Covid-19 Vaccines — Immunity, Variants, Boosters | NEJM – nejm.org

Prepare to become a physician, build your knowledge, lead a health care organization, and advance your career with NEJM Group information and services.
Stay connected to what’s important in medical research and clinical practice
Subscribe to the most trusted
and influential source of
medical knowledge
Already a subscriber? Renew or Sign in
Subscribe
or Renew

Selected specialties
View all specialties
Selected Topics
View all topics
Selected Multimedia
View all multimedia
Current Issue
Recent Issues
Browse full issue index
Recently Published Articles
Browse recently published
View all learning/CME
Other NEJM Group Learning
Subscribe
or Renew

Review ArticleFranklin H. Epstein Lecture
Metrics

The coronavirus disease 2019 (Covid-19) pandemic has claimed an estimated 15 million lives, including more than 1 million lives in the United States alone. The rapid development of multiple Covid-19 vaccines has been a triumph of biomedical research, and billions of vaccine doses have been administered worldwide. Challenges facing the Covid-19 vaccine field include inequitable vaccine distribution, vaccine hesitancy, waning immunity, and the emergence of highly transmissible viral variants that partially escape antibodies. This review summarizes the current state of knowledge about immune responses to Covid-19 vaccines and the importance of both humoral and cellular immunity for durable protection against severe disease.
The immune system is broadly divided into the innate and adaptive immune systems. Innate immune responses are the first line of defense against viruses and are rapidly triggered when cellular pattern-recognition receptors, such as toll-like receptors, recognize pathogen-associated molecular patterns. Innate antiviral immunity includes secretion of type I interferons, antiviral cytokines, and certain cellular responses, including neutrophils, monocytes and macrophages, dendritic cells, and natural killer cells.1
Adaptive immune responses, the second line of defense against viruses, involve antigen-specific recognition of viral epitopes. Adaptive immunity includes two complementary branches of the immune system: humoral immunity and cellular immunity. Humoral immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) includes antibodies that bind the SARS-CoV-2 spike protein and either neutralize the virus or eliminate it through other effector mechanisms.2,3 Cellular immunity to SARS-CoV-2 includes virus-specific B cells and T cells, which provide long-term immunologic memory and rapidly expand on reexposure to antigen. B cells produce antibodies, CD8+ T cells directly eliminate virally infected cells, and CD4+ T cells provide help to support the immune responses.
Humoral and cellular immune responses contribute to protection against SARS-CoV-2 infection and coronavirus disease 2019 (Covid-19). Plus signs denote the relative importance of antibodies and T cells for protection in each category of disease severity, with more plus signs indicating greater importance.
For acute viral infections, including SARS-CoV-2, it is likely that neutralizing antibodies are critical for blocking acquisition of infection, whereas a combination of humoral and cellular immune responses most likely controls viral replication after infection and prevents progression to severe disease, hospitalization, and death (Figure 1).4-7 For a highly transmissible SARS-CoV-2 variant that largely escapes neutralizing antibodies, cellular immunity may be particularly important for long-term protection against severe disease.
Early data from nonhuman primate studies showed the protective efficacy of both natural immunity8 and vaccine immunity9-12 against experimental SARS-CoV-2 challenge. These findings provided preclinical support for the rapid clinical development of SARS-CoV-2 vaccines. The World Health Organization (WHO) reported that more than 300 Covid-19 vaccines were in preclinical or clinical development as of May 6, 2022. Ten Covid-19 vaccines, reflecting eight distinct vaccine products, have been approved by the WHO for global use (see Table S1 in the Supplementary Appendix, available with the full text of this article at NEJM.org). These vaccines involve four distinct vaccine platforms: inactivated virus vaccines (Sinopharm’s Covilo, Sinovac’s CoronaVac, and Bharat Biotech’s Covaxin), messenger RNA (mRNA) vaccines (Moderna’s Spikevax mRNA-1273 and Pfizer–BioNTech’s Comirnaty BNT162b2), adenovirus vector–based vaccines (AstraZeneca’s Vaxzevria and Covishield ChAdOx1 and Johnson & Johnson–Janssen’s Ad26.COV2.S), and adjuvanted protein vaccines (Novavax’s Nuvaxovid and Covovax NVX-CoV2373). Additional vaccines have been approved by other regulatory bodies and are also in widespread clinical use, but a comprehensive summary of all Covid-19 vaccines is beyond the scope of this review. It has been estimated that global Covid-19 vaccination saved approximately 20 million lives during the first year of the vaccine rollout.13
Shown is the percentage of fully vaccinated persons by country as of July 8, 2022. Data are from the New York Times (www.nytimes.com/interactive/2021/world/covid-vaccinations-tracker.html).
In the United States, four vaccines have been authorized for either full approval or emergency use: the mRNA vaccines BNT162b2 and mRNA-1273, the adenovirus vector–based vaccine Ad26.COV2.S, and most recently the adjuvanted protein vaccine NVX-CoV2373. Randomized, placebo-controlled, phase 3 trials in the United States, conducted before the emergence of the omicron variant, showed initial protective efficacy of 94 to 95% against symptomatic Covid-19 infection with the two-shot BNT162b2 vaccine, the two-shot mRNA-1273 vaccine, and the two-shot Ad26.COV2.S vaccine, as well as 72% efficacy with the one-shot Ad26.COV2.S vaccine14-17 (Table 1). The mRNA vaccines have been used most widely in the United States and Europe, but their use in developing countries has been relatively limited — in part because of their cost, freezing requirements, distribution logistics, and business priorities — and has resulted in stark global health inequities. More than 70% of eligible persons in the United States and most other developed countries have been fully vaccinated, whereas less than 15% of persons in Africa have been fully vaccinated (Figure 2). A more equitable vaccine rollout that achieved the WHO target of 40% vaccination coverage in developing countries in 2021 would have saved an estimated 600,000 lives.13 The adenovirus vector–based vaccines have greater stability than the mRNA vaccines and no freezing requirements and have been used more extensively in developing countries.
The Food and Drug Administration and the Centers for Disease Control and Prevention (CDC) have recently restricted the use of Ad26.COV2.S in the United States because of the rare but serious occurrence of vaccine-induced immune thrombotic thrombocytopenia (VITT), also called thrombosis with thrombocytopenia syndrome (TTS). VITT has developed in 54 persons (9 of whom died), reflecting a rate of 3 to 4 cases per 1 million vaccinated persons.19,20 However, adenovirus vector–based vaccines remain first-line vaccines in much of the developing world, and the rate of VITT may be lower in South Africa than in the United States.18,21,22 VITT has also been reported in Europe with ChAdOx1, at a rate of 13 to 39 cases per 1 million vaccinated persons.23,24 In the United States, VITT has also been reported in 3 patients who received mRNA-1273 (1 of whom died).19,25
Myocarditis and pericarditis have been reported as complications with BNT162b2 and mRNA-1273, at a rate of 52 to 137 cases per 1 million vaccinated adolescent boys and young men after the second dose,26-29 with at least 10 reported deaths.27,28,30,31 The incidence rate of myocarditis within 7 days after the second mRNA dose has been reported as 566 cases per 1 million person-years.32 Although most cases of vaccine-induced myocarditis are mild, severe complications can occur, and cardiac magnetic resonance imaging changes have been reported to persist in a substantial fraction of young men for at least 3 to 8 months after recovery.33 Both thrombosis and myocarditis occur far more frequently after Covid-19 infection than after Covid-19 vaccination.
The BNT162b2 and mRNA-1273 vaccines induce outstanding short-term neutralizing antibody responses and protective efficacy.14,15 However, the high initial serum neutralizing antibody titers induced by mRNA vaccines wane by 3 to 6 months and decline further by 8 months, with a half-life of approximately 60 days.34-38 In contrast to BNT162b2 and mRNA-1273, Ad26.COV2.S induces lower initial neutralizing antibody titers,16,39,40 but these neutralizing antibody responses and clinical effectiveness are fairly durable for at least 8 months.38,41-43 At 6 to 8 months, antibody responses are fairly similar with BNT162b2, mRNA-1273, and Ad26.COV2.S.38,43 Data from real-world effectiveness studies are largely concordant with these immunologic data and have shown initially higher protection with BNT162b2 and mRNA-1273 than with Ad26.COV2.S, but these differences narrowed after several months.41,44-46 Thus, BNT162b2 and mRNA-1273 induce high initial antibody titers that wane after a few months, whereas Ad26.COV2.S induces lower initial antibody responses with greater durability.
The waning of immunity with mRNA vaccines is correlated with increased breakthrough infections in vaccinated persons, initially exemplified by the large cluster of breakthrough infections with the SARS-CoV-2 delta variant in July 2021 in Provincetown, Massachusetts.47 In vaccinated persons with breakthrough infections, particularly robust immune responses, known as hybrid immunity, have been shown to develop. These findings suggest that population immunity to SARS-CoV-2 will continue to increase through a combination of widespread vaccination and infection.48 Genomic and epidemiologic data from this outbreak showed evidence of transmission between fully vaccinated persons.49
Cellular immune responses are induced by both mRNA vaccines and adenovirus vector–based vaccines and have shown greater durability than serum antibody titers. Germinal center B cells have been reported to persist for at least 6 months after BNT162b2 vaccination.50,51 CD8+ T-cell responses are particularly high after Ad26.COV2.S vaccination, with durability for at least 6 to 8 months.38,52 Because CD8+ T-cell responses control viral replication after infection,4,6,53-55 it is likely that SARS-CoV-2 vaccines will continue to provide substantial protection against severe disease even after serum neutralizing antibody titers wane.
In immunocompromised persons, both antibody and T-cell responses to Covid-19 vaccines are reduced, with the degree of reduction dependent on the extent and type of immunosuppression.56,57 In these populations, additional vaccine doses and prophylactic treatment with monoclonal antibodies are recommended.
Panel A shows global and weekly average case counts and frequencies of SARS-CoV-2 variants over time, and Panel B shows regional frequencies. The data are based on Pango lineage designations of sequences shared through the global collection of SARS-CoV-2 sequences and made available through the GISAID database from March 1, 2020, to July 8, 2022 (www.gisaid.org). Data are from Bette Korber and James Theiler at Los Alamos National Laboratories (https://cov.lanl.gov/content/index).
In the spring of 2020, the predominant global form of the original virus rapidly transitioned to a variant that carried four mutations in the SARS-CoV-2 genome, including a single D614G point mutation in the spike protein that conferred a fitness advantage.58,59 Subsequently, multiple waves of SARS-CoV-2 variants have emerged that replaced prior variants, with new variants often showing increased transmissibility and greater antibody escape (Figure 3). In late 2020, the alpha (B.1.1.7), beta (B.1.351), and gamma (P.1) variants emerged in the United Kingdom, South Africa, and Brazil, respectively. These variants were then replaced globally by the delta (B.1.617.2) variant, which emerged in India in the summer of 2021. In late 2021, the highly transmissible omicron (B.1.1.529) variant emerged in Africa and abruptly became the most prevalent virus globally. In contrast to the 4 mutations in delta, omicron has more than 50 mutations, including more than 30 mutations in the spike protein, which result in substantial escape from neutralizing antibody responses elicited by vaccination or prior infection with a non-omicron variant.60-64 The omicron lineage has rapidly splintered into subvariants BA.1, BA.1.1, BA.2, BA.2.12.1, BA.4, and BA.5 (Figure 3). Neutralizing antibody titers against BA.5, which is currently the predominant variant in the United States, are decreased by a factor of approximately 3 as compared with titers against BA.1 and BA.2.65-67
Multiple studies have shown that neutralizing antibodies induced by all primary vaccine regimens show little cross-reactivity with omicron but that boosting leads to a substantial increase in omicron neutralizing antibodies.68,69 However, these increased neutralizing antibody titers, as well as clinical effectiveness, have been shown to wane by 4 months after a third mRNA immunization.62,70,71 After a fourth mRNA immunization, protection against infection with SARS-CoV-2 omicron has been reported to wane after just 4 weeks, although protection against severe disease lasts longer.72 Hybrid immunity from both vaccination and infection provides greater and more durable protection than either alone.73,74
In contrast with the limited cross-reactivity of vaccine-induced neutralizing antibodies to omicron, T-cell responses induced by vaccines have very good (>80%) cross-reactivity to omicron75-77 and to prior variants.6,39,53 These data suggest that cellular immunity to SARS-CoV-2 variants remains largely intact. During the omicron surge in South Africa, the two-shot Ad26.COV2.S and the two-shot BNT162b2 vaccines provided 72% and 70% protection against hospitalization and 82% and 70% protection against admission to an intensive care unit, respectively18 (Table 1). This robust protection, essentially in the absence of high titers of omicron neutralizing antibodies, suggests the importance of other immune measures, including CD8+ T-cell responses and possibly other functional antibody responses, in providing protection against severe disease with a viral variant that largely escapes neutralizing antibodies.
Covid-19 breakthrough rates (no. of cases per 100,000 vaccinated persons) are shown from April 4, 2021, to May 21, 2022, for persons who received the BNT162b2, mRNA-1273, or Ad26.COV2.S vaccines in the United States. Data are from the Centers for Disease Control and Prevention (https://covid.cdc.gov/covid-data-tracker/#rates-by-vaccine-status).
In the United States, composite data from the CDC show that the BNT162b2, mRNA-1273, and Ad26.COV2.S vaccines all provided substantial protection against the delta surge in the fall of 2021 and against the omicron surges in the winter of 2021–2022 and in the spring of 2022 (Figure 4). Breakthrough rates per 100,000 vaccinated persons were higher with Ad26.COV2.S than with the mRNA vaccines during the delta surge but were lower with Ad26.COV2.S than with the mRNA vaccines during the omicron surge, potentially reflecting the durability of Ad26.COV2.S-induced immunity. These data are consistent with real-world effectiveness studies that have shown that the mRNA vaccines are initially more effective than the Ad26.COV2.S vaccine but that these differences diminish or disappear after several months.18,41,44-46
Early preclinical studies in nonhuman primates identified both neutralizing and other functional antibodies as correlates of vaccine protection against SARS-CoV-2 challenge.8,9,12 Adoptive transfer studies with purified IgG confirmed that antibodies alone were sufficient to block infection in both nonhuman primates and hamsters, provided that the antibodies were administered at a sufficiently high dose.54,78,79
In vivo CD8 depletion studies in nonhuman primates also showed that CD8+ T cells contributed to protection when antibody titers were subprotective.54 Moreover, vaccine failure against an omicron challenge in nonhuman primates was associated with low levels of both omicron neutralizing antibodies and CD8+ T cells.80 These data suggest that antibodies alone can block infection if antibody titers against the infecting virus strain are sufficiently high but that a combination of humoral and cellular immunity is critical for virologic control after breakthrough infection.
Analyses of immune correlates from the phase 3 clinical trials of mRNA-1273 and Ad26.COV2.S confirmed that antibody titers correlated with protection against symptomatic Covid-19 infection.81,82 However, these studies were performed before the emergence of the omicron variant, and T-cell responses were not included in these correlate analyses. It is possible that correlates of protection against highly transmissible viral variants that largely escape neutralizing antibody responses, such as omicron, skew more heavily toward cellular immunity. Moreover, CD8+ T cells have been shown to correlate with survival among patients with Covid-19 and hematologic cancer.7 As discussed above, both BNT162b2 and Ad26.COV2.S provided robust protection against severe disease during the omicron surge in South Africa in the absence of high titers of omicron neutralizing antibodies.18 Moreover, Ad26.COV2.S provided protection against hospitalization and death during the beta and delta surges in South Africa in the absence of high titers of neutralizing antibodies against these variants.21
Taken together, these data suggest that neutralizing antibodies are primarily responsible for blocking acquisition of SARS-CoV-2 infection but that both antibody and CD8+ T-cell responses are critical for preventing severe disease (Figure 1). Current vaccines provide only modest protection against infection and transmission with the omicron variant, even at peak immunity after boosting. Moreover, it is likely that neutralizing antibody titers may need to be substantially higher to protect against infection with the highly transmissible omicron variant than were needed to protect against infection with prior variants. In contrast to neutralizing antibodies, vaccine-induced CD8+ T-cell responses are highly cross-reactive against omicron and most likely contribute substantially to protection against severe disease. Future research should focus on the role of mucosal humoral and cellular immunity at the site of inoculation, which may play a critical role in protection against SARS-CoV-2 infection.
The expectation that Covid-19 vaccines would prevent acquisition of infection and block onward transmission was based on initial data in 2020 (before the emergence of viral variants) that showed high neutralizing antibody titers and robust protective efficacy at peak immunity after mRNA vaccination. However, given the substantial waning of serum neutralizing antibody titers and the emergence of variants with increased transmissibility and antibody escape, it would be reasonable now to recalibrate goals for Covid-19 vaccines. Current vaccines may not provide high-level, sustained protection against infection or transmission with omicron, even after multiple boosts and also after the introduction of updated omicron-specific vaccines. Instead, the most important goal of Covid-19 vaccination should be to provide long-term protection against severe disease, hospitalization, and death from current and future variants.
Booster recommendations should therefore take into account not only peak neutralizing antibody titers but also durable prevention of severe Covid-19 disease. Such protection will probably require a combination of humoral and cellular immunity, with an emphasis on long-term rather than short-term immune responses. However, to date, the field has focused largely on short-term neutralizing antibody responses. The potential role of an omicron-containing booster is currently being explored, but a study in nonhuman primates showed that an omicron-specific mRNA vaccine was not better than the original mRNA-1273 vaccine for protection against omicron challenge.83 Early clinical studies have shown that boosting with bivalent mRNA vaccines containing both ancestral and omicron BA.1 spike immunogens induced peak omicron neutralizing antibody titers that were less than twice the peak titers induced by boosting with the original mRNA vaccines. Thus, clinical benefits of the updated boosters as compared with the current vaccines are not clear. Heterologous prime-boost (“mix-and-match”) regimens, which involve combinations of mRNA and Ad26 vaccines, are also being investigated as a strategy for improving the magnitude and durability of humoral and cellular immunity, as compared with either type of vaccine alone.52,84,85 In addition, early research on the development of pan-sarbecovirus and pan-betacoronavirus vaccines is under way.
Boosting every 4 to 6 months to maintain high serum neutralizing antibody titers may not be a practical or desirable long-term strategy. Boosting with mRNA vaccines is also not risk-free. Moreover, frequent boosting recommendations may worsen “booster fatigue” in the general population, given that to date only 47% of eligible persons in the United States have received any booster dose. Expert opinion on the benefits of frequent boosters remains divided, communications from public health authorities have been viewed as confusing and overpromising, and vaccine hesitancy remains a major challenge. Frequent booster recommendations may also distract from the critical goal of vaccinating the large number of unvaccinated persons in the United States and throughout the world and may further exacerbate global health inequities.
Plans for boosters should therefore be based on robust scientific data that show substantial and sustained increases in prevention of severe disease rather than on short-term increases in neutralizing antibody titers. Enhanced community engagement and implementation research may also reduce vaccine misinformation. Ideally, Covid-19 boosters should be recommended no more than annually and preferably less frequently, and a diversity of booster options should be available to the public. The use of vaccine platforms with improved durability would be highly desirable.
The Covid-19 pandemic appears to be transitioning from a hyperacute phase to an endemic phase. Current Covid-19 vaccines are less effective at blocking infection with the omicron variant than at blocking infection with prior variants, but protection against severe disease remains largely preserved. The primary goal of Covid-19 vaccines should be to provide long-term protection against severe disease, hospitalization, and death. It is therefore important for studies of Covid-19 vaccines and boosters to evaluate not only short-term neutralizing antibody titers but also durability of antibody responses, memory B-cell responses, and cross-reactive T-cell responses.
Disclosure forms provided by the author are available with the full text of this article at NEJM.org.
This article was published on August 31, 2022, at NEJM.org.
I thank Mark Zeidel, Bette Korber, James Theiler, Stanley Plotkin, Nelson Michael, and Bruce Walker for helpful discussions, assistance with earlier versions of figures, and critical review of the manuscript.
From Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, and the Ragon Institute of MGH, MIT, and Harvard, Cambridge — both in Massachusetts.
Dr. Barouch can be contacted at or at the Center for Virology and Vaccine Research, E/CLS-1043, 330 Brookline Ave., Beth Israel Deaconess Medical Center, Boston, MA 02115.
1. Gasteiger G, D’Osualdo A, Schubert DA, Weber A, Bruscia EM, Hartl D. Cellular innate immunity: an old game with new players. J Innate Immun 2017;9:111125.
2. Bartsch YC, Tong X, Kang J, et al. Omicron variant Spike-specific antibody binding and Fc activity are preserved in recipients of mRNA or inactivated COVID-19 vaccines. Sci Transl Med 2022;14(642):eabn9243eabn9243.
3. Chung AW, Kumar MP, Arnold KB, et al. Dissecting polyclonal vaccine-induced humoral immunity against HIV using systems serology. Cell 2015;163:988998.
4. Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021;184:861880.
5. Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 2020;183(4):9961012.e19.
6. Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 2021;374(6572):abm0829abm0829.
7. Bange EM, Han NA, Wileyto P, et al. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med 2021;27:12801289.
8. Chandrashekar A, Liu J, Martinot AJ, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science 2020;369:812817.
9. Mercado NB, Zahn R, Wegmann F, et al. Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature 2020;586:583588.
10. Corbett KS, Flynn B, Foulds KE, et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N Engl J Med 2020;383:15441555.
11. van Doremalen N, Lambe T, Spencer A, et al. ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature 2020;586:578582.
12. Yu J, Tostanoski LH, Peter L, et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science 2020;369:806811.
13. Watson OJ, Barnsley G, Toor J, Hogan AB, Winskill P, Ghani AC. Global impact of the first year of COVID-19 vaccination: a mathematical modelling study. Lancet Infect Dis 2022 June 23 (Epub ahead of print).
14. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med 2020;383:26032615.
15. Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med 2021;384:403416.
16. Sadoff J, Gray G, Vandebosch A, et al. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med 2021;384:21872201.
17. Hardt K, Vandebosch A, Sadoff J, et al. Efficacy and safety of a booster regimen of Ad26.COV2.S vaccine against Covid-19. January 31, 2022 (https://www.medrxiv.org/content/10.1101/2022.01.28.22270043v1). preprint.
18. Gray G, Collie S, Goga A, et al. Effectiveness of Ad26.COV2.S and BNT162b2 vaccines against omicron variant in South Africa. N Engl J Med 2022;386:22432245.
19. See I, Lale A, Marquez P, et al. Case series of thrombosis with thrombocytopenia syndrome after COVID-19 vaccination — United States, December 2020 to August 2021. Ann Intern Med 2022;175:513522.
20. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA 2021;325:24482456.
21. Bekker L-G, Garrett N, Goga A, et al. Effectiveness of the Ad26.COV2.S vaccine in health-care workers in South Africa (the Sisonke study): results from a single-arm, open-label, phase 3B, implementation study. Lancet 2022;399:11411153.
22. Takuva S, Takalani A, Garrett N, et al. Thromboembolic events in the South African Ad26.COV2.S vaccine study. N Engl J Med 2021;385:570571.
23. Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and thrombocytopenia after ChAdOx1 nCoV-19 vaccination. N Engl J Med 2021;384:21242130.
24. Pavord S, Scully M, Hunt BJ, et al. Clinical features of vaccine-induced immune thrombocytopenia and thrombosis. N Engl J Med 2021;385:16801689.
25. Sangli S, Virani A, Cheronis N, et al. Thrombosis with thrombocytopenia after the messenger RNA-1273 vaccine. Ann Intern Med 2021;174:14801482.
26. Ling RR, Ramanathan K, Tan FL, et al. Myopericarditis following COVID-19 vaccination and non-COVID-19 vaccination: a systematic review and meta-analysis. Lancet Respir Med 2022;10:679688.
27. Oster ME, Shay DK, Su JR, et al. Myocarditis cases reported after mRNA-based COVID-19 vaccination in the US from December 2020 to August 2021. JAMA 2022;327:331340.
28. Mevorach D, Anis E, Cedar N, et al. Myocarditis after BNT162b2 mRNA vaccine against Covid-19 in Israel. N Engl J Med 2021;385:21402149.
29. Witberg G, Barda N, Hoss S, et al. Myocarditis after Covid-19 vaccination in a large health care organization. N Engl J Med 2021;385:21322139.
30. Heymans S, Cooper LT. Myocarditis after COVID-19 mRNA vaccination: clinical observations and potential mechanisms. Nat Rev Cardiol 2022;19:7577.
31. Verma AK, Lavine KJ, Lin C-Y. Myocarditis after Covid-19 mRNA vaccination. N Engl J Med 2021;385:13321334.
32. Klein NP, Lewis N, Goddard K, et al. Surveillance for adverse events after COVID-19 mRNA vaccination. JAMA 2021;326:13901399.
33. Schauer J, Buddhe S, Gulhane A, et al. Persistent cardiac magnetic resonance imaging findings in a cohort of adolescents with post-coronavirus disease 2019 mRNA vaccine myopericarditis. J Pediatr 2022;245:233237.
34. Falsey AR, Frenck RW Jr, Walsh EE, et al. SARS-CoV-2 neutralization with BNT162b2 vaccine dose 3. N Engl J Med 2021;385:16271629.
35. Widge AT, Rouphael NG, Jackson LA, et al. Durability of responses after SARS-CoV-2 mRNA-1273 vaccination. N Engl J Med 2021;384:8082.
36. Doria-Rose N, Suthar MS, Makowski M, et al. Antibody persistence through 6 months after the second dose of mRNA-1273 vaccine for Covid-19. N Engl J Med 2021;384:22592261.
37. Pegu A, O’Connell SE, Schmidt SD, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science 2021;373:13721377.
38. Collier AY, Yu J, McMahan K, et al. Differential kinetics of immune responses elicited by Covid-19 vaccines. N Engl J Med 2021;385:20102012.
39. Alter G, Yu J, Liu J, et al. Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans. Nature 2021;596:268272.
40. Stephenson KE, Le Gars M, Sadoff J, et al. Immunogenicity of the Ad26.COV2.S vaccine for COVID-19. JAMA 2021;325:15351544.
41. Polinski JM, Weckstein AR, Batech M, et al. Durability of the single-dose Ad26.COV2.S vaccine in the prevention of COVID-19 infections and hospitalizations in the US before and during the delta variant surge. JAMA Netw Open 2022;5(3):e222959e222959.
42. Barouch DH, Stephenson KE, Sadoff J, et al. Durable humoral and cellular immune responses 8 months after Ad26.COV2.S vaccination. N Engl J Med 2021;385:951953.
43. Zhang Z, Mateus J, Coelho CH, et al. Humoral and cellular immune memory to four COVID-19 vaccines. March 21, 2022 (https://www.biorxiv.org/content/10.1101/2022.03.18.484953v1). preprint.
44. Zheutlin A, Ott M, Sun R, et al. Durability of protection against COVID-19 breakthrough infections and severe disease by vaccines in the United States. January 6, 2022 (https://www.medrxiv.org/content/10.1101/2022.01.05.22268648v1). preprint.
45. Rosenberg ES, Dorabawila V, Easton D, et al. Covid-19 vaccine effectiveness in New York State. N Engl J Med 2022;386:116127.
46. Lin D-Y, Gu Y, Wheeler B, et al. Effectiveness of Covid-19 vaccines over a 9-month period in North Carolina. N Engl J Med 2022;386:933941.
47. Gharpure R, Sami S, Vostok J, et al. Multistate outbreak of SARS-CoV-2 infections, including vaccine breakthrough infections, associated with large public gatherings, United States. Emerg Infect Dis 2022;28:3543.
48. Collier AY, Brown CM, McMahan KA, et al. Characterization of immune responses in fully vaccinated individuals after breakthrough infection with the SARS-CoV-2 delta variant. Sci Transl Med 2022;14(641):eabn6150eabn6150.
49. Siddle KJ, Krasilnikova LA, Moreno GK, et al. Transmission from vaccinated individuals in a large SARS-CoV-2 Delta variant outbreak. Cell 2022;185(3):485492.e10.
50. Turner JS, O’Halloran JA, Kalaidina E, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature 2021;596:109113.
51. Kim W, Zhou JQ, Horvath SC, et al. Germinal centre-driven maturation of B cell response to mRNA vaccination. Nature 2022;604:141145.
52. Atmar RL, Lyke KE, Deming ME, et al. Homologous and heterologous Covid-19 booster vaccinations. N Engl J Med 2022;386:10461057.
53. Dan JM, Mateus J, Kato Y, et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 2021;371(6529):eabf4063eabf4063.
54. McMahan K, Yu J, Mercado NB, et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature 2021;590:630634.
55. Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell 2020;181(7):14891501.e15.
56. Collier AY, Yu J, McMahan K, et al. Coronavirus disease 2019 messenger RNA vaccine immunogenicity in immunosuppressed individuals. J Infect Dis 2022;225:11241128.
57. Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody response to 2-dose SARS-CoV-2 mRNA vaccine series in solid organ transplant recipients. JAMA 2021;325:22042206.
58. Korber B, Fischer WM, Gnanakaran S, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell 2020;182(4):812827.e19.
59. Hou YJ, Chiba S, Halfmann P, et al. SARS-CoV-2 D614G variant exhibits efficient replication ex vivo and transmission in vivo. Science 2020;370:14641468.
60. Cele S, Jackson L, Khoury DS, et al. Omicron extensively but incompletely escapes Pfizer BNT162b2 neutralization. Nature 2022;602:654656.
61. Iketani S, Liu L, Guo Y, et al. Antibody evasion properties of SARS-CoV-2 omicron sublineages. Nature 2022;604:553556.
62. Liu L, Iketani S, Guo Y, et al. Striking antibody evasion manifested by the Omicron variant of SARS-CoV-2. Nature 2022;602:676681.
63. Yu J, Collier AY, Rowe M, et al. Neutralization of the SARS-CoV-2 omicron BA.1 and BA.2 variants. N Engl J Med 2022;386:15791580.
64. Viana R, Moyo S, Amoako DG, et al. Rapid epidemic expansion of the SARS-CoV-2 omicron variant in southern Africa. Nature 2022;603:679686.
65. Hachmann NP, Miller J, Collier AY, et al. Neutralization escape by SARS-CoV-2 omicron subvariants BA.2.12.1, BA.4, and BA.5. N Engl J Med 2022;387:8688.
66. Cao Y, Yisimayi A, Jian F, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by omicron infection. Nature 2022 June 17 (Epub ahead of print).
67. Khan K, Karim F, Ganga Y, et al. Omicron sub-lineages BA.4/BA.5 escape BA.1 infection elicited neutralizing immunity. May 1, 2022 (https://www.medrxiv.org/content/10.1101/2022.04.29.22274477v1). preprint.
68. Carreño JM, Alshammary H, Tcheou J, et al. Activity of convalescent and vaccine serum against SARS-CoV-2 omicron. Nature 2022;602:682688.
69. Nemet I, Kliker L, Lustig Y, et al. Third BNT162b2 vaccination neutralization of SARS-CoV-2 omicron infection. N Engl J Med 2022;386:492494.
70. Pajon R, Doria-Rose NA, Shen X, et al. SARS-CoV-2 omicron variant neutralization after mRNA-1273 booster vaccination. N Engl J Med 2022;386:10881091.
71. Ferdinands JM, Rao S, Dixon BE, et al. Waning 2-dose and 3-dose effectiveness of mRNA vaccines against COVID-19-associated emergency department and urgent care encounters and hospitalizations among adults during periods of delta and omicron variant predominance — VISION Network, 10 states, August 2021–January 2022. MMWR Morb Mortal Wkly Rep 2022;71:255263.
72. Bar-On YM, Goldberg Y, Mandel M, et al. Protection by a fourth dose of BNT162b2 against omicron in Israel. N Engl J Med 2022;386:17121720.
73. Altarawneh HN, Chemaitelly H, Ayoub HH, et al. Effects of previous infection and vaccination on symptomatic omicron infections. N Engl J Med 2022;387:2134.
74. Goldberg Y, Mandel M, Bar-On YM, et al. Protection and waning of natural and hybrid immunity to SARS-CoV-2. N Engl J Med 2022;386:22012212.
75. Liu J, Chandrashekar A, Sellers D, et al. Vaccines elicit highly conserved cellular immunity to SARS-CoV-2 omicron. Nature 2022;603:493496.
76. Keeton R, Tincho MB, Ngomti A, et al. T cell responses to SARS-CoV-2 spike cross-recognize omicron. Nature 2022;603:488492.
77. Tarke A, Coelho CH, Zhang Z, et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from alpha to omicron. Cell 2022;185(5):847859.e11.
78. Tostanoski LH, Chandrashekar A, Patel S, et al. Passive transfer of Ad26.COV2.S-elicited IgG from humans attenuates SARS-CoV-2 disease in hamsters. NPJ Vaccines 2022;7:22.
79. Corbett KS, Nason MC, Flach B, et al. Immune correlates of protection by mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. Science 2021;373(6561):eabj0299eabj0299.
80. Chandrashekar A, Yu J, McMahan K, et al. Vaccine protection against the SARS-CoV-2 omicron variant in macaques. Cell 2022;185(9):15491555.e11.
81. Gilbert PB, Montefiori DC, McDermott AB, et al. Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science 2022;375:4350.
82. Fong Y, McDermott AB, Benkeser D, et al. Immune correlates analysis of a single Ad26.COV2.S dose in the ENSEMBLE COVID-19 vaccine efficacy clinical trial. April 12, 2022 (https://www.medrxiv.org/content/10.1101/2022.04.06.22272763v1). preprint.
83. Gagne M, Moliva JI, Foulds KE, et al. mRNA-1273 or mRNA-omicron boost in vaccinated macaques elicits similar B cell expansion, neutralizing responses, and protection from omicron. Cell 2022;185(9):15561571.e18.
84. Tan CS, Collier AY, Yu J, et al. Durability of heterologous and homologous COVID-19 vaccine boosts. JAMA Netw Open 2022;5(8):e2226335e2226335.
85. Liu X, Munro APS, Feng S, et al. Persistence of immunogenicity after seven COVID-19 vaccines given as third dose boosters following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK: three month analyses of the COV-BOOST trial. J Infect 2022;84:795813.

10.1056/NEJMra2206573-t1
Protective Efficacy of Coronavirus Disease 2019 (Covid-19) Vaccines against the Ancestral Viral Strain in the United States and against the Omicron Variant in South Africa.
Data on protective efficacy of vaccines against symptomatic Covid-19 in the United States are from randomized, placebo-controlled phase 3 clinical trials.14-17 Interim efficacy data before the emergence of the omicron variant are shown for each vaccine. The global efficacy of Ad26.COV2.S was lower, at 66% for the one-shot vaccine and 75% for the two-shot vaccine, as a result of the beta, lambda, and mu variants in Africa and South America.
Shown are data on clinical effectiveness of BNT162b2 and Ad26.COV2.S against hospitalization and admission to the intensive care unit (ICU) during the omicron surge in South Africa (November 15, 2021, to January 14, 2022).18 Data are for effectiveness 1 to 2 months after the second immunization. ND denotes no data.
Humoral and cellular immune responses contribute to protection against SARS-CoV-2 infection and coronavirus disease 2019 (Covid-19). Plus signs denote the relative importance of antibodies and T cells for protection in each category of disease severity, with more plus signs indicating greater importance.
Shown is the percentage of fully vaccinated persons by country as of July 8, 2022. Data are from the New York Times (www.nytimes.com/interactive/2021/world/covid-vaccinations-tracker.html).
Panel A shows global and weekly average case counts and frequencies of SARS-CoV-2 variants over time, and Panel B shows regional frequencies. The data are based on Pango lineage designations of sequences shared through the global collection of SARS-CoV-2 sequences and made available through the GISAID database from March 1, 2020, to July 8, 2022 (www.gisaid.org). Data are from Bette Korber and James Theiler at Los Alamos National Laboratories (https://cov.lanl.gov/content/index).
Covid-19 breakthrough rates (no. of cases per 100,000 vaccinated persons) are shown from April 4, 2021, to May 21, 2022, for persons who received the BNT162b2, mRNA-1273, or Ad26.COV2.S vaccines in the United States. Data are from the Centers for Disease Control and Prevention (https://covid.cdc.gov/covid-data-tracker/#rates-by-vaccine-status).
September 15, 2022
N Engl J Med 2022; 387:1011-1020
DOI: 10.1056/NEJMra2206573

Tap into groundbreaking research and clinically relevant insights
Already a subscriber? Sign In or Renew
Back to top

source