Global regulators agree on way forward to adapt COVID-19 vaccines to emerging variants


International regulators have published a report today highlighting the outcomes of their discussions on COVID-19 vaccines and the need for and strategy to update their composition based on the emerging evidence on coronavirus SARS-CoV-2 variants and lessons learned from previous vaccine updates. The workshop, co-chaired by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA), was organised under the umbrella of the International Coalition of Medicines Regulatory Authorities (ICMRA)

and took place on 8 May 2023.

Currently authorised vaccines continue to be effective at preventing hospitalisation, severe disease and death due to COVID-19. However, protection against infection wanes over time and as new SARS-CoV-2 variants emerge. Preliminary data show that COVID-19 vaccines adapted to the currently circulating strains improve immunity to recently emerged variants, such as XBB descendent lineages. 

Meeting participants discussed the available scientific evidence on epidemiology, seroprevalence (i.e. the number of persons in a population who test positive for a specific disease based on blood serum measurements) and vaccine performance, and key regulatory considerations related to the adaptation of authorised or new COVID-19 vaccines against emerging coronavirus variants. There is a broad agreement that vaccine formulations for the upcoming winter season in the northern hemisphere should include only one virus strain and be based on the XBB family of Omicron subvariants (such as XBB.1.5). International regulators also highlighted that such monovalent vaccines could be used for both booster and primary vaccinations (the latter, for example, only in young children below 4-5 years of age). They noted that only data on manufacturing and quality of the vaccine and laboratory data would be required for the authorisation or approval of strain changes for the already authorised COVID-19 vaccines, provided that post-authorisation data regarding vaccine quality, effectiveness, immunogenicity and safety data are collected. 

The meeting built on the experience and knowledge gained from a series of ICMRA workshops on COVID-19 vaccine development and virus variants held over the past three years. Participants included representatives of international regulators as well as experts from the World Health Organization (WHO).

COVID-19 vaccines caused DECLINE in life expectancy in the U.S.


Life expectancy across the U.S. has declined, and this drop coincides with the introduction of the Wuhan coronavirus (COVID-19) vaccines.

According to Elizabeth Arias, a researcher for the Centers for Disease Control and Prevention (CDC), the COVID-19 pandemic impacted the U.S. life expectancy. “[It will take] some time before we’re back where we were in 2019,” she said.

The Daytona Beach News-Journal reported that according to CDC numbers, life expectancy for the entire U.S. was 77 years in 2020. However, this dropped to 76.4 years by 2021 – in time for the introduction of the COVID-19 injections that year.

Heart disease was the leading cause of death in the U.S. for 2021, and this is also linked to the COVID-19 injections. It can be noted that many of the most widely reported side-effects from COVID-19 injections have been related to the cardiovascular system. Countless people have suffered from either a heart attack or a stroke after being injected. (Related: New “smoking gun” analysis shows dramatic excess mortality rise linked to COVID-19 vaccines.)

One study by the Cedars-Sinai Medical Center in California found that heart attack deaths climbed for all age groups during 2020 and 2021. But the biggest jump in heart attack deaths was seen in the group aged 25 to 44 at 29 percent.

COVID-19 injections shorten men’s lives by 24 years

The CDC’s own data also disclosed the true dangers of the vaccines. According to the Daily Expose, men injected with the mRNA COVID-19 vaccines could see as much as 24 years taken off their lifespan as a result.

The public health agency’s all-cause mortality data shows that each dose of the COVID-19 vaccine a person got raised their mortality rate by seven percent in 2022, compared to 2021. In other words, people injected with five doses were 35 percent more likely to die in 2022 than in 2021.

The Expose compared the COVID-19 vaccines to “slow-acting genetic poison” based on this data, given the fact that people do not appear to be recovering from the damage caused by earlier vaccines when it comes to excess mortality. It ultimately warned that a person injected with five doses would be 350 percent more likely to pass away in 2031, and a shocking 700 percent more likely to die in 2041 than an unvaccinated person.

Dr. Robert Califf, commissioner of the Food and Drug Administration (FDA), acknowledged this reduced life expectancy. “We are facing extraordinary headwinds in our public health with a major decline in life expectancy,” he wrote on X. “The major decline [of life expectancy] in the U.S. is not just a trend; I’d describe it as catastrophic.”

Not surprisingly, Califf stopped short of pinning the blame on the COVID-19 vaccines. Many of those who dare to suggest that the injections are responsible for excess deaths find themselves being censored.

In one instance, whistleblower Barry Young from New Zealand shared data from the country’s health agency that pointed to a strong link between the COVID-19 injections and excess mortality. According to the data he shared, the vaccines killed more than 10 million around the world.

But this revelation came with a steep price, as Young was arrested and now faces prison time. Nevertheless, the whistleblower said he shared the data as it blew his mind. He also wanted experts to analyze it and make people aware of what is happening.

COVID-19 vaccines caused DECLINE in life expectancy in the U.S.


Life expectancy across the U.S. has declined, and this drop coincides with the introduction of the Wuhan coronavirus (COVID-19) vaccines.

According to Elizabeth Arias, a researcher for the Centers for Disease Control and Prevention (CDC), the COVID-19 pandemic impacted the U.S. life expectancy. “[It will take] some time before we’re back where we were in 2019,” she said.

The Daytona Beach News-Journal reported that according to CDC numbers, life expectancy for the entire U.S. was 77 years in 2020. However, this dropped to 76.4 years by 2021 – in time for the introduction of the COVID-19 injections that year.

Heart disease was the leading cause of death in the U.S. for 2021, and this is also linked to the COVID-19 injections. It can be noted that many of the most widely reported side-effects from COVID-19 injections have been related to the cardiovascular system. Countless people have suffered from either a heart attack or a stroke after being injected. (Related: New “smoking gun” analysis shows dramatic excess mortality rise linked to COVID-19 vaccines.)

One study by the Cedars-Sinai Medical Center in California found that heart attack deaths climbed for all age groups during 2020 and 2021. But the biggest jump in heart attack deaths was seen in the group aged 25 to 44 at 29 percent.

COVID-19 injections shorten men’s lives by 24 years

The CDC’s own data also disclosed the true dangers of the vaccines. According to the Daily Expose, men injected with the mRNA COVID-19 vaccines could see as much as 24 years taken off their lifespan as a result.

The public health agency’s all-cause mortality data shows that each dose of the COVID-19 vaccine a person got raised their mortality rate by seven percent in 2022, compared to 2021. In other words, people injected with five doses were 35 percent more likely to die in 2022 than in 2021.

The Expose compared the COVID-19 vaccines to “slow-acting genetic poison” based on this data, given the fact that people do not appear to be recovering from the damage caused by earlier vaccines when it comes to excess mortality. It ultimately warned that a person injected with five doses would be 350 percent more likely to pass away in 2031, and a shocking 700 percent more likely to die in 2041 than an unvaccinated person.

Dr. Robert Califf, commissioner of the Food and Drug Administration (FDA), acknowledged this reduced life expectancy. “We are facing extraordinary headwinds in our public health with a major decline in life expectancy,” he wrote on X. “The major decline [of life expectancy] in the U.S. is not just a trend; I’d describe it as catastrophic.”

Not surprisingly, Califf stopped short of pinning the blame on the COVID-19 vaccines. Many of those who dare to suggest that the injections are responsible for excess deaths find themselves being censored.

In one instance, whistleblower Barry Young from New Zealand shared data from the country’s health agency that pointed to a strong link between the COVID-19 injections and excess mortality. According to the data he shared, the vaccines killed more than 10 million around the world.

But this revelation came with a steep price, as Young was arrested and now faces prison time. Nevertheless, the whistleblower said he shared the data as it blew his mind. He also wanted experts to analyze it and make people aware of what is happening.

Research: COVID-19 Vaccines May Trigger Takotsubo Cardiomyopathy


Side effects and adverse events related to COVID-19 vaccines continue to be identified.

Research: COVID-19 Vaccines May Trigger Takotsubo Cardiomyopathy
COVID-19 vaccines may trigger Takotsubo cardiomyopathy. (Freepik)

Various vaccines have been rapidly developed and administered in response to the COVID-19 pandemic. However, accompanying the widespread vaccination efforts, there has been a notable increase in the occurrence of side effects and adverse events related to COVID-19 vaccines. Research has unveiled a potential association between COVID-19 vaccines and Takotsubo cardiomyopathy, with two fatalities among 16 patients.

The clinical presentation of Takotsubo cardiomyopathy resembles that of acute myocardial infarction, with common symptoms including acute chest pain and breathlessness. Its hallmark is impaired left ventricular function, typically occurring after intense emotional or physical stressors such as the death of a loved one, traumatic events, or severe illness. This condition, first identified by Japanese physician Dr. Hikaru Sato in 1990, is named “Takotsubo” due to the balloon-like bulging of the left ventricle, resembling the octopus-catching pot used in Japan.

Outside of Japan, Takotsubo cardiomyopathy is also referred to as stress cardiomyopathy, apical ballooning syndrome, or broken heart syndrome.

A Case Study

In August, a case report was published in the journal Cureus detailing the experience of a 59-year-old woman who developed Takotsubo cardiomyopathy after receiving a booster dose of the COVID-19 vaccine. The patient experienced persistent dyspnea for six hours, prompting her visit to the emergency department. According to the patient’s account, she had been experiencing intermittent chest pain for the past two days, described as a stabbing sensation that progressively intensified with each episode but did not radiate to other areas. Exertion worsened the pain, and there was no relief method. The patient had received the Moderna vaccine booster dose three days prior.

The patient did not have a fever and remained conscious, with a blood oxygen saturation of 89 percent and blood pressure at 150/90 mmHg. Crackling and rattling sounds known as crepitations were detected in her lungs. A COVID-19 polymerase chain reaction test yielded a negative result. The emergency electrocardiogram showed ST-segment elevation, a chest X-ray revealed pulmonary edema and an ultrasound indicated reduced left ventricular systolic function, with an estimated ejection fraction of 30 percent. Additionally, there was moderate hypokinesia (abnormally diminished motor activity) in the apex and anterior wall of the heart.

The patient continued to experience tachycardia and blood pressure fluctuations, leading to hemodynamic instability due to fluid overload and ultimately resulting in cardiac shock. The medical team administered intravenous injections of norepinephrine and dobutamine for treatment. As no other etiologies were identified, she was diagnosed with Takotsubo cardiomyopathy.

The patient showed improvement and was discharged on the sixth day but continued to experience persistent tachycardia, requiring treatment with metoprolol, a medication for the treatment of high blood pressure.

Before receiving the vaccine, the patient had a history of hyperlipidemia, hypothyroidism, and celiac disease. Additionally, she had smoked for five years but quit 15 years ago, and she had no history of alcohol consumption or drug abuse.

The researchers stated that the pathophysiology of COVID-19 vaccine-induced Takotsubo cardiomyopathy is not yet clear, but that  “several theories have been proposed.” The immune response triggered by COVID-19 vaccines may, for some individuals, “result in an exaggerated inflammatory cascade, leading to endothelial dysfunction, microvascular dysfunction, and myocardial injury.” Vaccination may also stimulate the release of pro-inflammatory factors such as interleukin-6. Additionally, the stress response induced by COVID-19 vaccination could potentially “dysregulate the autonomic nervous system, contributing to the development of cardiac dysfunction.”

Takotsubo Cardiomyopathy Poses a Life-Threatening Risk

The association between COVID-19 vaccines and Takotsubo cardiomyopathy is not widely known, with only a few reported cases. On Dec. 11, a peer-reviewed study published in the journal Cureus consolidated and analyzed the evidence concerning COVID-19 vaccine-induced Takotsubo cardiomyopathy.

The researchers conducted a literature search and included 15 case reports involving a total of 16 patients. Among them, 14 individuals received mRNA vaccines (Pfizer, Moderna), while two received viral vector vaccines (AstraZeneca). Seven patients developed Takotsubo cardiomyopathy after the first dose and seven after the second dose.

All patients exhibited elevated cardiac troponin levels, abnormal electrocardiogram findings, and reduced left ventricular ejection fraction on echocardiograms. The most predominant symptom among patients was chest pain, followed by dyspnea and nausea. Eventually, 14 patients recovered and were discharged, while two of the patients died.

The researchers noted that 87.5 percent of patients recovered and were discharged, indicating that Takotsubo cardiomyopathy occurring after vaccination is mostly “transient and reversible.” However, the death of 2 of the patients highlights the “potentially life-threatening nature of this vaccine-related adverse event.”

The paper’s authors urge clinicians to consider the possibility of Takotsubo cardiomyopathy, especially among recipients of mRNA vaccines when presented with patients experiencing chest pain or dyspnea symptoms after vaccination.

The study also mentioned that vaccines developed for COVID-19 have various side effects, including pain and swelling at the injection site, fever, headache, myalgia (muscle pain), fatigue, and nausea.

FDA Authorizes Updated COVID Boosters to Target Newest Variants


The FDA on Wednesday granted emergency use authorization to Omicron-specific COVID-19 vaccines made by Pfizer/BioNTech and Moderna.

The agency cited data to support the safety and efficacy of this next generation of mRNA vaccines targeted toward variants of concern.

If you’ve been waiting to get a variant-specific booster shot, you may be in luck as early as next week.

The Pfizer EUA corresponds to the company’s combination booster shot that includes the original COVID-19 vaccine as well as a vaccine designed to protect against the most recent Omicron variants, BA.4 and BA.5.

The Moderna combination vaccine will contain both the firm’s original COVID-19 vaccine and a vaccine to protect specifically against Omicron BA.4 and BA.5 subvariants.

As of Aug. 27, BA.4 and BA.4.6 accounted for about 11% of circulating variants, and BA.5 made up almost all the remaining 89%, CDC data show.

The next step will be a review of the scientific data by the CDC’s Advisory Committee on Immunization Practices, which is set to meet Thursday and Friday. The final hurdle before distribution of the new vaccines will be a sign-off on CDC recommendations for use by agency Director Rochelle Walensky, MD.

“If you’ve not yet received a booster dose or it’s been several months since your last booster dose, now is the time to consider getting one,” Peter Marks, MD, director, FDA Center for Biologics Evaluation and Research, said during a virtual FDA news conference. 

“Also, if you’ve not yet vaccinated your children, now is a great time to consider taking them along to receive their vaccination as well,” he said.

“Unfortunately, COVID-19 continues to cause devastating consequences throughout the country with nearly 400 deaths and over 5,000 new hospitalizations every day,” FDA Commissioner Robert M. Califf, MD, said. “And just yesterday provisional CDC data indicated that US life expectancy fell again in 2021, In large part due to COVID deaths.”

“Regrettably only about half of eligible Americans have received their first booster,” he continued. “So, this is a remarkable opportunity to improve our life expectancy.”

How Do COVID-19 mRNA Vaccines Work?

Some of the COVID-19 vaccines are known as mRNA shots. How are they different from traditional vaccines? And do they contain the real virus? Share on Facebook Share on Twitter

CDC data indicate that those who are up to date on their vaccines are 13 times less likely to die from COVID compared to those who have not received the vaccine and are 3 times less likely to die from COVID compared to those who only had one booster instead of two.

“It’s just painful to see people dying unnecessarily when there’s a free treatment that would prevent their death,” said Califf, noting that protection against death associated with the COVID-19 vaccines “is much more clear than anything I’ve ever seen.”

Protection Now and In the Future

Scientific modeling suggests “that we are looking at a possible fall wave with a peak around Dec. 1,” Marks said. “By giving the booster now, we will hopefully both control the current plateau that we’re in — we’re dropping off very slowly — as well as address this future potential wave that looms out there.”

Califf noted that the new vaccines have another potential long-term benefit, protection against long COVID, “which for young people is increasingly a major concern.”

“I want to make clear that these updated boosters present us with an opportunity to get ahead of the next wave of COVID-19,” Califf said. “And for those who may be wondering, CDC says you may get a COVID-19 booster at the same time as your annual flu shot.”

The FDA will continue to study how well the new vaccines protect again COVID going forward, Marks said.

And another hope is that these next generation vaccines will provide stronger protection, Marks said.

“The idea here is not just to increase the antibodies right now, but also to hopefully give us a longer duration of protection,” he said.

If this holds true, then Americans might need fewer booster shots in the future.

“Hopefully [this] holds us for as much of the entire season.”

Covid-19 Vaccines —Covid-19 Vaccines — Immunity, Variants, Boosters


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.

Antiviral Immunity

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.Figure 1. Immune Responses for Protection against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2).

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.

Current Covid-19 Vaccines

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.

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.

Vaccine Durability

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.

SARS-CoV-2 Variants of Concern

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.Figure 4. Protective Efficacy of Covid-19 Vaccines in the United States.

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

Immune Correlates of Protection

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.

Proposed Framework for Covid-19 Vaccine Boosters

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.

Conclusions

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.

soucre: NEJM

Fast-evolving COVID variants complicate vaccine updates


COVID-19 vaccines are due for an upgrade, scientists say, but emerging variants and fickle immune reactions mean it’s not clear what new jabs should look like.

Syringes containing the Moderna Covid-19 vaccination sit on a paper-covered tray
Vaccines might become more effective if they target newer variants of the SARS-CoV-2 virus, not just the original one.Credit: Joseph Prezioso/AFP/Getty

As countries brace for another Omicron wave driven by the variants BA.4 and BA.5, calls to update COVID-19 vaccines are growing louder.

Existing vaccines based on the version of the virus SARS-CoV-2 that emerged in Wuhan, China, in late 2019 are a poor match to current Omicron strains. As a result, the vaccines now offer only short-lived protection from infection — although they seem to be holding up against severe disease.

This week, an advisory panel to the US Food and Drug Administration (FDA) will meet to discuss whether COVID-19 vaccines should be updated — and what the upgraded vaccines should look like.

Many — although by no means all — scientists agree that COVID-19 vaccines are overdue for change. But constantly emerging variants and hard-to-predict immune responses mean that it’s far from clear what the new jabs ought to look like.

“I think it’s time,” says Meagan Deming, a virologist and vaccine scientist at the University of Maryland School of Medicine in Baltimore. “The virus is changing, and what worked two years ago may not work for future variants.” But she and other scientists caution that updating COVID-19 vaccines won’t be as simple as swapping genetic material based on the Wuhan strain for that matching Omicron.

Shifting sands

Omicron has altered the course of the pandemic and spawned a series of offshoots, with BA.4 and BA.5 the latest. Each has eroded the immunity earnt from vaccination and infection with previous strains, including earlier versions of Omicron.

So if upcoming vaccines are based on the original Omicron, called BA.1, there is a real possibility that by the time they are rolled out later this year, circulating Omicron strains will be different. “BA.1 is yesterday’s news,” says John Beigel, a physician-scientist at the US National Institute of Allergy and Infectious Diseases (NIAID) in Bethesda, Maryland, who is leading a trial of potential vaccine updates.

It is also possible — and some scientists say likely — that an entirely new variant will pop up from a distant part of the SARS-CoV-2 family tree. “My concern is that there’s this huge focus on Omicron, and the assumption that Omicron is what we will be dealing with in the future,” says Penny Moore, a virologist at the University of the Witwatersrand in Johannesburg, South Africa. “We have a strong track record of getting that wrong.”

As a result of such uncertainty, scientists say the next COVID-19 vaccines need to cast a wide net, ideally eliciting an immune response that can recognize variants past, present and future. “The broadest response is definitely what I want,” says Deming.

Mix and match

How to achieve such breadth is the million-dollar question. Moderna, the biotechnology company in Cambridge, Massachusetts, that co-developed a successful mRNA-based vaccine with NIAID, is trialling an updated jab that encodes two versions of the SARS-CoV-2 spike protein: the original formulation and a version based on BA.1.

On 25 June, the company posted results from the trial1, which gives a booster of this ‘bivalent’ vaccine to people who have had three doses of the original Moderna vaccine, and compares their immune responses with those seen in people who receive a fourth dose of the original vaccine. But data announced this month suggest that the updated vaccine triggered antibody responses that were 75% more potent against BA.1 and 24% stronger against a version of SARS-CoV-2 from the early months of the pandemic, compared with an extra dose of the original vaccine. “This is a clearly superior booster,” the company’s president, Stephen Hoge, said in an 8 June investors’ call.

And last week, Moderna added that the bivalent vaccine generates antibodies that still block BA.4 and BA.5, although their levels were about three times lower than those against BA.1. However, the company did not provide a comparison with responses triggered by an extra dose of the original vaccine.

Other vaccine manufacturers, including Pfizer of New York City and its collaborator BioNTech of Mainz, Germany, as well as Novavax of Gaithersburg, Maryland, are testing their own Omicron-based vaccines. In a 25 June press release, Pfizer–BioNTech reported that an Omicron BA.1-only vaccine generated neutralizing antibody responses against BA.1 that were around 2–3 times more potent than an extra dose of the original vaccine; their bivalent vaccine, similar to Moderna’s, generated BA.1 responses that were about 1.5 to 2-fold stronger. BA.4 and BA.5 sapped these responses similarly to the Moderna vaccine.

Beigel says that the Moderna trial shows why now is the time to update COVID-19 vaccines. “We should be moving away from the prototype because the Omicron looks so much better,” he says.

But John Moore, a vaccine scientist at Weill Cornell Medicine in New York City, wonders whether the improvements the updated vaccines offer are worth it. “The question the FDA advisers have to decide on is whether this modest increase is enough to justify the expense and complexity of a composition switch,” Moore says. “I’ve seen nothing in the Pfizer and Moderna data to obviously justify a composition switch to Omicron.”

Beigel and his colleagues will soon report the first results from a NIAID-funded trial that is testing combinations of vaccines based on a range of variants, including Omicron, Beta, Delta and the original strain. This trial, called COVAIL, includes mRNA vaccines manufactured by Moderna and Pfizer–BioNTech, as well as an experimental protein-based booster developed by Sanofi in Paris and GSK in London.

Surprise entrant

Beigel says that we shouldn’t presume that the original vaccine is the best way to trigger a response against earlier non-Omicron strains. He hopes his study will shine a light on the ideal combinations. Another trial found that Sanofi–GSK’s booster, which is based on the Beta variant, triggered strong neutralizing-antibody responses against all variants, including BA.1 and Delta2. This hints that Beta shouldn’t be ruled out as a component of future updates, scientists say.

The quest for an updated formulation is also complicated by the possibility that vaccines based on a particular strain, such as Omicron, might not always trigger a potent immune response against that strain. Some recent studies3 have found that Omicron infections after vaccination recall the same antibodies that vaccines triggered against earlier strains, instead of eliciting all new responses to Omicron. But it’s not yet clear whether updated vaccines will behave in the same way. Pre-clinical studies of Omicron-based vaccines in animals showing little difference between Omicron and original-strain boosters suggest that they might, says John Moore.

A similar phenomenon, known as imprinting, affects how people respond to influenza vaccination and infection, causing levels of protection to vary between people and from year to year. Nonetheless, health officials attempt to match the make-up of seasonal vaccines to the strains most likely to be in circulation.

This strategy makes sense with SARS-CoV-2, says Jesse Bloom, an evolutionary biologist at the Fred Hutchinson Cancer Center in Seattle, Washington. “We can safely assume that having the vaccine as close as possible to the circulating virus will generally be better.”

But decisions surrounding the composition of influenza vaccines are based on a solid understanding of how those viruses evolve, says Beigel, something that researchers can’t yet claim for SARS-CoV-2. “We know the rules of flu and we can predict that very well. For COVID, we don’t.”

Which COVID boosters to take and when: a guide for the perplexed


A diverse menu of vaccine options leaves people searching for the best route to protection.

Two medical workers in PPE await patients to receive a dose of the anti-Covid-19 vaccine on March 20, 2021 in Belgrade, Serbia.
Health-care workers wait to administer doses of COVID-19 vaccine.

The next generation of COVID-19 vaccines is on its way, but those shots will be looking to take a seat at an already crowded table.

On the menu in some countries this autumn will be the familiar standards — mRNA and protein vaccines based on the spike protein from the ancestral version of SARS-CoV-2, which ushered in the pandemic. Alongside them will be a smattering of new specials, including mRNA vaccines with spike sequences both from ancestral virus and from Omicron variants.

It is a luxury of choice that many countries don’t have. But the range of options, which will be available at different times, has left people wondering which vaccines to take, and when. “These are hard questions, and there are no real right answers,” says Kathryn Edwards, a paediatrician and director of the Vanderbilt Vaccine Research Program at Vanderbilt University Medical Center in Nashville, Tennessee.

Nature asked specialists what evidence is on hand to help make the decision.

What do we know about the Omicron-specific versions of the COVID vaccine?

Relatively little.

On 15 August, the United Kingdom authorized the use of a two-pronged, or ‘bivalent’, vaccine containing both ancestral and Omicron BA.1 sequences of the spike protein, which the virus uses to latch onto human cells. The country’s Joint Committee on Vaccination and Immunisation recommended the bivalent vaccine as one option, alongside the first-generation mRNA vaccines, in the country’s autumn booster programme.

But BA.1 has been largely replaced by other Omicron variants in many countries, and the BA.4 and BA.5 variants now dominate in the United States and Europe. As a result, US regulators have said they intend to bypass BA.1-specific vaccines and instead authorize COVID-19 vaccines — expected to be available this autumn — that include spike sequences from BA.4 and BA.5.

Laboratory data show that the BA.1 bivalent vaccines stimulate the production of antibodies that can ‘neutralize’ the virus — that is, stop it from infecting host cells. Those data suggest that the inclusion of BA.1 sequences boosts neutralization of Omicron by about twofold1,2, but it’s unclear how much, if any, extra protection against illness this will produce. Neutralization data for BA.4- and BA.5-specific vaccines aren’t expected until around mid-September.

The underwhelming results for the bivalent vaccine are probably due to a phenomenon known as immune imprinting, says microbiologist John Moore at Weill Cornell Medicine in New York City. By now, much of the population has either been vaccinated or infected with an earlier variant of SARS-CoV-2. The immune system has been trained to remember this variant — and a dose of vaccine, even one with Omicron-specific components, will tend to boost those earlier immunological memories. The degree of Omicron-specific response will be relatively small, says Moore.

“If we had an immunologically naive population of people who had not been infected or vaccinated, it would make absolute sense for the vaccine to be from the Omicron lineage,” he says. “But how many people are neither infected nor vaccinated?”

If I’m due to get a booster now, should I get a regular booster or wait for one that is Omicron specific?

In countries such as the United States, people eligible for a booster in the summer have been wrestling with a decision: take a booster shot of the original vaccine, or wait a few more months for a version containing Omicron-specific spike.

Several physicians told Nature that the decision should be a personal one, and people should factor in whether they’re at risk of serious disease, their community’s infection rate and how well they can shield themselves from SARS-CoV-2. “It’s a lot of individual decision-making,” says Meagan Deming, an infectious-disease specialist at the University of Maryland School of Medicine in Baltimore.

It also depends on how long the wait is for the Omicron-specific vaccine, says Angela Branche, an infectious-disease specialist at the University of Rochester Medical Centre in New York. “The answer to that question is constantly changing,” she says. “If you asked me two or three months ago, I’d have said the Omicron-specific version of the vaccine is several months away, get your booster now.” But now, the wait is potentially only about a month, so she is more willing to advise her low-risk patients to hold out for new vaccines.

Yet others argue that there is still no need to delay a booster even a few weeks for the sake of receiving a new vaccine that hasn’t been shown to offer a significant advantage over the old ones. “There’s so little potential advantage to having an Omicron booster,” says Moore. “Why bother, when you can use the existing booster sooner?”

How long should I wait between COVID-19 vaccine boosters?

Here, researchers are largely in agreement: it’s best to wait at least four months between doses. Although receiving a COVID-19 booster sooner does not cause harm, there’s probably little benefit.

One study found that the antibody response to vaccination was weaker in people who had high antibody levels prior to getting the shot than in people with lower levels of pre-vaccination antibodies3. This is not a surprise to immunologists, says study author and viral immunologist Pablo Penaloza-MacMaster at Northwestern University Feinberg School of Medicine in Chicago: circulating antibodies might clear the spike protein in the vaccine before the immune response has a chance to be boosted.

“If the vaccines are given in a very short period of time, without allowing a resting period, you’re minimizing the effect,” he says.

Can you get too many boosters?

As long as the boosters are sensibly spaced, there’s really no such thing as “too many” from an individual standpoint, says Moore. From a public-health standpoint, however, a focus on boosting everyone could shift attention and resources away from the people who most need boosters: those over 50 years old, and people with pre-existing health conditions.

Boosters can significantly decrease the risk of serious disease for these groups. For younger people without risk factors, the benefits of a booster are less pronounced, but it is possible that a person who was boosted shortly before an infection might clear the virus faster— and shed less virus into their community — than someone whose antibody levels are lower when they are infected, says Penaloza-MacMaster.

Still, such a benefit is likely to be more transient and less meaningful than the benefit for people at risk of serious illness, says Moore. “The value for the under-50s in good health is far less certain,” he says. “That’s going to be a lower priority from a public-health perspective.”

Frequently asked questions about COVID-19 vaccines for infants, toddlers


COVID-19 vaccines are now available for children as young as age 6 months in the United States.

The FDA has authorized and the CDC has recommended Moderna’s two-dose series at 25 g per dose for children aged 6 months through 5 years and Pfizer-BioNTech’s three-dose series at 3 g per dose for children aged 6 months to 4 years.

Below, Healio Pediatrics Editorial Board Member Leonard R. Krilov, MD, FAAP, FIDSA, FPIDS, chief of pediatric infectious diseases at NYU Langone Hospital – Long Island, answers some frequently asked questions about the vaccines.

Leonard R. Krilov

Healio: Is one vaccine preferred over the other?

Krilov: I think it’s a little too soon to answer that. I think there are pros and cons to each of the two.

Moderna has the advantage that it is only two doses and will be completed in a month. On the counter side, there did seem to be more adverse reactions in terms of fever and local pain, as well as fatigue and headache or irritability and sleep disturbances with this vaccine. Will that impact compliance with the second dose or how the vaccine will be perceived? Additionally, even though it is approved as a two-dose series with follow-up, there may well be an indication for a booster for this vaccine.

On the other side of the coin, the Pfizer vaccine at one-tenth of the adult dose seems to be associated with lower rates of local reactions and fever compared with placebo. However, the Pfizer vaccine requires three doses, and would take potentially up to almost 3 months to complete the series. The preliminary data suggested minimal, if any, protection after just the first two doses.

As in any clinical trial, you cannot put two experimental products head to head. The studies were conducted at different times with differing SARS-CoV-2 infection rates in the community and the data may not be totally comparable for the two vaccines. I think it is too soon to say where the favorability is going to be. Both vaccines are recommended equally. I’ll switch hats and put my parent hat on (although my children are grown) and suggest I would probably lean toward achieving protection sooner, but the most important thing is to get the children vaccinated.

Healio: As you mentioned, Pfizer’s vaccine is three doses. What are some ways a pediatrician can make sure a parent brings their child back for two follow-up vaccine appointments?

Krilov: Ensuring compliance with the vaccine series is important and needs to be emphasized to achieve the best level of protection. In a way, compliance is inversely proportional to number of doses or number of visits, so it is a concern that needs to be addressed.

I think we need to work with parents on demonstrating the significance of the infection, as well as the safety and benefits of the vaccine. The communication with the family should address the misconception that COVID-19 is a benign disease for young children and infants. Although we have not seen the mortality in young children from COVID-19 observed in the elderly and those with significant medical conditions, there have been more than 30,000 children aged younger than 5 years hospitalized with COVID-19 and more than 440 deaths in children aged younger than 4 years from this infection. More than one-half of these cases occurred in children with no pre-existing medical conditions.

The phrase I have used in these discussions is that although the infection is milder in children, it is not necessarily mild. It can still be a significant disease in terms of the acute illness and long COVID. Symptoms persisting for months have been described in children as well as in adults. Beyond this, discussing the safety and effectiveness of the vaccine in preventing severe disease should be included.

As we are trying to return to some degree of normalcy with no masks in school and increased in-person events, which is especially important for younger children’s speech development and socialization, I think vaccination is a critical part of our toolbox to help us get there safely. Telephone or text message reminders for second or third doses can be helpful in achieving compliance.

Healio: How can pediatricians counsel parents who may be hesitant about getting their young children vaccinated?

Krilov: I think there are two components to that discussion. One is certainly to emphasize that it is a safe and beneficial vaccine for their child. The other is that we need to spend some time addressing the misconception that this is a benign disease for the younger child, as described in the previous answer. In other words, we need to educate about what we’re vaccinating against. We have to be able to convincingly demonstrate that what we’re vaccinating against is worth preventing.

I think, in a way, for the other routine childhood vaccines, we are the victim of our own success. When people don’t see a lot of the vaccine-preventable diseases, they lose track of how significant they are, or were, and so therefore, can focus on potential concerns about reactions or the vaccines not being necessary. Maybe some of that’s true for COVID-19 as well, in the sense that the focus has been on older individuals and those with underlying medical issues. But I think it’s still significant for young children.

We want children to be able to do more and more and interact — that this is an important part of our toolbox help get there, maintain that. So, I think that is the track we need to take in order to be able to work with parents on showing the significance of the infection, as well as the safety and benefits of the vaccine.

Healio: Should parents start scheduling appointments now, or can the COVID-19 vaccine wait until they being their children in for a well visit or some other appointment?

Krilov: Because the vaccination is going to be administered over two or three visits, it might be hard to just totally dovetail into regular scheduled visits. And given, you know, the high levels of community spread in most areas of the country right now, I see no benefit to waiting. I understand it’s an extra visit, but I think the benefits are clear and the benefits should be achieved as soon as possible. It also should be noted that the COVID-19 vaccine can be given at the same time as other scheduled vaccines to avoid delays in getting vaccinated.

Healio : Do you think young children will eventually need a booster?

Krilov: Yes, I do think it is likely a booster for these children will be needed at some point, but it will require follow-up to determine if or when. Is this going to become like influenza, requiring periodic boosters for everyone? Are we going to settle into a more seasonal endemic pattern that will make timing of or need for boosters more predictable? Will there be additional waves this fall or winter, leading to recommendations for booster doses? Two and a half years ago we were talking about no hospital beds and having portable morgues in hospital parking lots, and now it’s a much more manageable disease. But again, there are still large numbers of cases.

I think the likelihood is this virus will continue to circulate, and with that, probably we will need periodic boosters. Is it going to be the same booster or, like influenza, is it going to be modified to match circulating variants? For example, omicron variant vaccines are in development now, so will the booster be with a different variant even to boost the immunity better? Will it turn out in a way down the road like influenza if we have multiple circulating variants to be a multivariant vaccine? It is still a bit premature to know the answer, but based on how immunity seems to wane for older children and adults, I do expect we will be looking at boosters for young children.

FDA sets new dates to review pediatric COVID-19 vaccines


The FDA this week revised the dates for several upcoming meetings of its vaccine advisory board to discuss pediatric COVID-19 vaccines.

The announcement came the same day that Pfizer and BioNTech announced promising data on a third dose of their pediatric vaccine and said they will submit the results to the FDA this week as part of the rolling emergency use application they began in February.

Source: Adobe Stock.
Pfizer and BioNTech have announced results of a COVID-19 vaccine for children aged 6 months to 4 years. Source: Adobe Stock

The FDA postponed its review of the vaccine for children aged 6 months to 4 years in January, citing a need for more data.

Pfizer and BioNTech announced in December that they were evaluating the addition of a third 3 µg dose — one-tenth the size of an adult dose — of the vaccine after a two-dose series failed to produce the expected level of protection among study participants aged 2 to 4 years, although protection among participants aged 6 to 24 months matched that seen in adolescents and young adults.

In the new data, companies reported that a third 3 ug dose of the vaccine given 2 months after a second dose was well tolerated among 1,678 children aged younger than 5 years of age, with a vaccine efficacy of 80.3%.

“The study suggests that a low 3 ug dose of our vaccine, carefully selected based on tolerability data, provides young children with a high level of protection against the recent COVID-19 strains,” BioNTech CEO Ugur Sahin, MD said in a press release. “We are preparing the relevant documents and expect completing the submission process to the FDA this week, with submissions to [the European Medicines Agency] and other regulatory agencies to follow within the coming weeks.”

The FDA’s Vaccines and Related Biological Products Advisory Committee (VRBPAC) will tentatively meet June 14 to discuss Moderna’s EUA request for children aged 6 to 17 years, then again on June 15 to discuss Moderna’s EUA request for children aged 6 months to 5 years and Pfizer-BioNTech’s EUA request for children aged 6 months to 4 years.

The FDA said the dates are tentative because none of the EUA submissions are complete.

A Pfizer-BioNTech vaccine has been available under an EUA for children aged 5 to 11 since December. Last week, the FDA endorsed a booster shot this age group.

References:

Coronavirus (COVID-19) update: FDA announces tentative advisory committee meeting schedule regarding COVID-19 vaccines. https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-announces-tentative-advisory-committee-meeting-schedule-regarding. Updated May 23, 2022. Accessed May 23, 2022.

Pfizer-BioNTech COVID-19 vaccine demonstrates strong immune response, high efficacy and favorable safety in children 6 months to under 5 years of age following third dose. https://www.pfizer.com/news/press-release/press-release-detail/pfizer-biontech-covid-19-vaccine-demonstrates-strong-immune. Published May 23, 2022. Accessed May 24, 2022.