done.
Tal Zaks:
Let me answer -- if I understood correctly your question on the Chikungunya monoclonal antibody, these data are emerging. We're giving you an execution update on to in terms of where we are, once I have a full sense of the data set there, I will, of course, update everybody.
Ross Weinreb:
Great. And then just lastly, so outside of the Phase 2 CMV update in 3Q, what programs are you expecting to have data this year?
Lorence Kim:
We -- as you know, we don't guide to specific timing on various milestones. I'd refer back to the slide which had the rich catalyst calendar and clinical data calendar that we referred to. That list of events that included data readouts as well as advancement of programs is a list that comprises all the next steps. Some of those have advanced fairly far along. For instance, I would note that the phase one vaccine studies for RSV and Zika running since last year. And other instances you'll recall that as you pointed out, the Chikungunya antibody program is a relatively small study in healthy volunteers. But again, we're focused on advancing all these programs as rapidly as we can towards data.
Ross Weinreb:
Great. Thanks.
Operator:
Next question is from Geoff Meacham of Bank of America. Go ahead, please.
Alec Stranahan:
Hey, guys. This is Alec on for Jeff. Thanks for taking our questions. Two questions for me. My first is on your PCV program and I guess the KRAS vaccine as well, have you guys seen any updated data from these studies, including the ongoing Phase 1 for the PCV since ASCO? Just trying to get a sense of why this modality hasn't made the cut for your core franchises, given it's one of the more advanced in terms of clinical development? And then I've got one more.
Tal Zaks:
Thanks, Alec. It's Tal. Look, on the personalized cancer vaccine, it's a Phase 1. So, data continues to come in once we have a full cogent dataset there, we will be disclosing it. And the question of why we don't consider this a core? I think for us core is one that we have gotten the requisite pharmacology to believe it will translate into a clinical benefit. And I think that's true both for the vaccines clearly, and it's also true for the secreted and the surface protein expression, because that you can give the monoclonal antibody actually reached what would otherwise be therapeutic levels of a protein. So that's why it's core. I think in oncology, until you actually show that you’re -- the pharmacology that you're describing in this case immunology, in T cell immunology, until you actually demonstrate that, that translates into a clinical benefit for patients. It's hard for me to call it core.
Alec Stranahan:
Got it. That's very helpful. Thanks. And then secondly, do you have any updates on the CF partnership with Vertex? We've seen Vertex partner with some other gene therapy approaches from CRISPR Therapeutics and others. So any color you can give on this partnership would be great. Thanks.
Stephen Hoge:
You know, we don't generally comment on research. We continue to work with Vertex, we're pleased to be working with them in the CFTR space, but we don't have any updates at this time.
Alec Stranahan:
All right.
Operator:
Our next question is from Cory Kasimov from JPMorgan. Go ahead, please.
Cory Kasimov:
Good morning, guys. Thank you for taking my questions. Two of them for you. First is another one on coronavirus. Thinking a little bit further out, just curious what kind of manufacturing capacity you anticipate having, say, looking out to 2021 event? Should you need to manufacture mRNA-1273 for coronavirus? And then secondly for the CMV program. I mean, if you guys could kind of broadly talk about what a win would look like in that interim update for the Phase 2 we will get in 3Q? Is this more or less kind of looking to replicate the Phase 1 results in a larger group of patients or other nuances we should be thinking about? Thanks.
Stéphane Bancel:
So, Corey, good morning. It's Stéphane. So I will take the first one on manufacturing capacity. I think the [indiscernible] is just too early to know precisely. First, we don’t have those, then fill finish, there's Norwood CMO capacity actually new sites. So just way too early to comment on that. But we’re working out to get as much as we can.
Tal Zaks:
Hi, Corey, it's Tal. Let me answer your question on CMV Phase 2. So I think in a nutshell, the goal here is, as you say, to replicate what we saw in the Phase 1, but I'd like to be able to do that, of course, in the larger data set so that our confidence in picking the right dose for Phase 3 is there. And that has to do both with replicating the nice immunogenicity that we've seen, but being able to kind of plan a clear flag on what we expect in terms of the safety and tolerability profile that would enable us to go into Phase 3 trial.
Cory Kasimov:
Okay. Thank you.
Operator:
Our next one is from Hartaj Singh of Oppenheimer and Company. Go ahead, please.
Hartaj Singh:
Great. Thank you. I’ve just a question on CMV. With the Phase 2 results coming up in the third quarter and then the Phase 3 getting going, could you talk a little bit about how you could speed it to market? I know that a lot will be depending on the Phase 2 results. But can you give us some sort of -- kind of a confidence interval as to when you think the Phase 3 could read out and by which time you could be in the market, 2024, '25, '26? And then just a follow on to that, which is that, you've [indiscernible] as [technical difficulty] $5 billion, which seems to make a lot of sense because as you get to broader and broader immunity, can you just broadly walk us through what kind of patient populations would you want to sort of have the vaccine brought into to get to that higher point of that range? Thank you very much.
Tal Zaks:
Hi, Hartaj. This is Tal. Let me start by answering the first question. So, look, the timeline for CMV roughly speaking, once we get in, we anticipate fairly aggressively to be able to complete enrollment within 18 months. I anticipate the duration of the trial to be 2 years. Now, that still needs, of course, vetting with regulatory authorities. So I want to make sure I caveat that appropriately. Once that we have 2 years on everybody on study, then it's a matter of analyzing, looking at the results and filing and that's where I think the timelines are pretty well understood for what's achievable in our industry. So you can do the math from there.
Stéphane Bancel:
Yes. And -- Stéphane, I will take the second one on CMV, what we take to get to $5 billion number. I think a few things. The first one is, as we explained, the R&D Day in September, it will take -- get an indication approval in women in childbearing age, which, as you know, is a first one. Then to getting to an adolescent approval like the HPV GARDASIL. And third is to getting to pediatric. As we shared, humans are [indiscernible] for CMV. We believe it is a very important public health opportunity here to vaccinate newborns in the pediatric setting. That has been done, as you know, successfully, with Rubella to eradicate the virus and to make sure that humans don't get infected by this virus, which has a lot of long-term negative impact on health at a population as well as for individuals and their own immune system and their own health. Another dimension, of course, is competitive landscape. We will be the only one on market for next 10 years or we’re going to be with one competitor, or two competitor or five competitors. I can come back on that if you adjust it moving further. And then it's the population growth. There is a lot of emerging markets that's not only growing in size with our population growth, but also in terms of dollar invested in [indiscernible], if you take about 5-year, 10-year, 15-year timeframe. And that’s what impact the model that we’ve for getting to around $5 billion annual peak sales.
Hartaj Singh:
Great. Thank you.
Operator:
Our next question is from Yasmeen Rahimi of ROTH Capital Partners. Go ahead, please.
Yasmeen Rahimi:
Hi, team. Thank you for taking my questions and thank you for the tremendous progress that you're making quarter-over-quarter. Few questions for you, all related on CMV. The first one is can you give us a little bit more color on how we think about how current the numbers are in regards to infection rate? If there are differences between U.S. and Europe, how current they are as it guides you sort of for powering assumptions in your Phase 3? And then a second question that was often that is, as you’re in a predominant part of being able to scale up and we’re going to learn more in manufacturing on March 4. Can you enlighten us what are aspects that are unique when you're scaling up in mRNA therapeutics versus other R&A modality? Just we have a little bit of color, do you want [indiscernible] here. And thank you for taking the questions.
Tal Zaks:
Hi, Yasmeen. It's Tal. Let me take your first question. It's a great question and one that obviously keeps me up at night. I think the literature is out there in terms of incidence rates and infections. But it's also clear from that literature that there a high level of variability. And it's not just on a continental level, U.S. versus Europe, that is actually local geography, socio economic status, a lot of things that play into that. So how are we thinking about it in terms of designing the trial, which is obviously, I think where your question is going to, I think the goal here is to design both a large trial and a broad enough trial in terms of size and population, so that on average we are able to hit the incidence rate that people have described. And I'm pretty confident in the ballpark of where we are powering this study to be able to reach it. And finally, I would note that in the trial of this type, you have the ability to actually on an ongoing basis, monitor the incidence in real time. So that the only risk you're really taking if you're missing it is you follow subjects for longer and you catch up the cases. So it will -- ultimately the trial size will come down to the number of cases. And that's one that you can monitor in almost real time.
Stéphane Bancel:
So -- good morning, Yasmeen. It's Stéphane. On the manufacturing process and why mRNA is such a powerful molecule. I think a few things. I mean the first thing is, it's a liquid based process to make mRNA, which is cell free. So that drives to a very small reactors compared to other technologies, especially compared to recombinant is the most staggering changes to forgiven up just the size of our reactors, which of course has a big impact on your CapEx, including all your purification technologies because you just have less details [indiscernible]. So everything is much, much cheaper across the board. As we talked in the past, because mRNA is a new formation molecule. It is the same process for Zika or for [indiscernible] CMV or for coronavirus. So drives incredible flexibility and incredible time to the clinic because we do not have to invent the process for every vaccine or every molecule as the team has shown in the last two weeks with coronavirus. If we’ve had like traditional technologies to invest a new process just for corona, we still be working at it as we speak. And we most probably not even have started to make the product. In that case we are able to just do a tiny bit of optimization for a very large molecule that this mRNA is. And then go right into production. Thanks to this aspect of a platform that we have. And definitely it is time which is -- the separate time to make mRNA is days, not weeks. And so when you think about that, we can use an asset, I mean, once you make with [indiscernible] you can basically change with this possible equipment and use the same room or the same team to make another product. And so if you can deliver in a few days, make mRNA versus a few weeks to make component before you’re going to go into fill finish. That’s a massive use of your capital infrastructure in terms of just CapEx turnover.
Yasmeen Rahimi:
Thank you, team and look forward to seeing you in Boston.
Stéphane Bancel:
Right.
Operator:
Thank you. [Operator Instructions] And next one is from Alan Carr of Needham. Please go ahead.
Alan Carr:
Hi. Thanks for taking my questions. A couple of them. One of them is, can you clarify between your exploratory and your core modalities? Does this mean that you don't plan to add any more new programs to your exploratory until they become core? And then also around RSV, 1345 versus 1172, how are they different and how does this fall outside of the agreement that you already have with Merck around RSV? And the last thing is, can you get over the -- your overall manufacturing capacity at the Norwood facility right now across all programs, the total capacity? And without regard to coronavirus, what were your -- what are your long-term plans in terms of your needs for capacity, in terms of adding manufacturing capacity in the long-term? Thanks. As you go commercial.
Stéphane Bancel:
Good. Thanks for those three questions. So let me take the first one on exploratory and core. So, yes, if you go back to the strategy, we started with six modalities in the clinic to say we cannot manage the unknown, unknown. And so because of the exciting opportunity to create a new class of medicine, we are very focused on managing the unknown technology risk. And so we tried those six technology in parallel. So we could learn from a clinic where to invest more, because mRNA is a new formation molecule making it to platform. And wherever you wanted to fix, if you learn something about the science, fix that science if you can or decide that you’re stop investing in that opportunity, you want to deploy your capital wisely, where you know the technology is working. So that was kind of a premise as we started. And so what is happening with this pivot in the company history that now we’ve clinical data we gathered for prophylactic vaccine and systemic therapeutics. We believe those are core, i.e., in our opinion, we believe the technology risk is off the table, meaning we want to deploy our capital to make innovative medicine that goes to the clinic, because its exactly the same technology, same manufacturing process than the ones we already have the positive clinical data. On the exploratory front, yes, we want to be very cautious and that has been the case for years. We say doing only one program per exploratory modality seems too risky for us, because you have variety risk and there's also variety risk. But we always say that few two, three programs kind of makes sense for us. As such you see so the reason to invest more shareholder capital on more infrastructure, more -- for example -- an example one over four exploratory right now. The answer is clearly no. we want to see what we get from [indiscernible] IL-12 and the triplet. But like we’ve done with those two modalities, that’s migrated from exploratory to core in the last few months, if we get signal the team has a lot of ideas of what other things we could do. And, of course, then those program where we get positive note, we will take those as fast as we can to BLA because those medicine will be needed for patients. Stephen, you want to talk about RSV.
Stephen Hoge:
Yes. Just quickly on RSV. So, Alan, as you reference, we have a partnership with Merck in respiratory syncytial virus, just a monotherapy vaccine. There are two candidates in Phase 1 study. V172 is the one that is currently being conducted and those are targeting the elderly target product profile. And so there is a nearly equal burden of disease in the elderly, 170,000 hospitalizations a year in this country. We're excited to be working with Merck in that elderly population. We have a right in our agreements as we disclose to conduct the development of an RSV vaccines towards a respiratory combination and that has been our intent. And so we are -- that is separate from Merck's progress in RSV.
Stéphane Bancel:
Thanks, Stephen. And on the -- last question on manufacturing capacity. So let me try to [indiscernible] pre-coronavirus world, which we talked about in previous calls, in previous discussion, which is our manufacturing long-term plan is to use Norwood to make development material like we are doing today and to launch our commercial product like CMV, Zika and the others out of Norwood, because of [indiscernible] Norwood design we’ve always said to manage financing risks, we do not want to invest in the big manufacturing capacity, commercial plant until we have, of course, BLA approved, clearly a risk management. But we’ve always said, our long-term vision is to have Norwood focus on development. So that we have a commercial site and hopefully down the road as we scale the company several around the planet that are just focused on commercial products. We believe, based on our experience in previous pharmaceutical companies, that there is dedicated focus per site is really important for success. Development requires nimbleness. Commercial requires scale and efficiencies, very different worlds. So that’s kind of a pre-coronavirus world, our previous plans. And the post coronavirus, while in the last few weeks I go back to buy [indiscernible] a few minutes ago, which is we are looking at the law options, both internally and externally, CMO partners to figure out what's the right path forward and when we have a better picture, we will share it.
Alan Carr:
Great. Thanks for taking my questions.
Operator:
Thank you. That ends our Q&A session. I would now like to hand the call back to Stéphane Bancel.
Stéphane Bancel:
Well, thank you for your question, especially thank you for your trust into our ability to make mRNA an important new class of medicines. We hope to see many of you next week in Norwood for what I believe will be an exciting manufacturing and Digital Day. Thank you.
Operator:
Thank you. This concludes today's conference call. Thank you all for attending. You may now disconnect.
Operator:
Good morning and welcome to the Moderna's Third Quarter 2019 Conference Call. At this time, all participants are in listen-only mode. Following the formal remarks, we will open up the call up for your questions. Please be advised that the call is being recorded. At this time, I'd like to turn the call over to Lavina Talukdar, Head, Investor Relations in Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning and welcome to Moderna's third quarter 2019 conference call to discuss business updates and financial results. You can access the press release issued this morning as well as the slides that we'll be reviewing by going to the Investors section of our website. Today on this call we have Stéphane Bancel, our Chief Executive Officer; Tal Zaks, our Chief Medical Officer; Stephen Hoge, our President; and Lorence Kim, our Chief Financial Officer. Before we begin I would like to remind everyone that this conference call will include forward-looking statements. Please see slide two of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future developments or updates. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina, and good morning, everyone. We are committed to building mRNA as a new class of medicines with Moderna positioned to remain the leader in the field. As you know, we believe our mRNA medicine have a potential to help patients by addressing large unmet medical needs and treating diseases that are not addressable by recombinant proteins or small molecules. Due to the platform nature of mRNA, we believe mRNA medicines provide a higher probability of technical success and faster timelines to clinical trials compared to traditional medicines. We also believe that the manufacturing capital intensity of mRNA is materially lower of a recombinant protein and the cost of manufacturing at commercial scale will be similar to small molecule injectables. We continue to focus on managing risk across the portfolio, especially technology and biology risk. In the quarter, we were proud to share positive Phase I data for two important milestones of CMV vaccine and our antibody against the chikungunya virus-delivered IV. We show that we successfully immunized seronegative subjects with CMV and boosted seropositives. We plan to start a Phase 2 study very soon. The GMP material has already been made and the study protocol has been submitted to the FDA. As a frame of interim results of our Phase 2 trial, we should be able to make the decision for Phase 3 dose. The team is already spending a lot of energy to plan for our Phase 3 study. We believe our CMV vaccine could, if approved, change the future of thousands of children around the world by preventing birth defects. Moderna owns the global commercial rights to the CMV vaccine. We also presented data from the first-ever mRNA-encoded antibody in a human study. We are very proud of this major scientific achievement and we are eager to move forward with the MMA and PA clinical studies soon. Both of these rare disease programs use the same delivery technology as the one in the antibody program. If we step back and look at the body of clinical data of over the last three-and-a-half years we have come a long way. We started 16 Phase 1 trials. We dose and maintained 1,400 subjects and patients. We repeat dose up to eight doses of mRNA up to three weeks apart in our OX40 Ligand and PCV trials. Additionally, across the five modalities for which we have human data, we have consistently shown that our mRNA medicines are well tolerated, results in consistent protein expression in humans, encode for protein that are functional in humans and translate from pre-clinical models into humans. These findings are very important. They assure that our technology investments are paying off. We believe we are at an important inflection point in the company's history. Five out of the first five modalities have found success in the clinic. That was not easy. Nobody is obvious that this would be the case. Two critical factors explained it for me
Tal Zaks:
Thank you, Stéphane. We're advancing our pipeline of medicines in six different modalities. So let me start with the prophylactic vaccines where we've treated over 1,000 healthy volunteers across seven programs to-date. We're pleased with the emerging consistent safety and tolerability profile and of course with the repeated demonstration of peptide immunogenicity, which we had expected based both on the fundamentals of expressing a viral antigen using our body's own cells and the wealth of preclinical data that continued to translate well in the clinic. The CMV Phase 1 success is the latest addition here. These data have exceeded our expectations. But before I review them, let me first briefly update you on other progress. Both the RSV and Zika programs continued to dose patients in their respective Phase 1 studies. For Zika, we also presented preclinical data at the International Society for Vaccines conference in late October, where vaccination with mRNA-1893 was shown to be fully protective at the lowest dose tested. On the hMPV/PIV3 program, I'm pleased to report that the age de-escalation study as Stéphane noted has started with the first subject dosed. We also presented 7-month immunogenicity data at Infectious Disease Week in October. And finally, with our chikungunya vaccine program, we continued to see good durability with 78.6% of the subjects seropositive to chikungunya virus at 12 months post vaccination. So let's get back to CMV. CMV is a common pathogen that is a member of the herpes family of viruses. And like other herpes viruses, it's a lengthened virus, which means once we're infected it's for life. The largest associated with CMV is birth defects and disease in babies born to mothers who are infected during pregnancy. The burden of disease is significant with approximately 25,000 newborns infected each year and is infected each year in the U.S. Currently there aren't any vaccines against CMV on the market despite repeated attempts over the last 50 years. Now we believe that a reason for prior failures has been the inadequate immunization against the pentameric complex which the virus uses to infect epithelial cells, either the cells that line body surfaces and so are the first port of entry for the virus. This pentamer is made of five distinct proteins that have to assemble inside the cell before they are transported to the membrane. So trying to make this structure one protein at a time, using traditional recombinant technologies is obviously challenging. However, this challenge can be overcome with our mRNA-based vaccine, because our technology enables a simultaneous translation of five components. As shown on the bottom half of this slide, mRNA-1647 actually contains six mRNA sequences, five of which encode for this pentamer and one that encodes for gB. Now, gB is a relatively simple single-protein receptor. And the important thing is that prior studies with the recombinant vaccine that only included gB already showed prevention of half the cases of CMV infection, which provides us with a very good starting point for demonstrating efficacy. So let me frame the next two data slides. Every one of us has either been infected with CMV in the past or not. And if we have we're lifelong carriers and our immune system is constantly fighting this chronic infection. So we have antibodies in the serum of our blood and that is called seropositive. If we've never been infected we're seronegative. Now both seropositive and seronegative subjects were enrolled in our Phase I study. So we'll be looking at the levels of neutralizing antibody titers for each. mRNA-1647 encodes for the two antigen, the pentamer and the gB that in general mediate the ability of the virus to infect two different cell types, the epithelial cells and fibroblast. So we will look at titers in the blood of subject that can neutralize the virus' ability to infect either type of cell. That is important, because we're talking about a functional readout. So all-in-all, we are looking at four data sets from this trial. Let's start with the seronegative subjects on slide 13. The top half of the table shows neutralizing antibody titers against epithelial cell infection. I'm going to draw your attention to two rows in the top half of the table. The fourth row down labeled GMT post second vaccination, where you will see the titer levels for placebo in each dose tested, which were basically zero for the placebo group and roughly 3,000, 15,000 and 31,000 for the 30- 90- and 180-microgram dose levels. The second row to focus on is the sixth row that shows the CMV seropositive titer level of 5588. This is the level of neutralizing titers seen in the people, who have been previously infected by CMV the seropositives. The row right above shows the ratio between the neutralizing antibody levels achieved with our vaccine relative to that level seen from seropositive subjects in the trial. At the 90 and 180 microgram doses mRNA-1647 elicited an immune response against epithelial cell infection that is 2.7 and 5.5 fold higher than what is seen in seropositive subjects with natural infection. The reason why this is an important point is that because of the CMV seropositive pregnant women have been observed to have a lower rate of CMV transmission to their unborn children. So these results are striking and that previous vaccines have not exceeded seropositive levels, and it demonstrates the strong immunological potency of our mRNA vaccine platform. The bottom half of the table shows a similar comparison for neutralizing antibody titers against fibroblast. Here, we see our vaccine-induced neutralizing antibody titer levels similar to levels in seropositive subjects. So we've achieved the goal here as well. Taken together, these interim data shows that mRNA-1647 effectively immunizes seronegative subjects to level at/or well above seropositive individuals. Slide 14 shows the same representation of the data only this time for the seropositive subjects in the trial. Here, placebo subjects started off with neutralizing antibody titers. The average level with as I described was 5588. After both the first and second vaccination, we see marked increases in neutralizing antibody titers. So, both vaccinations boost the neutralizing antibody levels, well above the baseline. And this is true for epithelial cells as well as fibroblast. With epithelial cells, we see a 10 to 19 fold increase above the CMV-seropositive benchmark. And with fibroblast we see a 2 to 4 fold increase. It's worth noting that this boosting of seropositive individuals has not been observed with prior vaccines. In terms of safety and tolerability, the vaccine was generally safe and well tolerated with no serious adverse events. Local adverse events included pain, redness and swelling at the injection site. And the most common systemic adverse events were flu-like symptoms of fatigue fever chills and myalgia. The adverse events seem to be more common and tend to be more severe in seropositive subjects in after more than one vaccination. In other words, they tend to correlate with the potency of this vaccine to elicit a specific immune response. That said, the adverse events are of a type that one expects to see in a healthy volunteer vaccine trial and are consistent with the safety and tolerability profile that we've seen in our other vaccine trials. As a reminder, the data I've described thus far the interim data as analyzed one month after the second dose. Our regimen is a three-dose regimen at zero, two and six months. And on this slide you can see preliminary results from a small cohort of seronegative subjects for which we have data out to a year following the 3-dose regimen. The solid gray line is the level of seropositive-neutralizing titers against epithelial cell infection. And you can see that, everyone starts at zero gets to the level, I previously described by month three and are further boosted at month six, so that they remain at/or above the level of seropositive at least out to a year, with a relatively shallow decline over time. My expectation is that, this durability is a function of eliciting strong T-cell help, which we know is an inherent feature of our mRNA vaccine platform. So in summary, we're very pleased with these CMV data. We've shown seronegative subjects, who were successfully immunized to generate neutralizing titers, while seropositive subjects were boosted to level above their baseline something that I don't believe the vaccine community has seen before. mRNA-1647 was generally safe and well tolerated, and we have early evidence of durability out to a year. We're excited by these results and are quickly moving to start a Phase 2 dose confirmation study. Phase 3 preparations are well underway, and importantly, FDA feedback suggests a regulatory path to approval using prevention of primary infection in a population that would include women of childbearing age as a basis for licensure as opposed to a licensure that would be based only on demonstrating the prevention of infection in newborns. We believe a phase 3 trial with this endpoint is achievable with less than 8,000 subjects. Prevention of CMV in women of childbearing age is a very significant unmet need, which we expect will translate into a large commercial opportunity. Currently, there aren't any vaccines on the market for CMV. And we're targeting a highly motivated population with clear opportunities down the road for label expansion. The sales effort should be focused calling on OB/GYNs and pediatricians, and assuming pricing similar to other innovative vaccines one can quickly get to a multi-billion dollar annual sales potential. Moving to the cancer vaccines modality. Recall that both we and NCI demonstrated at ASCO the ability of our personalized cancer vaccine to induce T-cells against the new antigens that we encode. The NCI has now completed enrollment of five subjects in their trial. They used our personalized cancer vaccine as a single agent in an attempt to further boost the expansion of tumor-infiltrating lymphocytes that they had given to a heavily pretreated patient population. They have not seen any responses to the vaccine as a single agent in these five patients. In contrast, our PCV program is designed primarily towards testing the ability of a personalized cancer vaccine to work in synergy with a PD-1 inhibitor and in earlier stages of disease. And to that end, we're actively enrolling patients with melanoma in the randomized Phase 2 trial where we will compare our personalized cancer vaccine in combination with KEYTRUDA versus KEYTRUDA alone for the treatment of patients in the adjuvant setting. The Phase 1 PCV trial also continues to enroll. And finally, KRAS as a target is getting a lot of attention these days. And our approach is to use the four most prevalent KRAS mutations as a cancer vaccine. The trial for our cancer – for our KRAS vaccine in combination with KEYTRUDA is being run by our partner Merck. And the Phase 1 targeting pancreatic colorectal and lung cancer is ongoing. Moving to the intratumoral modality. We have three programs in this sphere with OX40 ligand the Triplet and interleukin 12. Starting with mRNA-2416, which encodes OX40 ligand a potent co-stimulator of immune activation. The Phase 1 is completing the monotherapy arm and currently dosing patients with a combination of OX40 ligand and durvalumab. We're no longer moving forward with just monotherapy of OX40 ligand, but we'll focus our efforts on the combination with durvalumab, which we intend to move into a Phase 2 cohort in patients with advanced ovarian cancer, once the dose escalation and confirmation cohort is complete. The Phase 1 trial for mRNA-2752 which encodes for OX40 ligand and two pro-inflammatory cytokines IL-23 and IL-36 is ongoing. We intend to test the combination of this triplet with durvalumab in several tumor-specific cohorts. Finally, our IL-12 program partnered with AstraZeneca based on a similar rationale is also ongoing. In our localized regenerative modality the Phase 2 VEGF trial is ongoing. Recall that, our partner AstraZeneca has opened additional sites in Europe with the goal of increasing enrollment here. So let me spend the last few minutes on our systemic therapeutics starting with recent data we shared at the R&D Day. As a reminder, mRNA-1944 encodes for an antibody against chikungunya virus. Antibodies are complex proteins consisting both of a heavy and a light chain that come together. So mRNA-1944 includes two mRNAs, one that encodes for the heavy chain and one that encodes for the light chain. Once formed, we would expect the antibody to be secreted in the bloodstream, where we can measure it. So, here are the Phase 1 data. We saw a translation of CHKV-24 the antibody in a dose-dependent manner meaning the more mRNA-1944 you gave the more CHKV-24 antibody was made. Administering mRNA-1944 at the middle and high doses show protein production well in excess of the one microgram per ml level that we had pre-specified as the level predicted to be protective against the chikungunya virus. And the half-life behavior of the protein is exactly as predicted, such that the amount of antibody is expected to stay above the one microgram level for at least four months at the 0.3 mg per kg dose. As you can see in the inset, protein production starts within hours. This is relevant as we think of using this delivery technology to direct the synthesis of intracellular enzymes in liver cells of children born with rare genetic disease as is the case in methylmalonic acidemia and propionic acidemia. The final point to make on this slide is the remarkably low inter subject variability as measured by the coefficient of variance in the range of 20% to 40%. This is similar to what is seen with recombinant proteins. In other words, our delivery technology can direct protein expression without apparent increase in the variability between subjects, which is important as it relates to our ability to define a dose response relationship not just within a given subject, but within the population as a whole. We also show that the antibody made using mRNA-1944 is active as expected. Here the antibody is collected from the serum of subjects and tested against virus in an assay. And it is shown here as the proportion of patients in whom you can dilute the blood at least a hundredfold and still be able to neutralize the virus. Let me review the safety information. Based on the preclinical toxicology, we were able to test mRNA-1944 without steroid premedications though of course the investigator had the option of using steroids if needed. For the low and middle doses, we could not distinguish a difference between the drug arm and placebo as it relates to safety. And recall that at the middle dose, we reached potentially therapeutic levels of protein. And at the high dose we started to see infusion-related reactions that are typically associated with lipid nanoparticles and in fact with recombinant proteins at large. These were anticipated in the protocol. They occurred within hours of the infusion were clinically resolved by the time the subject went to sleep or got up in the morning and did not require medical intervention. We did not see any serious adverse events and we did not see changes in liver or kidney function tests. To understand the performance of our technology, I'd ask our clinical pharmacology team to model the exposure that we would expect in humans based on the preclinical animal model experience. So here in the shaded areas you can see the 90% confidence intervals for where we would expect human protein levels to fall at each dose level. And the solid dots are the individual clinical data landing exactly where expected. Our deliberate technology in other words translates from preclinical species to human with no loss of potency. And this is important because it increases our confidence in the predicted activity of the 0.2 mg per kg dose level for MMA our first rare disease program because it is using the same delivery lipid nanoparticle. Given that we explicitly did not include steroid premedication in the first part of the study the question is whether we can add steroids to the 0.6 mg per kg dose and further reduce or completely eliminate the infusion-related reactions that we've seen. So we're continuing to explore the pharmacology of mRNA-1944 at the 0.6 mg per kg dose level and intend to do so with both the inclusion of steroid premedication as well as by splitting the dose and giving two doses of 0.3 mg per kg one week apart without steroids. So I look forward to continuing to show the data as it emerges from this trial. In summary, we're really happy with the results of our Chikungunya antibody program mRNA-1944. We saw a dose-dependent increase in levels of antibodies against the chikungunya virus. The antibodies were functional with a predicted translation profile from species to species and no loss of potency when we bring it into humans. mRNA-1944 was also well-tolerated in a healthy volunteer population at a dose where we saw therapeutic levels of antibodies being produced. And importantly, these results bode well for our rare disease programs like MMA that use the same LNP delivery technology as mRNA-1944. So this brings me to the intracellular therapeutics. And in this modality let me provide you a quick update on MMA and PA. These are similar diseases caused by inborn error of protein metabolism and are caused by MUT enzyme deficiency or PCC deficiency both of which act intracellularly. Our MMA and PA programs aim to replace these proteins and bring the protein metabolism and the associated acidemia levels closer to normal. From a regulatory standpoint, mRNA-3704 and mRNA-3927 have similar designations. Both compounds have FDA orphan drug designation, EMA orphan disease status, FDA fast-track status and FDA rare pediatric disease designation which upon approval will qualify the two programs for rare pediatric disease vouchers. The Phase I study for MMA is currently active recruiting patients and the PA program recently had its IND open. And we're preparing to start a Phase 1/2 trial there shortly. Now regarding MMA the initial cohort was limited by FDA to adolescents between the ages of 12 and 18. And during the summer we had gone back to the agency and they allowed for the expansion of this age bracket to include patients older than 8. While we're continuing the dialogue with FDA regarding further modification to the enrollment criteria in the past few weeks this amended protocol has been approved by the IRBs of the first few institutions. So I look forward to updating you on our progress soon. PA has a similar design and similar initial age restrictions. And following the opening of the IND and subsequent fast track designations, we have begun start-up activities at the leading academic centers. Let me close with slide 32 which lists the anticipated next steps and upcoming clinical catalysts. And I'll focus you on the CMV Phase 2 start as well as the eventual readout and on the MMA and PA Phase 1/2 trials. With that let me turn the call over to Lorence.
Lorence Kim:
Thank you, Tal. In today's press release, we reported our third quarter 2019 financial results. Please note these results are unaudited. We ended Q3 2019 with cash, cash equivalents and investments of $1.34 billion. This compares to $1.69 billion at the end of 2018. Net cash used in operating activities was $363 million for the first nine months of 2019 compared with $240 million in 2018. And cash used for repurchases of property and equipment was $25 million for the first nine months of 2019 compared to $92 million in 2018, which was the first year that we put our Norwood manufacturing facility into service. Now recall that on January 1, 2019 we adopted the mandated revenue recognition standard ASC 606 using the modified retrospective transition method applied to those contracts, which were not completed as of January 1, 2019. And so the decreases in total revenue for Q3 and for the first nine months of 2019 compared to 2018 were mainly attributable to the adoption of this new revenue standard. Revenue for Q3 2019 was $17 million as compared to $42 million for Q3 of 2018. And for the first nine months of 2019 revenue was $46 million compared to $100 million in 2018. Total revenue under the previous revenue recognition standard would have been $25 million for Q3 2019 and $80 million for the first nine months of 2019. R&D expenses for Q3 2019 were $120 million compared to $109 million for Q3 2018. For the first nine months of 2019, R&D expenses were $379 million compared to $304 million in 2018. The increase in Q3 and for the first nine months of 2019 compared to prior year was mainly driven by an increase in personnel-related costs including stock-based compensation with additional increases for the first nine months of 2019 being driven by higher clinical trial, manufacturing costs and increase in lab supplies and materials, and an increase in consulting and outside services. G&A expenses for Q3 2019 were approximately $28 million compared to $19 million in Q3 2018. And for the first nine months of 2019 G&A expenses were $84 million compared to $56 million in 2018. The increases in Q3 and the first nine months of 2019 compared to the prior year were mainly due to the additional costs of operating as a publicly traded company including an increase in personnel-related costs and stock-based compensation, consulting and outside services, legal and insurance costs. Let me now drill down on our balance sheet strength. We ended Q3 as I mentioned with cash, cash equivalents and investments of $1.34 billion. That balance sheet is augmented by our access to a number of grants. We are fortunate to have established strategic alliances with government-sponsored and private organizations including DARPA, BARDA and the Bill & Melinda Gates Foundation. And so as of September 30, 2019 we've recognized $59 million in revenue to-date, while we have a total additional available funding of $187 million comprising $99 million of committed funding and another $88 million of non-committed funding that we can tap into. I'll turn now to where we expect to end 2019. We ended 2018 with $1.69 billion in cash, cash equivalents and investments. And early this year, we guided to a year-end 2019 cash balance of $1.15 billion to $1.20 billion. We've reiterated that guidance on subsequent quarterly calls. And today, we are guiding the year-end cash at the high end of that range. That is we expect to have approximately $1.2 billion at the end of 2019. This works out to a change in cash of less than $500 million for 2019. I also want to highlight two specific cash flow line items our cash used in operating activities and purchases of property and equipment by quarter. In Q1, we used $152 million of cash in these two items. That number included $22 million of in-licensing payments, but we have no further in-licensing payment obligations to these entities going forward. And then you can see the decline in our quarter-over-quarter cash used for these items and a year-to-date number of $388 million. And so when we look ahead to the full year on these cash flow metrics we can expect 2019 net cash used in operating activities and purchases of property and equipment to total approximately $500 million. As we've said in the past, this reflects our ongoing focus on allocation of our shareholders' capital toward value-driving investments in our portfolio and platform. Lastly, as we look at 2020, we are issuing guidance now for next year around these same cash flow metrics. I'll start by noting that we actually expect the trend to be relatively flat year-over-year. This trend is really due to a combination of factors. First, we had non-recurring costs such as the $22 million of in-licensing payments in 2019. Also at this time with Norwood operating well, our need for significant new facilities and associated capital expenditures is limited. And our pipeline while it's advancing well is actually not burdened with very large trials in 2020. For instance, while we are planning for a substantial Phase III CMV vaccine study that program only requires pre-investment in 2020. And lastly, I'll note that our partners fund significant development costs for programs such as RSV, Zika, KRAS, IL-12, VEGF and Chikungunya antibody. And so for these reasons, we do not expect growth in our cash needs for next year, while being able to fund significant pipeline advancement and ongoing platform development. As a result our guidance is that, we expect net cash used in operating activities and for purchases of property and equipment to total $490 million to $510 million for 2020 very similar to where we will end 2019. As I hand it back to Stéphane, I'll close again with our pipeline which reiterated my last point on the number of programs that we've highlighted that our partner-funded. Stéphane?
Stéphane Bancel:
Thank you Lorence. To close I would like to spend a couple of minutes on the development products and why we are so excited about them and what they can do for patients. Let's start with our innovative vaccines. We now have in the clinical studies four important innovative vaccines
Operator:
[Operator Instructions] And our first question comes from the line of Matthew Harrison with Morgan Stanley. Your line is now open.
Matthew Harrison:
Great. Good morning. Thanks for taking the question and thanks for the detail on the update here. I guess two parts for me. So first, could you just talk about the progress in being able to enroll patients in MMA? I think you've had sites opened for a bit of time here and now you've been able to expand the age group but it doesn't sound like you've enrolled a patient yet. So maybe you can just talk about what's going on there? And then second on, chikungunya maybe just talk about when we should expect to see some information about dosing profile and if you're able to drive dose higher with premedication or split dosing?
Tal Zaks:
Thank you Matthew. This is Tal. Let me address both of your questions. You're right on MMA. We have not yet dosed our first subject. We as noted initially FDA had requested that we limit the first three subject to ages 12 and above and before we can proceed to the children that are young as – as young as one year old, where the highest unmet need is. And I think we found in opening the sites and trying to enroll this that there are several factors that come into play. By the time somebody gets to be an adolescent, if their disease has been severe they've often had a liver transplant. In fact liver transplants in the U.S. are recommended between the age of a 1 year in preschool. And when a kid with MMA shows up for liver transplant they move to the top of the queue, so there's not much waiting. Those who don't get a liver transplant often suffer from kidney deterioration and do a Phase I trial with really abnormal kidney function. So we have gone back in the summer as noted to discuss with FDA, lowering the age criteria. They had agreed to take it down to the ages of eight and above. And while we're still in dialogue with them about that the amendment to enable us to do that has just recently opened at some of the leading institutions. And I look forward to enrolling the first subject soon. It has been a challenge for all those factors that I alluded to. But I think between lowering the age and now being open at some of the select institutions we should see progress soon. Now you'd asked me about the chikungunya and when do you expect to see data? This is a healthy volunteer trial. So we had gone back to the safety monitoring committee aligned with them on the next steps and we're actively working with the site to get the next cohorts enrolled. So as soon as data will become available and we understand what it is, we have we'll be sharing it with you. Being a healthy volunteer trial it should move relatively quickly.
Operator:
Thank you. And our next question comes from the line of Ted Tenthoff with Piper Jaffray. Your line is now open.
Ted Tenthoff:
Great. Thanks for the update and all of the great progress. I want to ask about the CMV vaccine, really just get a little bit better clarity on what you see as the path forward here, appreciating a Phase II dose confirmation start next year. Can you give us a sense of the size of that study and maybe how long it would take? And then looking kind of forward how soon do you think you could actually initiate pivotal studies in women of childbearing age?
Tal Zaks:
Thank you, Ted. This is Tal. I'll take that question. So a couple of points. First in terms of size and timing, the Phase II is about 250 subjects or so. The design is really meant to confirm the dose and give us more color on both immunogenicity and safety and tolerability, so that we are able to pick the right dose for Phase III. That being said, the time should be relatively quickly because the -- we'll make the decision based on a 3-month endpoint once everybody has been dosed similar to what the interim data that we've shown in the Phase I. By then we will have the further follow-up post the third dose in the Phase I, so we'll have overall more data to be able to share and substantiate that decision. Now the path to Phase III I think the big change here for me has been the fact that FDA has provided guidance that speaks of the potential for licensure based on the prevention of primary infection. And that's very significant because that means that it is imminently doable with the trial of the size that we've discussed. In terms of launching that, I would say the two gating factors that I think to actually launching that study are going to be the Phase II data and some regulatory discussions. This was just very preliminary initial guidance of course before one launches a Phase III trial here, we'd need to have those more deep discussions with FDA and other agencies to make sure we're launching the right study. Now once we get their agreement, I think the design is fairly straightforward, one would expect an incident rate of about 1.5% a year. So hopefully with a couple of years of follow-up once everybody is enrolled, you could see the effect that you're looking for and wrap it up. So that gives you a rough sense of the overall time line here.
Ted Tenthoff:
Great. That's super helpful. And then if I may just ask a second question. Appreciating that it's with Merck, when could we get data from the enhanced RSV vaccine? Thanks so much.
Tal Zaks:
Yes. That's a great question. I ask it as well. The truth is that they are pretty efficient in running these trials. They've got a long track record of running a healthy volunteer vaccine trials and of analyzing data quickly. So as soon as they have something material to share with us we'll of course share it with you.
Ted Tenthoff:
Awesome. Thanks so much guys.
Operator:
Thank you. And our next question comes from the line of Salveen Richter with Goldman Sachs. Your line is now open.
Salveen Richter:
Good morning. Thanks for taking my questions. Maybe a first question here on how your chikungunya vaccine clinical development plan reflects the guidance that was provided by the FDA. And then a second question on the chikungunya antibody program here. Could you just walk us through your plans for evaluating repeat dosing recognizing that you do have this cohort study coming up with two doses about a week apart?
Tal Zaks:
Thank you Salveen. This is Tal. I'll take that. So the plan to develop a chikungunya vaccine is a great question. It's true that FDA is giving a potential path. In fact, there's a big discussion in a couple of days time down at FDA where our team is presenting alongside others to flesh out what that could look like. To-date, it hasn't been clear how one could develop it and do it in a commercially viable setting. To the degree that the discussions with the agency in the near term will actually paint a path where it would be worth our capital and time to go there we'll obviously be looking at it carefully. So I don't have a clear answer because I think this is a work in progress. To date, we've not seen a commercial path. I'm happy with the recent agency guidance and we'll see how the conversations go. Your question on repeat dosing for the monoclonal antibody program. The goal really here is to substantiate our ability to stack pharmacology. In other words to show that the antibody level, the protein level production of two doses given a week apart behave as expected in the clinic. We have seen that in the preclinical toxicology in the nonhuman primate. But I think being able to show that and leveraging the fact that the adverse event profile seems to be a very quick infusion-related reactions that come on quickly and come off quickly, we would expect that there would -- there should not be any stacking in terms of safety, while there would be stacking in terms of pharmacology. And that's the rationale of doing two doses one week apart at a dose where we actually have not seen any significant adverse events. Did that answer the question?
Salveen Richter:
Yes. That was helpful. Thanks.
Tal Zaks:
Thank you.
Operator:
Thank you. And our next question comes from the line of Cory Kasimov with JPMorgan. Your line is now open.
Cory Kasimov:
Hey, good morning guys. Thanks for taking my questions. So two as well. So first is just following up on CMV. And as you get the Phase 2 sites up and running and plan for your Phase 3, are there any additional processes or challenges we should be thinking about given the new modality you're introducing? Or is this something that you think you can go pretty quickly from a patient accrual point of view? And then my second question is just regarding your natural history studies for MMA and PA. What's the age range you have there for the natural history? And do you think that provides any read-through to how enrollment might go once you have a broader age range introduced for your clinical trials? 87 subjects at this stage, it seems like a pretty decent number for rare diseases? Thanks.
Tal Zaks:
Thank you, Cory. It's Tal. Let me take those two in sequence. I don't foresee any additional challenge for enrolling the CMV out of the Phase 2 or the Phase 3 that are platform-dependent. And I'll sort of give you the two reasons for that. The first is the totality of safety and tolerability data that we've seen across the portfolio are consistent with what one would expect. We haven't seen anything surprising. There's no scientific reason to expect anything untoward and we haven't seen anything untoward in preclinical. So I think this is going to be a vaccine platform that will behave as such. In fact, the CMV data suggests that the more potent the vaccine is, the more adverse event you get there were flu-like symptoms. But the adverse event the type of adverse events as I've noted is what you'd expect. I think the other reference point is the fact that this is -- the platform as a platform is no longer considered as an adjuvanted vaccine by the agency. And I think that is also reassuring in that respect. So I don't see any hurdles that are a function of the platform to enrolling CMV or getting in the market frankly. Your question about the natural history study is a good one. You're right. I was pleased by the rate of enrollment and the numbers that we've gotten there to date. The age distribution is pediatric. We are seeing I think a median age of around five to eight roughly speaking, looking at the totality of the data so far. It is providing us I think good information as it relates to the variability, the biomarkers, the clinical endpoints all the things we've expected to see. And I think it is substantiating our sense that the older the children are at least those that are found at any given time points in terms of prevalence, certainly in the U.S. tend to have either undergone transplant or have deteriorating kidney function. So it all -- it is very coherent I would say overall with what we're seeing in starting up sites now in trying to enroll the first subjects into the MMA trial.
Cory Kasimov:
Okay. That’s helpful. Thanks, Tal.
Operator:
Thank you. And our next question comes from the line of Alec Stranahan with Bank of America. Your line is now open.
Alec Stranahan:
Hey, guys. Thanks for taking my question. So on the ongoing follow-up from the Phase 1 trial of the personalized cancer vaccine. I appreciate that the objective of the Phase 1 is primarily to assess safety and immunogenicity. But do you intend to present any outcomes-type data from the study beyond immunogenicity, say with this next six months? Or will we need to wait on the Phase 2 study for this sort of analysis? And then on the KRAS vaccine, is this something that Merck will report since they are the ones leading the study?
Tal Zaks:
Hi, Alec it's Tal. So yes, yes to both questions. The Phase 1 PCV data, we will share that data once we've got a sort of a cogent totality of it. Every Phase 1 or every clinical trial that we do as a matter of policy and ethics in the company we will publish and this is no different. So at the next opportune moment when we have a body of evidence there we will share it, including the totality of the clinical data. And in fact, I think it is the clinical data that is important to me and I think the rest of the world to assess activity here. In that regard, recall that one of the cohorts there is testing patients with previously refractory to PD-1 inhibitor and so I think that will be meaningful data once we have it. KRAS, you are correct. Merck is running it. And so Merck will be I think the party reporting out the data and I look forward to that as well.
Alec Stranahan:
Okay. Great. Thanks. And I had one more question if I may. So I noticed that the follow-on Phase 1b for the hMPV/PIV3 vaccine was posted at ct.gov last week. Now looks like you've dosed the first patient in the study. So a few questions. What kind of safety metrics are required for gating between the adult and pediatric cohort? What dose levels of the vaccine will be included in the study? How quickly do you foresee the study completing? And from your conversations with the FDA, do you have a sense of potential registrational path following the study? Thanks.
Tal Zaks:
Those are great questions. Let me answer them in turn. The safety metrics are your typical vaccine safety metrics, so you look for safety tolerability. And we have a safety monitoring -- an independent safety monitoring committee that reviews that and we've actually had our recent inaugural meeting with them. And so, it won't be just us making that. We've got some of the world's experts in this, actually looking over our shoulders and helping make those decisions. The dose level honestly on the top of my mind, I don't remember. It's the honest truth. I'm happy to get back to you. If it's not on the trial let's double check that. The path to approval, that's a great question. I think as we had -- as disclosed in the past, we had an initial interaction with the agency. We had a Type C meeting in response. And we had a line of sight to the possibility to have a pivotal trial that would test both viruses together. So, roughly speaking the path there is demonstrate safety in the seropositive toddlers and then go on to a larger Phase 3 study that would demonstrate efficacy in seronegative toddlers, because remember we're trying to establish immunogenicity from before anybody has been exposed. And so I can't give you a clear time to completion. And as with CMV before one goes into a Phase 3 trial, we're going to have to have more in-depth discussion with the regulatory authorities before we do that.
Alec Stranahan:
Great. Thanks for the color.
Operator:
Thank you. And our next question comes from the line of Hartaj Singh with Oppenheimer. Your line is now open.
Hartaj Singh:
Great. Thank you for the questions. I just want to ask one question about an asset that you got the furthest along AZD8601. I know that's in partnership with AstraZeneca, the CABG study. I mean that's when you're treating patients for almost about a year and half now. Just any thoughts there? And could -- with the readout of this Phase 2 basically a study could A, move that along faster or into a pivotal clinical trial? So any thoughts there? And then I just have a couple of quick follow-ups.
Tal Zaks:
All right, Hartaj. It's Tal. Let me take that. Two points on that trial. It is -- I mean to give AZ credit, they've designed a very informative pharmacology trial because they're using sort of state-of-the-art as you may recall oxygen labeled CAD imaging to really very finally map the heart and the areas at risk. Now the challenge for them has been that the half-life of that isotope is extremely short and so you have to find a hospital that basically has a cyclotron next door. And so what they've done, I think in this past year has been to go and find additional sites where they can launch that trial. So they're now open not just in Finland as they were but actually in Germany and in the Netherlands and are actively looking for patients. The good thing about that trial is that the endpoint once they enroll the subject should be relatively quickly because it is a six months endpoint. And the trial overall is not very large. I think that's as much insight as I have in terms of enrollment. Could they move faster? I think there are other ways that one could think of how you would develop an intracardially injected molecule. They've got a ton more expertise in this space than I do and they're actively thinking and working along those fronts. But frankly I'd have to defer to them to give you more color on what that could look like. And I think you had a few follow-ups. Yes.
Hartaj Singh:
Yeah. A quick one, I'll just mention them quickly. I know you had mentioned that the seropositive patients there's -- the data seen in those in the CMV trial that had not been seen previously in other trials. For your Phase 2 and your Phase 3 will you stratify by seropositives? And could those be a group that you could get approval just on their own? Just any thoughts there?
Tal Zaks:
Yeah. So a couple of thoughts. I think the fact that one sees this effect on seropositives, I think is really scientifically interesting and I've got a deep curiosity to try and figure out where it could translate to benefit. That being said, in terms of enrolling on the Phase 3 and the eventual label, I'd make two points, one I think the Phase 3 will be focused on seronegatives because that's where you can see the obvious effect of preventing infection. It's hard if not impossible to actually demonstrate a clinical benefit in the seropositive, although of course I remain intensely curious about that. The second point to note is that FDA in their response had told us that they understand that one is not expected to demonstrate the benefit in seropositives. But in order for this vaccine to be viable in terms of access to patients and having an impact on the population, you would need a label that would encompass both populations. So the expectation is that we would demonstrate immunogen -- I'm sorry prevention of primary infection in the seronegatives, but we would demonstrate safety and tolerability in the entire population that is all-inclusive of seropositives and that could lead then to a label irrespective of sero status. Does that answer your…
Hartaj Singh:
That's great Tal. Yeah, great. Thank you.
Operator:
Thank you. And our next question comes from the line of Yasmeen Rahimi with Roth Capital Partners. Your line is now open.
Yasmeen Rahimi:
Hi, Tal, thank you for taking our questions. Questions are all around PA. Congrats on recently receiving fast track designation. So the first one is for you, can you tell me when you're thinking about dosing strategy for MA -- I'm sorry PA, how it will differ from your strategies in 3704? And then when we think about sort of key PD biomarker, what key biomarker in your view proves established proof-of-concepts? And then part three of the question is, what do we know from the literature in regards to what level of expression of PCC one needs to achieve to provide therapeutic benefit?
Tal Zaks:
Thank you ,Yasmeen. Great questions. Let me take them one at a time. The dosing strategy is roughly similar to what we expect to see in MMA. It is based on our translation of the preclinical data. We've done long-term models in animals and have used that to extrapolate. So far the technology has extrapolated nicely from all the preclinical species in the various modalities. And so I would expect this one to be similar. So, I expect the dosing strategy to be once every two or every three weeks. So, I think we're going to have to define that and see the effect we have on Phase I. There is a whole slew of biomarkers here that one looks at and I think it's premature to specify which one is going to be the one. It is true. I think what you're indirectly alluding to is that the biomarkers on PA are not maybe as clear-cut as they are in MMA. I think that remains to be proven. I think, the information we hope to achieve from the natural history study in terms of biomarker levels where we're collecting it should also be informative. So, I mean we'll continue to update that as we learn more. I think our understanding of the literature and of our natural history level here is evolving. In terms of the level of expression needed, I think it's hard to quantify. What we've seen from our preclinical models suggest that it is a similar level that we need of MMA in order to have the effect in the mice be effective as profound in the PA models as it is in the MMA models. And so, we expect a similar dosing level to be required for both. It may be a little bit higher given that in PA we are encoding for two proteins, not one, right? So, on a model basis, we would expect similarity on a mg per kg. It may be a tad higher, but it's clearly within the same ballpark.
Yasmeen Rahimi:
Thank you. And then if I may have a quick follow-up in regards to the antibody against chikungunya. And we're excited to see the data from the two doses of 0.3 mg per kg. How far out are you going to assess safety? So, what are you planning to do? And what has regulators asked you to do?
Tal Zaks:
Just to clarify the question. You are asking how far are we looking at safety?
Yasmeen Rahimi:
Yes. Yes. Just monitoring the safety, since you're giving for the first time two doses. How far out will you actually monitor these patients and therefore will provide for us the safety results?
Tal Zaks:
Several weeks I would say. I don't think it's going to be much longer than that. The reality is you've seen the adverse event profile to-date. It really is a function of the LNP as I understand it more than the protein that we make. We'll be continuing to follow these patients -- I'm sorry these subjects for weeks, but I've not seen anything that leads me to expect that there's a long-term challenge here. I think the safety profile within a week we'll describe what it is.
Yasmeen Rahimi:
Thank you.
Operator:
Thank you. And our last question comes from the line of Alan Carr with Needham. Your line is now open.
Alan Carr:
Hi, thanks for taking my questions. Can you give us an update on where the other rare disease programs stay? And might those be a little bit easier to enroll? And then also, do you have any update on AZ's program with Relaxin?
Tal Zaks:
Yes. So this is Tal. Let me take them. They're in their preclinical phases. Once we file the IND and we get rolling, we'll update you accordingly. And I think, we don't typically comment on where exactly in the early research space they are. I think the same holds true for Relaxin which is partnered with AstraZeneca.
Alan Carr:
All right. And then, you also mentioned in the press release some interest in more partnerships and that sort of thing. I was wondering if you were getting at more in terms of with academic institutions or talking about partnering programs with pharma and that sort of thing.
Stéphane Bancel:
Yes. So, thank you. This is Stéphane. So, I mean if you look at the company history, as you know we have done a lot of partnership. On the last 12 months, you might have observed we have been quiet a lot of issues because we are very busy. We're preparing the IPO and then spending a lot of time on the road as a newly public company. But if you look at what the team has achieved and what we believe a platform can provide in term of new medicines for patients, we have a primal abundance which is a wonderful point to have in this industry. And so, we think that we cannot take all the drugs that are coming from Stephen's team on the platform into the clinic by ourselves. And so, we will continue to have dialogue with biopharmaceutical companies. If you look at the company history, we work around the clock there of our capital since inception of the company through partnerships. And we think this is a wonderful way to tap capabilities that we don't have to tap capital that we don't have and to just increase the opportunities of getting Moderna's mRNA medicines to patients and to a finish line. And so, we anticipate to continue to have new partnership set up in the quarters to come.
Alan Carr:
And what's the profile of programs that you might partner versus keep in-house when you're looking at this?
Stéphane Bancel:
So, I mean speaking to what we have done before is looking at where do we have expertise and keep us looking at biology risk. So, this is what we're selling for.
Alan Carr:
Thanks for taking my questions.
Stéphane Bancel:
Thank you.
Operator:
Thank you. And this concludes today's question-and-answer session. I would now like to turn the call back to Stéphane Bancel for any further remarks.
Stéphane Bancel:
Yes. Thank you very much for joining us today and for your questions. We look forward to seeing many of you in the coming weeks and at the JPM conference in January. We also wanted to share with you that we are planning to start an Annual Day focused on manufacturing and digital. This new Investor Day will complement our Science Day which usually happen in the spring and the R&D Day usually in September. So, 2020 manufacturing and digital events will be held at our Norwood plant on the afternoon of March 4 on the back end of a Cowen conference in Boston. Juan Andres who leads our Technical Development, Manufacturing and Quality; as well as Marcello Damiani our Chief Digital Officer will host the event. They will share with you what the progress their team have made in 2019 and the new initiatives they will do in 2020. We hope to see many of you on the afternoon March 4, in Norwood. Have a nice day.
Operator:
Ladies and gentlemen, this concludes today's conference call. Thank you for participating. You may now disconnect.
Operator:
Good morning, and welcome to Moderna Second Quarter 2019 Conference Call. At this time, all participants are in listen-only mode. Following the formal remarks, we will open the call up for your questions. Please be advised that the call is being recorded. At this time, I’d like to turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, operator. Good morning, and welcome to Moderna's second quarter 2019 conference call to discuss business updates and financial results. You can access the press release issued this morning as well as the slides that we’ll be reviewing by going to the Investors section of our Web site at www.modernatx.com. Today, on this call, we have Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Tal Zaks, our Chief Medical Officer; and Lorence Kim, our Chief Financial Officer. Before we begin, I would like to remind everyone that this conference call will include forward-looking statements. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. I will now turn the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina, and good morning, everyone. We believe mRNA has the potential to be a new class of medicines. We believe mRNA medicines have the potential to address large unmet medical needs and to treat diseases that are not addressable by recombinant proteins or small molecules. Due to the platform nature of mRNA, we believe our mRNA medicines provide a higher probability of technical success and faster timelines to clinical trials and to the market relative to traditional medicines. We also believe that the manufacturing capital intensity of mRNA is materially lower than recombinant proteins and that the manufacturing costs at commercial scale will be similar to small molecule injectable. Because of this large potential, we continue to focus on managing the risk across our portfolio, especially technology risk and biology risk. We believe that programs will be in the same modality of similar technology risk, meaning that once we derisk a sentinel program, they are important read-throughs. As a key example in the near future, we believe our chikungunya antibody program will be an important clinical readout as it uses the same formulation technology as our MMA program, our most advanced rare disease candidate. Our corporate focus is on three priorities. First, to execute on the development pipeline; two, to move new development candidates in existing modalities from the lab into the clinic; and three, to invest new development candidates in new modalities. I will review now our most important progress since our last quarterly update in early May. Starting with PCV, personalized cancer vaccine. We presented positive interim Phase 1 data at the ASCO meeting in June and we are happy to report today that since ASCO, we started a Phase 2 head-to-head trial in the adjuvant melanoma setting. We look forward to the readout of this important immuno-oncology program to assess if PCV plus Merck’s KEYTRUDA can increase recurrence-free survival versus KEYTRUDA monotherapy. We’re happy to report today that our Phase 1 for CMV has completed enrolling healthy subjects at doses up to 300 micrograms. We believe CMV is a large unmet medical need and we look forward to reviewing and sharing the Phase 1 trial data in the near term. The team continued to execute at a rapid pace in the last 90 days. We advanced four new programs into Phase 1 since our May call. Two programs in immuno-oncology started dosing in cancer patients. Our KRAS vaccine which is partnered with Merck and the IL12 intratumoral program which is partnered with AstraZeneca dosed their first patients. Two programs in infectious vaccines started dosing as well, our RSV vaccine mRNA-1172 partnered with Merck and our Zika vaccine mRNA-1893 which is funded by the U.S. agency BARDA. Finally, I’m happy to report that clinical sites are now open and actively recruiting patients in our first rare disease program MMA. We have three open sites in the U.S. and in the UK, the clinical trial application, or CTA, was just opened by local authorities. I am very pleased with the company’s progress and I am very thankful for Kim’s dedication to this execution. We have five immuno-oncology programs in the clinic including PCV in Phase 2 and OX40 soon entering Phase 2. We have five important rare disease programs and are still working out to dose the first MMA patients and to submit INDs for all our rare disease programs. We have four vaccines in the clinic for major unmet medical needs; CMV, RSV, hMPV PIV3 combo and Zika. I want to remind you that there are no approved vaccines for any of these harmful pathogens that severely affect thousands each year. We are very pleased to have completed enrollment in our CMV trial and we’ll look forward to sharing the data with you soon. The company has never been as strong and we’re more focused on continuing to execute and share our progress in the months to come. With this, let me turn to Tal to give you some more color on the development pipeline.
Tal Zaks:
Thank you, Stéphane. As you know, we’re advancing our pipeline of medicines in six different modalities. In the next few slides I will highlight the progress we have made this quarter in each of these. So starting with prophylactic vaccines on Slide 13, you will see we have eight programs in this modality and we’ve made significant progress in the last quarter. In total to date, we have safety data from over a 100 healthy volunteers who have participated in our Phase 1 study and we remain pleased with the emerging safety and tolerability profile of our vaccines. I’m happy to report that our CMV program with mRNA-1647 is now fully enrolled in the Phase 1 trial and I’ll go over this opportunity in greater detail in just a moment. The RSV Phase 1 study testing mRNA-1172 dosed its first subjects in this quarter and recall that at the last quarterly update we reported that our partner Merck had just filed the IND. Our Zika program with mRNA-1893 also had the IND filed and open in the second quarter and I’m happy to report that the first subjects in the Zika Phase 1 trial was also dosed. In terms of emerging data, in hMPV and PIV3 or mRNA-1653, we continue to see neutralization titers above baseline at the second interim look seven months after the last vaccination. For context, in January we reported the two-month immunogenicity data. We plan to present the full data from this Phase 1 study at IDWeek in the fall. We’re also pleased with the feedback from FDA regarding the development plans for mRNA-1653 where we discussed the potential fast forward to evaluate protection against both hMPV and PIV3 in a single Phase 3 study. Consistent with these plans, we plan to enroll seropositive toddlers in our next trial. Finally, the Phase 1 data for our influenza vaccines against H7 and H10 were published in the journal Vaccine. Let me now spend a few minutes on CMV. As noted before the Phase 1 trial for CMV is fully enrolled, CMV is a common pathogen that is a leading cause of birth defects. The burden of disease is significant where approximately 25,000 newborns are infected each year in the U.S. alone. Currently there aren’t any vaccines against the CMV virus on the market. That’s because CMV has proven to be a challenging vaccine to manufacture using traditional technologies given the structure of one of the antigens, the pentamer, which we think is required to elicit a protective immune response. We believe these challenges can be overcome with our mRNA base vaccine as our technology lends itself to producing the pentameric viral antigen by encoding further simultaneous translation of its five components. As a reminder and as shown on Slide 15, mRNA-1647 actually contains six mRNA sequences, five of which encode for this pentamer and one that encodes for the gB protein. We believe the combination of these two antigens encoded by mRNA-1647 will produce potent and durable antibody titers against CMV that have the potential to protect against infection. We look forward to the Phase 1 results soon. Let me now turn to cancer vaccines. You will see the programs in the modality on Slide 17 and I’ll focus on mRNA-4157, our personalized cancer vaccine, and on the KRAS vaccine, mRNA-5671. Recall that we and our partner Merck announced the Phase 2 trial earlier this year. The Phase 2 design is randomized trial testing the combination of mRNA-4157 in combination with pembrolizumab against a pembrolizumab monotherapy control arm in high risk melanoma patients in the adjuvant setting. I’m happy to report today that the Phase 2 is up and running and that the first patients have consented to the trial. Interim safety, tolerability and immunogenicity data from our Phase 1 were the basis for the decision to move to Phase 2. We presented these interim data with mRNA-4157 either as monotherapy in a respected adjuvant population or in combination with pembrolizumab in the metastatic setting. These two arms represent arms A and arms B, respectively, of the Phase 1 study. We have a Part C and Part D that continue to enroll. Our T histologies include microsatellite stable or MSS, colorectal cancer and head and neck squamous cell carcinoma. We and our partner Merck have also added an additional cohort in Part B where we will be testing the combination of mRNA-4157 with pembrolizumab in patients who are refractory to PD1 inhibitors. Turning now to the interim results we presented at ASCO of this year, we showed that mRNA-4157 was safe and well tolerated but no reported DLTs and no grade 3 or grade 4 adverse events. We also showed that mRNA-4157 elicited neoantigen specific T-cell activation in 10 of the 18 class I neoantigens in the one patient treated at the top dose where apheresis was performed. While these data were obtained with the first version of our vaccine that included up to 20 neoantigens, we’re now selecting for up to 34 neoantigens using our proprietary algorithm. For the data presented at ASCO, the patients were dosed with the PCV that had the 20 neoantigens and all patients who have enrolled since April in both the Phase 1 and Phase 2 studies have been receiving the 34 neoantigen version. At ASCO, while the clinical data are early in preliminary, we did report six responses in Part B of the study in the metastatic setting. One of these was the complete responder to pembrolizumab at monotherapy prior to receiving the personalized cancer vaccine and five other dosed with the combination of mRNA-4157 and pembrolizumab had a partial response. Two of these five PRs were patients who were previously treated with checkpoint inhibitors. While these early signals are trending in the right direction, we believe that our Phase 2 trial will help us and Merck to definitively ascertain the incremental benefit of mRNA-4157. Moving now to the KRAS vaccine, I’m also pleased to announce that the first patient in our Phase 1 trial testing KRAS vaccine, mRNA-5671, was dosed. As a reminder, KRAS is a key regulator of cell proliferation and survival. Mutation in the KRAS gene caused dysregulated cell proliferation and its one of the best studied oncogenes. It is the most commonly mutated oncogene and it drives over 20% of human cancers predominately in the pancreatic, lung and colorectal cancers. Indeed, the team at the NCI led by Steve Rosenberg had shown at the end of 2016 that the recognition of a mutated KRAS epitopes by T-cells can lead to cancer regression. A quick overview of mRNA-5671 is shown on Slide 21. It encodes for the four most prevalent mutations of KRAS which together represent 80% to 90% of KRAS mutations. The generic sequences that span the mutations are combined into a single mRNA that encodes for all four neoantigens. When translated within the cell into a neoantigen protein chain, the cellular protozoan [ph] machinery is expected to clear the chain and present these neoantigens to the immune system to stimulate what we think will be an active anticancer T-cell response. The Phase 1 trial for this vaccine which is being run by our partner Merck has enrolled its first patient and this study will evaluate safety and tolerability of mRNA-5671 both as monotherapy and in combination with KEYTRUDA in patients with metastatic non-small cell lung, colorectal and pancreatic cancers that harbor the KRAS mutations. Of note, in this trial we are selecting for specific HLA subtypes that based on the signs are most likely to respond. For the intratumoral immuno-oncology programs, we’re progressing with all three of our development candidates; OX40 Ligand, the Triplet and interleukin-12. Starting with mRNA-2416 which encodes for OX40 Ligand, which you will recall is a potent co-stimulator that promotes T-cell proliferation. The Phase 1 is completing the dose confirmation cohort at 8 milligram and in parallel we’re progressing to start the Phase 2 cohort in patients with advanced ovarian cancer. Slide 26 shows the schematic of the Phase 1 trial and the Phase 2 cohort. Turning to mRNA-2752 or the Triplet, which encodes for OX40 Ligand and two pro-inflammatory cytokines interleukin- 23 and interleukin 36-gamma. The rationale here is to stimulate T-cells through the presence of OX40 Ligand while attracting the T-cells to the tumor site with the local expression of these cytokines. By injecting the tumors directly we expect the cytokines to act locally within the tumor microenvironment. The Phase 1 is ongoing and it has both the monotherapy arm and a combination arm with durvalumab. I’m pleased to report that the first patient in the combination arm with durvalumab has been dosed. We’ve also made progress with MEDI1191 in our interleukin-12 intratumor injection program partnered with AstraZeneca, as the first patient in this trial was dosed. Recall that interleukin-12 is a potent immune modulator associated with type 1 interferon response and production of interferon gamma. Its activity against cancer has been described in the literature but safety has been a problem when interleukin-12 has been administered systemically. We believe that the intratumoral mRNA approach should allow for interleukin-12 to act locally in the tumor microenvironment while avoiding the toxicities seen with systemic administration. Let me touch for a moment on the localized regenerative therapeutics and AZD8601. The Phase 2a in coronary artery bypass graft population is ongoing. Our partner AstraZeneca continues to open additional sites in Europe with a clinical trial application now also open in Germany. Let me move ahead to our systemic secreted therapeutics and I will focus on mRNA-1944, our antibody against the chikungunya virus. As of today, we’ve enrolled six of the eight subjects in the third dose cohort. Before I get to the trial design and the strategy on this program, I want to take a few minutes to highlight the program which is DARPA funded. mRNA-1944 encodes for an antibody against chikungunya. Now antibodies are a complex protein that require both heavy and light chain to come together to form an active protein. So mRNA-1944 actually includes two mRNAs and one that encodes for the heavy chain and one that encodes for the light chain. Once formed, we expect the antibody to be secreted into the bloodstream where we will be watching to see if it has some immunity against the chikungunya virus as expected. On Slide 36, you will see the trial design. The key objectives of the trial are to evaluate the safety and tolerability of four single ascending doses of mRNA-1944 and to evaluate the pharmacokinetics of the drug and the pharmacodynamics of the anti-chikungunya virus antibody levels which together will describe the dose response growth. We are collecting assay data to see if the antibody levels neutralize the virus which we believe will ultimately speak as to whether or not this antibody we encode for is indeed functional. Now the utility of this program is really twofold. First, as a product that could potentially protect against chikungunya infection by conferring passive immunity; and second, as this program uses the same lipid nanoparticle formulation that is shared with our other programs in the rare disease indications that we are pursuing, it could conform the risk profile of those other programs as well. Lastly on systemic intracellular therapeutics where we have four developing candidates, I’ll highlight our rare disease programs methylmalonic acidemia, or MMA, and the closely associated disease propionic academia, or PA. Both MMA and PA are inborn errors of protein metabolism that are caused by MUT enzyme deficiency and PCC deficiency, respectively. As you can see, these two acidemias are really on the same metabolic pathway. The prevalence of both is approximately 325 to 2,000 patients in the U.S. Patients are identified during newborn screening and current regimens are palliatives. They consist of strict diet restrictions and oral and IV medications. Really the best treatment that we currently have available for suitable patients today is a liver transplant. Both mRNA-3704 and mRNA-3927 encode for intracellular proteins that act within the liver cell and act on the mitochondria. Both programs also have FDA orphan drug designation, EMA orphan drug status and FDA rare pediatric disease designation which upon approval will qualify the two programs for rare pediatric disease vouchers. The Phase 1 study of our sentinel rare disease program in MMA with mRNA-3704 currently has three sites open and we are actively recruiting patients. In parallel, the Natural History Study continues to enroll well with the total of 71 patients across the MMA and PA enrolled. Let me close with Slide 42, which shows you the breadth of our pipeline. In one place you’ll see all the new updates we’ve announced since December 2018 when we became a public company. And with that, let me turn the call over to Lorence.
Lorence Kim:
Thanks, Tal. In today’s press release, we reported our second quarter 2019 financial results. Please note these results are unaudited. We ended Q2 2019 with cash, cash equivalents and investments of $1.44 billion. This compares to $1.69 billion at the end of 2018. We are reiterating today our expectation for cash, cash equivalents and investments at December 31, 2019 to be in the range of $1.15 billion to $1.20 billion consistent with the guidance given on our call in March. We remain focused on allocation of our shareholder capital towards value-driving investments in our portfolio and platform. Net cash used in operating activity was 256 million for the first six months of 2019 compared to 160 million in 2018. These numbers include 22 million and 25 million of in-licensing payments in the first quarters of 2019 and 2018, respectively, as stated in the footnote. After the first quarter of 2019, we have no further in-licensing payment obligations to Cellscript and its affiliate. Cash used for purchases of property and equipment was 18 million in the first six months of 2019 compared to 66 million in 2018. And then on revenue, recall that on January 1, 2019, we adopted the mandated revenue recognition standard ASC 606, using the modified retrospective transition method applied to those contracts which were not completed as of January 1, 2019. The decrease in total revenue for Q2 and the first six months of 2019 as compared to 2018 was mainly attributable to this adoption of the new revenue standard. Revenue or Q2 2019 was $13 million as compared to $29 million for Q2 2018 and for the first six months of 2019 revenue was $29 million compared to $58 million in 2018. Total revenue under the previous revenue recognition standard would have been 17 million for Q2 2019 and 55 million for the first six months of 2019. R&D expenses for Q2 2019 were approximately $128 million compared to $104 million for Q2 2018 and for the first six months of 2019, R&D expenses were $259 million compared to $195 million in 2018. The increases in Q2 and the first six months of 2019 as compared to 2018 were primarily due to an increase in personnel-related costs including stock-based compensation, an increase in clinical trial and manufacturing costs, an increase in lab supplies and materials, and an increase in consulting and outside services. G&A expenses for Q2 2019 were approximately $29 million compared to $21 million in Q2 of 2018. And for the first six months of 2019, G&A expenses were $56 million compared to $38 million in 2018. The increases in Q2 and the first six months of 2019 as compared to 2018 were mainly due to the additional costs of operating as a publicly traded company, including an increase in personnel-related costs and stock-based compensation, consulting and outside services and insurance costs. And with that, I'll hand the call back over to Stéphane.
Stéphane Bancel:
Thank you, Lorence. To close our remarks, I would like to reiterate that our team is focused on executing on our three priorities; advancing the development pipeline, investing in new development candidates in the existing six modalities and investing in new modalities. The team at Moderna executed across the board during the quarter. To summarize quickly the highlights of the quarter, we initiated the PCV Phase 2 trial with first patient consenting to participate in the trial. Our CMV vaccine study is fully enrolled. We started four new clinical trials; two in immuno-oncology and two in infectious diseases. Vertex’s cystic fibrosis research collaboration. As you might recall, in July 2016, Moderna and Vertex announced an exclusive research collaboration and licensing agreement aimed at the discovery and development of mRNA therapeutics for the treatment of CF. Based on the clinical work to date, Vertex has extended this collaboration for the first quarter of 2020 with options to extend further based on future progress. Preliminary mRNA delivery will present a potential new route administration for Moderna. I am pleased with the progress we made today and look forward to the rest of 2019 and 2020 as we approach clinical data results. I will particularly look for the CMV Phase 1 data and the chikungunya antibody Phase 1 data in the near term. As a reminder, the chikungunya antibody is the first monoclonal antibody encoded by mRNA technology to be dosed in a human. Because RSV and Zika are dosing healthy subjects, this trial should complete soon. And if positive, we intend to transition to Phase 2. We now have five immuno-oncology programs in the clinic, two of which are already dosing in combination with a proved checkpoint inhibitor, Merck’s KEYTRUDA for PCV and AstraZeneca IMFINZI for Triplet. All teams working on clinical trial sites are focused on the milestone of dosing of first patients with MMA. We believe that mRNA has the potential to be a new class of medicine. We see a large product opportunity ahead of us and we are energized that the potential to bring these important medicines to patients. Four vaccines for large unmet medical needs where there are no vaccines approved today. That is a unique opportunity towards millions and as such we have large commercial products. Five immuno-oncology programs which all have the potential to improve the response of PD1 or PD1 checkpoint inhibitors. Five rare disease programs for conditions like MMA and PA where children born with a missing or defective protein urgently need a treatment that targets the underlying cause of their disease. The cardiology program, VEGF, which could transform the care of patients who have [indiscernible] and MI. And it is the only first wave of innovative products. Stephen Hoge and his team are working hard to move new innovative development candidates from the labs into the clinic. The productivity of our mRNA platform is significant. We dosed our first clinical trial in December 2015. In just three and a half years, we started 16 programs in the clinic and we’ve had a high success rate. The team did get 19 IND of CTAs opened by local authorities. We know we have a special opportunity and we are committed to delivering on the promise of our science, and bringing forward a new class of medicines to patients. I would like to end our remarks by thanking the many people who participate in our clinical studies, including patients, healthy volunteers and physicians. I would also like to thank the great team of Moderna employees working hard every day to make our vision a reality. With that, we are now happy to take any questions.
Operator:
[Operator Instructions]. Our first question comes from Salveen Richter of Goldman Sachs. Your line is open.
Andrea Tan:
Great. Thanks for taking our questions. This is Andrea on for Salveen. My first one is, how are you thinking about positioning for your KRAS vaccine in the context of growing competition in disease? And then I have a follow up.
Tal Zaks:
Hi. This is Tal. Thanks for the question. First of all, I’m really happy that we finally have therapies that are emerging as effective against KRAS mutations. I think that progress for the field is tremendous. I think it’s still early days, so let me make two points. First, the exact nature of the activity and against which mutations and in our case which mutations and which HLA still needs to be defined. So I don’t see them even on – if you look at the patient distribution necessarily as competing. Second and I think more importantly on the fundamentals, I think what our vaccine is trying to do and what the emerging inhibitors are trying to do are very different things in terms of patient benefit. I think the history of small molecule targeted therapy has been terrific in the sense that it’s translated into real benefit for these patients. But we struggle to turn them into curative-intent treatments. I think on the other hand, the immuno-oncology approaches were successful, have translated into a much more durable effect. And so my expectation is down the road if both of these approaches are successful, you would expect them to have complementary benefits for the patients. And I’m really excited in the coming years to see how that story plays out.
Stéphane Bancel:
And maybe, it’s Stéphane, just to add one thing, because Tal described in his remarks the mRNA that we designed is actually coding for mutation; G12D, G12V, G13d, and G12C.
Andrea Tan:
Great. And then just on your MMA program, how many patients right now are enrolling in your clinical study that have been rolled over from the Natural History study?
Tal Zaks:
So in the clinical study we have not yet enrolled. We’re actively recruiting. In the Natural History study there have been 71 patients enrolled to-date.
Andrea Tan:
Sorry. So do you anticipate I guess rolling any patients over from that Natural History study or no?
Tal Zaks:
It is a possibility. We’re looking at it.
Andrea Tan:
Got it. Thank you so much.
Operator:
Our next question comes from Matthew Harrison of Morgan Stanley. Your line is open.
Matthew Harrison:
Good morning. Thanks for taking the question. Two for me. So the first one is, can you just comment broadly how we should think about safety so far in the chikungunya study given that you’re through almost a third cohort? I don’t know if you can comment on what the stop and gos are from a safety standpoint. And then second question just on OX40 Ligand. Can you talk about what you need to do in this space to probably be able to take that to the FDA? I guess what I’m asking is how should we think about potential regulatory tests forward with that molecule? Thanks.
Tal Zaks:
Sure. Let me start with the chikungunya. This study is ongoing so I can’t really comment on the data until we see the totality of the picture there and then we’ll describe it for you. It’s a healthy volunteer study, so stopping rules are what you would expect in these typical studies. In terms of OX40 Ligand, the regulatory path, if you look at where we’re expanding into the Phase 2 cohort, we’re going after ovarian cancer. I think in that setting checkpoint inhibitors are not yet approved and it’s because they really have marginal activity as monotherapy. If we could demonstrate that the combination has a clear benefit to patients, I think the path to approval will be relatively straightforward. So that’s how we’re looking at it. Did that answer your question, Matthew?
Matthew Harrison:
It did. Thanks, Tal.
Operator:
Our next question comes from Ted Tenthoff of Piper Jaffray. Your line is open.
Edward Tenthoff:
Great. Thank you very much and thank you for the updates; lots of good progress. So my question is and I apologize if this was asked, but with respect to the Triple my concern here is certainly not activity especially with durvalumab, but are you doing any special immune safety analysis or any special additional safety analysis just because of the potential potency of the Triple therapy? Thanks.
Tal Zaks:
Thanks. That’s a really good question and I wish I had a wider answer for you. The reality is that we’re looking at the safety I think in the traditional way that people do in clinical trials maybe colored by a better understanding over the years of what the safety profile of the checkpoint inhibitors alone is. And so we’re looking at whatever autoimmune phenomena, et cetera, and all the other adverse events that one would expect from checkpoint inhibitor monotherapy and assessing very carefully to see whether we exceeded. If there’s any other safety signal that is attributable to the Triplet, then I think we’ve got two ways of finding it. First, recall we’re dosing as monotherapy so that will give us a clear view on the safety profile just of the Triplet. And second, in the combination arm we’re looking carefully at all the clinical characteristics. And unfortunately I think in the field it’s very hard to predict the adverse reactions that one sees and they’re not very frequent. So all we can do at this point is maintain a careful visual for what to expect and make sure you’re not missing anything unexpected. I don’t know if that answers the question. I’m not sure I’ve got a better one.
Edward Tenthoff:
That’s all right. That makes a lot of sense. I appreciate that. Then just a really quick high-level question. With respect to the CF collaboration, are there any novel delivery modalities that are being incorporated for that disease or is this really not just treating lung but really systemic disease? Thank you.
Stephen Hoge:
Ted, it’s Stephen Hoge.
Edward Tenthoff:
Hi, Steve.
Stephen Hoge:
So first of all, it’s a research collaboration with Vertex and we’re excited to continue it based on the preclinical progress to date. As a part of our general research activities, we do look broadly at a range of different delivery modalities. We have obviously made progress in one direction here but we haven’t yet defined a development candidate at which point we’d probably provide specifics about that. Generally, our approach with Vertex and CF has been to address the unmet need in CF, particularly for those patients who are [indiscernible] for CFTR and focusing intensively on the pulmonary disease. But obviously without commenting specifically in the CF example, pulmonary delivery is a route of administration that could be valid for other systemic diseases or other applications as well.
Edward Tenthoff:
Thanks a lot. I’m looking forward to the – yes, sorry. Go ahead.
Stephen Hoge:
No. That’s it.
Edward Tenthoff:
Great. Awesome. I’m looking forward to the CMV data and see you guys in September. Thanks.
Tal Zaks:
Thank you.
Operator:
Our next question comes from Cory Kasimov of JPMorgan. Your line is open.
Cory Kasimov:
Hi. Good morning, guys. Thanks for taking my questions. I have two as well. So I guess first is can you just walk us through the cadence of what you see is the key validating clinical updates we should expect in the next 12 months or so? Beyond the CMV and chikungunya updates, what else has a change of recurring in that time period and would we see new clinical data at your R&D Day in September? And then I have one follow up.
Stéphane Bancel:
Hi, Cory. Good morning. It’s Stéphane. So I will not comment on the R&D Day. I hope you come to the R&D Day, but we’ll make sure that we give good updates on everything we know then. On the next 12 months, as you can see on Slide 46 on the presentation, as I discussed in my comments, CMV is really important. As you know, it really is a very large opportunity. We own 100% of the economic of this product. We believe it’s a very large medical need out there. And so the CMV data are going to be very important we believe for company. RSV and Zika, because they’re in Phase 1 in healthy subjects and they are dosing as we speak, should read pretty quickly. And as I shared, the plan is to move the dose to Phase 2 assuming we have good data into the clinic. I remind you that we have already in the past shown in a good translation from primates into humans, into our vaccines that have positive data. And so we look forward to this data in human. PCV, of course, will take a little bit of time because we started a Phase 2. That’s a very important study. We need to recruit for that Phase 2, in all 150 patients across the board. And then it’s basically of course looking at survival in four months. KRAS is going to be interesting. We all believe there’s a big medical need [indiscernible] tumor types. So we are dosing two patients in Phase 1, so we’ll be sharing observation at the planned clinical meetings of what we see in the clinic. In the intratumoral because it’s oncology and we’ve dosing, the Triplet is in combination with PD1. OX40 will be in Phase 2 soon and also will be in combination with a checkpoint. [Indiscernible] IL-12 forward, same thing we will update the different medical meetings and clinical observation except [indiscernible]. And then as we discussed, the chik antibody is very important for us and then getting the first body in the clinic. The [indiscernible] are going to go straight into patients. As we have commented before, we are starting MMA as a dose that has been showing in animal models having some benefit. And so I think the next few months and the next few quarters are going to be quite rich, data wise. We have [indiscernible] drugs for potential [ph] in the clinic. That’s a lot of potential data.
Cory Kasimov:
Okay, great. And then the follow up is regarding your personalized cancer vaccine, 4157 program. Any near-term plans for exploring indications beyond resected melanoma patient that are at high risk of relapse, or PD1 refractory? What do you see is the potential of this program and indications that may have considerably less neoepitopes?
Tal Zaks:
Thanks for that, Cory. It’s Tal. It’s a question that we’ve asked ourselves since the beginning of this program. I think strategically and philosophically, what we want to do with this program is first to go where the likelihood of success is the highest before we look for areas that are more challenging. And so that’s why we’ve focused in the histologies that we have in the Phase 1 and that’s why we went into an adjuvant setting even within melanoma for a definitive study into Phase 2. I think once we have a clear proof of concept, clearly we will begin to explore some of those additional indications. But there’s not any current plans to do that.
Stéphane Bancel:
And Cory, it’s Stéphane, to add to Tal’s remark. If you think about it, going back to Lorence’s comments, we are very disciplined in our capital allocation. So, of course, there could be a lot of different things one could think of trying with personalized cancer vaccine as you think about all of our patients that aren’t treated today. But unfortunately we cannot know – before we have an important derisking, we cannot expand too much because we have so many opportunities of products across the portfolio, we could be increasing the [indiscernible] that will not be unreasonable. And so we want to be very disciplined. That’s just one example. There’s a lot of things, trust me, that the clinical team – as you know Tal is oncologist by training who love to be trying in the clinic to help those patients. But we just have to be very disciplined in capital allocation and how much we spend and where we spend it, and when we spend it based on the risking.
Cory Kasimov:
Okay, makes a lot of sense. Thanks for taking the questions.
Stéphane Bancel:
Thank you.
Operator:
Our next question comes from Alan Carr of Needham. Your line is open.
Jennifer Shen:
Hi. This is Jennifer speaking for Alan. I have a couple of questions. First question is, I was wondering if the team can give us some color on the commercial strategy and possibly specific patient groups that you may be planning to target for the CMV assets. Thank you.
Stéphane Bancel:
So thank you, Jennifer, for the question. I think it would be great if you can join us at the R&D Day because we will spend quite some time on CMV commercial opportunity. As we discussed in the past, there are many populations from women in the age of bearing a child, adolescent women who might want to protect. There was also discussion about partners of those pregnant women. There’s also discussion because humans are very [indiscernible] CMV, do you put down an age to try to eradicate CMV. So there’s a lot of different segments that we’ll discuss quite at length on the R&D Day. But this is why I think we believe CMV, if you take a 10, 20-year timeframe and if you look at all the old vaccines, like the HPV vaccine and the [indiscernible] vaccine, those very important vaccines, the lifecycle management of those products can be very important and we have our eyes very much on how do we go about this. Again, we cannot do all the indications at the same time. It’s going back to very simply on capital allocation and investments. But we are very much in mind of how do we maximize with them this opportunity to get the largest label that we can for CMV, so it can be given to the largest population we can around the world and not only in the U.S.
Jennifer Shen:
Thank you so much. The other question is for the hMPV/PIV3 vaccine, could you possibly give us some comments or color, any new understanding of the titer level needed to progress this effort? Thank you.
Tal Zaks:
This is Tal. I think that our understanding of the titers there is going to be based mostly on the preclinical modeling and what we’ve seen to date that is protective. Unfortunately, there is no vaccine on the market, so we don’t really have a correlative protection like we have from influenza. But it is a respiratory virus, so one draws similar parallels from the experience with flu and you get a sense from the totality of our understanding in the science on the respiratory viruses what are the titers like. I think what we’ve seen in the Phase 1 is supportive of our ability to immunize. Recall though that the target population here is in seronegative infants, right. So ultimately we’re going to have to define our ability to reach significant titers and boost to the maximum the immune response capability to respond in that population down the road. So I think it may not be a very black and white answer, because I think it will take inference from multiple lines of reasoning, from science and from clinical studies and from other vaccines I think to come to that. Does that help you?
Jennifer Shen:
Yes. Thank you so much for taking my questions.
Tal Zaks:
Thanks, Jennifer.
Operator:
Our next question comes from Hartaj Singh of Oppenheimer & Company. Your line is open.
Hartaj Singh:
Great. Thank you for the questions. I just have two. One is; I know that you mentioned that the chik antibody is very important. Can you just talk maybe a little bit about that if you see the proof of concept manufacturing the antibody using an mRNA and then see efficacy on the vaccine side, antibodies are over 100 billion in sales per year. What other areas could you go into? Would you see yourself being in vaccines? Are there other types of antibodies, other types of diseases that would be amenable to your approach in that regards, or is that just looking too far into the future? And then I got a quick follow up.
Stéphane Bancel:
Good morning. It’s Stéphane. So as you know we have disclosed all 21 development candidates and we don’t comment on future plans in research. Obviously, as I said in my remarks, we think it’s a very important milestone. It’s the first time using a modern technology and antibody is being produced in a human. And so that’s an important technology that as you commented has a lot of different applications. What we try to do always as the portfolio of the assets that we develop with our shareholders’ capital is to be thoughtful about managing biology risk, technology risk and to create important innovative products for patients. That’s always a big driver for us. If you look back to one of my closing slides, if you look at the portfolio today, for most of the products in the pipeline, there is no product on the market with big enough medical need and there’s no solution on the market. And so we are always thoughtful about all those things. But it would of course become a very important tool in our Moderna toolbox. As you know, partnering is certainly an important part of our strategy. If you look back over time, we’ve done four partnerships with Merck, a few with AZ. We’re very, of course, happy with the decision by Vertex to expand the collaboration. And so that’s why technology can be made available to a partner. So this is an important piece of the Moderna toolbox.
Hartaj Singh:
Great. That helps a lot. And then I just had a question on your manufacturing strategy. I visited the Norwood facility; really, really kind of cool stuff going on there. It is clinical grade sort of material and research material. Can you just talk a little bit about how you’re thinking about your commercial-grade material? You’re getting to the point now where you might have one or two other rare diseases where you might be able to get to the clinic fairly rapidly and the regulators want to see a sort of clinical to commercial sort of strategy. Can you just talk a little bit about that? And then which of your modality actually requires more intensity from a commercial manufacturing perspective than others? Thank you.
Stéphane Bancel:
Those are great questions. So, on the commercial front, as we’ve shared in the past, Norwood is able to do commercial, but it’s not ready today. We have to do much work around validation and the quality systems and so on. But the infrastructure of the plant itself has been built so that the site can be brought to commercial readiness and be able to do pivotal studies, registration studies out of Norwood. What we also shared and it’s again going back to our focus on managing the risk, we will not have to be the commercial facility before we have our first commercial product approved. That’s a very important part of Moderna’s strategy to derisk the company. So we can launch products out of Norwood. We can do Phase 3 out of Norwood. We will get the site ready on time so that we can do that. We also always have a contract manufacturer strategy. We never want to be single sourced for the company. That would be way too risky. So we have as we speak contract manufacturers that have also commercial capabilities from their site and the quality system already. And if you think about the different products on the portfolio, which is a second question, the thinking path is mostly on the back end, which is on killing the virus. Because if you think about it, mRNA for vaccines, the doors are very, very tiny, so you don’t need a lot of mRNA drug substance to supply actually millions of virus. Because you go back to the data we have published, our vaccines show efficacy at 2,500 micrograms per human. So in half a kilo or kilo you can do a lot of doses that can do enough. And so – and [indiscernible] because the end of patients is low. Don’t go into gigantic quantities. Of course, oncology is a different ballgame. But again, that would be a very happy program to manage when we get there. On the back end, no, it does not have the capability to do millions of vials of fillings, but that’s something that is readily available to contract manufacturing. So that is why we feel very confident that we have – with the current infrastructure that we have, we have the ability to do pivotal, to do commercial. If we’re to manage the back end, we need more vial capacity. We get and contract that out with an existing partner of a new partner, we have time for that. And if we needed more capacity, one thing to remember about Norwood is that we can increase the capacity of Norwood tremendously versus the current capacity. We are only working with two-day shifts. We could put of course a night shift. We don’t have a shift on weekends. So right there you have a lot of capacity available. We can move the warehouse out of the site, the warehouse doesn’t have to be on the site and you have right away more GMP facilities that you can access your utilities. The QC lab can also be moved off of the plant. It can be moved in the parking lot. You just build a new building. And here, again, you go with more GMP capacity. The preclinical – all the robotics are preclinical. Same thing, it’s currently in a GMP suite but doesn’t have to be, because it’s preclinical material. So there again, you can move it to the parking lot or in a new building. So if you think about the manufacturing strategy of Moderna, Norwood was a big investment. We think it’s a strategic investment. We cannot deliver on the mission of the company or the pipeline without Norwood. But we really need Norwood so that this becomes the central node for us that it there for a very long term so that we do not have to invest CapEx in the years to come at a high level. We will not be in the commercial plant until our products are approved. That would be way too risky.
Hartaj Singh:
Great. Thank you, Stéphane. That’s great.
Operator:
Our next question comes from Alec Stranahan from Bank of America Merrill Lynch. Your line is open.
Alec Stranahan:
Hi, guys. Thanks for taking my questions and congrats on the progress. I just had a couple. So maybe first on the hMPV/PIV3 combination vaccine, do you have a sense of the sort of data we’ll likely see in October? And will we see data outside of antibody titer comparisons? And is the Phase 1b toddler study necessary as per your conversations with the FDA before you begin a Phase 3? And then I have one more.
Tal Zaks:
Thanks, Alex. It’s Tal. So in October, we’ll present the totality of the data as we have it, so you’ll see the antibody titers, you’ll see the safety, you’ll see what you typically see when we describe the totality of the study. And I believe that’s been accepted to IDWeek. In terms of the seropositive toddlers, yes, I think that is consistent with the development that one would expect and that the agency concurs in terms of the next step in the development path here. So ultimately remember that the target population here is infants, so there’s a pretty structured and rigorous way by which you work your way down into that population.
Alec Stranahan:
Okay, great.
Tal Zaks:
Now given the sensitivity to the pediatric population, we wanted to make sure that we’ve got clarity from the agency in terms of designing that study and that’s why we put it in the press release and we discussed that interaction.
Alec Stranahan:
Right, I understand that’s a sensitive patient population. And then shifting gears to your KRAS vaccine, 5671, we’ve seen data from Amgen and others that are pretty encouraging on G12C, although it seems maybe there’s a subgroup that requires additional combination therapies. So I was just curious on the KRAS vaccine what your thoughts are for it as monotherapy and also in terms of combination with checkpoint inhibitors? Thanks.
Tal Zaks:
Yes, so I’ll give you two versions of the answer, one on the science and one as a drug developer. On the science, unquestionably, one would want to combine these as early as possible because we think there’s orthogonal benefit, as I described previously, and one would expect they’d get combined with checkpoint inhibitors in addition. As a drug developer, you want to get confidence first that each individual has merit on its own before you go into the combination. I think for us it’s critical to demonstrate that the cancer vaccine is such in combination with the PD1 inhibitor can actually mediate responses. I think once we get to that stage, we will obviously have a keen interest in pursuing the right combinations with the inhibitors depending on where they are at that point in time. Alec, did that answer your question?
Alec Stranahan:
Yes, that’s great. Thank you.
Operator:
There are no further questions. I’d like to turn the call back over to Stéphane Bancel for any closing remarks.
Stéphane Bancel:
Thank you for joining us today and for your questions. We look forward to seeing you during our upcoming third annual R&D Day in New York City. This meeting will be held during the morning of September 12. Have a wonderful day. Thank you.
Operator:
Ladies and gentlemen, thank you for participating in today’s conference. This does conclude the program and you may all disconnect. Everyone, have a great day.
Operator:
Good morning, and welcome to the Moderna First Quarter 2019 Conference Call. [Operator Instructions]. Please be advised that the call is being recorded. At this time, I would like to turn the call over to Lavina Talukdar, Head, Investor Relations at Moderna. Please proceed.
Lavina Talukdar:
Thank you, Operator. Good morning, and welcome to Moderna's First Quarter 2019 Conference Call to discuss financial results and business update. You can access the press release issued this morning as well as the slides that we will be reviewing by going to the Investors section of our website at www.modernatx.com. Today, on this call, we have Stéphane Bancel, our Chief Executive Officer; Stephen Hoge, our President; Tal Zaks, our Chief Medical Officer; and Lorence Kim, our Chief Financial Officer. Before we begin, I would like to remind everyone that this conference call will include forward-looking statements. Please see Slide 2 of the accompanying presentation and our SEC filings for important risk factors that could cause our actual performance and results to differ materially from those expressed or implied in these forward-looking statements. We undertake no obligation to update or revise the information provided on this call as a result of new information or future results or developments. I will now pass the call over to Stéphane.
Stéphane Bancel:
Thank you, Lavina. Good morning or good afternoon. As you know, we believe that mRNA has the potential to become a new class of medicines. And since Moderna's inception, we have worked hard to be the company that could become the leader in this field. We believe our mRNA medicines have a potential to address large unmet medical needs and to treat diseases that are not addressable by recombinant proteins or small molecules. Due to the platform nature of mRNA, we believe our mRNA medicines provide a higher probability of technical success and faster timelines to clinical trials and to the market relative to traditional medicines. We also believe that the manufacturing capital intensity of mRNA is materially lower than recombinant proteins. We have, of course, the manufacturing at commercial scale. We will be similar to small molecule injectable in order to achieve and deliver on our goals, we continue to focus on managing the risk, especially technology risk and biology risk. We believe that programs will be in the same modality of similar technology risk, meaning that once we derisk a sentinel program, they are important breakthroughs. We believe our chikungunya antibody program will be an important clinical readout, as it uses the same formulation technology as our MMA program, our most advanced rare disease candidate. As articulated before, our corporate focus is on 3 priorities
Tal Zaks:
Thank you, Stéphane. Let me begin with reviewing our progress by modality and I'm going to start with the prophylactic vaccines. We're capping in our mRNA vaccine for respiratory syncytial virus or RSV, one of the most common causes of respiratory disease in both the very young and the elderly. In collaboration with Merck, we designed our vaccine to encode a membrane-anchored version of stabilized pre-fusion F protein, which elicits a neutralizing antibody response. In the inset, we show the intended design of the mRNA formulated in an LNP. We worked with Merck to identify and advance improvements to the RSV vaccine, which has led us to the identification of mRNA-1172. This second vaccine has an improved RSV antigen resulting in an enhanced potency in preclinical models as well as a proprietary formulation developed by Merck. On the right, we have outlined data from a preclinical study in African green monkeys comparing serum neutralization titers of mRNA-1172 to mRNA-1777. In this study, mRNA-1172 was shown to be significantly more potent than mRNA-1777. As Stéphane mentioned, the IND for this new vaccine was filed earlier this month and mRNA-1777 will not move forward into the planned Phase IIa study at this time. Let me now turn to the rest of the prophylactic vaccines. Overall to date, approximately 1,000 healthy volunteers have been enrolled across 7 Phase I trials at doses of up to 400 micrograms. The emerging safety and tolerability profile remains consistent with that of marketed adjuvant vaccines. We've shown promising data from 5 Phase I programs within the prophylactic vaccines modality and we continue to make progress. For CMV, the first 3 dose levels are now fully enrolled, and healthy volunteers have started to being dosed at 300 micrograms for the fourth dose level in that study. And VZV, based on an assessment of the commercial opportunity, research priorities and other factors, as Stéphane has discussed, Merck has discontinued preclinical development of mRNA-1278. Merck has returned the rights back to us and we will not continue development at this time. Turning to our cancer vaccines. We announced that we will present Phase I updates for personalized cancer vaccine at the upcoming American Society of Clinical Oncology meeting or ASCO. As a reminder, our personalized cancer vaccines are being studied as a single agent in patients with resected tumors and in combination with pembrolizumab in patients with unresectable solid tumors. The National Cancer Institute will also present data from its Phase I study of PCV, NCI-4650, a monotherapy for patients with advanced metastatic cancers. The abstract titles and presentation times can be found in the press release that went out this morning. In addition, we're continuing enrollment for our PCV trial and preparing for the randomized Phase II, while the team at Merck is gearing up to enroll patients on the KRAS Phase I trial. For intratumoral immuno-oncology, we continue to enroll our OX40 ligand and triplet trials and prepare for the OX40 ligand Phase II cohort in ovarian cancer. Also, at AACR, our partner AstraZeneca shared preclinical data that supported advancing MEDI1191 or the IL12 program into a Phase I trial. AstraZeneca will lead an open-label multicenter study of intratumoral injections of IL12 alone or in combination with a checkpoint inhibitor. Finally, the local regenerative therapeutic modalities, which includes AZD8601, our mRNA encoding for VEGF-A, AstraZeneca recently obtained clinical trial authorization to open a clinical site in the Netherlands. As a reminder, the Phase I study showed that AZD8601 was well tolerated and led to the translation of functional VEGF-A protein, which caused a dose-dependent increase in blood flow where it was injected. Moving next to our systemic secreted therapeutics. Let me provide some context on our first program in this modality mRNA-1944, including for chikungunya antibody. As we were assessing the biology risk for our systemic secreted therapeutics, we began the development efforts by encoding a monoclonal antibody with the goal of inducing transient passive immunity in the recipient. An antibody would represent a complex protein as it requires the co-translation of 2 separate mRNAs in the correct intracellular folding and eventual secretion into the bloodstream. From a safety perspective, you would expect an antiviral monoclonal antibody to be fixed, and pharmacologically, we should be able to easily measure its blood levels. This would then allow us to quantify the PK/PD relationship. In other words, how much mRNA is required to produce how much protein in a relatively straightforward manner. The lipid nanoparticle delivery system used for chikungunya antibody or mRNA-1944 is shared with our sentinel rare disease program, methylmalonic acidemia. The Phase I design for this study is shown on Slide 18. We're conducting it in healthy volunteers, so it should give us a read on the safety and tolerability of this formulation. The pharmacological goal that I mentioned is to determine how much antibody we can produce as a result of systemically administered mRNA and whether we may be able to offer people passive immunity against infection by this virus, which today has neither a vaccine nor specific therapies available. We recently completed enrollment of all 8 subjects in the second dose cohort in healthy volunteers treated at 0.3 milligram per kilogram in this Phase I study and I look forward to sharing more once we have a complete picture of the data. Also, our chikungunya antibody preclinical data was just accepted for publication in the Journal of Science Immunology. We're continuing to move our Fabry and Relaxin programs towards clinical trials and look forward to providing you with an update at a later date. Finally, let me turn to our systemic intracellular therapeutics. From methylmalonic acidemia or MMA, we're currently in the process of initiating sites for the Phase I trial. We have 45 patients enrolled in our MaP natural history study for methylmalonic acidemia and propionic acidemia. 26 of these are MMA patients and 19 are PA patients. In addition, the European Commission has adopted the recommendation from the Committee of Orphan Medicinal Products for orphan designation for mRNA-3927, our PA development candidate, further supporting its advancement. We continue to progress mRNA-3927 for propionic acidemia and mRNA-3283 for phenylketonuria with the goal of bringing them to the clinic. I'm really excited about the opportunity for the new development candidate within our systemic intracellular therapeutic modalities and will now turn it over to Stephen who will introduce this candidate for glycogen storage disease type 1a or GSD1a.
Stephen Hoge:
Thanks, Tal. So glycogen storage disease type 1a or GSD1a is a rare inherited metabolic disorder resulting from a deficiency in an enzyme called glucose 6-phosphatase, or G6Pase for short. The G6Pase enzymes involved in the metabolic pathways to allow the liver and, to a lesser extent the kidney, to maintain the level of glucose in the blood during fasting. After a couple of hours in a normal healthy person, most of the glucose from your last meal have been consumed by tissues. So as glucose levels start to fall, most of the tissues in our bodies will convert to using -- should convert from using glucose for energy to other sources such as lipids or triglycerides. But cells in our brains are unable to make that switch and are dependent upon glucose to survive. So our livers take over the responsibility for making and releasing glucose into the bloodstream to keep the brain alive. This process involves two different metabolic processes
Lorence Kim:
Thank you, Stephen. In today's press release, we reported our first quarter 2019 financial results. Please note these results are unaudited. We ended Q1 2019 with cash, cash equivalents and investments of $1.55 billion, and this compares to $1.69 billion at the end of 2018. Let me highlight two cash flow metrics, which provide a direct view on cash use. These are better given the quantity of deferred revenue stock-based compensation and depreciation that's embedded in our operating income. Net cash used in operations was approximately $144 million for the first quarter of 2019, that compares to $111 million for the first quarter of 2018. Please note that Q1 of 2019 and 2018 include $22 million and $25 million, respectively of in-licensing payments. Secondly, cash used for purchases of the property and equipment was $8 million for Q1 2019 compared to $32 million for Q1 '18. Revenue for Q1 2019 was $16 million as compared to $29 million for Q1 '18. Note that on January 1, 2019, we adopted the new revenue recognition standard ASC606, using the modified retrospective transition method applied to those contracts, which were not completed as of January 1. The decrease in total revenue was mainly attributable to the decrease in collaboration revenue across all of our strategic alliances, particularly AstraZeneca and Merck, driven by the adoption of the new revenue standard. Total revenue under the previous revenue recognition standard would have been $38 million for Q1 2019. R&D expenses for Q1 2019 were approximately $131 million compared to $90 million for Q1 '18, the increase was primarily due to an increase in personnel-related costs, including stock-based comp, mainly driven by an increase in number of employees supporting research and development programs, an increase in clinical trial and manufacturing costs, an increase in lab supplies and materials for preclinical studies and clinical trials, and an increase in consulting and outside services costs. G&A expenses for Q1 2019 were approximately $27 million compared to $16 million in Q1 of '18. That increase was mainly attributable to an increase in personnel-related costs, including stock-based compensation, primarily driven by an increase in the number of employees and consulting and outside services cost, both of which were related to operating as a publicly traded company. So with that, I'll hand the call back over to Stéphane.
Stéphane Bancel:
Thank you, Lorence. To close our remarks, I would like to reiterate that our team is very focused on the execution of all the 3 priorities
Operator:
[Operator Instructions]. Our first question comes from Matthew Harrison of Morgan Stanley.
Matthew Harrison:
I guess, two from me. So on RSV, can you maybe just clarify a little bit what specifically is Merck hoping to achieve with the new formulation and the neoantigen selection relative to what you've achieved so far? And then, I guess, just remind us when you started a similar point with 1777, how long did it take you to progress to being Phase II ready? And then I have a follow-up.
Stéphane Bancel:
Sure, Stephen?
Stephen Hoge:
Sure. Obviously, we're pretty excited about that one, the 1172 and we're really pleased that Merck's filed the IND already, and it's moving quickly. You saw the summary of the data in primates. It's been hard to argue with the substantial improvement that we demonstrated there. 1777, just referencing that slide, shows superior titers to a natural RSV infection, which was there in gray. But 1172 was qualitatively and quantitatively several-fold higher. And so in the face of those improvements, we felt compelled and that it makes a ton of sense to move forward. Now what are those improvements just quickly, I think, you hit on them, Matt, but the first is antigen design. This is still a pre-fusion F protein, but it's been further stabilized to improve potency and immunogenicity, so that's a desirable feature. And then the second scientific improvement went into the program, obviously, is the switch from a legacy lipid nanoparticle to a Merck proprietary formulation, that has some improved features. Those improved features, I'll let Merck talk about at a future date, but we think they are a part of what's happening in terms of the improvement here. And ultimately, the combination of antigen and formulation leads to the qualitative improvement you see there. It does, at this point, mean a pause to 1777 moving to Phase IIa as described, but as far -- our goal is the best RSV vaccine possible, with the highest probability of success and as much as we wanted to see the 1777 data, it's clearly the right answer to look for 1172 first, given the multifold improvement that's been demonstrated in African green monkeys. In general, our experience has translated and you know that well. Things that we've been able to see in terms of preclinical species like primates for vaccines has translated into humans. So if this translates in Phase I relatively quickly for 1172, which we hope and expect it will, this is the vaccine that we should advance further in development to Phase II and Phase III.
Tal Zaks:
And Matthew, this is Tal. In terms of time lines, if you look historically, it typically takes you 12 to 18 months for a Phase I vaccine trial. It's done in healthy volunteers, so it's a fairly straightforward simple design. I can't give you a forward-looking guidance here, but I would note that Merck has recently filed its IND and they will be leading the execution of this trial. Obviously, the team there knows what they're doing in terms of vaccine trials.
Matthew Harrison:
Okay. Great. That's helpful. And then the second question is on chikungunya. I know you've entered the second cohort. Do you have any view on how many cohorts you need to get through before you feel like you have enough data to talk about with that modality?
Tal Zaks:
This is Tal. The trial that's currently designed has 4 dose levels, that was predicted based on our expectation that translates the data from the nonhuman primate that you've seen. I think, we're on track with that design. We've been enrolling fairly consistently, and as we said, we're at the second dose level.
Operator:
And our next question comes from Salveen Richter of Goldman Sachs.
Ross Weinreb:
It's Ross on for Salveen. Just in terms of mRNA-1172 and RSV and the decision to move forward the more optimized program, is this something that we should kind of think that may occur in other programs going forward as you continue to test new formulations of your current compounds in clinical candidates?
Stephen Hoge:
So we continue -- with any program we advance, we're always looking at whether there are improvements. And yes, there are always -- there sometimes can be small improvements you can make. I think the question gets more complicated where you start to see 5-fold improvements kind of like we're talking here with 1172 where qualitatively that improvement is so strong and in the background of what's been happening in RSV vaccines more generally, it makes sense to bring the best vaccine forward first and quickly, particularly when you are this close after the lead, in this case 1777. That isn't something that we can predict happening broadly across many programs or efforts. And in general, as you can see with other vaccines in our pipeline, we haven't been doing this. And so just pointing at the history here, it's more of by exception a situation where this has been happening and that's probably what we would expect about the future.
Tal Zaks:
This is Tal. I mean, just to add a couple of points. I think it's one of the fascinating things of beauty of our platform that we have relatively rapid improvements and that the coding-like nature of mRNA lends itself to those improvements. So if you look at our history, for example, in the personalized cancer vaccine, we've been able to take it to the next level by increasing the number of neoantigens from 20 to 34. And as the team worked through what that means both in terms of mRNA length and sequence optimization, we found a way within the clinic to actually introduce that within the current clinical paradigm. Now, of course, the PCV is a unique type of drug in that regard and I give the FDA a lot of credit for working with us to enable that. So where we can, we always try to optimize for the best medicine that we can and it depends on the specifics of the product as to what's the best way to do it.
Ross Weinreb:
Great. And then I just have one follow-up on the chikungunya antibody. How can we think about the clinical bar here as we approach the data readout, like what are the level -- the antibody levels that we should expect to see to confer this passive immunity in humans?
Tal Zaks:
So as you will see in the preclinical data that as we said that's recently been accepted for publication, so it should come out soon, the target expression for this antibody is about 1 microgram per mL in terms of what we think is going to be a therapeutic concentration or a concentration that will -- should prevent from infection based on the totality of the preclinical data. And that was easily achieved within the dose levels that we've cleared for in the toxicology studies and the preclinical studies. So from a target product profile for this application what we're shooting for is 1 microgram per mL. If you look at where most therapeutic antibodies live in terms of concentrations, if you pull the FDA label for REMICADE, the trough levels are between 0.5 per microgram per mL and about 6 microgram per mL. So it's within the range of what monoclonal antibodies have achieved for therapeutic levels from many indications.
Operator:
And our next question comes from Cory Kasimov from JPMorgan.
Cory Kasimov:
I have two of them for you as well. So first one is I'm curious how this RSV decision changes how you're thinking about your CMV program, if at all? And maybe what the latest in terms of time lines for when we should expect data from that fourth dosing cohort from CMV? And then I have one follow-up.
Tal Zaks:
Yes, so this is Tal. It does not actually change, in my view, one iota how we think of CMV or what we're doing about CMV. I think if you look at the totality of our data, to date, we've shown a pretty consistent safety and tolerability profile across the 1,000 or so healthy volunteers that have been dosed across the various programs. And so now, it's about figuring out the best vaccine and the best dose for any given application. In that regard, CMV -- the dose for CMV and the activity for CMV needs to be defined in the context of a CMV clinical trial. I can't give you a forward guidance. Obviously, clinical trials are at the mercy of a lot of external factors, but if you look at our past performance and if you see where this trial is, you can sort of connect the dots. And we're at the fourth dose level here. The goal with this vaccine is really to -- since it's a Phase I and you're dose escalating, we want to make sure, we're getting a good sense of what would be a tolerable profile to take forward. And so as we continue to dose escalate, at some point, we will say, okay, we think we've reached the bar for what should be tolerable. That's how you defined it in the Phase I; at which point, we'll wait and make sure that we understand the immunogenicity and then we'll show the data.
Cory Kasimov:
Okay. And then, the follow-up question is on MMA and curious when we should expect the first patient to be dosed in that program? And can you just kind of give maybe a little overview on what that dose escalation strategy might be there?
Tal Zaks:
Yes. So as we said, we're -- the IND has been open for a couple of months now. It takes you time to talk to sites, initiate the sites. The first cohort here following discussions with regulators is going to be adolescents, aged 12 to 18. So obviously, we're in active discussion with the sites to get them initiated, get the study ongoing. And at the same time, talking to the investigators and even the patient advocacy groups here to try and identify the patients. In terms of the dosing regimen that you asked, this is sort of a traditional dose exploration. We're going in from the very first dose in this disease at a dose that can be effective. It's one of the requirements to do a pediatric development. And so even at the initial starting dose, which is 0.2 milligrams per kilogram, we think we -- there's an opportunity to have benefit and we'll dose escalate from there based on the totality of tolerability and what we see in terms of biomarker effect.
Operator:
And our next question comes from Ted Tenthoff of Piper Jaffray.
Edward Tenthoff:
Thanks for hosting us yesterday up in Cambridge, that was really interesting. I want to pick up on Cory's first question just with respect to CMV development path, and maybe you can kind of outline sort of where you would expect to take the Phase I data sets and what next studies would be? And then I just have one quick follow-up please.
Tal Zaks:
So I think the Phase I -- the goal is to demonstrate obviously tolerability, safety and immunogenicity. In our case, the critical pieces to be able to demonstrate the immunogenicity against both components of this vaccine. As you would recall, we both have a complex pentameric protein here, which I think is a unique feature you can do with mRNA and against gB, so we need to figure that out. Now in terms of a full development path, I think there is work ahead of us to talk to FDA and other regulators to understand this is obviously a very high unmet need, but it is an unmet need within a context that's obviously challenging to develop, which is trying to prevent maternal transmission to babies in utero is what we're trying to achieve here at the end of the day. Getting there is not simple. Over the years, there have been various discussions of other companies with the FDA, so there is a good body of precedent for what full development looks like. We're just starting down the path of those conversations. I will say that the effort on our side is led by Wellington Sun as you may recall, he was the prior Division Director for Vaccines for FDA for the past decade before joining our team. So he is very busy these days actually putting this together, discussing with other experts in the field and planning on that next set of conversations to understand what that full development path would look like.
Edward Tenthoff:
Great. That's super helpful. And then one more for you Tal, if I may. Just with respect to the triplet program for IL, for cancer, so I was pleased to see that you guys are expanding to the second dose. I think this thing is going to really carry a big hammer and safety is going to be the one of the primary focus areas. Was there anything you saw to date that gives you any pause there? Or how is that looking from an early standpoint from a safety side?
Tal Zaks:
So look, it's hard for me to comment on a trial that's in progress. The data continues to come in, and we're obviously evaluating it on an ongoing basis. I can tell you that we're at the second dose level. As a reminder, for OX40 ligand alone, for the membrane protein, we got up to 8-milligram, which was the intended dose and that seemed to be tolerated well. So in terms of the contribution of OX40 ligand itself and the lipids nanoparticle delivery technology, I'm pretty comfortable with their tolerability profile. I think your question is correct with locally secreted cytokines, do you get to a level that is worrisome? I don't know yet. I would point that the preclinical data in totality was pretty supportive of going to much higher doses. I think that's the unique ability of mRNA to set up this local paracrine effect within the tumor with minimal spillover into the blood. These are obviously things we'll measure, so we will be looking at the expression of these proteins, both in the tumors and systemically to see if they're detectable. We will be looking at the typical safety parameters [indiscernible] labs, et cetera. It's frankly, too early to say.
Operator:
And our next question comes from Alan Carr of Needham & Company.
Alan Carr:
I wish you could -- I'm hoping you can tell us a little bit more about some of these rare disease programs that -- or preclinical programs that you presented just recently. It looks like a bit of a shotgun strategy in terms of the number that you have running in parallel, how do you decide which ones to bring forward? And maybe give us a sense of expectation in terms of when these might move forward in the clinic? And then the second one, I'm wondering if you can elaborate a bit more about this proprietary formulation developed by Merck for 1172? To what extent do you, I guess, work with Merck on developing new delivery formulations for in both those specific programs? And how do they -- with Merck and then they translate to other programs that are not licensed during collaboration with Merck?
Stephen Hoge:
Sure. So the two questions. So first on the ASGCT abstracts, 5 of which were presented to a wide range of academic collaborators and institutions, as you saw. We do have a very active research collaborations with academia across a very wide range of rare diseases. What you saw was a sampling of that activity, but it was ready for presentation by many of the academic labs. By participating with those folks, we absolutely commit to them being able to present that data at the appropriate fora as well as publish it. And so you will see a steady flow of those sorts of things, that doesn't necessarily translate into our portfolio strategy or programs that we're taking forward into development. I'll point to our own publication record there, even in the last couple of years, there was a number of programs that we published in high-impact journals that are not currently in our development pipeline. What we choose to take into development goes through a different series of filters in terms of prioritization as well as our view on unmet need and developability. So I can't comment on whether or not you should expect those things to be translating forward, and in fact, I would only say that you should expect to continue to see a large amount of that activity both in publication and presentation collaborations with academia. Onto the 1172 delivery vehicle, so we -- obviously, we've had a multiyear collaboration with Merck where we do talk a lot about things we're learning to enable the best development of the programs we're partnering with on them, including the vaccines like RSV. We -- I can't comment specifically on the improvements that went into the 1172 formulations that Merck was developing, but -- or is developing, but obviously we published last year in a paper on our website, Tasset et al, some of the deficiencies with the legacy lipid nanoparticle formulations in terms of local tolerability in the vaccine context. That underpinned a lot of our efforts to -- several years ago to develop our own proprietary lipid nanoparticle delivery technologies that are actually already in programs like CMV and hMPV PIV. RSV had started before that as an effort and so as we shared that data with Merck long before we published it, they made their own determinations about what that might mean for that development program. And as I said, I'll let them, at the appropriate time, talk about any improvements in specifics, but obviously, they are aware of all of work together. Now we do share information back and forth. I think part of your question was do we collaborate on these things, and obviously, we do. We were sharing that information with Merck long before the publication, and as they make improvements and step-forwards in formulation then that becomes a two way street, and so we're excited about it.
Operator:
And our next question comes from Geoff Meacham of Barclays.
Geoffrey Meacham:
For the new GSP program, the disease ideology is pretty complicated with multiple organs involved. I guess, the question is, just help us with how you're thinking about the regulatory path down the road post proof of concept? I imagine there's been some discussion of endpoints already. And I have a follow-up.
Tal Zaks:
Geoff, this is Tal. It's a great question. This disease is a different one, as you rightly point out. The unmet need here is -- it's not just the multiple organs that are affected, if you think about the patient experience and the quality of life, it's the frequent feeding at night, but if you actually meet one of these patients, which I had the opportunity to sit down, when I sit for a meeting for 2 hours, I typically take a cup of coffee. A GSD1a patient who sits for a 2-hour meeting needs to have a cup of cornstarch that they're drinking. And if you think about that day in, day out your entire life, there's obviously a high unmet need there that's manifest in many dimensions. Now to your point about regulatory path and endpoints, I think it's still early days. This is just the declaration of our intent in the development candidate, there's work ahead of us. We have to go talk to the agency. We have to go figure out the strategy for the Phase I first and then we have to understand what is going to be the development path to bring and demonstrate that we can actually impact not just the basic biology of these diseases, as you've shown quite remarkably, I think in the mouse model, but actually, how do we demonstrate that we're able to address all those manifold manifestations of this disease spanning from the biochemistry all through the quality of life, preventing relatively rare but very severe events, but at the same time actually changing their day-to-day quality of life in a much more consistent fashion. That's a lot of work that's still ahead of us.
Geoffrey Meacham:
Got you. Okay. That's helpful. And then, on the cancer vaccine and the IO programs, I know you're in an enrollment ramp at this point, but maybe can you speak to whether we'll see any clinical updates at the upcoming ASCO meeting?
Tal Zaks:
So the -- you know it's a medical meeting. You like to show the data that you have, I'll point to you that the -- to the abstracts once they will be live I think in a couple of weeks, so they will describe the contents of the data that we will be sharing there.
Operator:
[Operator Instructions]. Our next question comes from Ying Huang of Bank of America.
Ying Huang:
I want to ask about the GSD1a program. First of all, I guess, now we have 5 rare disease programs in the pipeline. Does that suggest an increasing focus on this modality compared to others? Secondly, there is a competitor, which is conducting a trial in gene therapy for this disease specifically. Can you talk about maybe pros and cons versus the mRNA approach? And given the data we obtained recently from [indiscernible], what do you think you can improve on?
Stéphane Bancel:
This is Stéphane, I am going to take your first question and Stephen will be talk about gene therapy. So as we indicated since early 2018, rare disease is an important point of focus for the company, obviously because of the unmet medical need, because of a quick path to approval, and also because what we believe is a lower biology risk against some of the things we're trying to do. So as you know, as we beat the pipeline of the company, as I said in my introduction remarks, we focus very much on managing risk. And so the way we think about our pipeline now in development is, we have some very interesting vaccine in infectious disease that got a big unmet medical need that I think we have a [indiscernible] to be able to help a lot of people. With five other programs now we think we have a nice portfolio of IO and what we were looking for is great execution in the clinic to see what we can do for patients with those programs. And given the more "straightforward" biology of rare disease where clearly missing instruction for the DNA, and as you have seen in a lot of publication work that we have done, there's very exciting preclinical model data. We really want to focus on rare diseases and this is consistent with the strategy we are outlining now and 1.5 years ago. And also what you saw at the ASGCT a few weeks ago, there's a lot of work going on there. This is a very important area for the company.
Stephen Hoge:
Yes. And on GSD1a, and gene therapy specifically -- or generally, we -- starting with mRNA, our focus is on dose-dependent pharmacology. I think the defining feature of our platform is our ability to have predictable dose-dependent pharmacology. You give a dose today, you get a response. You give it next week, you get the same response. If you need a more response tomorrow, I give twice the dose, you should expect to get twice the response and that's what we mean by sort of dose-dependent pharmacology functions like a traditional drug. We focus a lot of our rare disease efforts in metabolic diseases where you would expect the needs of the patient based on metabolism will change over time. Sometimes in a week, sometimes over years as they age. And in particular, as they're growing and young can be highly dynamic. And so we focus on looking at the disease like GSD1a, mRNA is another example of this, where we are expecting to need to titrate to the dose response that's desirable to manage that disease. Now there are, as you mentioned, a gene therapy approach going after GSD1a as we speak and we really don't view those as necessarily competitive. In fact, we think these are different tools that physicians will use hopefully in combination for different purposes to help patients manage this disease. We can easily imagine a world where sort of analogous to insulin is sort of basal long-acting forms, but you're still managing the disease day in, day out with -- by testing blood glucose for week in, week out by controlling for it. That would give, frankly, the physician we hope that -- personally, I would hope that they're successful and we're successful because more tools for clinicians as they are treating patients will be a great thing. So we don't necessarily view it as competitive. We view it as adjunctive and we're focusing on different areas of pharmacology.
Operator:
And I'm not showing any further questions at this time. I would now like to turn the call back over to Stéphane Bancel for any further remarks.
Stéphane Bancel:
Well, thank you very much for joining us today and for your questions. We look forward to talking and to seeing many of you in the coming weeks, including at the ASCO around the PCV posters. Have a great day.
Operator:
Thank you. Ladies and gentlemen, thank you for participating in today's conference. This does conclude today's program and you may all disconnect. Everyone, have a wonderful day.