Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3233 2022-04-20 12:09:41 |
2 update references and layout -2 word(s) 3231 2022-04-21 03:44:05 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Souza Machado, B.A.; Saraiva Hodel, K.; Fonseca, L.; , .; Badaro, R. The Importance of Vaccination during COVID-19 Pandemic. Encyclopedia. Available online: https://encyclopedia.pub/entry/22005 (accessed on 02 July 2024).
Souza Machado BA, Saraiva Hodel K, Fonseca L,  , Badaro R. The Importance of Vaccination during COVID-19 Pandemic. Encyclopedia. Available at: https://encyclopedia.pub/entry/22005. Accessed July 02, 2024.
Souza Machado, Bruna Aparecida, Katharine Saraiva Hodel, Larissa Fonseca,  , Roberto Badaro. "The Importance of Vaccination during COVID-19 Pandemic" Encyclopedia, https://encyclopedia.pub/entry/22005 (accessed July 02, 2024).
Souza Machado, B.A., Saraiva Hodel, K., Fonseca, L., , ., & Badaro, R. (2022, April 20). The Importance of Vaccination during COVID-19 Pandemic. In Encyclopedia. https://encyclopedia.pub/entry/22005
Souza Machado, Bruna Aparecida, et al. "The Importance of Vaccination during COVID-19 Pandemic." Encyclopedia. Web. 20 April, 2022.
The Importance of Vaccination during COVID-19 Pandemic
Edit

The rapid spread and contagion of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), raised concern among the public and health authorities worldwide. Shortly after the first case was reported in Wuhan (China), the World Health Organization (WHO) defined COVID-19 as a pandemic. Since the pandemic began, one of the main effective and feasible ways to contain the spread of the SARS-CoV-2 has been through vaccination.

COVID-19 vaccines SARS-CoV-2 COV pandemic

1. Overview of COVID-19 Vaccines and Variants

One of the biggest challenges for vaccination, especially when it comes to airborne viruses—such as flu viruses, for example—is the emergence of new variants caused by mutations in the virus genome. Mutations occur naturally during the replication of any RNA virus due to the instability of the RNA molecules [1]. In the case of COVID-19, even before it was considered a pandemic, data related to the genomic surveillance of SARS-CoV-2 were available as a useful tool for investigating outbreaks and tracking evolution and possible new waves [2][3]. As a result, more than 25 billion sequences of SARS-CoV-2 have been performed worldwide, and it is estimated that at least two mutations in the viral genome occur per month [3][4]. These additional mutations often result in distinct immune-evasion mechanisms and lead to the appearance of different variants and lineages [5]. Currently, there are 21 variants of SARS-CoV-2; among them, variants Alpha a (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529) are considered as variants of concern (VOCs), while Lambda (C.37) and Mu (B.1.621) are considered as variants of interest (VOIs). It is emphasized that, in addition to these two categories, there are also variants under monitoring (VUMs), which are those strains that have genetic mutations that may pose a risk in the future, but for which phenotypic and epidemiological changes are currently unclear [6].
Because one of their characteristics is lower susceptibility to vaccines and other therapeutic alternatives, VOCs have been intensively monitored [7]. Modifications found in the structure of the S protein of SARS-CoV-2 have been attributed to the greater ability of the virus to escape the action of neutralizing antibodies [6][8]. Important examples of mutational mechanisms that lead to increased antigenic properties of protein S are the amino acid substitutions that alter the protein epitope, increase receptor-binding avidity, and lead to changes in glycosylation, deletion or insertion of residues, and allosteric structural effects [9]. These factors strongly contribute to the increased mortality and morbidity of SARS-CoV-2 [9][10][11], where the transmissibility can be up to 74% higher when compared to the original strain [12][13].
Since the SARS-CoV-2 S protein is the main target of COVID-19 vaccines, the mutations in this protein are of great concern, especially those which have the corresponding sequences of reference strain Wuhan-Hu-1—that is, no antigens based on different variants are used [14][15]. Thus, the appearance of variants with modifications in the SARS-CoV-2 S protein structure raises questions about the effectiveness of the vaccines available to the population; antibodies derived from the original strain (Wuhan-Hu-1) may have only a partial neutralizing effect against these viruses [16]. The vaccines BNT162b2 (brand name Comirnaty), mRNA-1273 (brand name Spikevax), CoronaVac, BBIBP-CorV, AZD-1222 (brand name Vaxzevria or Covishield), and Ad26.COV2-S (brand name Janssen COVID-19 Vaccine) are the most widely used around the world for COVID-19 prophylaxis, since all of them use the S protein as the main activator of the immune system [17]. Therefore, different studies have been published or are being conducted to analyze the efficacy or effectiveness of each of these vaccines against SARS-CoV-2 variants of concern.
Analysis of several studies shows that the efficiency or effectiveness of vaccines against SARS-CoV-2 variants depends on many factors, including the sample size, demographic factors, host factors, the type of vaccine, the number of doses, a heterologous or homologous booster vaccination scheme, and the time after primary vaccination is completed [7][18][19]. Different authors demonstrated that the application of a booster dose after a certain period is able to increase the humoral immune response against SARS-CoV-2, resulting in increased efficacy or effectiveness of vaccines against the VOC [20][21][22][23][24]. Bruxvoort et al. [25] found data that reinforced the need for booster dose administration, since primary immunization with mRNA-1273 shows limited protection against the Delta, Alpha, Gamma, and Mu variants. Andrews et al. [26] reported that administration of the booster dose with BNT162b2 after primary immunization of ChAdOx1, mRNA-1273, or BNT162b2 was able to significantly increase protection against Omicron. Other authors reported that the homologous booster regimen of BNT162b2, mRNA-1273, CoronaVac and BBIBP-CorV vaccines and the heterologous booster with Ad26.COV2.S associated with mRNA-1273 vaccines showed similar performance against Omicron [27][28].
Data provided by the Control and Prevention (CDC) show that the number of cases and deaths caused by COVID-19 in the United States is higher among unvaccinated individuals when compared to individuals with a full primary vaccination scheme and/or those who have already received a booster dose, regardless of the vaccine administered (BNT162b2, mRNA-1273, or Ad26.COV2-S) [29]. Among those vaccinated, although the three vaccines showed similar efficacy in reducing COVID-19 infection in the period evaluated (April 2021 to February 2022), it was observed that the number of deaths among individuals vaccinated with Ad26.COV2-S was higher when compared to the mRNA vaccines (BNT162b2 and mRNA-1273) [29]. In early January 2022, during the wave caused by the Omicron variant, the highest incidence per 100,000 population occurred, where the rates reached 5.44, 2.34, and 1.79 for the Ad26.COV2-S, BNT162b2, and mRNA-1273 vaccines, respectively, after the primary vaccination scheme [29]. However, it is important to note that the mortality rate among those who received the Ad26.COV2-S vaccine was lower compared to unvaccinated individuals, where the rate was ~9 times higher. Although it has already been reported that the Ad26.COV2-S vaccine can elicit a stable humoral and cellular response over time, data reported by the CDC suggest that the immune response induced by mRNA vaccines may be more effective in reducing mortality rates [30][31]. The greater efficacy observed in mRNA vaccines may be a reflection of their advantages as a technological platform; unlike adenovirus (DNA)-based vaccines, once administered, RNA molecules do not need to cross the nuclear membrane or transcription to start the protein-synthesis process [32]. Hence, these data encouraged discussions about the immune protection of the Ad26.COV2-S vaccine, particularly with a view to the possible implementation of the intervals required for the application of new booster doses.
Because of this, since the second half of 2021, health authorities around the world have been recommending booster doses for different vaccines to contain the spread of SARS-CoV-2 and consequently control the pandemic in terms of the number of hospitalizations and deaths [33][34][35][36]. Andrews et al. [37] conducted in England demonstrated that the administration of a booster dose with mRNA vaccines (BNT162b2 or mRNA-1273) compared to the primary immunization showed an efficiency between 94 and 97% in reducing symptomatic cases of the disease, while against hospitalization or death, the value found ranged between 97% and 99%. Studies conducted in Israel have also shown the same trend, which strengthens the importance of this new vaccination strategy [38][39][40]. Furthermore, studies involving computer modeling have shown that the administration of the booster dose will be able to decrease the effective reproduction number (R0), while authors have pointed out that the predicted increase in antibody titers induced by the booster dose provides important protection from infection with SARS-CoV-2 variants [41][42][43].
Currently, the application of a new booster dose (usually the fourth dose) has been recommended in different countries due to the emergence of the Omicron variant, which has a greater transmission capacity than the other variants and, consequently, has been associated with an increase in the number of reinfection cases [44][45][46]. Initially, the administration of this new dose was directed towards priority groups, such as healthcare workers, immunocompromised individuals, and the elderly; however, new groups are expected to be included soon [47][48]. A preliminary one released by the Israeli Ministry of Health in adults aged >60 years demonstrated that a fourth dose of the BNT162b2 vaccine was able to increase immune protection up to two-fold against SARS-CoV-2 infection and up to three-fold against severe disease when compared to individuals who received only the third dose [49]. The expectation that new VOCs will emerge over time exposes one of the main challenges associated with developing COVID-19 vaccines, that of using products capable of inducing a robust and/or long-lasting immune response against different variants [50][51].
In early November, a new COVID-19 sublineage named BA.2 was first reported [52] (2 months after variation BA.1, which rapidly became dominant due to immune-escape mechanisms [53][54]). In a public statement, WHO defined it as an Omicron sublineage, classifying it as a variant of concern. According to the organization, the amino acids differences in structural proteins have possibly conferred a growth advantage when compared to other Omicron sublineages (BA.1, BA. 1.1.), but not greater severity [55]. In later March, the US Centers for Disease Control and Prevention (CDC) reported that BA.2 was responsible for 55% (50.8–59.1%—95% PI) COVID-19 cases [56], followed by BA. 1.1 (40.4%, 36.4–44.5%—95% PI). The rapid spread of BA.2 has raised discussions about reinfection and vaccine efficacy. Although BA.2’s ability to evade neutralizing antibodies is unclear, authors have demonstrated evidence that the increasing frequency of BA.2 is probably related to increased transmissibility rather than to enhanced immunologic escape [57]. On the other hand, initial data from population-level reinfection studies suggest that infection with BA.1 provides strong protection against reinfection with BA.2 [58]. Recent studies have demonstrated that mRNA vaccines (BNT162b2 and mRNA-1273) provide similar protection against BA.1 (46.6% (95% CI: 33.4–57.2%) and BA.2 51.7% (95% CI: 43.2–58.9%) in the first three months, and this declines to about 10% after 4–6 months. These findings show that protection against BA.2 did not seem to wane any faster than protection against BA.1. Furthermore, in both cases, a second dose was able to recuperate immune protection levels [59]. Therefore, until now, recent data supported the need for a vaccine targeting the Omicron variant.
Within this context, in January 2022 the companies Pfizer and BioNTech, developers of the BNT162b2 vaccine, started a clinical trial to evaluate the safety, immunogenicity, and tolerability of an Omicron-based vaccine candidate. To this end, it will be conducted with healthy adults between the ages of 18 and 55 who may be allocated into three distinct cohorts with different dose regimens of the vaccine candidate [60]. Similarly, the pharmaceutical company Moderna (developer of the mRNA-1273 vaccine) is expected to start a clinical trial for the analysis of a new vaccine candidate against the Omicron variant in the first half of 2022 [61]. In addition, different institutes have supported the idea of investing in the development of a pan-coronavirus vaccine capable of protecting against several coronaviruses, including the different strains of SARS-CoV-2 [61][62].
Importantly, the development of Omicron-specific vaccines should not completely rule out the use of previously approved vaccines made available to the population, as robust data are still needed to elucidate the induction of the immune response after the first booster dose and its role in controlling infection or disease progression by this variant. Furthermore, one cannot exclude the possible “selective pressure” exerted by vaccines and even by monoclonal antibody therapy in targeting the S protein, since this may have influenced the appearance of new variants with mutations in this region, thus conferring escape mechanisms [63]. Therefore, new therapeutic targets must be considered for the development of new vaccines.

2. Safety of COVID-19 Vaccines

Due to the pandemic nature of COVID-19 and the various impacts generated in the global health, social and economic sectors, the vaccines against SARS-CoV-2 infection were made available in record time [64]. This was also because many scientists, manufacturers, and research institutions were already developing innovative technology platforms for new vaccines, which were eventually adapted for COVID-19 prevention. However, since no coronavirus vaccine had been licensed and approved for use in humans previously, the rapid development associated with the limited follow-up time post-vaccination and lack of information about long-term side effects of the vaccines aroused great public concern about the safety profile of the available vaccines [65]. It is important to note that as mass vaccination progresses, more post-vaccination adverse events are reported [66]. This demonstrates that vaccine safety information from ongoing clinical trials and surveillance data is important not only for building public confidence, but also for making evidence-based health-policy decisions [67]. The safety of vaccines is evaluated through adverse event monitoring in randomized controlled trials and safety post-licensure surveillance data after immunization campaigns [68]. Determining the safety profile of a vaccine is a critical step at the global level and is monitored by the WHO along with manufacturers, health officials, and national regulatory agencies [69][70][71], since they are drugs administered in healthy populations. According to the WHO, all available vaccines, including the COVID-19 vaccines, have been rigorously assessed for safety for diverse groups of people, according to age, sex, ethnicity, and medical conditions.
For the mRNA vaccines, the most commonly reported adverse events are local reactions at the injection site, such as pain, redness, and swelling, and systemic reactions, such as headache, myalgia, arthralgia, and chills [72]. In clinical studies evaluating the mRNA-1273 and BNT162b2 vaccines, the frequency and severity of these adverse events were higher after the administration of the second dose. When it comes to adenoviral vector vaccines, in the case of the AZD 1222 vaccine, pain, fever, chills, muscle ache, headache, and malaise were the most common adverse reactions. Regarding serious adverse events, seven have been associated with the AZD-122 vaccine, including transverse myelitis [73]. Overall, inactivated virus vaccines such as CoronaVac, BBIBP-CorV, and COVXIN have a good safety profile, with few grade 3 adverse reactions. In the elderly population, studies with vaccines of distinct technologies, such as AZD 1222 (modified adenovirus) and NVX-CoV2373 (protein adjuvant), for example, showed a good antibody response and low reactogenicity events after administration, with a higher incidence and severity of adverse events observed in younger subjects [74].
Another rare manifestation after vaccination, but which has been reported in different studies, is multisystem inflammatory syndrome (MIS) [75]. MIS has still poorly understood pathophysiology, however, it is believed to occur due to an exaggerated immune response against SARS-CoV-2 infection due to persistently high levels of IgG and activation of CD8+ T cells [76][77]. Considering the adult population, MIS may result from a delayed and dysregulated immune response and is characterized by the onset of symptoms such as fever, elevated inflammatory markers, as well as multiple-organ involvement (especially of the heart, stomach, and intestines) [78][79]. Different case studies have reported the onset of MIS in adults after immunization with the vaccine based on mRNA [80], inactivated virus [81], and adenovirus [82]. However, a crucial question was demonstrated by Belay et al. [83], in which the report of MIS after vaccination in adult patients was also associated with prior SARS-CoV-2 infection. These data demonstrate the need for further studies to elucidate the real association between MIS caused purely by vaccination or whether there is a direct relationship with previous viral infection.
It is interesting to highlight that most of the side effects that people experience after COVID-19 vaccination can be attributed to the “nocebo” effect. Nocebo refers to the non-pharmacological adverse effects reported after exposure to a placebo substance, which are usually motivated by the individual’s expectation that, after exposure to a vaccine, drug, or other medical intervention, some disagreeable event will occur [84][85]. Haas et al. [86] evaluated the frequency of adverse events in the placebo arm in 12 clinical studies of COVID-19 vaccines (mRNA-1273, CoV2 preS dTM, NVX-CoV2373, AZD-1222, BNT162b2, BNT162b1, and SCB-2019) at different phases of clinical development, and the results found demonstrate that while adverse events were mostly reported in the arms receiving the experimental vaccines, subjects receiving the placebo reported a significant frequency of adverse events. These results highlight the importance of critically evaluating the safety of experimental vaccines, especially when some minority groups are known to be resistant to COVID-19 vaccination [87][88].
When it comes to serious adverse events, particular concern has emerged related to the safety of COVID-19 during pregnancy due to reported cases of thrombosis and thrombocytopenia syndrome after vaccination with AZD 1222 in early 2021 [89]. Despite the devastating consequences of COVID-19 infection in pregnant women and the availability of vaccine safety and efficacy data in different populations, data related to vaccine safety in pregnant women are still limited, since most of the ongoing clinical trials do not include pregnant women [90]. However, preclinical and toxicological COVID-19 vaccine studies have found no safety concerns with no adverse effect on female reproduction, fertility, fetal or embryonal, or postnatal development, or miscarriage [90][91][92]. For mRNA vaccines, surveillance data demonstrated that vaccine-related adverse events in pregnant women were similar to those in non-pregnant women, with pain in the local area of the injection, fatigue, headache, and myalgia being the most frequent local and systemic reactions after vaccination [90]. Regarding the safety of COVID-19 vaccines for the fetus or breastfeeding infant, various expert panels suggest that mRNA-based and adenovirus vector vaccines do not possess any significant risk [93][94].

2.1.COVID-19 Vaccines and Serious Adverse Events: Myocarditis and Pericarditis

Currently, the main safety concerns for COVID-19 vaccines are related to mRNA vaccines, with the emergence of cases of myocarditis and pericarditis. Myocarditis is the inflammation of the heart muscle, while pericarditis is the inflammation of the outer lining of the heart. In both cases, the immune system causes inflammation in response to an infection or some other factor. Both can occur during infections, including SARS-CoV-2 infection. For inflammation caused by mRNA vaccines, one of the first one involving the evaluation of the incidence of myocarditis after vaccination with Pfizer’s vaccine (BNT162b2 or Comirnaty) was published in October 2021 in the New England Journal of Medicine. It was conducted with patients in a large Israeli healthcare system who had received at least one dose of the vaccine. It was reported an estimated incidence of myocarditis of 2.13 cases per 100,000 people; the highest incidence was among male patients between the ages of 16 and 29. Most of myocarditis were mild or moderate in severity [95]. According to the CDC, myocarditis is a rare and serious adverse event that has been associated with mRNA-based COVID-19 vaccines, in this case BNT162b2. The reporting rates of vaccine-associated myocarditis appear to be highest among males aged 12–29 years. As of 31 December 2021, myocarditis among children aged 5–11 years is classified as rare, where 11 Vaccine Adverse Event Reporting System (VAERS)-verified reports were received after the administration of approximately eight million doses of vaccine, and in an active vaccine safety surveillance system, no confirmed reports of myocarditis were observed during the 1–21 days or 1–42 days after 333,000 doses of vaccine were administered to children of the same age. Two deaths following the BNT162b2 vaccine were reported in children with multiple chronic medical conditions, where, in the initial one, no data were found to suggest a causal association between death and vaccination.
It is important to compare the cases among vaccinated and infected ones. The incidence of COVID-19-associated cardiac injury or myocarditis can be 100 times higher than COVID-19 mRNA-vaccine-related myocarditis [96][97]. Another relevant observation is that cases of myocarditis and pericarditis have been reported mainly for mRNA vaccines [98]; in Brazil, no cases have been reported related to inactivated virus-based vaccines [99]. The possible mechanisms underlying heart injury side-effects in specific groups were brightly hypothesized by Heymans and Cooper [96], where, according to the authors, mRNA vaccines might trigger immune hyper immunity in a minority population that is genetically susceptible to developing acute myocarditis after viral injury [100]. In summary, they indicated three potential mechanisms: hormonal differences (the sex-specific distinction can be explained by hormone-related factors); mRNA immune reactivity (genetic variants in HLA genes); and antibodies to SARS-CoV-2 spike glycoproteins cross-reacting with myocardial contractile proteins (genotypes in desmosomal, cytoskeletal or sarcomeric protein).

References

  1. Singh, D.; Yi, S.V. On the origin and evolution of SARS-CoV-2. Exp. Mol. Med. 2021, 53, 537–547.
  2. Deng, X.; Gu, W.; Federman, S.; du Plessis, L.; Pybus, O.G.; Faria, N.R.; Wang, C.; Yu, G.; Bushnell, B.; Pan, C.-Y.; et al. Genomic surveillance reveals multiple introductions of SARS-CoV-2 into Northern California. Science 2020, 369, 582–587.
  3. Li, J.; Lai, S.; Gao, G.F.; Shi, W. The emergence, genomic diversity and global spread of SARS-CoV-2. Nature 2021, 600, 408–418.
  4. Song, S.; Ma, L.; Zou, D.; Tian, D.; Li, C.; Zhu, J.; Chen, M.; Wang, A.; Ma, Y.; Li, M.; et al. The Global Landscape of SARS-CoV-2 Genomes, Variants, and Haplotypes in 2019nCoVR. Genomics. Proteom. Bioinform. 2020, 18, 749–759.
  5. Rathinasamy, M.; Kandhasamy, S. An exploratory study on the propagation of SARS-CoV-2 variants: Omicron is the most predominant variant. J. Med. Virol. 2022, 1–8.
  6. Jiang, Y.; Wu, Q.; Song, P.; You, C. The Variation of SARS-CoV-2 and Advanced Research on Current Vaccines. Front. Med. 2022, 8, 2908.
  7. Tregoning, J.S.; Flight, K.E.; Higham, S.L.; Wang, Z.; Pierce, B.F. Progress of the COVID-19 vaccine effort: Viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat. Rev. Immunol. 2021, 21, 626–636.
  8. Harvey, W.T.; Carabelli, A.M.; Jackson, B.; Gupta, R.K.; Thomson, E.C.; Harrison, E.M.; Ludden, C.; Reeve, R.; Rambaut, A.; Peacock, S.J.; et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat. Rev. Microbiol. 2021, 19, 409–424.
  9. Starr, T.N.; Greaney, A.J.; Hilton, S.K.; Ellis, D.; Crawford, K.H.D.; Dingens, A.S.; Navarro, M.J.; Bowen, J.E.; Tortorici, M.A.; Walls, A.C.; et al. Deep Mutational Scanning of SARS-CoV-2 Receptor Binding Domain Reveals Constraints on Folding and ACE2 Binding. Cell 2020, 182, 1295–1310.e20.
  10. Weisblum, Y.; Schmidt, F.; Zhang, F.; DaSilva, J.; Poston, D.; Lorenzi, J.C.C.; Muecksch, F.; Rutkowska, M.; Hoffmann, H.-H.; Michailidis, E.; et al. Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Elife 2020, 9, e61312.
  11. McCarthy, K.R.; Rennick, L.J.; Nambulli, S.; Robinson-McCarthy, L.R.; Bain, W.G.; Haidar, G.; Duprex, W.P. Recurrent deletions in the SARS-CoV-2 spike glycoprotein drive antibody escape. Science 2021, 371, 1139–1142.
  12. Volz, E.; Mishra, S.; Chand, M.; Barrett, J.C.; Johnson, R.; Geidelberg, L.; Hinsley, W.R.; Laydon, D.J.; Dabrera, G.; O’Toole, Á.; et al. Assessing transmissibility of SARS-CoV-2 lineage B.1.1.7 in England. Nature 2021, 593, 266–269.
  13. Davies, N.G.; Abbott, S.; Barnard, R.C.; Jarvis, C.I.; Kucharski, A.J.; Munday, J.D.; Pearson, C.A.B.; Russell, T.W.; Tully, D.C.; Washburne, A.D.; et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science 2021, 372, eabg3055.
  14. Dai, L.; Gao, G.F. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 2021, 21, 73–82.
  15. Heinz, F.X.; Stiasny, K. Distinguishing features of current COVID-19 vaccines: Knowns and unknowns of antigen presentation and modes of action. npj Vaccines 2021, 6, 104.
  16. Ding, C.; He, J.; Zhang, X.; Jiang, C.; Sun, Y.; Zhang, Y.; Chen, Q.; He, H.; Li, W.; Xie, J.; et al. Crucial Mutations of Spike Protein on SARS-CoV-2 Evolved to Variant Strains Escaping Neutralization of Convalescent Plasmas and RBD-Specific Monoclonal Antibodies. Front. Immunol. 2021, 12, 3231.
  17. Holder, J. COVID World Vaccination Tracker. Available online: https://www.nytimes.com/interactive/2021/world/covid-vaccinations-tracker.html (accessed on 6 February 2022).
  18. Hafiz, I.; Illian, D.N.; Meila, O.; Utomo, A.R.H.; Susilowati, A.; Susetya, I.E.; Desrita, D.; Siregar, G.A.; Basyuni, M. Effectiveness and Efficacy of Vaccine on Mutated SARS-CoV-2 Virus and Post Vaccination Surveillance: A Narrative Review. Vaccines 2022, 10, 82.
  19. Huang, Z.; Su, Y.; Zhang, T.; Xia, N. A review of the safety and efficacy of current COVID-19 vaccines. Front. Med. 2022, 16, 39–55.
  20. Pajon, R.; Doria-Rose, N.A.; Shen, X.; Schmidt, S.D.; O’Dell, S.; McDanal, C.; Feng, W.; Tong, J.; Eaton, A.; Maglinao, M.; et al. SARS-CoV-2 Omicron Variant Neutralization after mRNA-1273 Booster Vaccination. N. Engl. J. Med. 2022, 386, 1088–1091.
  21. Sablerolles, R.S.G.; Rietdijk, W.J.R.; Goorhuis, A.; Postma, D.F.; Visser, L.G.; Geers, D.; Schmitz, K.S.; Garcia Garrido, H.M.; Koopmans, M.P.G.; Dalm, V.A.S.H.; et al. Immunogenicity and Reactogenicity of Vaccine Boosters after Ad26.COV2.S Priming. N. Engl. J. Med. 2022, 386, 951–963.
  22. Zhao, X.; Li, D.; Ruan, W.; Chen, Z.; Zhang, R.; Zheng, A.; Qiao, S.; Zheng, X.; Zhao, Y.; Dai, L.; et al. Effects of a Prolonged Booster Interval on Neutralization of Omicron Variant. N. Engl. J. Med. 2022, 386, 894–896.
  23. Sester, M.; Becker, S.L. Boosting immunity after CoronaVac. Lancet 2022, 399, 496–497.
  24. del Rio, C.; Malani, P.N.; Omer, S.B. Confronting the Delta Variant of SARS-CoV-2, Summer 2021. JAMA 2021, 326, 1001.
  25. Bruxvoort, K.J.; Sy, L.S.; Qian, L.; Ackerson, B.K.; Luo, Y.; Lee, G.S.; Tian, Y.; Florea, A.; Aragones, M.; Tubert, J.E.; et al. Effectiveness of mRNA-1273 against delta, mu, and other emerging variants of SARS-CoV-2: Test negative case-control study. BMJ 2021, 375, e068848.
  26. Andrews, N.; Stowe, J.; Kirsebom, F.; Toffa, S.; Rickeard, T.; Gallagher, E.; Gower, C.; Kall, M.; Groves, N.; O’Connell, A.-M.; et al. Effectiveness of COVID-19 vaccines against the Omicron (B.1.1.529) variant of concern. medRxiv 2021.
  27. Garcia-Beltran, W.F.; St. Denis, K.J.; Hoelzemer, A.; Lam, E.C.; Nitido, A.D.; Sheehan, M.L.; Berrios, C.; Ofoman, O.; Chang, C.C.; Hauser, B.M.; et al. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell 2022, 185, 457–466.e4.
  28. Yu, X.; Qi, X.; Cao, Y.; Li, P.; Lu, L.; Wang, P.; Feng, Y.; Yang, J.; Wei, H.; Guo, L.; et al. Three doses of an inactivation-based COVID-19 vaccine induces cross-neutralizing immunity against the SARS CoV-2 Omicron variant. Emerg. Microbes Infect. 2022, 11, 749–752.
  29. CDC COVID Data Tracker: Rates of COVID-19 Cases and Deaths by Vaccination Status. Available online: https://covid.cdc.gov/covid-data-tracker/#rates-by-vaccine-status (accessed on 5 April 2022).
  30. Barouch, D.H.; Stephenson, K.E.; Sadoff, J.; Yu, J.; Chang, A.; Gebre, M.; McMahan, K.; Liu, J.; Chandrashekar, A.; Patel, S.; et al. Durable Humoral and Cellular Immune Responses 8 Months after Ad26.COV2.S Vaccination. N. Engl. J. Med. 2021, 385, 951–953.
  31. Johnson, A.G.; Amin, A.B.; Ali, A.R.; Hoots, B.; Cadwell, B.L.; Arora, S.; Avoundjian, T.; Awofeso, A.O.; Barnes, J.; Bayoumi, N.S.; et al. COVID-19 Incidence and Death Rates Among Unvaccinated and Fully Vaccinated Adults with and Without Booster Doses During Periods of Delta and Omicron Variant Emergence—25 U.S. Jurisdictions, April 4–December 25, 2021. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 132–138.
  32. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines-a new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279.
  33. Krause, P.R.; Fleming, T.R.; Peto, R.; Longini, I.M.; Figueroa, J.P.; Sterne, J.A.C.; Cravioto, A.; Rees, H.; Higgins, J.P.T.; Boutron, I.; et al. Considerations in boosting COVID-19 vaccine immune responses. Lancet 2021, 398, 1377–1380.
  34. COVID-19 Vaccine Booster Shots. Available online: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/booster-shot.html (accessed on 8 February 2022).
  35. Comirnaty and Spikevax: EMA Recommendations on Extra Doses and Boosters. Available online: https://www.ema.europa.eu/en/news/comirnaty-spikevax-ema-recommendations-extra-doses-boosters (accessed on 8 February 2022).
  36. Ministério da Saúde Ministério da Saúde Anuncia dose de reforço para Vacinação Contra a COVID-19 na Segunda Quinzena de Setembro. Available online: https://www.gov.br/saude/pt-br/assuntos/noticias/ministerio-da-saude-anuncia-dose-de-reforco-para-vacinacao-contra-a-covid-19-na-segunda-quinzena-de-setembro (accessed on 9 February 2022).
  37. Andrews, N.; Stowe, J.; Kirsebom, F.; Toffa, S.; Sachdeva, R.; Gower, C.; Ramsay, M.; Bernal, J.L. Effectiveness of COVID-19 booster vaccines against COVID-19 related symptoms, hospitalisation and death in England. Nat. Med. 2022, 1.
  38. Bar-On, Y.M.; Goldberg, Y.; Mandel, M.; Bodenheimer, O.; Freedman, L.; Kalkstein, N.; Mizrahi, B.; Alroy-Preis, S.; Ash, N.; Milo, R.; et al. Protection of BNT162b2 Vaccine Booster against COVID-19 in Israel. N. Engl. J. Med. 2021, 385, 1393–1400.
  39. Arbel, R.; Hammerman, A.; Sergienko, R.; Friger, M.; Peretz, A.; Netzer, D.; Yaron, S. BNT162b2 Vaccine Booster and Mortality Due to COVID-19. N. Engl. J. Med. 2021, 385, 2413–2420.
  40. Barda, N.; Dagan, N.; Cohen, C.; Hernán, M.A.; Lipsitch, M.; Kohane, I.S.; Reis, B.Y.; Balicer, R.D. Effectiveness of a third dose of the BNT162b2 mRNA COVID-19 vaccine for preventing severe outcomes in Israel: An observational study. Lancet 2021, 398, 2093–2100.
  41. Gardner, B.J.; Kilpatrick, A.M. Third doses of COVID-19 vaccines reduce infection and transmission of SARS-CoV-2 and could prevent future surges in some populations: A modeling study. medRxiv 2021.
  42. Keeling, M.J.; Thomas, A.; Hill, E.M.; Thompson, R.N.; Dyson, L.; Tildesley, M.J.; Moore, S. Waning, Boosting and a Path to Endemicity for SARS-CoV-2. medRxiv 2021.
  43. Cromer, D.; Steain, M.; Reynaldi, A.; Schlub, T.E.; Wheatley, A.K.; Juno, J.A.; Kent, S.J.; Triccas, J.A.; Khoury, D.S.; Davenport, M.P. Neutralising antibody titres as predictors of protection against SARS-CoV-2 variants and the impact of boosting: A meta-analysis. Lancet Microbe 2022, 3, e52–e61.
  44. Reardon, S. How well can Omicron evade immunity from COVID vaccines? Nature 2022.
  45. Li, M.; Lou, F.; Fan, H. SARS-CoV-2 variant Omicron: Currently the most complete “escapee” from neutralization by antibodies and vaccines. Signal Transduct. Target. Ther. 2022, 7, 28.
  46. Burki, T.K. Fourth dose of COVID-19 vaccines in Israel. Lancet Respir. Med. 2022, 10, E19.
  47. Iacobucci, G. COVID-19: Fourth vaccine doses—Who needs them and why? BMJ 2022, 376, o30.
  48. Ministry of Health of Canada COVID-19 Vaccine Third Dose Recommendations. Available online: https://www.health.gov.on.ca/en/pro/programs/publichealth/coronavirus/docs/vaccine/COVID-19_vaccine_third_dose_recommendations.pdf (accessed on 6 April 2022).
  49. Preliminary Data Analysis: Effectiveness of the Fourth Dose for Older Adults 60 Years of Age and Older. Available online: https://www.gov.il/en/Departments/news/23012022-01 (accessed on 25 January 2022).
  50. Watson, C. Three, four or more: What’s the magic number for booster shots? Nature 2022, 602, 17–18.
  51. Callaway, E. Beyond Omicron: What’s next for COVID’s viral evolution. Nature 2021, 600, 204–207.
  52. Pango Cov-Lineages. Available online: httpshttps://cov-lineages.org/lineage_list.htmlsublineage-ba.2 (accessed on 5 April 2022).
  53. Cele, S.; Jackson, L.; Khoury, D.S.; Khan, K.; Moyo-Gwete, T.; Tegally, H.; San, J.E.; Cromer, D.; Scheepers, C.; Amoako, D.G.; et al. Omicron extensively but incompletely escapes Pfizer BNT162b2 neutralization. Nature 2022, 602, 654–656.
  54. Liu, L.; Iketani, S.; Guo, Y.; Chan, J.F.W.; Wang, M.; Liu, L.; Luo, Y.; Chu, H.; Huang, Y.; Nair, M.S.; et al. Striking antibody evasion manifested by the Omicron variant of SARS-CoV-2. Nature 2022, 602, 676–681.
  55. WHO Statement on Omicron Sublineage BA.2. Available online: https://www.who.int/news/item/22-02-2022-statement-on-omicron-sublineage-ba.2 (accessed on 5 April 2022).
  56. Centers of Disease and Control, C. CDC COVID Data Tracker. Available online: https://covid.cdc.gov/covid-data-tracker/#variant-proportions (accessed on 5 April 2022).
  57. Yu, J.; Collier, A.-R.Y.; Rowe, M.; Mardas, F.; Ventura, J.D.; Wan, H.; Miller, J.; Powers, O.; Chung, B.; Siamatu, M.; et al. Neutralization of the SARS-CoV-2 Omicron BA.1 and BA.2 Variants. N. Engl. J. Med. 2022.
  58. Stegger, M.; Edslev, S.M.; Sieber, R.N.; Ingham, A.C.; Ng, K.L.; Tang, M.-H.E.; Alexandersen, S.; Fonager, J.; Legarth, R.; Utko, M.; et al. Occurrence and significance of Omicron BA.1 infection followed by BA.2 reinfection. medRxiv 2022.
  59. Chemaitelly, H.; Ayoub, H.H.; AlMukdad, S.; Coyle, P.; Tang, P.; Yassine, H.M.; Al-Khatib, H.A.; Smatti, M.K.; Hasan, M.R.; Al-Kanaani, Z.; et al. Duration of mRNA vaccine protection against SARS-CoV-2 Omicron BA.1 and BA.2 subvariants in Qatar. medRxiv 2022.
  60. Pfizer Pfizer and BioNTech Initiate Study to Evaluate Omicron-Based COVID-19 Vaccine in Adults 18 to 55 Years of Age. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizer-and-biontech-initiate-study-evaluate-omicron-based (accessed on 8 February 2022).
  61. Waltz, E. Does the world need an Omicron vaccine? What researchers say. Nature 2022, 602, 192–193.
  62. Morens, D.M.; Taubenberger, J.K.; Fauci, A.S. Universal Coronavirus Vaccines—An Urgent Need. N. Engl. J. Med. 2022, 386, 297–299.
  63. Sheridan, C. COVID-19 vaccine makers chase variant-ready vaccines. Nat. Biotechnol. 2022, 40, 141–143.
  64. Kim, Y.C.; Dema, B.; Reyes-Sandoval, A. COVID-19 vaccines: Breaking record times to first-in-human trials. npj Vaccines 2020, 5, 34.
  65. Dodd, R.H.; Pickles, K.; Nickel, B.; Cvejic, E.; Ayre, J.; Batcup, C.; Bonner, C.; Copp, T.; Cornell, S.; Dakin, T.; et al. Concerns and motivations about COVID-19 vaccination. Lancet Infect. Dis. 2021, 21, 161–163.
  66. Blumenthal, K.G.; Phadke, N.A.; Bates, D.W. Safety Surveillance of COVID-19 mRNA Vaccines Through the Vaccine Safety Datalink. JAMA 2021, 326, 1375.
  67. Albalawi, O.M.; Alomran, M.I.; Alsagri, G.M.; Althunian, T.A.; Alshammari, T.M. Analyzing the U.S. Post-marketing safety surveillance of COVID-19 vaccines. Saudi Pharm. J. 2022, 30, 180–184.
  68. Kim, J.H.; Marks, F.; Clemens, J.D. Looking beyond COVID-19 vaccine phase 3 trials. Nat. Med. 2021, 27, 205–211.
  69. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416.
  70. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615.
  71. Coronavirus Disease (COVID-19): Vaccines Safety. Available online: https://www.who.int/news-room/questions-and-answers/item/coronavirus-disease-(covid-19)-vaccines-safety (accessed on 11 February 2022).
  72. Pfizer-BioNTech COVID-19 Vaccine Reactions & Adverse Events. Available online: https://www.cdc.gov/vaccines/covid-19/info-by-product/pfizer/reactogenicity.html (accessed on 11 February 2022).
  73. European Medicines Agency Committee for Medicinal Products for Human Use (CHMP) Assessment Report; European Medicines Agency: Amsterdam, The Netherlands, 2021.
  74. Soiza, R.L.; Scicluna, C.; Thomson, E.C. Efficacy and safety of COVID-19 vaccines in older people. Age Ageing 2021, 50, 279–283.
  75. Sacco, K.; Castagnoli, R.; Vakkilainen, S.; Liu, C.; Delmonte, O.M.; Oguz, C.; Kaplan, I.M.; Alehashemi, S.; Burbelo, P.D.; Bhuyan, F.; et al. Immunopathological signatures in multisystem inflammatory syndrome in children and pediatric COVID-19. Nat. Med. 2022, 1–13.
  76. Vogel, T.P.; Top, K.A.; Karatzios, C.; Hilmers, D.C.; Tapia, L.I.; Moceri, P.; Giovannini-Chami, L.; Wood, N.; Chandler, R.E.; Klein, N.P.; et al. Multisystem inflammatory syndrome in children and adults (MIS-C/A): Case definition & guidelines for data collection, analysis, and presentation of immunization safety data. Vaccine 2021, 39, 3037–3049.
  77. Bartsch, Y.C.; Wang, C.; Zohar, T.; Fischinger, S.; Atyeo, C.; Burke, J.S.; Kang, J.; Edlow, A.G.; Fasano, A.; Baden, L.R.; et al. Humoral signatures of protective and pathological SARS-CoV-2 infection in children. Nat. Med. 2021, 27, 454–462.
  78. Weatherhead, J.E.; Clark, E.; Vogel, T.P.; Atmar, R.L.; Kulkarni, P.A. Inflammatory syndromes associated with SARS-CoV-2 infection: Dysregulation of the immune response across the age spectrum. J. Clin. Investig. 2020, 130, 6194–6197.
  79. Bastug, A.; Aslaner, H.; Aybar Bilir, Y.; Kemirtlek, N.; Gursoy, F.M.; Bastug, S.; Bodur, H. Multiple system inflammatory syndrome associated with SARS-CoV-2 infection in an adult and an adolescent. Rheumatol. Int. 2021, 41, 993.
  80. Nune, A.; Iyengar, K.P.; Goddard, C.; Ahmed, A.E. Multisystem inflammatory syndrome in an adult following the SARS-CoV-2 vaccine (MIS-V). BMJ Case Rep. 2021, 14, e243888.
  81. Uwaydah, A.K.; Hassan, N.M.M.; Abu Ghoush, M.S.; Shahin, K.M.M. Adult multisystem inflammatory syndrome in a patient who recovered from COVID-19 postvaccination. BMJ Case Rep. 2021, 14, e242060.
  82. Park, J.W.; Yu, S.N.; Chang, S.H.; Ahn, Y.H.; Jeon, M.H. Multisystem Inflammatory Syndrome in an Adult after COVID-19 Vaccination: A Case Report and Literature Review. J. Korean Med. Sci. 2021, 36, e312.
  83. Belay, E.D.; Godfred Cato, S.; Rao, A.K.; Abrams, J.; Wyatt Wilson, W.; Lim, S.; Newton-Cheh, C.; Melgar, M.; DeCuir, J.; Webb, B.; et al. Multisystem Inflammatory Syndrome in Adults After Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection and Coronavirus Disease 2019 (COVID-19) Vaccination. Clin. Infect. Dis. 2021, ciab936.
  84. Arrow, K.; Burgoyne, L.L.; Cyna, A.M. Implications of nocebo in anaesthesia care. Anaesthesia 2022, 77, 11–20.
  85. Graham, F. Daily briefing: ‘Nocebo’ effect underlies most COVID vaccine side effects. Nature 2022.
  86. Haas, J.W.; Bender, F.L.; Ballou, S.; Kelley, J.M.; Wilhelm, M.; Miller, F.G.; Rief, W.; Kaptchuk, T.J. Frequency of Adverse Events in the Placebo Arms of COVID-19 Vaccine Trials. JAMA Netw. Open 2022, 5, e2143955.
  87. Sever, P.P. Nocebo affects after COVID-19 vaccination. Lancet Reg. Health-Eur. 2022, 12, 100273.
  88. Razai, M.S.; Chaudhry, U.A.R.; Doerholt, K.; Bauld, L.; Majeed, A. COVID-19 vaccination hesitancy. BMJ 2021, 373, n1138.
  89. Baker, A.T.; Boyd, R.J.; Sarkar, D.; Teijeira-Crespo, A.; Chan, C.K.; Bates, E.; Waraich, K.; Vant, J.; Wilson, E.; Truong, C.D.; et al. ChAdOx1 interacts with CAR and PF4 with implications for thrombosis with thrombocytopenia syndrome. Sci. Adv. 2021, 7, 8213.
  90. Fu, W.; Sivajohan, B.; McClymont, E.; Albert, A.; Elwood, C.; Ogilvie, G.; Money, D. Systematic review of the safety, immunogenicity, and effectiveness of COVID-19 vaccines in pregnant and lactating individuals and their infants. Int. J. Gynecol. Obstet. 2022, 156, 406–417.
  91. Goncu Ayhan, S.; Oluklu, D.; Atalay, A.; Menekse Beser, D.; Tanacan, A.; Moraloglu Tekin, O.; Sahin, D. COVID-19 vaccine acceptance in pregnant women. Int. J. Gynecol. Obstet. 2021, 154, 291–296.
  92. Meyyazhagan, A.; Pushparaj, K.; Balasubramanian, B.; Kuchi Bhotla, H.; Pappusamy, M.; Arumugam, V.A.; Easwaran, M.; Pottail, L.; Mani, P.; Tsibizova, V.; et al. COVID-19 in pregnant women and children: Insights on clinical manifestations, complexities, and pathogenesis. Int. J. Gynecol. Obstet. 2022, 156, 216–224.
  93. Sarwal, Y.; Sarwal, T.; Sarwal, R. Prioritizing pregnant women for COVID-19 vaccination. Int. J. Gynecol. Obstet. 2021, 155, 57–63.
  94. Sarwal, Y.; Sarwal, T.; Sarwal, R. Vaccination of pregnant women against COVID-19 in India and Indonesia: Moving beyond the opt-in to the opt-out option. Int. J. Gynecol. Obstet. 2021, 155, 549–550.
  95. Witberg, G.; Barda, N.; Hoss, S.; Richter, I.; Wiessman, M.; Aviv, Y.; Grinberg, T.; Auster, O.; Dagan, N.; Balicer, R.D.; et al. Myocarditis after COVID-19 Vaccination in a Large Health Care Organization. N. Engl. J. Med. 2021, 385, 2132–2139.
  96. Heymans, S.; Cooper, L.T. Myocarditis after COVID-19 mRNA vaccination: Clinical observations and potential mechanisms. Nat. Rev. Cardiol. 2021, 19, 75–77.
  97. Maiese, A.; Frati, P.; Del Duca, F.; Santoro, P.; Manetti, A.C.; La Russa, R.; Di Paolo, M.; Turillazzi, E.; Fineschi, V. Myocardial Pathology in COVID-19-Associated Cardiac Injury: A Systematic Review. Diagnostics 2021, 11, 1647.
  98. Block, J.P.; Boehmer, T.K.; Forrest, C.B.; Carton, T.W.; Lee, G.M.; Ajani, U.A.; Christakis, D.A.; Cowell, L.G.; Draper, C.; Ghildayal, N.; et al. mm7114e1 Cardiac Complications After SARS-CoV-2 Infection and mRNA COVID-19 Vaccination—PCORnet, United States, January 2021–January 2022. MMWR Morb. Mortal. Wkly. Rep. 2022, 71, 517–523.
  99. Por Unanimidade, CoronaVac é Aprovada Pela Anvisa para uso Emergencial em Crianças de Seis a 17 Anos. Available online: https://butantan.gov.br/noticias/por-unanimidade-coronavac-e-aprovada-pela-anvisa-para-uso-emergencial-em-criancas-de-seis-a-17-anos- (accessed on 25 January 2022).
  100. Heymans, S.; Eriksson, U.; Lehtonen, J.; Cooper, L.T. The Quest for New Approaches in Myocarditis and Inflammatory Cardiomyopathy. J. Am. Coll. Cardiol. 2016, 68, 2348–2364.
More
Information
Subjects: Others
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 427
Revisions: 2 times (View History)
Update Date: 21 Apr 2022
1000/1000
Video Production Service