Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Heliyon. 2023 Jun; 9(6): e16813.
Published online 2023 Jun 7. doi: 10.1016/j.heliyon.2023.e16813
PMCID: PMC10245239
PMID: 37303517

A comprehensive insight on the challenges for COVID-19 vaccine: A lesson learnt from other viral vaccines

Associated Data

Data Availability Statement

Abstract

The aim of this study is to comprehensively analyze previous viral vaccine programs and identify potential challenges and effective measures for the COVID-19 vaccine program. Previous viral vaccine programs, such as those for HIV, Zika, Influenza, Ebola, Dengue, SARS, and MERS, were evaluated. Paramount challenges were identified, including quasi-species, cross-reactivity, duration of immunity, revaccination, mutation, immunosenescence, and adverse events related to viral vaccines. Although a large population has been vaccinated, mutations in SARS-CoV-2 and adverse events related to vaccines pose significant challenges. Previous vaccine programs have taught us that predicting the final outcome of the current vaccine program for COVID-19 cannot be determined at a given state. Long-term follow-up studies are essential. Validated preclinical studies, long-term follow-up studies, alternative therapeutic approaches, and alternative vaccines are necessary.

Keywords: COVID-19, HIV, Influenza, Dengue, Ebola, Zika, SARS, MERS, Omicron, Delta variant, Vaccine

Highlights

  • The currently available data on the COVID-19 vaccine development program is limited.
  • Comprehensive analysis of previous vaccine programs has presented challenges.
  • The identified common challenges are also applicable to the COVID-19 vaccine program.
  • Longevity, adverse effects, revaccination, and mutation are significant barriers.
  • Validated preclinical and long-term follow-up studies are necessary.

1. Introduction

On December 31, 2019, several cases of pneumonia caused by a novel coronavirus were observed in Wuhan, China. The virus is believed to have been transmitted from animals to humans in a local market in China. The novel coronavirus began as human-to-human transmission, resulting in the spread of respiratory illness throughout the Wuhan region of China [1]. On February 11, 2020, the World Health Organization (WHO) identified the disease and named it COVID-19, while the virus itself was named SARS-CoV-2 [2]. COVID-19 is characterized by symptoms such as fever, cough, loss of taste or smell, fatigue, severe acute respiratory illness (SARI), organ failure, and mortality. Among the elderly population with underlying chronic illnesses, including hypertension, cardiovascular diseases, and diabetes, the disease resulted in a high number of fatalities [3]. The molecular diagnosis of SARS-CoV-2 involves both general and advanced polymerase chain reaction (PCR)-based techniques using various types of biological samples from the patient [4]. SARS-CoV-2 has a high transmission rate, with a reproductive number (R0) of 2.2 [1]. COVID-19 triggered a devastating pandemic that strongly shows the urgent need for a therapeutic approach to control and prevent it. Although repurposed drug strategies and medicinal plants have shown potent activity, studies are still under investigation [4]. Two conventional methods are used for vaccine development: inactive and live-attenuated vaccines [5]. Other vaccine platforms, such as viral vectors, virus-like particles (VLPs), DNA-based, protein-based, and mRNA-based vaccines, are in the preliminary stages of development [6]. Previous vaccine development strategies for similar viral family members have yielded promising results, paving the way for speedy and efficient vaccine development against newly emerged viruses like SARS-CoV-2 [7]. Several antigenic targets (surface-based or internal), vaccine development platforms, and clinical trial approaches were based on prior studies of various targets used for influenza, Zika, Ebola, HIV, and dengue viruses after their emergence [8]. The S-protein (spike protein) is a potential vaccine candidate target in pre-clinical or experimental models for a diversity of novel viral categories, including SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV) [9,10]. Several SARS-CoV and MERS-CoV vaccine strategies, such as DNA plasmids and inactivated virus, have been evaluated in human trials, but none showed efficacy and safety in late-phase clinical trials, and no vaccine candidate is available for the treatment of SARS-CoV and MERS-CoV diseases [11]. The failure of a particular vaccine candidate can be attributed to various confounders, such as issues with participant enrolment, administrative reasons, safety, efficacy, and immunogenicity [12]. No effective vaccines have been approved against other human coronaviruses [13]. Extensive in-silico, in-vitro, and in-vivo research and well-organized clinical trials are required for the successful development of a vaccine against viral diseases. A race among vaccine makers has started to develop an efficacious vaccine in a short span of time by bypassing the conventional protocols for evaluating long-term safety parameters.

The availability of relevant literature and previous experience with vaccine development against other viral diseases can potentially aid in the quick development of a vaccine against COVID-19. By comparing the methods and technical platforms used for previous emerging and re-emerging infectious illnesses and pandemics, valuable lessons can be learned to expedite the development of a vaccine for COVID-19. The success of vaccine development heavily depends on prior knowledge about the antigen, use of animal models, testing of animal models, and use of adjuvants and immunogenic response. However, studies that focus on learning strategies and lessons from previous viral vaccine development programs are limited. Therefore, the current study aims to evaluate vaccine development programs for other viral diseases, including HIV, Influenza, Dengue, Ebola, Zika, SARS, and MERS, to provide critical insights into the challenges faced by these programs. The findings of the study will be beneficial in understanding the long-term safety and efficacy of the COVID-19 vaccine development program.

2. Methods

To identify relevant studies, extensive searches were conducted on PubMed and Google Scholar databases using scientific keywords, including ‘Human immunodeficiency virus' OR ‘HIV’ OR ‘Dengue’ OR ‘Influenza, Human’ OR ‘Zika Virus' OR ‘Ebola virus' OR ‘Middle East Respiratory Syndrome Coronavirus' OR ‘SARS Virus' OR ‘COVID-19′ OR 'coronavirus' AND 'vaccines' OR ‘Therapeutic vaccine’ OR ‘immunotherapy’. Additionally, the progress and prospects for clinical development of immunotherapies aimed at curing HIV, Dengue, Influenza, Zika, Ebola, and Coronavirus-related infections were reviewed by searching the ClinicalTrial.gov database.

2.1. Challenges for HIV vaccine development

A total of 861 clinical trials have been registered for the HIV vaccine so far, out of which 708 were interventional trials. Of these, 603 clinical trials have been completed successfully, but 48 clinical trials failed due to suspension, withdrawal, or termination. Although the failure rate is minimal, there is currently no approved vaccine for HIV. The following are important challenges that need to be overcome for the development of an HIV vaccine.

  • 1)
    HIV - a chronic disease not acute

The current preventive vaccine strategies for HIV focus on targeting the virus and immune response produced during the late stage of the disease, which is not an effective approach. This is because a highly mutated strain of HIV is identified during the chronic disease stage, which results in neutralizing antibodies with unique characteristics. The current strategies are focused on developing immunogens that stimulate these broadly neutralizing antibodies, which are only conserved to the individual infected host and the result of long-term evolutionary within-host pressure. To design a preventive HIV vaccine, it is critical to understand the early stage interaction of the HIV viral variables and the immune response [14].

  • 2)
    HIV as Quasispecies

Quasispecies are recombinant and mutant genomes of viruses that are closely related and undergo progressive genetic variations, forming a unit that can be selected [15]. HIV also behaves as a quasispecies [16]. There is significant genetic diversity during the initial phase of HIV infection in the host and in the virus during the terminal stage. During transmission, HIV undergoes an evolutionary bottleneck event resulting in a reduction of fitness and mutations. However, there are conserved sequences in the transmitted/founder (T/F) viruses that are resistant to evolutionary mucosal bottleneck events and are the primary targets for vaccine research [17]. This means that T/F virus variants remain homogenous just before and just after transmission and can be targeted using neutralizing antibodies towards the conserved sequences [18].

  • 3)
    Ignorance of immunological principles

The interaction of HIV epitopes was predicted by structure-based reverse vaccinology with neutralizing antibodies that produce a similar immune response as produced against the vaccine antigen. Efforts were made to identify the pathways of affinity maturation that occur during the production of broadly neutralizing antibodies [19]. However, this approach completely ignores the significant complex interactions of the HIV virus and the host immune system. The polyspecific nature of antibodies, B-cell receptors, and the degeneracy of the immune system have also not been considered [20]. Errors during reverse transcription lead to the diversification of the HIV-1 M group into various recombinant forms. Within a particular clade, there is a difference of nearly 20% in Env amino acid sequences, and the difference is more than 35% between the various clades. Therefore, an immunogen for a vaccine will have to deal with a very high level of viral variety [9].

  • 4)
    Glycosylation of HIV envelope

Developing vaccines that induce broad-spectrum neutralizing antibodies (bnAbs) against different HIV strains is an important strategy [21]. However, a major challenge is to optimize the immunogen to present the precise epitopes identified by bnAbs [22]. Another obstacle is the high glycosylation of the HIV envelope. The region where monoclonal antibodies (mAbs) bind is conserved and covered with glycans derived from the host, which have low immunogenicity and complexity, making it difficult for mAbs to recognize and interact with them [23].

2.1.1. Future prospects of HIV vaccine development program

Clinical trials focused on certain scientific theories, such as better antibody and T-cell-based vaccines, may benefit the HIV-1 vaccine sector more than those solely focused on product development. However, there is a current debate over whether the field can “withstand” another vaccine efficacy study failure. Therefore, the decision to progress a vaccine candidate into effectiveness trials should be made with great care and be based on a thorough study of preclinical and clinical data. To successfully develop an effective and safe HIV-1 vaccine, one must be prepared to overcome obstacles and accept failures [24].

2.2. Challenges of zika virus vaccine

There are currently 30 registered clinical trials for the Zika vaccine, 20 of which are interventional studies. Of these, 16 have already been completed, but none of the vaccines have been approved for Zika virus. The following are the key challenges in the development of a Zika vaccine.

  • 1)
    Antigen Identification

Guillain-Barre syndrome (GBS) occurs in approximately one out of every 4000 to 5000 Zika-infected patients. It is important to identify the antigen that attacks the peripheral nerves in these patients [25].

  • 2)
    Cross-reactivity

Zika virus vaccine-induced antibodies can significantly increase dengue viremia and other flavivirus infections due to the challenge of cross-reactivity among antibodies. The severity of Zika virus infection is also influenced by cross-reactive T-cell responses from earlier dengue infections. Therefore, the challenge is to develop a Zika virus vaccine that can produce more specific viral antibodies. Another important challenge is to identify more specific epitopes that can generate more specific neutralizing antibodies [26].

  • 3)
    Human Infection models

Testing the efficacy of Zika virus vaccinations in both pregnant and non-pregnant populations is difficult due to the decline in active cases of Zika virus infection. This has created an unmet need for developing human infection models that can accelerate vaccine research and other countermeasures [27].

  • 4)
    Sexual transmission risk

The Zika virus can persist in the male urogenital tract for 3–8 months, indicating a risk of sexual transmission of the disease. Additionally, there is a potential risk of infection during pregnancy [28].

2.2.1. Future prospects of ZIKV vaccine development program

Using knowledge of the atomic structure of flavivirus E dimers, the overall structure of the virion, mechanisms of neutralization, and other facets of flavivirus biology and immunology, ZIKV vaccines were developed quickly, and candidate vaccines advanced to the point of efficacy testing with unprecedented speed. Due to this, some other programs have had to be delayed. Despite funding efforts, the incidence of new ZIKV infections is declining in many places, which may make it more difficult to assess the vaccine's effectiveness in field trials. Alternate routes to licensure might be required. In the future, it would be desirable to have pre-assigned budgets accessible for emergency responses [24]. It has been demonstrated that a number of ZIKV vaccine candidates are safe, well-tolerated, and immunogenic in humans. Neutralizing antibody titers similar to those found to be protective in pre-clinical models were elicited in the majority of trial participants. Efforts are being made to create animal models for testing vaccine effectiveness in preventing congenital Zika syndrome. Thanks to the astoundingly quick development of its vaccines, we now understand much more about the ZIKV virus. However, there are still significant obstacles to conducting clinical effectiveness trials and obtaining vaccine licensure. A vaccine is urgently required due to the congenital Zika syndrome and the potential lifelong effects on infants born to mothers who have the virus [24].

2.3. Challenges in influenza vaccine development program

There are currently 1703 clinical trials registered for the influenza vaccine, of which 1412 are related to vaccine intervention and the rest are observational or non-interventional. Only 61 clinical trials have been suspended, withdrawn, or terminated so far. There are 26 FDA-approved vaccines for influenza. Although there were a few challenges in the development of the influenza vaccine, addressing those challenges can certainly be helpful for the development of other vaccines.

  • 1)
    Availability issue of embryonated eggs

The production of inactivated (IIV) and attenuated (LAIV) Influenza vaccines primarily relies on embryonated eggs. However, during a pandemic or a rise in the pathogenic strain against poultry, their use may be limited. Nevertheless, the emergence of several new types of vaccines that are now developed in mammalian cell lines, recombinant HA vaccine, and DNA vaccine provide an alternative to this challenge.

  • 2)
    Time lag in vaccine production

The production of Influenza vaccines targeting specific strains is a time-consuming process that involves various stages. These stages include global surveillance of circulating strains, antigenic characterization, prediction of the upcoming season, production of vaccine seed virus, amplification, inactivation, purification, and dispensing [29]. During the 2009 pandemic of the Influenza virus, there was a challenge in producing and distributing a newly evolved viral vaccine in a short period of time [30].

  • 3)
    Revaccination

There is a need for revaccination against Influenza due to a decrease in viral strain-specific antibodies and the phenomenon of antigenic drift. Strategies such as the use of computational HA sequence analysis, generation of specific viral strain antibodies, and immunization with conserved proteins targeting T-cell responses may be helpful in addressing this issue [31].

  • 4)
    Novel virus

The current vaccines available against seasonal flu are effective. However, the emergence of novel strains or antigenic drift poses a great challenge to the current strategies. Four previous novel strains of Influenza (in 1918, 1957, 1968, and 2009) caused pandemics [32].

  • 5)
    Immunosenescence

The gradual decrease in immune response with increasing age is known as “immunosenescence.” The current standard Influenza vaccines are found to be less effective in elderly populations, who are more susceptible to Influenza and its complications. Increasing the antigenic dose in the vaccine and adding an adjuvant can be a useful approach [33].

  • 6)
    Adverse effect

The preclinical studies in animals showed that vaccines were associated with enhanced respiratory illness in pigs but this has not been reported in humans [34]. The AS03-adjuvanted Influenza A (H1N1) pdm09 vaccine has been reported to induce sleep disorder (narcolepsy) in Northern Europe [35].

  • 7)
    Underestimation of the disease burden

The estimated Influenza-related mortality is between 291,234 and 645,832 [35]. Elderly individuals, children, and pregnant women are more susceptible to Influenza-related critical illness [36]. In most countries, Influenza vaccination is not mandatory [37], but it is essential to educate the general population about the importance of receiving the vaccine.

2.3.1. Future prospects of influenza vaccine development program

A coordinated immune response that targets conserved areas of various influenza virus proteins can activate both B and T cells. Currently, numerous groups are assessing vaccines targeting various proteins, using various administration platforms, and investigating the influence of immunological history or imprinting on vaccine response. Additionally, innovative techniques for producing vaccine viruses that are not dependent on eggs or cell culture are being explored. These research findings will help researchers develop better seasonal flu vaccinations and act as models for the development of broadly protective, enduring immunity that could offer protection from emerging influenza strains [24]. Studies have shown that influenza vaccines can reduce the risk of serious complications, hospitalization, and death, albeit at varying rates depending on the host and other variables such as storage, handling, and proper administration within the allotted time frame that results in protective levels of immunity. Improved study designs are needed to address the urgent need for funding to carry out seasonal studies, develop better vaccines with greater immunogenicity and efficacy, and to generate specific research outcomes. Universal vaccines and more recent adjuvants for vaccines could contribute to the solution. The benefits of existing influenza vaccines outweigh any known hazards and should be regularly administered as a crucial component of influenza prophylaxis. Validated data demonstrating actual vaccination failure and its underlying cause(s) are essential for guiding further investigation into the molecular underpinnings of vaccine failure. In turn, the findings of such research are likely to inspire fresh, targeted efforts to develop vaccines that are more highly immunogenic and effective. The development of higher-dimensional systems biology and vaccineomics techniques is necessary to comprehend the maturation of immunity following vaccination, as well as the identification of molecular signatures and biomarkers for vaccine immunogenicity [24].

2.4. Challenges in dengue vaccines development

Almost 133 clinical trials have been registered, out of which 115 studies are interventional. In Mexico, in December 2015, Dengvaxia (CYD-TDV) became the first licensed Dengue vaccine with an age restriction of 9–45 years for individuals residing in endemic regions. CYD-TDV is a three-dose series of a live recombinant tetravalent Dengue vaccine administered according to a 0/6/12-month schedule, and it has been developed by Sanofi Pasteur [38]. There are several challenges in the path of developing a Dengue vaccine, which are listed below.

  • 1)
    Antibody-Dependent Enhancement (ADE)

The Dengue vaccine is designed to be effective against all four serotypes (Type 1, 2, 3, 4). The primary role of antibodies is to neutralize the antigen, but sometimes they can enhance the entry of the antigen, a phenomenon known as antibody-dependent enhancement (ADE) [39]. ADE occurs due to the cross-reactivity of antibodies produced during some other heterologous infection, resulting in increased viral entry by escaping the endosomal-dependent pathway [40]. This phenomenon has been observed in HIV, Ebola, and SARS-CoV-2-related infections.

The first theory suggests that the Fc region of antibodies binds to the FcR receptor present on immune cells like macrophages, monocytes, and B-cells, producing an immune synapse that enhances the entry of the virus [41]. The second theory proposes that ADE occurs via the complement-dependent pathway, where virus-C3 fragments bind with the gp120 protein in HIV, and this complex binds to receptors present on the cell, thereby increasing viral entry [43]. The third theory suggests that neutralizing antibodies bind to the natural receptor of the virus (spike receptor) and cause ADE [44]. In cases where the natural receptors are not present, viruses can also enter via FcR-mediated ADE, as observed in foot and mouth diseases. ADE can also occur when there is a poor response to vaccination [45]. The majority of ADE cases have been reported in people who were previously infected with a different serotype of Dengue or another flavivirus.

  • 2)
    Cross-reactivity

There is a structural similarity between DENV and other flaviviruses, such as Japanese encephalitis virus (JEV), Zika virus (ZIKV), and yellow fever virus (YFV). The envelope (E) protein in these flaviviruses is highly conserved and is the main target of the immune response. The DENV E protein is almost 50% similar to that of ZIKV. This close structural homology between these viruses causes cross-reactivity, which can provide protection or pathogenic enhancement for other infections [46].

In 2016, a ZIKV outbreak occurred in regions of Brazil and Mexico that were also endemic for DENV [47]. Both humoral and cellular immune responses contribute to the pathology, but humoral immunity is the primary cause of ADE.

  • 3)
    Lack of adequate animal model

One of the most challenging aspects of vaccine research and development in the early stages is the lack of a suitable animal disease model. Experimental animal models of infection play a significant role in understanding disease pathology, the mechanisms of action of different therapeutic strategies, and the safety and efficacy profiles of vaccines [48]. To study the mechanism, effectiveness, and immunogenicity of the Dengue vaccine, a suitable and validated animal model is urgently needed.

Non-human primates develop Dengue-like infections that result in acute neutralizing antibodies and high viremia, but they lack hemorrhage, fever, and shock [49]. A recent study observed promising clinical symptoms of Dengue virus infection in the tree shrew model, but further validation is necessary [50].

2.4.1. Future prospects in dengue vaccine development program

While TAK-003 has the dengue virus serotype 2 backbone and the US NIH vaccine contains three complete genomes of the four dengue virus serotypes, CYD-TDV (the first licensed vaccine for Dengue) does not contain the non-structural proteins of dengue. Intense research is being done to determine whether second-generation vaccinations will offer enhanced protection against all four serotypes by including non-structural proteins from the dengue backbone and offering more convenient dosing with fewer doses required. While CYD-TDV (Dengvaxia) is licensed for three doses with an interval of six months for each dose, Takeda's TAK-003 shows better efficacy for serotypes 3 and 4 compared to serotypes 1 and 2. TAK-003 is being considered for two doses with a gap of three months. Sequential vaccination should be taken into consideration in addition to a heterologous prime-boost strategy to enhance tetravalent protection [24].

2.5. Challenges in the development of ebola vaccines

There are 78 clinical trials registered for Ebola, out of which 71 are interventional. None of the trials have been suspended, withdrawn, or terminated. ERVEBO, the Ebola Zaire Vaccine Live developed by Merck Sharp & Dohme Corp, is designed for individuals aged 18 years and older for the prevention of disease caused by Zaire ebolavirus [51]. However, there are certain challenges in the development of Ebola vaccines.

  • 1)
    Presence of virus in bodily fluids

Previous studies have observed the prolonged presence of the Ebola virus in bodily fluids, such as semen and breast milk. This poses an important concern as the risk of sexual transmission of the disease and transmission during feeding of neonates is high [52].

Vaccine virus has been detected in urine, saliva, and fluid from skin vesicles, and there is a possibility of vaccine virus transmission.

  • 2)
    Duration of the immunity

The duration of immunity developed through the use of Ebola vaccines is currently unknown, and there is a need for long-term studies to evaluate the persistence of immune response [53].

  • 3)
    Limitation of vaccine effectiveness

While the approved vaccine ERVEBO can provide protection against Ebola, it is not fully effective, and adherence to infection control practices is still necessary. Additionally, the vaccine has not been evaluated in immunocompromised populations.

2.5.1. Future prospects of ebola vaccines development program

Although several Ebola vaccine platforms' mechanisms of protection are becoming more obvious, further research is still required to completely understand these mechanisms. A common indicator of protection across various Ebola virus (EBOV) vaccine methods is the titers of anti-GP (glycoprotein) IgG antibodies. Future trials should confirm and expand on the investigation of this crucial correlation. Glycoprotein sequence analysis amongst Ebola virus species reveals a considerable degree of variability in terms of cross-protection between the various EBOV types. To elicit enhanced cross-protection against different Ebola virus species, GP targeted vaccines would either require strong immunogenicity and proper presentation of glycoproteins from one species, or vaccines should be multivalent, encoding specific GPs for each species [24].

2.6. Challenges in the development of vaccines for coronavirus (SARS, MERS, and COVID-19)

There are 46 clinical trials registered for the SARS vaccine and 9 clinical trials registered for the MERS vaccine, but none of the approved vaccines are available for these diseases. For COVID-19, 1246 clinical trials were registered in a short amount of time, out of which 701 are vaccine interventions. Although several COVID-19 vaccines have been approved, challenges remain in the path of coronavirus vaccine development. The challenges faced in developing vaccines for HIV, Ebola, Dengue, and Zika are also applicable to coronavirus vaccine development. Mutation poses a significant challenge that the COVID-19 vaccine development program is still facing.

2.6.1. Quasispecies

Under the effect of host and virus mechanisms, RNA viruses undergo genetic variability referred to as quasispecies. One study conducted a deep analysis using a Next-generation sequencing platform to evaluate the phenomenon of quasispecies in COVID-19 patients with mild to severe pathologies. The study revealed that mutations induced by nsp 12 mainly occurred in patients with high viral loads and long-term infections. These genetic variations are the main source of new SARS-COV-2 variants [54].

Another study highlighted several single nucleotide variations (SNVs) affecting 70% of the viral genome with varying densities, revealing the colocalization of strain-specific and intra-host SNVs. The locations of genomic rearrangement are in the poly-A/poly-U regions in ORF1ab and spike S gene. The identified SNVs and rearrangements point out the genetically diverse quasispecies' intra-patient capacity, which may arise rapidly during the outbreak and allow the virus to evade the immune system [55] (refer to Fig. 1).

Fig. 1

Common Challenges faced by different Viral Vaccines programs & COVID-19.

2.6.2. Post-covid vaccine adverse events

Most of the world population has been safely vaccinated with COVID-19 vaccines. While studies of these vaccines have demonstrated significant safety data, there have been reported cases of adverse events related to COVID-19 vaccines, including autoimmune syndromes like Guillain-Barré syndrome and thrombotic thrombocytopenia. The exact mechanism of the development of these adverse events has not yet been established. Adjuvants used in vaccines may be associated with these cases [56]. Further studies are needed to investigate the association of COVID-19 vaccines with the innate-adaptive immune response and to better understand the underlying mechanism of vaccine-induced autoimmune syndromes [57].

2.6.3. Vaccine in older & immune-compromised people

The immune response of the older population with comorbidities towards vaccination varies compared to adults due to immunosenescence, inflammation, and altered immune responses to infections and vaccines [58]. Similarly, immunocompromised patients, who are suffering from dermatologic immune disorders treated with IL-17 inhibitors or IL-4/13 inhibitors, presented with a normal seroconversion rate after COVID-19 vaccination [59]. Renal transplant patients, who were on treatment with alkylating agents and antimetabolites, presented with a delay in immune response [60]. The safety and efficacy of COVID-19 vaccination have not been checked in the immunocompromised population in any clinical trial. Investigating how vaccination affects this particular patient population will be difficult, and special vaccination protocols need to be created. To determine how long seroconversion and immunity last after vaccination, regular activity of T and B cells as well as the antibody response must be examined.

2.6.4. Difficulty in cross-over study

The most effective design for a clinical trial involving a vaccine or a placebo is a crossover double-blind study, where all participants receive the intervention they were not given at the beginning of the trial [24]. However, the ability of trials sponsored by Pfizer-BioNTech, Moderna, AstraZeneca, and Janssen to enroll and retain patients is impacted by the temporary authorization of these four vaccines for adult and older patients. Placebo-controlled randomized controlled trials in areas where these licensed vaccines are accessible are further impacted by the deployment of additional vaccines in early 2021, such as Sputnik V (Russia), Sinovac (China), Bharat (India), and Sinopharm (China) [24].

2.6.5. Under developing countries

Unfortunately, in many low- and middle-income countries, only a small portion of the population received the SARS-CoV-2 vaccine in 2021–2022 [1,61]. For many COVID-19 vaccine candidates, conducting trials in under-resourced environments may be the only practical or reasonable course of action going forward. This would help to fulfill the justifiable desire for trials to take place in low- and middle-income countries' territories [61]. However, this directly contradicts what is mentioned in the Sustainable Development Goals, as many low- and middle-income countries are not given equal access to vaccines. The prime issue in such countries is the availability and cost of vaccines.

2.6.6. Infrastructure

Vaccine candidates for COVID-19 that will be examined in nations with restricted access to and affordability of vaccines for COVID-19 should be those that could ultimately be used at a low cost in the nations where the trials were conducted. Vaccine candidates that require extremely low temperatures (−70 °C) should not be tested in nations with low economies, as the storage conditions of these vaccines require high maintenance [62].

2.6.7. Mutation in novel coronavirus

Since 2003, the outbreak of Coronaviruses has posed a significant challenge to global health. SARS-CoV-2 is an RNA virus, and studies show that RNA viruses have higher mutation rates than most DNA viruses. A mutation in the sequence can cause changes in amino acids, and mutations in the surface (S) protein of viruses can significantly alter viral interactions with receptors and functions with broad-spectrum neutralizing antibodies [63]. For example, the A226V mutation in the Chikungunya virus E1 protein strengthened the virus's adaptability in the vector Aedes albopictus, ultimately leading to increased transmissibility and infectivity [64]. The A82V mutation in the GP protein of the Ebola virus leads to enhanced infectivity, transmissibility, and mortality [27]. The mutation in four amino acids in highly pathogenic Avian Influenza H5N1 reported enhanced transmissibility [65], whereas the A143 V/R148K (combined amino acid alteration) mutation of hemagglutinin in H7N9 increases resistance towards broadly neutralizing antibodies by ten times [66].

The SARS-CoV-2 spike (S) protein is highly glycosylated and is undergoing frequent mutations. These mutations may have significant biological effects that require investigation. Most of the variant changes in the receptor-binding domain due to amino acid alterations are less infectious, but some variants, such as V483A, A475V, F490L, and L452R, are resistant and can escape from neutralizing antibodies [67]. A SARS-CoV-2 variation expressing a D614G mutation in the S protein has affected the globe, implying a fitness advantage for the mutant. The infectivity was significantly higher in the case of D614G mutated variants as well as other variants containing both D614G and another amino acid alteration. The mutation led to a high infectivity rate, increased S-protein incorporation into the virion, and increased ACE2 binding affinity [68].

The UK consortium dataset on COVID-19 genomics presented a substitution of an amino acid in the receptor-binding domain (RBD) of the SARS-CoV-2 B.1.1.7 mutant strain (N501Y mutation). In-silico studies depicted that the N501Y mutation caused a more significant number of interactions by increasing electrostatic interactions due to the formation of hydrogen bonds between SARS-CoV-2-T500 and ACE-2 D355 near the mutation site [69]. The Delta variant (SARS-CoV-2 variant: B.1.617.2) appeared during the second wave of infections and has become widespread globally, although it is constantly evolving [70]. The distribution of the Delta variant was comparable to that of other variants of concern (VOCs) [70]. The D950 N mutation, like the D614G mutation, is localized to the trimer interface, suggesting that it may play a role in the control of spike protein dynamics [70]. It has a 156–157 deletion and a G158R mutation [70]. The ACE2 patch has just one mutation, at position 501, which improves the affinity of the receptor-binding domain (RBD) for ACE2 and is also implicated in antibody escape [70].

The emergence of the most mutated SARS-CoV-2 variant, B.1.1.529 (named Omicron), has sparked panic throughout the world and was first reported from South Africa on November 24, 2021 [71]. The spike protein of this variant has undergone 50 mutations and has about 30 alterations, including 13 mutations in the RBD in the S gene [71]. At both the ACE2 and antibody binding locations, 5 new mutations were discovered, indicating an improved immune escape capacity [71]. The Omicron variant has a higher antibody escape, transmissibility, and viral binding affinity [72]. Silent mutations in the RNA binding domain of a gene coding for the N-protein of SARS-CoV-2 have been reported in the Iranian strain compared to the Chinese strain. The silent mutation results in the replacement of thymine with cytosine so that the TTG in the Chinese strain becomes CTG in the Iranian strain, which codes for leucine. The missense mutation was reported during thymine DNA glycosylase action in humans and the base excision repair mechanism, resulting in the conversion of another thymine to cytosine. The codon after mutation is CCG, which codes for proline. This results in an enhanced affinity of the N-protein (through an RNA binding domain) to viral RNA, leading to an increase in transcription rate, pathogenicity, and transmission [73].

According to the US Centers for Disease Control and Prevention (CDC), vaccinations authorized by the US Food and Drug Administration (FDA) are “polyclonal,” meaning they develop antibodies that target several regions of the spike protein. This suggests that the virus would need numerous mutations to accumulate in the spike protein to avoid neutralizing antibodies and evade natural infection or vaccine-induced immunity. According to WHO [74], laboratory investigations are currently underway to investigate the biological features of these mutant viruses and how these variants may impact the efficiency of vaccines. There is currently insufficient data to identify whether such polymorphisms are associated with any significant changes in the severity of clinical aspects of disease development, general antibody response, or vaccination efficacy. The common challenges of all vaccine development programs are summarized in Fig. 1.

WHO - EUA approved vaccines (as shown in Fig. 2).

Fig. 2

WHO - EUA approved vaccines.

2.7. COVD-19 mRNA vaccine (COMIRNATY)

The BioNTech-Pfizer COVID-19 vaccine is a lipid-based nanoparticle, nucleoside-modified mRNA vaccine that expresses the viral perfused spike glycoprotein [75]. It was approved for Emergency Use Authorization (EUA) by the World Health Organization (WHO) on December 31st, 2020 [76]. It is authorized in 118 countries, and 49 clinical trials in 23 countries are still ongoing [77]. The BioNTech-Pfizer COVID-19 vaccine provides 95% protection against COVID-19 after two doses [75]. After receiving the vaccine, around 0.2% of adverse events were reported to the Vaccine Adverse Event Reporting System (VAERS) in the United States [75]. The most common side effects associated with the vaccination were fatigue, headache, muscular discomfort, joint pain, chills, nausea, and vomiting [78].

2.8. COVID-19 (ChAdOx1-S) vaccine (VAXZEVRIA/COVISHIELD)

Covishield is a recombinant, replication-deficient chimp adenovirus containing the S protein antigen of SARS-CoV-2 [79]. It was approved for Emergency Use Authorization (EUA) by the World Health Organization (WHO) on February 15th, 2021 [76]. It has been authorized for EUA in 133 countries and 52 clinical trials in 23 countries are currently ongoing [77]. The efficacy of the Oxford-AstraZeneca vaccine is 70.4% after two doses have been administered [80].

2.9. COVID-19 vaccine (Ad26.COV2·S)

The Ad26.COV2·S vaccine is a recombinant adenoviral vector that carries the sequence for the spike protein (S) of the SARS-CoV-2 virus [80]. The WHO granted Emergency Use Authorization on March 12, 2021 [76]. It is approved in 91 countries, and 16 clinical trials have already been registered in 18 countries [77]. After one dose, the Johnson & Johnson COVID-19 vaccine has an efficacy rate of 85% [81].

2.10. COVID-19 mRNA vaccine (SPIKEVAX)

Spikevax is based on a lipid nanoparticle-based nucleoside-modified mRNA vaccine that expresses the stabilized perfusion spike (S) glycoprotein of SARS-CoV-2 [82]. The WHO granted Emergency Use Authorization on April 30, 2021 [76]. It is authorized in 81 countries, and 35 clinical trials in nine countries are ongoing [77]. The efficacy of the Moderna vaccine for COVID-19 has been found to be 94.1% effective against COVID-19 after taking two doses in clinical trials [83]. The most prevalent side effects of the Moderna vaccination were fatigue, headache, muscular discomfort, joint pain, chills, nausea, and vomiting [84].

2.11. Inactivated COVID-19 (Vero Cell) vaccine (COVID-19 vaccine BIBP)

The COVID-19 (Vero Cell) vaccine, inactivated with β-propiolactone, is generated in Vero Cells [85]. The WHO granted Emergency Use Authorization on May 7, 2021 [76]. It has been approved in 89 countries, and 22 clinical trials have been registered in 11 countries [77]. The COVID-19 Vaccine BIBP has an effectiveness rate of 79% after the second dose [86].

2.12. COVID-19 (Vero Cell) vaccine (CoronaVac)

CoronaVac is a COVID-19 vaccine based on an inactivated virus [87]. The WHO granted Emergency Use Authorization to this vaccine on June 1, 2021 [76]. It has been approved in 48 countries, and 27 clinical trials have been registered in eight countries [77]. The efficacy of the Sinovac-Coronavac vaccine is 63.09% [88].

2.13. Covaxin (BBV152) vaccine

The Covaxin (BBV152) vaccine is based on an inactivated virus [89]. The Central Drugs and Standards Committee (CDSCO) in India authorized it for Emergency Use Authorization on January 3, 2021 [90], and it was also approved for EUA by the WHO on December 3, 2021 [76]. It has been approved in 12 countries, and seven clinical trials have been registered in seven countries [77]. The Covaxin COVID-19 vaccine has an effectiveness rate of 78% after the second dose [91].

2.14. COVOVAX vaccine

COVOVAX is a COVID-19 vaccine that is adjuvanted with Matrix-M1 and based on the S protein of SARS-CoV-2 [92]. Matrix-M1 contains Fraction A and Fraction C of Quillaja Saponaria Molina extract [92]. WHO approved it for Emergency Use Authorization on December 17, 2021 [76]. It is approved in 3 countries and registered in 2 clinical trials in one country [77]. The efficacy of the COVOVAX COVID-19 vaccine has been found to be 90.4% effective in protecting against COVID-19 after taking two doses [93].

2.15. Nuvaxovid vaccine

Nuvaxovid is a COVID-19 vaccine that uses Matrix-M as an adjuvant and is based on the SARS-CoV-2 spike protein [94]. It was granted Emergency Use Authorization (EUA) by the WHO on December 20th, 2021 [76]. Nuvaxovid is approved in 34 countries and registered in 11 clinical trials in 11 countries [77]. The vaccine has been found to have an efficacy of 90.4% against COVID-19 after taking two doses [93].

2.16. Other vaccines

  • 1.
    Sputnik V Vaccine

The Sputnik V vaccine is a COVID-19 adenovirus viral vector vaccination [95], authorized for Emergency Use Authorization (EUA) in 74 countries. Currently, 22 clinical trials are registered in seven countries [77]. The vaccine has shown efficacy in providing 91.6% protection against COVID-19 after administering two doses [96].

  • 2.
    Sputnik Light Vaccine

The Sputnik Light vaccine is a single-dose COVID-19 vaccine based on the Ad26 vector [95]. It has been authorized for Emergency Use Authorization (EUA) in 22 countries, and four clinical trials are registered in two countries [77]. The efficacy of the Sputnik V vaccine is reported to be 79.4% [97].

2.16.1. Future prospects of COVID-19 development vaccine program

The current study critically analyzes the vaccine development programs for HIV, Zika, Ebola, influenza, MERS, and SARS infections and presents all the challenges faced by these programs. Su et al. have identified vaccine-associated disease enhancement (VADE) as one of the pivotal challenges in SARS, MERS, and RSV vaccine development programs. The study raises concerns for vaccine safety and efficacy. In preclinical testing, appropriate validation criteria for animal models and COVID-19 viral strains should be used. Other safety and efficacy testing recommendations include using specific neutralization titers, assessing eosinophil infiltration in the lungs, and measuring the response time of neutralizing antibodies in preclinical studies. The study recommends an optimized receptor-binding domain (RBD) as the most appropriate antigen for a COVID-19 vaccine [98].

Another study has identified challenges concerning the COVID-19 vaccine, such as long-term safety, efficacy, duration of immunity, resistance, and viral mutation [99]. Addressing the challenges faced by other viral vaccine programs can be helpful for the COVID-19 vaccine program in the future. The development of life-saving vaccine shots for the 8 billion people on the planet in just two years was made possible by the development and upscaling of emergency vaccine discovery. Vaccine formulations against COVID-19 were created in record time during this pandemic by referring to earlier viral vaccines and skipping some initial preclinical (in vitro and in vivo) steps of vaccine design and development. The NVX-CoV2373 or mRNA-based vaccines produced circulating follicular helper T cells (cTfhs) specific for the SARS-CoV-2 spike, memory CD4+ T cells, and CD4-CTLs. Ad26.COV2. S and mRNA vaccinations both produced equivalent CD8+ T-cell responses. After receiving mRNA injections, antibodies drastically diminished, but memory T- and B-cells were largely unaltered. Booster shots, especially those against novel COVID-19 variants, have been successful in preventing infection and sickness [100]. More studies are required to determine the longevity of immunity following booster doses, as well as the impact of booster doses on the efficacy of COVID-19 vaccines in reducing illness incidence, mortality, and hospitalization rates. It is strongly advised to conduct additional studies on the effectiveness of booster doses using other vaccines and novel variations [101]. For high-risk individuals, immunization may be required to prevent the severe form of the disease. Individuals with allergies cannot be prohibited from vaccination. Vaccination should be recommended because it helps prevent a potentially fatal condition, and adverse events following immunization (AEFI) with substantial case fatalities are quite rare. If a patient experiences an anaphylactic incident, they must be transported to another facility to receive necessary medical care, and physicians must be alert to spot, treat, and record any such incidents [102].It's also conceivable that SARS-CoV-2 will become endemic and persist in the population for some time to come. Due to declining immunity and emerging infections from new variants, variant-specific or pan-coronavirus vaccinations, as well as future boosters, may be necessary. Establishing immunological correlates of protection, utilizing nAb titers and additional biomarkers like cellular immunity or memory B-cell levels, is crucial for the creation of next-generation vaccines [103].

3. Conclusion

There are ten WHO-approved vaccines for COVID-19 currently in emergency use, developed within a relatively short period of time. Both DNA and RNA-based vaccines have received FDA approval. To identify potential barriers, previous viral vaccine programs such as HIV, Zika, Influenza, Ebola, Dengue, SARS, and MERS were comprehensively evaluated. While a large portion of the population has been vaccinated, there remain numerous challenges ahead for the COVID-19 vaccine, including long-term efficacy, safety, tolerability in special populations, duration of the immune response, and adverse events related to vaccination. Additionally, the mutation of SARS-CoV-2, the longevity of the immune response, and adverse events related to vaccines all pose significant challenges. Long-term follow-up studies, alternative therapeutic approaches, or alternative vaccines may be necessary in the future.

Author's contribution

RSS- Conception or design of the work, Data collection, Data analysis and interpretation.

AS- Data collection, Data analysis and interpretation.

GDM, GB, ARS, RJ- Data collection, Data analysis and interpretation.

AP, BS, PS, MP, HK- Critical revision of the article, Data analysis.

AB, SU- Data collection, Data analysis.

BM- Conception or design of the work, Drafting the article, Critical revision of the article, Final approval of the version to be published.

Data availability statement

The authors are unable or have chosen not to specify which data has been used.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

1. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., Zhang L., Fan G., Xu J., Gu X., Cheng Z., Yu T., Xia J., Wei Y., Wu W., Xie X., Yin W., Li H., Liu M., Xiao Y., Gao H., Guo L., Xie J., Wang G., Jiang R., Gao Z., Jin Q., Wang J., Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
2. Director-General W. vols. 6–9. 2020. https://www.who.int/director-general/speeches/detail/who-director-general-s-remarks-at-the-media-briefing-on-2019-ncov-on-11-february-2020 (WHO Director-General’s Remarks at the Media Briefing on 2019-nCoV on 11 February 2020, World Heal. Orgnatization). [Google Scholar]
3. Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z., Xiang J., Wang Y., Song B., Gu X., Guan L., Wei Y., Li H., Wu X., Xu J., Tu S., Zhang Y., Chen H., Cao B. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet (London, England) 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
4. Mahalmani V., Mahendru D., Semwal A., Kaur S., Kaur H., Sarma P., Prakash A., Medhi B. COVID-19 pandemic: a review based on current evidence. Indian J. Pharmacol. 2020;52:117. doi: 10.4103/ijp.IJP_310_20. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
5. Reed S.G., Orr M.T., Fox C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013;19:1597–1608. doi: 10.1038/nm.3409. [PubMed] [CrossRef] [Google Scholar]
6. Wang N., Shang J., Jiang S., Du L. Subunit vaccines against emerging pathogenic human coronaviruses. Front. Microbiol. 2020;11:298. doi: 10.3389/fmicb.2020.00298. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
7. Martin J.E., Louder M.K., Holman L.S.A., Gordon I.J., Enama M.E., Larkin B.D., Andrews C.A., Vogel L., Koup R.A., Roederer M., Bailer R.T., Gomez P.L., Nason M., Mascola J.R., Nabel G.J., Graham B.S. A SARS DNA vaccine induces neutralizing antibody and cellular immune responses in healthy adults in a Phase I clinical trial. Vaccine. 2008;26:6338–6343. doi: 10.1016/j.vaccine.2008.09.026. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
8. Rauch S., Jasny E., Schmidt K.E., Petsch B. New vaccine technologies to combat outbreak situations. Front. Immunol. 2018;9:1963. doi: 10.3389/fimmu.2018.01963. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
9. Singh A., Singh R.S., Sarma P., Batra G., Joshi R., Kaur H., Sharma A.R., Prakash A., Medhi B. A comprehensive review of animal models for coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV. Virol. Sin. 2020;35:290–304. doi: 10.1007/s12250-020-00252-z. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
10. Zhou Y., Jiang S., Du L. Prospects for a MERS-CoV spike vaccine. Expert Rev. Vaccines. 2018;17:677–686. doi: 10.1080/14760584.2018.1506702. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
11. Han Y.J., Lee K.H., Yoon S., Nam S.W., Ryu S., Seong D., Kim J.S. Treatment of severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and coronavirus disease 2019 (COVID-19): a systematic review of in vitro, in vivo, and clinical trials. Theranostics. 2021;11:1207–1231. doi: 10.7150/thno.48342. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
12. Tai W., Zhang X., He Y., Jiang S., Du L. Identification of SARS-CoV RBD-targeting monoclonal antibodies with cross-reactive or neutralizing activity against SARS-CoV-2. Antivir. Res. 2020;179 doi: 10.1016/j.antiviral.2020.104820. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
13. Padron-Regalado E. Vaccines for SARS-CoV-2: lessons from other coronavirus strains. Infect. Dis. Ther. 2020;9:255–274. doi: 10.1007/s40121-020-00300-x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
14. Smith S.A., Kilgore K.M., Kasturi S.P., Pulendran B., Hunter E., Amara R.R., Derdeyn C.A. Signatures in simian immunodeficiency virus SIVsmE660 envelope gp120 are associated with mucosal transmission but not vaccination breakthrough in rhesus macaques. J. Virol. 2016;90:1880–1887. doi: 10.1128/jvi.02711-15. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
15. Domingo E., Schuster P. What is a quasispecies? Historical origins and current scope. Curr. Top. Microbiol. Immunol. 2016;392:1–22. doi: 10.1007/82_2015_453. [PubMed] [CrossRef] [Google Scholar]
16. Holland J., Spindler K., Horodyski F., Grabau E., Nichol S., VandePol S. Rapid evolution of RNA genomes. Science. 1982;216(80):1577–1585. doi: 10.1126/science.7041255. [PubMed] [CrossRef] [Google Scholar]
17. Couch R.B., Cate T.R., Douglas R.G., Gerone P.J., Knight V. Effect of route of inoculation on experimental respiratory viral disease in volunteers and evidence for airborne transmission. Bacteriol. Rev. 1966;30:517–529. doi: 10.1128/br.30.3.517-529.1966. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
18. Gonzalez M.W., DeVico A.L., Lewis G.K., Spouge J.L. Conserved molecular signatures in gp120 are associated with the genetic bottleneck during simian immunodeficiency virus (SIV), SIV-human immunodeficiency virus (SHIV), and HIV type 1 (HIV-1) transmission. J. Virol. 2015;89:3619–3629. doi: 10.1128/jvi.03235-14. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
19. Andrabi R., Voss J.E., Liang C.H., Briney B., McCoy L.E., Wu C.Y., Wong C.H., Poignard P., Burton D.R. Identification of common features in prototype broadly neutralizing antibodies to HIV envelope V2 apex to facilitate vaccine design. Immunity. 2015;43:959–973. doi: 10.1016/j.immuni.2015.10.014. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
20. Van Regenmortel M.H.V. Structure-based reverse vaccinology failed in the case of HIV because it disregarded accepted immunological theory. Int. J. Mol. Sci. 2016;17 doi: 10.3390/ijms17091591. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
21. Burton D.R., Hangartner L. Broadly neutralizing antibodies to HIV and their role in vaccine design. Annu. Rev. Immunol. 2016;34:635–659. doi: 10.1146/annurev-immunol-041015-055515. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
22. Bonsignori M., Liao H.X., Gao F., Williams W.B., Alam S.M., Montefiori D.C., Haynes B.F. Antibody-virus co-evolution in HIV infection: paths for HIV vaccine development. Immunol. Rev. 2017;275:145–160. doi: 10.1111/imr.12509. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
23. Haynes B.F., Burton D.R. Developing an HIV vaccine. Science. 2017;355(80):1129–1130. doi: 10.1126/science.aan0662. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
24. Barouch D.H. Challenges in the development of an HIV-1 vaccine. Nature. 2008;455:613–619. doi: 10.1038/nature07352. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
25. dos Santos T., Rodriguez A., Almiron M., Sanhueza A., Ramon P., de Oliveira W.K., Coelho G.E., Badaró R., Cortez J., Ospina M., Pimentel R., Masis R., Hernandez F., Lara B., Montoya R., Jubithana B., Melchor A., Alvarez A., Aldighieri S., Dye C., Espinal M.A. Zika virus and the guillain–barré syndrome — case series from seven countries. N. Engl. J. Med. 2016;375:1598–1601. doi: 10.1056/nejmc1609015. [PubMed] [CrossRef] [Google Scholar]
26. Bardina S.V., Bunduc P., Tripathi S., Duehr J., Frere J.J., Brown J.A., Nachbagauer R., Foster G.A., Krysztof D., Tortorella D., Stramer S.L., García-Sastre A., Krammer F., Lim J.K. Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity. Science. 2017;356(80):175–180. doi: 10.1126/science.aal4365. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
27. Kirkpatrick B.D., Whitehead S.S., Pierce K.K., Tibery C.M., Grier P.L., Hynes N.A., Larsson C.J., Sabundayo B.P., Talaat K.R., Janiak A., Carmolli M.P., Luke C.J., Diehl S.A., Durbin A.P. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model. Sci. Transl. Med. 2016;8 doi: 10.1126/scitranslmed.aaf1517. 330ra36. [PubMed] [CrossRef] [Google Scholar]
28. Mead P.S., Duggal N.K., Hook S.A., Delorey M., Fischer M., Olzenak McGuire D., Becksted H., Max R.J., Anishchenko M., Schwartz A.M., Tzeng W.-P., Nelson C.A., McDonald E.M., Brooks J.T., Brault A.C., Hinckley A.F. Zika virus shedding in semen of symptomatic infected men. N. Engl. J. Med. 2018;378:1377–1385. doi: 10.1056/nejmoa1711038. [PubMed] [CrossRef] [Google Scholar]
29. Smith D.J., Lapedes A.S., De Jong J.C., Bestebroer T.M., Rimmelzwaan G.F., Osterhaus A.D.M.E., Fouchier R.A.M. Mapping the antigenic and genetic evolution of influenza virus. Science. 2004;305(80):371–376. doi: 10.1126/science.1097211. [PubMed] [CrossRef] [Google Scholar]
30. Lee Y.-T., Kim K.-H., Ko E.-J., Lee Y.-N., Kim M.-C., Kwon Y.-M., Tang Y., Cho M.-K., Lee Y.-J., Kang S.-M. New vaccines against influenza virus. Clin. Exp. Vaccine Res. 2014;3:12. doi: 10.7774/cevr.2014.3.1.12. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
31. Houser K., Subbarao K. Influenza vaccines: challenges and solutions. Cell Host Microbe. 2015;17:295–300. doi: 10.1016/j.chom.2015.02.012. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
32. Taubenberger J.K., Kash J.C. Influenza virus evolution, host adaptation, and pandemic formation. Cell Host Microbe. 2010;7:440–451. doi: 10.1016/j.chom.2010.05.009. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
33. Haq K., McElhaney J.E. Immunosenescence: influenza vaccination and the elderly. Curr. Opin. Immunol. 2014;29:38–42. doi: 10.1016/j.coi.2014.03.008. [PubMed] [CrossRef] [Google Scholar]
34. Khurana S., Loving C.L., Manischewitz J., King L.R., Gauger P.C., Henningson J., Vincent A.L., Golding H. Vaccine-induced anti-HA2 antibodies promote virus fusion and enhance influenza virus respiratory disease. Sci. Transl. Med. 2013;5:200ra114. doi: 10.1126/scitranslmed.3006366. [PubMed] [CrossRef] [Google Scholar]
35. Miller E., Andrews N., Stellitano L., Stowe J., Winstone A.M., Shneerson J., Verity C. Risk of narcolepsy in children and young people receiving AS03 adjuvanted pandemic A/H1N1 2009 influenza vaccine: retrospective analysis. BMJ. 2013;346:f794. doi: 10.1136/bmj.f794. [PubMed] [CrossRef] [Google Scholar]
36. Nair H., Brooks W.A., Katz M., Roca A., Berkley J.A., Madhi S.A., Simmerman J.M., Gordon A., Sato M., Howie S., Krishnan A., Ope M., Lindblade K.A., Carosone-Link P., Lucero M., Ochieng W., Kamimoto L., Dueger E., Bhat N., Vong S., Theodoratou E., Chittaganpitch M., Chimah O., Balmaseda A., Buchy P., Harris E., Evans V., Katayose M., Gaur B., O'Callaghan-Gordo C., Goswami D., Arvelo W., Venter M., Briese T., Tokarz R., Widdowson M.A., Mounts A.W., Breiman R.F., Feikin D.R., Klugman K.P., Olsen S.J., Gessner B.D., Wright P.F., Rudan I., Broor S., Simões E.A., Campbell H. Global burden of respiratory infections due to seasonal influenza in young children: a systematic review and meta-analysis. Lancet. 2011;378:1917–1930. doi: 10.1016/S0140-6736(11)61051-9. [PubMed] [CrossRef] [Google Scholar]
37. King C., Leask J. The impact of a vaccine scare on parental views, trust and information needs: a qualitative study in Sydney, Australia. BMC Publ. Health. 2017;17:1–10. doi: 10.1186/s12889-017-4032-2. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
38. Thomas S.J., Yoon I.K. A review of Dengvaxia®: development to deployment. Hum. Vaccines Immunother. 2019;15:2295–2314. doi: 10.1080/21645515.2019.1658503. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
39. Huang K.-J., Yang Y.-C., Lin Y.-S., Huang J.-H., Liu H.-S., Yeh T.-M., Chen S.-H., Liu C.-C., Lei H.-Y. The dual-specific binding of dengue virus and target cells for the antibody-dependent enhancement of dengue virus infection. J. Immunol. 2006;176:2825–2832. doi: 10.4049/jimmunol.176.5.2825. [PubMed] [CrossRef] [Google Scholar]
40. Tirado S.M.C., Yoon K.J. Antibody-dependent enhancement of virus infection and disease. Viral Immunol. 2003;16:69–86. doi: 10.1089/088282403763635465. [PubMed] [CrossRef] [Google Scholar]
41. Hawkes R.A. Enhancement of the infectivity of arboviruses by specific antisera produced in domestic fowls. Aust. J. Exp. Biol. Med. Sci. 1964;42:465–482. doi: 10.1038/icb.1964.44. [PubMed] [CrossRef] [Google Scholar]
43. Lund O., Hansen J., Søorensen A.M., Mosekilde E., Nielsen J.O., Hansen J.E. Increased adhesion as a mechanism of antibody-dependent and antibody-independent complement-mediated enhancement of human immunodeficiency virus infection. J. Virol. 1995;69:2393–2400. doi: 10.1128/jvi.69.4.2393-2400.1995. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
44. Wan Y., Shang J., Sun S., Tai W., Chen J., Geng Q., He L., Chen Y., Wu J., Shi Z., Zhou Y., Du L., Li F. Molecular mechanism for antibody-dependent enhancement of coronavirus entry. J. Virol. 2020;94 doi: 10.1128/jvi.02015-19. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
45. Huang K.-J., Yang Y.-C., Lin Y.-S., Huang J.-H., Liu H.-S., Yeh T.-M., Chen S.-H., Liu C.-C., Lei H.-Y. The dual-specific binding of dengue virus and target cells for the antibody-dependent enhancement of dengue virus infection. J. Immunol. 2006;176:2825–2832. doi: 10.4049/jimmunol.176.5.2825. [PubMed] [CrossRef] [Google Scholar]
46. Montecillo-Aguado M.R., Montes-Gómez A.E., García-Cordero J., Corzo-Gómez J., Vivanco-Cid H., Mellado-Sánchez G., Muñoz-Medina J.E., Gutiérrez-Castañeda B., Santos-Argumedo L., González-Bonilla C., Cedillo-Barrón L. Cross-reaction, enhancement, and neutralization activity of dengue virus antibodies against zika virus: a study in the mexican population. J. Immunol. Res. 2019;2019 doi: 10.1155/2019/7239347. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
47. Nogueira M.L., Nery Júnior N.R.R., Estofolete C.F., Bernardes Terzian A.C., Guimarães G.F., Zini N., Alves da Silva R., Dutra Silva G.C., Junqueira Franco L.C., Rahal P., Bittar C., Carneiro B., Vasconcelos P.F.C., Freitas Henriques D., Barbosa D.M.U., Lopes Rombola P., de Grande L., Negri Reis A.F., Palomares S.A., Wakai Catelan M., Cruz L.E.A.A., Necchi S.H., Mendonça R.C.V., Penha dos Santos I.N., Alavarse Caron S.B., Costa F., Bozza F.A., Soares de Souza A., Brandão de Mattos C.C., de Mattos L.C., Vasilakis N., Oliani A.H., Vaz Oliani D.C.M., Ko A.I. Adverse birth outcomes associated with Zika virus exposure during pregnancy in São José do Rio Preto, Brazil. Clin. Microbiol. Infect. 2018;24:646–652. doi: 10.1016/j.cmi.2017.11.004. [PubMed] [CrossRef] [Google Scholar]
48. Cleary S.J., Pitchford S.C., Amison R.T., Carrington R., Robaina Cabrera C.L., Magnen M., Looney M.R., Gray E., Page C.P. Animal models of mechanisms of SARS-CoV-2 infection and COVID-19 pathology. Br. J. Pharmacol. 2020;177:4851–4865. doi: 10.1111/bph.15143. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
49. Chan K.W.K., Watanabe S., Kavishna R., Alonso S., Vasudevan S.G. Animal models for studying dengue pathogenesis and therapy. Antivir. Res. 2015;123:5–14. doi: 10.1016/j.antiviral.2015.08.013. [PubMed] [CrossRef] [Google Scholar]
50. Jiang L., Lu C., Sun Q. Tree shrew as a new animal model for the study of dengue virus. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.621164. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
51. Food and Drug Administration (Fda) 2020. Ervebo | Fda; p. 1.https://www.fda.gov/vaccines-blood-biologics/ervebo [Google Scholar]
52. Thorson A., Formenty P., Lofthouse C., Broutet N. Systematic review of the literature on viral persistence and sexual transmission from recovered Ebola survivors: evidence and recommendations. BMJ Open. 2016;6 doi: 10.1136/bmjopen-2015-008859. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
53. Arnemo M., Viksmoen Watle S.S., Schoultz K.M., Vainio K., Norheim G., Moorthy V., Fast P., Røttingen J.A., Gjøen T. Stability of a vesicular stomatitis virus-vectored Ebola vaccine. J. Infect. Dis. 2016;213:930–933. doi: 10.1093/infdis/jiv532. [PubMed] [CrossRef] [Google Scholar]
54. Gregori J., Cortese M.F., Piñana M., Campos C., Garcia-Cehic D., Andrés C., Abril J.F., Codina M.G., Rando A., Esperalba J., Sulleiro E., Joseph J., Saubí N., Colomer-Castell S., Martin M.C., Castillo C., Esteban J.I., Pumarola T., Rodriguez-Frias F., Antón A., Quer J. Host-dependent editing of SARS-CoV-2 in COVID-19 patients. Emerg. Microb. Infect. 2021;10:1777–1789. doi: 10.1080/22221751.2021.1969868. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
55. Karamitros T., Papadopoulou G., Bousali M., Mexias A., Tsiodras S., Mentis A. SARS-CoV-2 exhibits intra-host genomic plasticity and low-frequency polymorphic quasispecies. J. Clin. Virol. 2020;131 doi: 10.1016/j.jcv.2020.104585. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
56. Chen Y., Xu Z., Wang P., Li X., Shuai Z., Ye D., Pan H. New‐onset autoimmune phenomena post‐COVID‐19 vaccination. Immunology. 2022;165:386–401. doi: 10.1111/imm.13443. [PubMed] [CrossRef] [Google Scholar]
57. Jara L.J., Vera-Lastra O., Mahroum N., Pineda C., Shoenfeld Y. Autoimmune post-COVID vaccine syndromes: does the spectrum of autoimmune/inflammatory syndrome expand? Clin. Rheumatol. 2022;41:1603–1609. doi: 10.1007/s10067-022-06149-4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
58. Ciabattini A., Nardini C., Santoro F., Garagnani P., Franceschi C., Medaglini D. Vaccination in the elderly: the challenge of immune changes with aging. Semin. Immunol. 2018;40:83–94. doi: 10.1016/j.smim.2018.10.010. [PubMed] [CrossRef] [Google Scholar]
59. Gresham L.M., Marzario B., Dutz J., Kirchhof M.G. An evidence-based guide to SARS-CoV-2 vaccination of patients on immunotherapies in dermatology. J. Am. Acad. Dermatol. 2021;84:1652–1666. doi: 10.1016/j.jaad.2021.01.047. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
60. Boyarsky B.J., Werbel W.A., Avery R.K., Tobian A.A.R., Massie A.B., Segev D.L., Garonzik-Wang J.M. Immunogenicity of a single dose of SARS-CoV-2 messenger RNA vaccine in solid organ transplant recipients. JAMA. 2021;325:1784. doi: 10.1001/jama.2021.4385. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
61. Organization W.H. World Heal. Organ.; Geneva, Switzerland: 2021. COVAX: Working for Global Equitable Access to COVID-19 Vaccines; p. 1.https://www.who.int/initiatives/act-accelerator/covax [Google Scholar]
62. Dal-Ré R., Bekker L.-G., Gluud C., Holm S., Jha V., Poland G.A., Rosendaal F.R., Schwarzer-Daum B., Sevene E., Tinto H., Voo T.C., Sreeharan N. Ongoing and future COVID-19 vaccine clinical trials: challenges and opportunities. Lancet Infect. Dis. 2021;21 doi: 10.1016/S1473-3099(21)00263-2. e342–e347. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
63. Duffy S. Why are RNA virus mutation rates so damn high? PLoS Biol. 2018;16 doi: 10.1371/journal.pbio.3000003. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
64. Tsetsarkin K.A., Vanlandingham D.L., McGee C.E., Higgs S. A single mutation in Chikungunya virus affects vector specificity and epidemic potential. PLoS Pathog. 2007;3:1895–1906. doi: 10.1371/journal.ppat.0030201. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
65. Herfst S., Schrauwen E.J.A., Linster M., Chutinimitkul S., De Wit E., Munster V.J., Sorrell E.M., Bestebroer T.M., Burke D.F., Smith D.J., Rimmelzwaan G.F., Osterhaus A.D.M.E., Fouchier R.A.M. Airborne transmission of influenza A/H5N1 virus between ferrets. Science. 2012;336(80):1534–1541. doi: 10.1126/science.1213362. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
66. Petrie J.G., Lauring A.S. Influenza A(H7N9) virus evolution: which genetic mutations are antigenically important? J. Infect. Dis. 2019;219:3–5. doi: 10.1093/infdis/jiy409. [PubMed] [CrossRef] [Google Scholar]
67. Li Q., Wu J., Nie J., Zhang L., Hao H., Liu S., Zhao C., Zhang Q., Liu H., Nie L., Qin H., Wang M., Lu Q., Li X., Sun Q., Liu J., Zhang L., Li X., Huang W., Wang Y. The impact of mutations in SARS-CoV-2 spike on viral infectivity and antigenicity. Cell. 2020;182:1284–1294.e9. doi: 10.1016/j.cell.2020.07.012. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
68. Ahmadpour D., Ahmadpoor P., Rostaing L. Impact of circulating SARS-CoV-2 mutant G614 on the COVID-19 pandemic, Iran. J. Kidney Dis. 2020;14:331–334. [PubMed] [Google Scholar]
69. Voysey M., Clemens S.A.C., Madhi S.A., Weckx L.Y., Folegatti P.M. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2021;397:99–111. doi: 10.1016/S0140-6736(20)32661-1. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
70. Plante J.A., Mitchell B.M., Plante K.S., Debbink K., Weaver S.C., Menachery V.D. The variant gambit: COVID-19's next move. Cell Host Microbe. 2021;29:508–515. doi: 10.1016/j.chom.2021.02.020. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
71. World Health Organization . vol. 1. 2021. https://www.who.int/news/item/26-11-2021-classification-of-omicron-(b.1.1.529)-sars-cov-2-variant-of-concern,Classif.Omi.SARS-CoV-2Var.Concern (Classification of Omicron (B.1.1.529): SARS-CoV-2 Variant of Concern). [Google Scholar]
72. Wang Y., Zhang L., Li Q., Liang Z., Li T., Liu S., Cui Q., Nie J., Wu Q., Qu X., Huang W. The significant immune escape of pseudotyped SARS-CoV-2 variant Omicron, Emerg. Microb. Infect. 2022;11:1–5. doi: 10.1080/22221751.2021.2017757. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
73. Zolfaghari Emameh R., Eftekhari M., Nosrati H., Heshmatnia J., Falak R. Identification and characterization of a silent mutation in RNA binding domain of N protein coding gene from SARS-CoV-2. BMC Res. Notes. 2021;14:10. doi: 10.1186/s13104-020-05439-x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
75. 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., Bailey R., Swanson K.A., Roychoudhury S., Koury K., Li P., Kalina W.V., Cooper D., Frenck R.W., Hammitt L.L., Türeci Ö., Nell H., Schaefer A., Ünal S., Tresnan D.B., Mather S., Dormitzer P.R., Şahin U., Jansen K.U., Gruber W.C. Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615. doi: 10.1056/nejmoa2034577. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
76. World Health Organization . 2022. COVID-19 Vaccines WHO EUL Issued | WHO - Prequalification of Medical Products (IVDs, Medicines, Vaccines and Immunization Devices, Vector Control)https://extranet.who.int/pqweb/vaccines/vaccinescovid-19-vaccine-eul-issued [Google Scholar]
77. World Health Organization . World Heal. Organ.; 2022. WHO – COVID19 Vaccine Tracker.https://covid19.trackvaccines.org/agency/who/ (accessed June 4, 2021) [Google Scholar]
78. Shimabukuro T., Nair N. Allergic reactions including anaphylaxis after receipt of the first dose of pfizer-BioNTech COVID-19 vaccine. JAMA, J. Am. Med. Assoc. 2021;325:780–781. doi: 10.1001/jama.2021.0600. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
79. Parums D.V. Editorial: SARS-CoV-2 mRNA vaccines and the possible mechanism of vaccine-induced immune thrombotic thrombocytopenia (VITT) Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 2021;27 doi: 10.12659/MSM.932899. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
80. World Health Organization . WHO; 2021. WHO recommendation Moderna COVID-19 mRNA Vaccine (nucleoside modified)https://extranet.who.int/pqweb/vaccines/covid-19-mrna-vaccine-nucleoside-modified (WHO - Prequalification of Medical Products (IVDs, Medicines, Vaccines and Immunization Devices, Vector Control)). [Google Scholar]
81. CDC . COV2.S; 2021. Overview of Janssen ’ S Single-Dose COVID-19 Vaccine, Ad26; pp. 1–54. [Google Scholar]
82. Oliver S.E., Gargano J.W., Marin M., Wallace M., Curran K.G., Chamberland M., McClung N., Campos-Outcalt D., Morgan R.L., Mbaeyi S., Romero J.R., Talbot H.K., Lee G.M., Bell B.P., Dooling K. The advisory committee on immunization practices' interim recommendation for use of Moderna COVID-19 vaccine — United States, december 2020. MMWR Morb. Mortal. Wkly. Rep. 2021;69:1653–1656. doi: 10.15585/mmwr.mm695152e1. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
83. Mahase E. Covid-19: Moderna applies for US and EU approval as vaccine trial reports 94.1% efficacy. BMJ. 2020;371 doi: 10.1136/bmj.m4709. [PubMed] [CrossRef] [Google Scholar]
84. Ndwandwe D., Wiysonge C.S. COVID-19 vaccines; Bethesda (MD): 2021. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
85. World Health Organization . WHO; 2021. Capacity Building | WHO - Prequalification of Medical Products (IVDs, Medicines, Vaccines and Immunization Devices, Vector Control)https://extranet.who.int/pqweb/medicines/capacity-building [Google Scholar]
86. WHO . World Heal. Organ.; 2021. The Sinopharm COVID-19 Vaccine: what You Need to Know; pp. 1–5.https://www.who.int/news-room/feature-stories/detail/the-sinopharm-covid-19-vaccine-what-you-need-to-know (accessed June 5, 2021) [Google Scholar]
87. Who . World Heal. Organ.; 2021. pp. 19–22.https://www.who.int/news-room/feature-stories/detail/the-sinovac-covid-19-vaccine-what-you-need-to-know (The Sinovac-CoronaVac COVID-19 Vaccine : what You Need to Know). [Google Scholar]
89. Ella R., Vadrevu K.M., Jogdand H., Prasad S., Reddy S., Sarangi V., Ganneru B. Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBV152: a double-blind, randomised, phase 1 trial. Lancet Infect. Dis. 2021;21:637–646. doi: 10.1016/S1473-3099(20)30942-7. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
90. Welfare F., Ella K., Biotech B., Moh S., Shri F.W., Bhushan R., Ayog N., Paul V., Raghavan V., Swarup R., Biotech B., Icmr G., Bhargava B., Subject T., Committee E., Process A.A., Subject T., Committee E., General D.C., Iii T.P., Iii P., Ii P., Biotech B., Council I. Bharat biotech ’ s ‘ COVAXIN TM ’ emergency use authorization approval by DCGI-CDSCO , MoH & FW , a significant landmark in India ’ s scie ntific discovery , and scientists capability. Bharat Biotech. 2021;3 https://www.bharatbiotech.com/images/press/bharat-biotech-covaxin-emergency-use-authorization-approval-by-dcgi-cdsco-moh-and-fw.pdf [Google Scholar]
91. Sapkal G.N., Yadav P.D., Ella R., Deshpande G.R., Sahay R.R., Gupta N., Vadrevu K.M., Abraham P., Panda S., Bhargava B. Inactivated COVID-19 vaccine BBV152/COVAXIN effectively neutralizes recently emerged B.1.1.7 variant of SARS-CoV-2. J. Trav. Med. 2021;28 doi: 10.1093/jtm/taab051. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
92. World Health Organization . WHO; 2022. WHO Recommendation Serum Institute of India Pvt. Ltd. (SIIPL) – COVID-19 Vaccine (SARS-CoV-2 rS Protein Nanoparticle [Recombinant]) - COVOVAXTM | WHO - Prequalification of Medical Products (IVDs, Medicines, Vaccines and Immunization Devices, Vector Control.https://extranet.who.int/pqweb/vaccines/who-recommendation-siipl-covid-19-vaccine-rs-proteinnanoparticle [Google Scholar]
93. Heath P.T., Galiza E.P., Baxter D.N., Boffito M., Browne D., Burns F. 2019 nCoV-302 study group, safety and efficacy of NVX-CoV2373 covid-19 vaccine. N. Engl. J. Med. 2021;385:1172–1183. doi: 10.1056/NEJMoa2107659. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
94. NOVAVAX . 2021. https://www.novavax.com/our-pipeline#nvx-cov2373 (Pipeline Is Robust at Novavax). NVX-CoV2373. [Google Scholar]
95. Barouch D.H., Kik S.V., Weverling G.J., Dilan R., King S.L., Maxfield L.F., Clark S., Ng’ang’a D., Brandariz K.L., Abbink P., Sinangil F., de Bruyn G., Gray G.E., Roux S., Bekker L.G., Dilraj A., Kibuuka H., Robb M.L., Michael N.L., Anzala O., Amornkul P.N., Gilmour J., Hural J., Buchbinder S.P., Seaman M.S., Dolin R., Baden L.R., Carville A., Mansfield K.G., Pau M.G., Goudsmit J. International seroepidemiology of adenovirus serotypes 5, 26, 35, and 48 in pediatric and adult populations. Vaccine. 2011;29:5203–5209. doi: 10.1016/j.vaccine.2011.05.025. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
96. Lawton G. Sputnik V vaccine goes global. New Sci. 2021;250:10–11. doi: 10.1016/s0262-4079(21)00671-0. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
97. Nogrady B. Mounting evidence suggests Sputnik COVID vaccine is safe and effective. Nature. 2021;595:339–340. doi: 10.1038/d41586-021-01813-2. [PubMed] [CrossRef] [Google Scholar]
98. Su S., Du L., Jiang S. Learning from the past: development of safe and effective COVID-19 vaccines. Nat. Rev. Microbiol. 2021;19:211–219. doi: 10.1038/s41579-020-00462-y. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
99. Mohamed K., Rzymski P., Islam M.S., Makuku R., Mushtaq A., Khan A., Ivanovska M., Makka S.A., Hashem F., Marquez L., Cseprekal O., Filgueiras I.S., Fonseca D.L.M., Mickael E., Ling I., Arero A.G., Cuschieri S., Minakova K., Rodríguez‐Román E., Abarikwu S.O., Faten A., Grancini G., Cabral‐Marques O., Rezaei N. COVID‐19 vaccinations: the unknowns, challenges, and hopes. J. Med. Virol. 2022;94:1336–1349. doi: 10.1002/jmv.27487. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
100. Chavda V.P., Yao Q., Vora L.K., Apostolopoulos V., Patel C.A., Bezbaruah R., Patel A.B., Chen Z.-S. Fast-track development of vaccines for SARS-CoV-2: the shots that saved the world. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.961198. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
101. Rahmani K., Shavaleh R., Forouhi M., Disfani H.F., Kamandi M., Oskooi R.K., Foogerdi M., Soltani M., Rahchamani M., Mohaddespour M., Dianatinasab M. The effectiveness of COVID-19 vaccines in reducing the incidence, hospitalization, and mortality from COVID-19: a systematic review and meta-analysis. Front. Public Health. 2022;10 doi: 10.3389/fpubh.2022.873596. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
102. Shekhar N., Sarma P., Prajapat M., Avti P., Kaur H., Raja A., Singh H., Bhattacharya A., Sharma S., Kumar S., Prakash A., Medhi B. Silico structure-based repositioning of approved drugs for spike glycoprotein S2 domain fusion peptide of SARS-CoV-2: rationale from molecular dynamics and binding free energy calculations. mSystems. 2020;5 doi: 10.1128/MSYSTEMS.00382-20. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
103. Chi W.-Y., Li Y.-D., Huang H.-C., Chan T.E.H., Chow S.-Y., Su J.-H., Ferrall L., Hung C.-F., Wu T.-C. COVID-19 vaccine update: vaccine effectiveness, SARS-CoV-2 variants, boosters, adverse effects, and immune correlates of protection. J. Biomed. Sci. 2022;29:82. doi: 10.1186/s12929-022-00853-8. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from Heliyon are provided here courtesy of Elsevier

-