Next Article in Journal
Cross-Reaction or Co-Infection? Serological Discrimination of Antibodies Directed against Dugbe and Crimean-Congo Hemorrhagic Fever Orthonairovirus in Nigerian Cattle
Next Article in Special Issue
Emergence of E484K Mutation Following Bamlanivimab Monotherapy among High-Risk Patients Infected with the Alpha Variant of SARS-CoV-2
Previous Article in Journal
Viral RNA Metagenomics of Hyalomma Ticks Collected from Dromedary Camels in Makkah Province, Saudi Arabia
Previous Article in Special Issue
Potential Prophylactic Treatments for COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Appraisal of the Current Scenario in Vaccine Research for COVID-19

1
Department of Molecular and Translational Science, Monash University, Clayton 3168, Australia
2
Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur 57000, Malaysia
3
Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
4
School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia
5
Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, UP, India
6
School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia
7
School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 4 June 2021 / Revised: 13 July 2021 / Accepted: 16 July 2021 / Published: 18 July 2021
(This article belongs to the Special Issue Vaccines and Therapeutics against Coronaviruses)

Abstract

:
The recent coronavirus disease 2019 (COVID-19) outbreak has drawn global attention, affecting millions, disrupting economies and healthcare modalities. With its high infection rate, COVID-19 has caused a colossal health crisis worldwide. While information on the comprehensive nature of this infectious agent, SARS-CoV-2, still remains obscure, ongoing genomic studies have been successful in identifying its genomic sequence and the presenting antigen. These may serve as promising, potential therapeutic targets in the effective management of COVID-19. In an attempt to establish herd immunity, massive efforts have been directed and driven toward developing vaccines against the SARS-CoV-2 pathogen. This review, in this direction, is aimed at providing the current scenario and future perspectives in the development of vaccines against SARS-CoV-2.

1. Introduction

The current pandemic, coronavirus disease 2019 (COVID-19), has officially been confirmed in more than 187 million individuals and caused in excess of four million deaths globally (as of 12 July 2021) [1,2]. The causative pathogen, severe acute respiratory syndrome (SARS)-coronavirus 2 (SARS-CoV2) belongs to the coronavirus family, which is a large family of enveloped viruses that contain a positive sense, single-stranded RNA genome [3]. In the recent past, several members of the coronavirus family have been involved in significant disease outbreaks globally. The major diseases in this category include SARS and Middle East respiratory syndrome (MERS). In late 2019, SARS-CoV-2 was first detected in people who had visited the wet markets in the city of Wuhan, China. The viral particles soon engulfed major parts of the globe in a chaotic manner, resulting in an uncontrollable pandemic outbreak. A number of studies have reported that SARS-CoV-2 attacks the host’s respiratory system and thereby induces pneumonia-like syndrome, which may even lead to death in severe conditions [4]. To date, there are no effective and proven medications available to treat the disease in a synchronized manner, although a number of drugs and antiviral agents are being administered in an effort to contain the viral spread and alleviate the inflammatory response in patients presenting with severe complications.
Simultaneous efforts, to considerable success, are being made to anticipate the severity and transmission potential of SARS-CoV-2 and to develop efficacious prophylactic approaches, including public health messaging/advisories (lockdowns/hygiene/social distancing) and, more importantly, the development of vaccines. To date, a number of candidate vaccines are under various stages of development. A total of 8 of them have been approved for use, 121 are in clinical development, and 184 are in the preclinical development phase [5]. In this rapid review, we summarize the utility of vaccines to contain COVID-19 in the longer term and elaborate on major vaccine candidates currently in use in various countries.

2. Vaccination as a Promising Strategy Against COVID-19

Vaccination is a type of immunotherapy that introduces an immunogenic material to artificially stimulate the adaptive immune response of the body. Its applications and efficacy in combating and eradicating deadly infectious diseases such as smallpox, poliomyelitis, human papillomavirus, and several other infectious diseases have been well-documented [6,7,8,9]. Hence, it is by far the primary strategy in contesting the COVID-19 pandemic [10]. Findings from the recent next-generation sequencing studies have revealed approximately 5500 full-length genomes of SARS-CoV-2 that were isolated from various countries. These findings facilitate delineating the polymorphisms in the S protein and other important proteins of the virus that may serve as potential targets in the development of vaccines [11,12,13,14]. Interestingly, there is little to no cross-neutralization between the SARS-CoV and SARS-CoV-2, proposing that recovery from one infection may not necessarily provide any immune protection against the other [15]. Hence, there is a compelling need for a new vaccine agent that could exclusively target the SARS-CoV-2 pathogen. Notably, the development is highly possible with the reference and aid of the available next-generation sequencing database.
Besides the vaccine target, the production platform also plays an important role in determining the efficacy of the product in question. There are multiple distinct vaccine platforms that are currently available. These may include live-attenuated virus vaccine, inactivated virus vaccine, protein subunit vaccine, viral vector transduction-based vaccine, and nucleic acid-based vaccine, to name a few. Each platform has its own distinct advantages and disadvantages [16]. The classical vaccine platforms are the live-attenuated viral vaccine, the inactivated virus vaccine, and the protein subunit vaccine [16]. There exist a series of vaccines containing viral antigen, either live-attenuated virus, inactivated virus, synthetic or recombinant antigenic protein, that serve as immunogenic antigens to trigger a strong and long-lasting adaptive immune response. Live-attenuated vaccines typically use a living target virus, which had been weakened and no longer possesses the ability to infect, to stimulate the body’s immune system to elicit an adaptive immune response against the target virus [17]. As compared to an inactive virus or protein subunit vaccine, the live-attenuated vaccine is an exemplar vaccine model that mimics the actual infectious process of the infectious agent without causing a pathogenic infection [17,18]. However, the disadvantage of this vaccine platform is, it may not be suitable to the population who have weak or compromised immune systems, including those with long-term health conditions, the elderly, or who had organ transplantation performed before vaccination [19,20,21,22]. For these specific groups of the population, inactivated virus or protein subunit vaccine may be more suitable as vaccine candidates. Inactivated virus vaccine uses dead virus as the immunogenic antigen, whereas a protein subunit vaccine uses a fragment of the virus protein as the antigen to trigger an immune response [18,23,24]. As for COVID-19, the spike protein of the SARS-CoV-2 is the most suitable target, as it holds an essential role in regulating the binding of the virus with the host cells [15,25,26]. As compared to live-attenuated vaccines, inactivated virus and protein subunit vaccines do not essentially contain any living virus; hence, they are considered relatively safe [18,24]. However, the classical vaccine platform, including both types of the above-described vaccines, possess relatively lower immunogenicity and require additional adjuvants or boosters to enhance their biological half-lives and potentiate elicited immune responses [17,18,23,24,27].
With the emerging challenges and increasing drawbacks associated with the current vaccine development platforms, the global attention has now turned toward next-generation vaccine platforms, which essentially consist of, viral vector-based and nucleic acid-based vaccine platforms [16]. In particular, the viral vector transduction-based vaccine has been demonstrated to be a promising vaccine platform. Unlike the conventional vaccine, this type of vaccine uses host cells to produce target immunogenic antigens for adaptive immune response by viral transduction of the genetic code into host cells via modified viral vectors [28]. Viral vector transduction-based vaccines may be further differentiated into replicating or non-replicating types. A replicating vaccine may infect host cells and transduce gene sequences for both viral vectors and the target antigen to produce more infectious viral vectors that are able to infect more host cells; whereas a non-replicating vaccine only transduces gene sequences for the target antigen, hence, restrict self-replication of the viral vector [29,30,31]. As there is no living infectious agent that is involved during the vaccination process, this vaccine platform is relatively safe, and furthermore, prevents the need of handling the infectious agent [32]. Furthermore, this vaccine platform offers a long-term antigenic immune response against the target pathogens, as the genetic code for the immunogenic antigen of the pathogen is constitutively expressed in host cells after the vaccination [32]. However, the vaccine has relatively low efficiency, as the host may react against the viral vector during the first exposure and produce an unspecific immune response [30,31].
Meanwhile, the nucleic acid-based vaccine may be further classified as DNA or mRNA-based vaccines [16]. A DNA-based vaccine is a type of vaccine that consists of a synthetic DNA plasmid-construct encoding the target virus’s antigen [33]. Unlike the conventional vaccine administration, a DNA-based vaccine requires additional electroporation after the vaccine administration to facilitate the uptake of DNA-construct into the host cell [33]. On the other hand, mRNA-based vaccines use a similar principle, except in, they bypass the nuclear translocation and mRNA transcription process [34,35]. As both mRNA and DNA vaccines do not use any extracted viral material, the vaccination process is safe and is suitable for most of the population [35,36,37]. As synthetic DNA is temperature stable, it poses significant advantages from the point of view of mass production, delivery, and storage [36]. As compared to DNA, mRNA-based vaccines are relatively unstable and are temperature-sensitive, hence require extensive precautions in the developing and handling process [36,37]. Nevertheless, as both vaccine platforms are relatively new, there is an insufficient number of studies to determine their possible adverse effects on the receivers.

3. COVID-19 Vaccine Development

To further fast-track the development of counter-measures against the COVID-19 pandemic, the WHO has declared the outbreak as a Public Health Emergency of International Concern, as well as has initiated a Research and Development Blueprint under the recommendation of the WHO emergency committee [38]. The blueprint aims to “accelerate innovative research to help contain the spread of the epidemic and facilitate care for those affected”. It further emphasizes to “support research priorities that contribute to global research platforms in hopes of learning from the current pandemic response to better prepare for the next unforeseen epidemic” [38]. Since then, massive research funding and government support have been channeled toward the development of vaccines. In accordance with the blueprint, currently, there are more than 40 vaccine candidates that are prepared to progress until the clinical trial phase, as of 29 June 2021, as shown in Table 1.

3.1. BNT162b2

BNT162 is a series of mRNA-based vaccine candidates developed by Pfizer and BioNTech as a measure to contain COVID-19 spread. These vaccines are essentially lipid nanoparticles containing mRNA that encodes the SARS-CoV2 antigens and expresses 1 of 2 antigens for the SARS-CoV-2 full-length, P2 mutant, prefusion spike glycoprotein (P2 S) (version 9) (Genbank: MN908947); or a trimerized SARS-CoV-2 spike glycoprotein receptor-binding domain (RBD) (version 5) [39]. Notably, vaccine candidates, BNT162b1 (variantRBP020.3; nucleoside-modified messenger RNA (modRNA) with blunted innate immune sensor-activating capacity and augmented expression encoding the RBD) and BNT162b2 (variant RBP020.2; nucleoside-modified messenger RNA (modRNA) as above, but encoding P2S) have demonstrated their efficacy in vivo by “inducing protective antiviral effects rhesus macaques, with concomitant high neutralizing antibody titers and a TH1-biased cellular response in rhesus macaques and mice” [39,40]. Under a phase 3 clinical trial setting (NCT04368728) with 195 healthy adult participants, both BNT162b1 and BNT162b2 demonstrated similar immunogenicity [41]. However, the trial also discovered that participants who received the BNT162b1 vaccine suffered greater adverse effects as compared to those individuals who received the BNT162b2 variant, where the adverse effects ranged from mild fever (38–40 °C) to moderate systemic effects such as fatigue, headache, and chills. The observations and findings have led to rising concerns on the safety and tolerability of BNT162b1 [41]. Similarly, another clinical trial study with 60 healthy adult participants found that participants who received two doses between 1 and 50 µg of BNT162b1 had robust RBD-specific antibody, T cell, and favorable cytokine responses [42]. However, due to the relatively small population size, it is still obscure if the adverse effects exclusively occur only in a certain population who received BNT162b1. BNT162b2 was authorized by the Medicines and Healthcare products Regulatory Agency (MHRA) for use in the U.K. on 2 December 2020 after a rolling review of vaccine data submitted by Pfizer and BioNTech, despite the vaccine candidate having not completed its planned clinical trials assessment [43]. Following the approval, the United States of America Food and Drug Administration (USFDA) released their independent analysis of the clinical trial conducted on the vaccine candidate, in which they discovered the candidate has about 95% vaccination efficacy without eliciting serious adverse events [44]. Similarly, a recent clinical study in Israel also reported the high efficacy of BNT162b2 in protecting the receiver against COVID-19, with a vaccine efficacy of around 90% [45]. The observations and findings have further attracted global attention as it was one of the notable vaccine candidates against COVID-19, leading WHO to list BNT162b2 in Emergency Use Listing for COVID-19 [46]. So far, multiple countries have approved BNT162b2 as a COVID-19 vaccine, not limited to Argentina, Canada, Chile, Costa Rica, Ecuador, Jordan, Kuwait, Mexico, Panama, Singapore, Bahrain, Saudi Arabia, and Switzerland [47,48,49,50,51,52,53,54,55,56,57]. Notably, a recent observational study with a total number of 9876 participants who received a complete dosage of BNT162b2 (4938 vaccinated, 4938 unvaccinated) demonstrated a significant reduction in SARS-CoV-2 viral load (p < 1 × 10−17), further pointing toward the efficacy of this vaccine against SARS-CoV-2 [58]. So far, the BNT162b2 vaccine has passed phase 1, 2, and 3 clinical trials and currently undergoing a phase 4 clinical trial (NCT04760132) with 10,000 participants [59].

3.2. Sputnik V

Sputnik V, also known as Gam-COVID-Vac, is a non-replicating viral vector vaccine developed by the Gameleva Research Institute in collaboration with the Russian Direct Investment Fund and The Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation. It consists of a recombinant adenovirus type 26 (rAd26) vector and a recombinant adenovirus type 5 (rAd5) vector, in which both encoded the SARS-CoV-2’s spike glycoprotein (rAd26-S and rAd5-S). By using a replication-competent EGFP-reporter vesicular stomatitis virus system, it consists of recombinant DNA, rcVSV-CoV2-S, which encodes spike glycoprotein from SARS-CoV-2 (Genbank: MN908947.3), and packed in recombinant adenovirus type 26 (rAd26) vector and a recombinant adenovirus type 5 (rAd5) vector [60].
Unlike other vaccine candidates, the Sputnik V trial protocol has not been made public. Moreover, there are limited preclinical and clinical trial data available with regards to the Sputnik V vaccine. So far, there are a total of 2 phase 1/2 clinical trial studies (NCT04436471 and NCT04437875) that were conducted with a total of 76 healthy adult participants. The preliminary observations on the vaccine trial have been published [60]. According to both the concluded trials, participants has developed antibodies against SARS-CoV-2 glycoprotein after being administered with the Sputnik V vaccine, without any serious adverse events [60]. Currently, a phase 2 trial of 110 participants who are older than 60 years (NCT04587219) and a phase 3 trial of about 40,000 participants at multiple centers in Russia (NCT04530396) are being conducted for the Sputnik V vaccine to further determine its safety and efficacy profile under larger population trial [61,62]. Despite having a smaller pool of tested participants and limited clinical trial data, the Sputnik V vaccine was quickly approved and was registered on 11 August 2020 by Russian Federation [63]. This approval has drawn strong criticisms from various quarters from among the international scientific community for lack of data on safety and efficacy and for not following proper clinical trial safety guidelines [64,65]. So far, the vaccine candidate was granted approval in various countries, including Belarus, Argentina, Algeria, Bolivia, Serbia, and Palestine [66,67,68,69,70,71].

3.3. EpiVacCorona

EpiVacCorona is a peptide vaccine developed by the Russian Federal Budgetary Research Institution, the State Research Centre of Virology and Biotechnology. It consists of 3 distinct amino acids: (1) CRLFRKSNLKPFERDISTEIYQAGS, (2) CKEIDRLNEVAKNLNESLIDLQE, and (3) CKNLNESLIDLQELGKYEQYIK, which are artificially synthesized small fragment-peptide antigens of SARS-CoV-2 protein, conjugated to a carrier protein and adsorbed on an aluminum-containing adjuvant (aluminum hydroxide) [72]. Its phase 1/2 clinical trial (NCT04527575) was reported with excellent efficacy, with a 100% response rate and seroconversion with a neutralizing antibody titer >1:20, 21 days following complete vaccine administration. Moreover, all participants are reported free from any severe local or systemic adverse events [72,73]. Currently, it is under a phase 3 clinical trial targeted at 3000 participants [74]. EpiVacCorona was granted emergency approval from the Russian government on 15 October 2020 [75,76].

3.4. CoronaVac (PiCoVacc)

CoronaVac is a formalin-inactivated, alum-adjuvanted vaccine developed by the Sinovac Biotech Company. It is developed from purified inactivated SARS-CoV-2 virus strain CN2, which was isolated from the bronchoalveolar lavage fluid from one of the 11 hospitalized patients infected with SARS-CoV-2 [77]. The CN2 strain is selected as it was closely related to SARS-CoV-2 strain 2019-nCoV-BetaCoV Wuhan/WIV04/2019 (GISAID accession ID = EPI_ISL_402124), which has been firstly reported as the main source of the infectious agent for the coronavirus pandemic [77,78,79]. Under preclinical setting, CoronaVac vaccine demonstrated distinct safety and immunogenicity profile in various animal models, including Wistar rat and rhesus macaque monkey models, in which the animals survived and produced a high titer of neutralizing antibodies without developing serious adverse events [77]. Furthermore, a recent phase 1/2 clinical trial (NCT04352608) with 743 healthy participants demonstrated similar outcomes, with positive seroconversion in most participants (92.4% of participants after being administered with a 3 μg dose, on a 0–14 day schedule and 97.4% after being administered with a 3 μg the same dose on a 0–28 day schedule) [80]. Notably, another safety and tolerability trial targeting the elderly population (aged 65–68) (NCT04383574) further showed that the participants who received a complete dosage of CoronaVac did not show severe adverse reactions, and about 97% of participants showed at least four-fold neutralizing antibody titer, suggestive of the vaccine candidate well tolerable and effective among the elderly community [81]. Due to its distinct safety and immunogenicity efficacy profile, the Government of China has fast-tracked the production and has given emergency approval for the administration of CoronaVac exclusively to high-risk essential populations, such as health care workers, doctors, and nurses [82]. Currently, several international phase 3 clinical trials are in progress for the CoronaVac in Brazil (NCT04456595), Turkey (NCT04582344), and Indonesia (NCT04508075) [83,84,85].

3.5. mRNA-1273

mRNA-1273 is a vaccine candidate developed by Moderna Inc. in collaboration with the Coalition of Epidemic Preparedness Innovations and the National Institute of Allergy and Infectious Disease. It is a novel lipid nanoparticle-encapsulated mRNA-based vaccine developed against SARS-CoV-2. The mechanism is based on the principle that cells intake non-replicating mRNA, translate, and transiently express viral antigen protein on the cell surface without entering the cellular nucleus or interacting with the genome constitutively [86]. mRNA-1273 encodes for the full-length spike protein of SARS-CoV-2 (Genbank: MN908947.3), which gets further modified to introduce two proline residues that stabilize the spike protein in a prefusion conformation [86]. Several studies have shown that the spike protein regulates adherent and admission of the virus into host cells upon infection, hence presented as a potential target for the infection [87,88,89,90]. Under in vivo setting, the mouse models administered with the mRNA-1273 vaccine showed a reduction in viral replication and induced a potent neutralizing antibody against SARS-CoV-2 virus, as well as, CD8+T cell response. This mechanism has provided immune protection against SARS-CoV-2 infection in the lungs and noses of the mice, without evidence of immunopathology [91]. Similar events were also observed in non-human primate models, in which those vaccinated with the mRNA-1273 triggered a robust immune response against SARS-CoV-2 upon a challenge with the infection [92]. Meanwhile, under a clinical setting, a phase 1 trial demonstrated the efficacy of the vaccine candidate in upregulating SARS-CoV-2 neutralizing antibody titer in 45 healthy adult participants (aged 18–55 years old) and 40 healthy elderly (aged > 56 years old) [93,94]. In the cohort, three participants reported severe local adverse reactions (erythema and induration), and another three participants reported severe systemic adverse reactions (fever, fatigue, nausea, and myalgia) [93,94]. Nonetheless, all adverse reactions resolved within 24 h, proposing that the vaccine candidate is well tolerated by the healthy adult and elderly [38,95]. Meanwhile, in phase 3 efficacy, safety, and immunogenicity trial (NCT04470427) with 30,420 participants (15,210 participants each in vaccine receivers and placebo control group) demonstrated 94.1% of vaccine efficacy in preventing COVID-19 illness after receiving two complete dosages of vaccine, without having any severe adverse side effects [96,97,98]. This study supports that mRNA-1273 is safe to be used as an efficacious vaccine against COVID-19. On 30 November 2020, Moderna requested an emergency use authorization from the FDA and additionally a conditional marketing authorization from the Europen Medicines Agency (EMA) [99,100]. In the meantime, U.K.’s MHRA and Switzerland’s Swissmedic regulator initiated a review on mRNA-1273, which will allow a quick approval process for the vaccine candidate [101,102]. On 19 December 2020, FDA authorized mRNA-1273 for emergency use in the United States of America [103]. Following that, Canada, European Union, Israel, Switzerland, and United Kingdom approved the usage of mRNA-1273 [104,105,106,107,108].

3.6. AZD1222

AZD1222 (also known as Oxford-AstraZeneca vaccine, ChAdOx1 nCoV-19) is a COVID-19 vaccine candidate developed by the Oxford University and AstraZeneca. It is a non-replicating primate adenovirus vector containing the sequence of SARS-CoV-2 S protein (Genbank: MN908947) and tissue plasminogen activator. Its preclinical study with mice (BALB/c and CD1) and primate (rhesus macaque) models demonstrated its efficacy in eliciting an immune response against the SARS-CoV-2, including the inducing of viral-specific antibodies and the activation of T-helper 1 immune response (high level of IFN and TNF; low level of IL-4 and IL-10) post-vaccination [109]. Moreover, under a clinical setting, a phase 1/2 trial demonstrated the efficacy of the vaccine candidate in inducing an immune response against SARS-CoV-2 after vaccination [110]. In addition, all participants were found to be well tolerated to the vaccine, and no serious adverse events were reported [110]. However, during its phase 2/3 trial, several adverse cases were reported, such as an unexplained neurological symptom developed by one of the participants in the United Kingdom and a volunteer who died during the trial in Brazil, which forced the trial to be suspended for safety review [111,112]. On 23 October 2020, FDA approved the trial to be restarted after the trial passed its safety review [113]. The preliminary findings of the phase 2/3 trial were published, and it demonstrated 90% vaccination efficacy in 420 healthy adult volunteers without developing any significant adverse events [114]. However, the result raised concern as some participants involved were administered half the actual required dosage. AstraZeneca later admitted that it was a mistake [115]. Despite this, the vaccine candidate was granted emergency authorization by MHRA, Argentina’s National Administration of Drugs, Food, and Medical Devices (ANMT), and Mexico’s Health Regulator “Comision Federal para la Proteccion contra Riesgos Sanitarios” (COFEPRIS) [116,117,118]. On 1 January 2021, India approved the vaccine candidate, with the condition that the vaccine will be manufactured by the Serum Institute of India and will be released as “Covishield” vaccine [119]. Recently, a vaccine efficacy result for AZD1222 with 11,636 participants was reported with an overall vaccine efficacy of 90.0% (67.4%–97%) for those who received two standard doses and 70.4% for those who received one standard dose [120]. Notably, several patients were reported with prothrombotic immune thrombocytopenia after receiving AZD1222 [121,122,123,124]. Patients involved are presented with a thrombotic disorder, including cerebral venous sinus thrombosis, splanchnic vein thrombosis, arterial cerebral thromboembolism, or thrombotic microangiopathy [121,122,123,124]. Furthermore, the hematologic analysis found the presence of anti-platelet factor 4 autoantibodies in patients’ sera, which targets healthy donor platelet in an AZD1222 dependent manner, despite no previous history of heparin-induced thrombocytopenia [121,122,123,124]. Importantly, such aggregation can be resolved and suppressed by heparin [121,122,123,124]. Collectively, AZD1222 demonstrated vaccine efficacy against COVID-19, but its application is associated with severe autoimmune prothrombotic disorders.

3.7. Covaxin (BBV152)

Covaxin (also known as BBV-152) is a COVID-19 vaccine candidate developed by the Bharat Biotech and the Indian Council of Medical Research (ICMR) [125]. It is an inactivated vaccine that consists of the whole-virion SARS-CoV-2 inactivated via the β-propiolactone inactivation method [126]. Under preclinical setting, Covaxin demonstrated remarkable immunogenicity and protective efficacy against SARS-CoV-2 in both hamster and rhesus macaques models [126,127]. Within the hamster model, the vaccinated group developed an immune response against the SARS-CoV-2 virus with an increased titer of neutralizing antibodies and rapid clearance of the virus from the lower respiratory tract, reduced virus load in the upper respiratory tract, and absence of lung pathology after vaccination [126]. Similar events also were observed in rhesus macaques models [127]. On the other hand, under a clinical setting, a phase 1 trial with 375 healthy adult participants demonstrated the production of increased neutralizing antibodies against the SARS-CoV-2 virus without developing any serious adverse events [128]. The findings were further supported by its phase 2 trial that was conducted with 380 participants, where seroconversion rates of neutralizing antibodies reached up to 73% without causing any serious adverse events after vaccination [129]. On 3 January 2021, the Indian government approved Covaxin as a COVID-19 vaccine in India [130]. Such quick approval drew global criticism within the international scientific community for lack of data on safety and efficacy and for not following standard clinical trial safety guidelines [131,132,133].

3.8. Ad26.COV2

Ad26.COV2 (also known as Janssen COVID-19 vaccine or Johnson & Johnson COVID-19 vaccine) is a vaccine candidate developed by Janssen Pharmaceutical in collaboration with Johnson & Johnson pharmaceutical company [134]. It is a recombinant adenovirus serotype 26 vector encoding SARS-CoV-2 peplomers based on SARS-CoV-2 Wuhan-Hu-1 isolate strain (Genbank accession number: MN908947) [135]. Ad26.COV2 demonstrated high efficacy under preclinical setting in multiple animal models, including Syrian hamster, non-human primates, in which all vaccinated animals showed a rapid increase in neutralizing antibodies and reduction in lung viral load [136,137,138,139]. Notably, all animal models reported the absence of any adverse side effects or signs of vaccine-related respiratory disease, suggestive of a safe and tolerable vaccine candidate against SARS-CoV-2 [136,137,138,139]. Under clinical setting, safety and tolerability trial (NCT04436276) exhibited high safety and efficacy profile of Ad26.COV2 in human participants, with elevated neutralizing antibody response of 88% in the adult population (aged 18–55) and 93% in the elderly population (aged > 65) without any significant adverse events reported [140]. On the other hand, phase 3 trial (NCT04505722) with 43,783 (21,895 received vaccine; 21,888 received placebo) participants revealed vaccine efficacy of approximate 66.8% [141]. Moreover, there are only 18 participants who reported pyrexia, which is resolved by using analgesics or antipyretics 7 days after vaccination [141]. Due to the excellent efficacy, FDA issued an emergency use authorization for Ad26.COV2 for the prevention of COVID-19 on 27 February 2021 [142], followed by the conditional marketing authorization by EMA [143] and MHRA [144]. However, increasing case reports found that some patients who received the Ad26.COV2 vaccine develop thrombotic thrombocytopenia symptoms [145,146,147]. In response, the manufacturers acknowledge the presence of rare thrombotic thrombocytopenia among the vaccine receiver but argue that the reporting rate is less than 1:1,000,000, and more evidence and studies are needed to clarify such observations [146].

3.9. BBIBP-CorV

BBIBP-CorV is an inactivated viral vaccine manufactured by Sinopharm pharmaceutical company in collaboration with the Chinese Center for Disease Control and Prevention and the Beijing Institute of Biological Products. It is an aluminum hydroxide-based adjuvant, β-propiolactone-inactivated vaccine according to the 19nCOV-CDC-TAN-HB02 strain (HB02 strain) [148]. Under a preclinical setting, the BBIBO-CorV vaccine was able to induce active seroconversion, leading the host to develop a significantly high titer of neutralizing antibodies in immune-competent mice models [149]. Similar positive effects were also demonstrated in other non-human animal models, including rabbit, guinea pig, rat, and rhesus macaques models [149]. Furthermore, it also demonstrated a suitable safety profile, in which the animal models that received the vaccine did not show any sign of ethical endpoint or ill-health after vaccination, suggestive of the safe and efficacious vaccine candidate. On the other hand, under the clinical trial setting, its safety and tolerability trial (ChiCTR2000032459) demonstrated that participants who received the vaccine have significantly elevated neutralizing antibody titer levels in their sera; 14 days after vaccination, with the seroconversion rates of approximately 100% [150]. Furthermore, no patients were reported with severe adverse events within 28 days after vaccination [150]. Whereas, in immunogenicity trial (NCT04510207) with 45,000 healthy participants, the vaccine candidate demonstrated excellent efficacy in preventing against COVID-19, with the seroconversion of neutralizing antibody of approximate 99.5% among adult participants (aged 18–59) and 100% among elderly participants (aged > 65) [150]. In a preliminary analysis based on the available data, BBIBP-CorV has an efficacy of 78.1% in protecting against COVID-19 [150]. Similarly, no severe adverse events were reported in participants who received the vaccination, suggesting that BBIBP-CorV is safe and tolerable in healthy participants [150]. Due to its high safety and efficacy profile, WHO granted approval for it to be used in the COVID-19 Vaccine Global Access program, and it is currently being used by multiple nations [151,152].

4. Efficacy of Vaccine Against Emerging SARS-CoV-2 Variant Strains

Increasing case studies have reported new variants of SARS-CoV-2 virus strains diagnosed in people around the world. By definition, a SARS-CoV-2 variant is a mutated strain of SARS-CoV-2, in which the mutated strain may possess different replication, transmission, or virulence rate as compared to its parental strain [153,154,155,156,157]. Notably, the mutated strain was reported with enhanced resistance or immune escape mechanisms against current vaccine candidates; hence, the vaccine developed so far may lose partial or complete efficacy in combating these mutated variant strains, posing an extreme challenge in combating the COVID-19 pandemic [157,158,159,160,161,162]. The earliest variant discovered is known as the SARS-CoV-2 Alpha variant (B.1.1.7), which was found and documented in United Kingdom [163]. Since then, multiple variant strains, such as SARS-CoV-2 Beta variant (B.1.351, South Africa) [164], Gamma variant (P.1, Brazil) [165], or Iota variant (B.1.526, United States of America) [166] were reported across the countries. So far, there are at least 19 lineage variants of SARS-CoV-2 were discovered and documented, as summarized in Figure 1 [167].
Fortunately, some vaccine candidates were found to be efficacious against these variants. For instance, the BNT162b2 vaccine had an estimated vaccination efficacy of 75% against the Beta variant, 88% against the Delta variant (B.1.617.2), and 93% against the Alpha variant of SARS-CoV-2 [168,169]. BBIBP-CoV showed that patients who were diagnosed with variant strain GDPCC (501Y.V2) and BJ01 (D614G) produced a high titer value of neutralizing antibody after receiving BBIBP-CoV vaccine, although the neutralization titer is relatively lower than compared to its target strain (HB02 = 110.9 vs. BJ01 = 107.2 vs. GDPCC = 70.9), suggesting that the vaccine candidate provided certain protections against the variant strains [170]. On the other hand, the mRNA-1273 demonstrated a reduction in seroconversion rates and neutralizing antibody titer level against the Beta, Gamma, and Alpha variants, concerning its effectiveness in protecting against COVID-19 variant strains [171]. Collectively, these findings highlight the limited efficacy of the current vaccine candidates against the SARS-CoV-2 variant strains, urging for a need to develop a better vaccine candidate with higher vaccine efficacy against these variants.

5. Conclusions

In this review, we have appraised the current state of the COVID-19 pandemic and the vaccines that have been approved so far in the fight against this viral disease. However, as SARS-CoV-2 is a novel organism that was recently identified, the information regarding this infectious agent remains obscure. Nonetheless, recent genome sequencing data may shed more light with regards to the pathogenesis of SARS-CoV-2, which will prove useful in the development of effective treatments against it. On the other hand, thanks to the accelerated track in vaccine development, which has resulted in the development and approval of several COVID-19 vaccines to date. Furthermore, due to the large number of trials that are in progress currently, there will be more vaccines to be approved in the near future. Notably, due to the emergence of new SARS-CoV-2 variant strains, vaccine candidates showing effectiveness on all variants are urgently needed.

6. Opinion

The situation of the COVID-19 pandemic is constantly changing, and there are constant updates regarding SARS-CoV-2 and its disease pathology, as well as the information on vaccines developed against the virus. The cases of COVID-19 are constantly on the rise in several countries, which has become unavoidable. If unchecked, the global viral infectivity rates may prove catastrophic. This has led to the urgent need to fully vaccinate society against the COVID-19 pathogen. Currently, the available vaccines are under slow progression for mass immunization, as the supply has been largely limited. Therefore, effective measures and suitable interventions are needed to further accelerate the vaccine development to eradicate the COVID-19 infection.

7. Key Findings

  • SARS-CoV-2 has been the causative viral pathogen responsible for the COVID-19 outbreak;
  • Vaccination against SARS-CoV-2 has now become the main therapeutic strategy for eradicating COVID-19 from the community;
  • Numerous vaccine candidates have been introduced, of which mRNA 1273, BNT162, AZD1222, CoronaVac, Sputnik V, EpiVaCorona, and Covaxin have been the leading vaccine candidates against SARS-CoV-2.

Author Contributions

Conceptualization: W.C.C. and M.D.S.; Writing—original draft: W.C.C. and M.D.S.; Writing—review and editing: all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dong, E.; Du, H.; Gardner, L. COVID-19 Dashboard by the Center for Systems Science and Engineering (CSSE) at Johns Hopkins University (JHU). Available online: https://www.arcgis.com/apps/opsdashboard/index.html#/bda7594740fd40299423467b48e9ecf6 (accessed on 27 March 2021).
  2. Dong, E.; Du, H.; Gardner, L. An interactive web-based dashboard to track COVID-19 in real time. Lancet 2020, 20, 533–534. [Google Scholar] [CrossRef]
  3. NIH. Coronaviruses. Available online: https://www.niaid.nih.gov/diseases-conditions/coronaviruses (accessed on 27 March 2021).
  4. Shastri, M.D.; Shukla, S.D.; Chong, W.C.; Kc, R.; Dua, K.; Patel, R.P.; Peterson, G.M.; O’Toole, R.F. Smoking and COVID-19: What we know so far. Respir. Med. 2020, 106237. [Google Scholar] [CrossRef]
  5. Zimmer, C.; Corum, J.; Wee, S.-L. Coronavirus Vaccine Tracker. Available online: https://www.nytimes.com/interactive/2020/science/coronavirus-vaccine-tracker.html (accessed on 25 March 2021).
  6. Paavonen, J.; Naud, P.; Salmerón, J.; Wheeler, C.M.; Chow, S.-N.; Apter, D.; Kitchener, H.; Castellsagué, X.; Teixeira, J.C.; Skinner, S.R. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): Final analysis of a double-blind, randomised study in young women. Lancet 2009, 374, 301–314. [Google Scholar] [CrossRef]
  7. Giuliano, A.R.; Palefsky, J.M.; Goldstone, S.; Moreira, E.D., Jr.; Penny, M.E.; Aranda, C.; Vardas, E.; Moi, H.; Jessen, H.; Hillman, R. Efficacy of quadrivalent HPV vaccine against HPV Infection and disease in males. N. Engl. J. Med. 2011, 364, 401–411. [Google Scholar] [CrossRef] [Green Version]
  8. Riedel, S. Edward Jenner and the history of smallpox and vaccination. In Baylor University Medical Center Proceedings; Taylor & Francis: Abingdon, UK, 2005; Volume 18, pp. 21–25. [Google Scholar]
  9. Patel, M.; Zipursky, S.; Orenstein, W.; Garon, J.; Zaffran, M. Polio endgame: The global introduction of inactivated polio vaccine. Expert Rev. Vaccines 2015, 14, 749–762. [Google Scholar] [CrossRef] [PubMed]
  10. Masihi, K.N. Fighting infection using immunomodulatory agents. Expert Opin. Biol. Ther. 2001, 1, 641–653. [Google Scholar] [CrossRef] [PubMed]
  11. NCBI. NCBI SARS-CoV-2 Resources; National Library of Medicine: Bethesda, MD, USA, 2020. [Google Scholar]
  12. Forster, P.; Forster, L.; Renfrew, C.; Forster, M. Phylogenetic network analysis of SARS-CoV-2 genomes. Proc. Natl. Acad. Sci. USA 2020, 117, 9241–9243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Sah, R.; Rodriguez-Morales, A.J.; Jha, R.; Chu, D.K.; Gu, H.; Peiris, M.; Bastola, A.; Lal, B.K.; Ojha, H.C.; Rabaan, A.A. Complete genome sequence of a 2019 novel coronavirus (SARS-CoV-2) strain isolated in Nepal. Microbiol. Resour. Announc. 2020, 9, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Khan, M.I.; Khan, Z.A.; Baig, M.H.; Ahmad, I.; Farouk, A.-E.; Song, Y.G.; Dong, J.-J. Comparative genome analysis of novel coronavirus (SARS-CoV-2) from different geographical locations and the effect of mutations on major target proteins: An in silico insight. PLoS ONE 2020, 15, e0238344. [Google Scholar] [CrossRef]
  15. Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef] [Green Version]
  16. van Riel, D.; de Wit, E. Next-generation vaccine platforms for COVID-19. Nat. Mater. 2020, 19, 810–812. [Google Scholar] [CrossRef]
  17. Belshe, R.B.; Edwards, K.M.; Vesikari, T.; Black, S.V.; Walker, R.E.; Hultquist, M.; Kemble, G.; Connor, E.M. Live attenuated versus inactivated influenza vaccine in infants and young children. N. Engl. J. Med. 2007, 356, 685–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Monto, A.S.; Ohmit, S.E.; Petrie, J.G.; Johnson, E.; Truscon, R.; Teich, E.; Rotthoff, J.; Boulton, M.; Victor, J.C. Comparative efficacy of inactivated and live attenuated influenza vaccines. N. Engl. J. Med. 2009, 361, 1260–1267. [Google Scholar] [CrossRef] [PubMed]
  19. Siedler, A.; Koch, J.; Ultsch, B.; Garbe, E.; von Kries, R.; Ledig, T.; Mertens, T.; Überla, K.; Zepp, F.; Hengel, H. Background paper to the decision not to recommend a standard vaccination with the live attenuated herpes zoster vaccine for the elderly in Germany. Bundesgesundheitsbl 2017, 60, 1162–1179. [Google Scholar]
  20. Kamboj, M.; Sepkowitz, K.A. Risk of transmission associated with live attenuated vaccines given to healthy persons caring for or residing with an immunocompromised patient. Infect. Control Hosp. Epidemiol. 2007, 28, 702–707. [Google Scholar] [CrossRef] [PubMed]
  21. Shinjoh, M.; Miyairi, I.; Hoshino, K.; Takahashi, T.; Nakayama, T. Effective and safe immunizations with live-attenuated vaccines for children after living donor liver transplantation. Vaccine 2008, 26, 6859–6863. [Google Scholar] [CrossRef] [PubMed]
  22. Chong, P.P.; Avery, R.K. A comprehensive review of immunization practices in solid organ transplant and hematopoietic stem cell transplant recipients. Clin. Ther. 2017, 39, 1581–1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Moyle, P.M.; Toth, I. Modern subunit vaccines: Development, components, and research opportunities. ChemMedChem 2013, 8, 360–376. [Google Scholar] [CrossRef]
  24. Tan, M.; Jiang, X. Recent advancements in combination subunit vaccine development. Hum. Vaccines Immunother. 2017, 13, 180–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yang, J.; Wang, W.; Chen, Z.; Lu, S.; Yang, F.; Bi, Z.; Bao, L.; Mo, F.; Li, X.; Huang, Y. A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature 2020, 586, 572–577. [Google Scholar] [CrossRef]
  26. Chi, X.; Yan, R.; Zhang, J.; Zhang, G.; Zhang, Y.; Hao, M.; Zhang, Z.; Fan, P.; Dong, Y.; Yang, Y. A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science 2020, 369, 650–655. [Google Scholar] [CrossRef]
  27. Cao, Y.; Zhu, X.; Hossen, M.N.; Kakar, P.; Zhao, Y.; Chen, X. Augmentation of vaccine-induced humoral and cellular immunity by a physical radiofrequency adjuvant. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef]
  28. Rauch, S.; Jasny, E.; Schmidt, K.E.; Petsch, B. New vaccine technologies to combat outbreak situations. Front. Immunol. 2018, 9, 1963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Henao-Restrepo, A.M.; Longini, I.M.; Egger, M.; Dean, N.E.; Edmunds, W.J.; Camacho, A.; Carroll, M.W.; Doumbia, M.; Draguez, B.; Duraffour, S. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: Interim results from the Guinea ring vaccination cluster-randomised trial. Lancet 2015, 386, 857–866. [Google Scholar] [CrossRef]
  30. Ura, T.; Okuda, K.; Shimada, M. Developments in viral vector-based vaccines. Vaccines 2014, 2, 624–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. de Vries, R.D.; Rimmelzwaan, G.F. Viral vector-based influenza vaccines. Hum. Vaccines Immunother. 2016, 12, 2881–2901. [Google Scholar] [CrossRef]
  32. Condit, R.C.; Kim, D.; Robertson, J.S.; Excler, J.-L.; Gurwith, M.; Monath, T.P.; Pavlakis, G.; Fast, P.E.; Smith, J.; Smith, E.R. The Brighton Collaboration standardized template for collection of key information for benefit-risk assessment of viral vector vaccines. Vaccine 2020, 38, 7708–7715. [Google Scholar] [CrossRef] [PubMed]
  33. Ulmer, J.B.; Wahren, B.; Liu, M.A. Gene-based vaccines: Recent technical and clinical advances. Trends Mol. Med. 2006, 12, 216–222. [Google Scholar] [CrossRef] [PubMed]
  34. Pascolo, S. Messenger RNA-based vaccines. Expert Opin. Biol. Ther. 2004, 4, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
  35. Ulmer, J.B.; Mason, P.W.; Geall, A.; Mandl, C.W. RNA-based vaccines. Vaccine 2012, 30, 4414–4418. [Google Scholar] [CrossRef]
  36. Jahanafrooz, Z.; Baradaran, B.; Mosafer, J.; Hashemzaei, M.; Rezaei, T.; Mokhtarzadeh, A.; Hamblin, M.R. Comparison of DNA and mRNA vaccines against cancer. Drug Discov. Today 2020, 25, 552–560. [Google Scholar] [CrossRef] [PubMed]
  37. Wadhwa, A.; Aljabbari, A.; Lokras, A.; Foged, C.; Thakur, A. Opportunities and Challenges in the Delivery of mRNA-based Vaccines. Pharmaceutics 2020, 12, 102. [Google Scholar] [CrossRef] [Green Version]
  38. WHO. WHO R&D Blueprint Novel Coronavirus (nCov) Vaccine Prioritization for Clinical Trials. Available online: https://www.who.int/publications/i/item/who-r-d-blueprint-novel-coronavirus-ncov-vaccine-prioritization-for-clinical-trials (accessed on 17 December 2020).
  39. Pfizer. A Phase 1/2/3 Study to Evaluate the Safety, Tolerability, Immunogenicity, and Efficacy of RNA Vaccine Candidates against COVID-19 in Healthy Individuals; Pfizer: New York, NY, USA, 2020; pp. 1–137. [Google Scholar]
  40. Vogel, A.; Kanevsky, I.; Che, Y.; Swanson, K.; Muik, A.; Vormehr, M.; Kranz, L.; Walzer, K.; Hein, S.; Gueler, A. A prefusion SARS-CoV-2 spike RNA vaccine is highly immunogenic and prevents lung infection in non-human primates. bioRxiv 2020, 1–38. [Google Scholar] [CrossRef]
  41. Walsh, E.E.; Frenck, R.W., Jr.; Falsey, A.R.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Mulligan, M.J.; Bailey, R. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N. Engl. J. Med. 2020, 383, 2439–2450. [Google Scholar] [CrossRef] [PubMed]
  42. Sahin, U.; Muik, A.; Derhovanessian, E.; Vogler, I.; Kranz, L.M.; Vormehr, M.; Baum, A.; Pascal, K.; Quandt, J.; Maurus, D. COVID-19 vaccine BNT162b1 elicits human antibody and TH 1 T cell responses. Nature 2020, 586, 594–599. [Google Scholar] [CrossRef]
  43. MHRA (Ed.) Regulatory Approval of Pfizer/BioNTech Vaccine for COVID-19; Government of the United Kingdom: London, UK, 2020. [Google Scholar]
  44. Jansen, K.; Gruber, W. BNT162b2 Vaccine Candidate against COVID-19. FDA: Silver Spring, MD, USA, 2020; p. 64. Available online: https://stacks.cdc.gov/view/cdc/105431 (accessed on 17 December 2020).
  45. Hunter, P.R.; Brainard, J.S. Estimating the effectiveness of the Pfizer COVID-19 BNT162b2 vaccine after a single dose. A reanalysis of a study of’real-world’vaccination outcomes from Israel. Medrxiv 2021. [Google Scholar] [CrossRef]
  46. WHO. WHO Issues its First Emergency Use Validation for a COVID-19 Vaccine and Emphasizes Need for Equitable Global Access. Available online: https://www.who.int/news/item/31-12-2020-who-issues-its-first-emergency-use-validation-for-a-covid-19-vaccine-and-emphasizes-need-for-equitable-global-access (accessed on 31 December 2020).
  47. RioTimes. Argentina Grants Emergency Approval to Pfizer/BioNTech Vaccine. The RioTimes, 23 December 2020. [Google Scholar]
  48. HealthCanada. Pfizer-BioNTech COVID-19 Vaccine: Authorization Information. Available online: https://www.canada.ca/en/health-canada/services/drugs-health-products/covid19-industry/drugs-vaccines-treatments/vaccines/pfizer-biontech/authorization.html (accessed on 8 January 2021).
  49. Montes, R.; Espana, S. Chile y Ecuador se adelantan en Sudamérica y autorizan la vacuna de Pfizer. El Pais, 17 December 2020. [Google Scholar]
  50. Zuniga, A. Costa Rica authorizes Pfizer-BioNTech coronavirus vaccine. The TicoTimes, 16 December 2020. [Google Scholar]
  51. RoyaNews. Obeidat confirms arrival of first batch of Pfizer vaccine Monday. RoyaNews, 10 January 2021. [Google Scholar]
  52. ABCNews. Bahrain approves Chinese COVID-19 vaccine for use. ABC News, 13 December 2020. [Google Scholar]
  53. NewYorkTimes. Covid-19: F.D.A. Clears Pfizer Vaccine and Millions of Doses Will Be Shipped Right Away. The New York Times, 15 December 2020. [Google Scholar]
  54. Castrellon, T. Panamá aprueba uso por emergencia de la vacuna Pfizer-BioNTech contra COVID-19. Telemetro, 15 December 2020. [Google Scholar]
  55. HSA. HSA Grants Interim Authorisation for First COVID-19 Vaccine in Singapore; Government of Singapore: Singapore, 2020. [Google Scholar]
  56. Radwan, R. Saudi Arabia ready to launch vaccine campaign. Arab News, 12 December 2020. [Google Scholar]
  57. FOPH (Ed.) Coronavirus: Vaccination; Swiss Government: Bern, Switzerland, 2021. [Google Scholar]
  58. Levine-Tiefenbrun, M.; Yelin, I.; Katz, R.; Herzel, E.; Golan, Z.; Schreiber, L.; Wolf, T.; Nadler, V.; Ben-Tov, A.; Kuint, J. Initial report of decreased SARS-CoV-2 viral load after inoculation with the BNT162b2 vaccine. Nat. Med. 2021, 27, 790–792. [Google Scholar] [CrossRef] [PubMed]
  59. ClinicalTrial. National Cohort Study of Effectiveness and Safety of SARS-CoV-2/COVID-19 Vaccines (ENFORCE) (ENFORCE). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04760132?term=vaccine%2C+phase+4&cond=Covid19&draw=2 (accessed on 6 July 2021).
  60. Logunov, D.Y.; Dolzhikova, I.V.; Zubkova, O.V.; Tukhvatullin, A.I.; Shcheblyakov, D.V.; Dzharullaeva, A.S.; Grousova, D.M.; Erokhova, A.S.; Kovyrshina, A.V.; Botikov, A.G. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: Two open, non-randomised phase 1/2 studies from Russia. Lancet 2020, 396, 887–897. [Google Scholar] [CrossRef]
  61. ClinicalTrial. The Study of “Gam-COVID-Vac” Vaccine against COVID-19 with the Participation of Volunteers of 60 y.o and Older. Available online: https://clinicaltrials.gov/ct2/show/NCT04587219?term=NCT04587219&draw=2&rank=1 (accessed on 17 December 2020).
  62. ClinicalTrial. Clinical Trial of Efficacy, Safety, and Immunogenicity of Gam-COVID-Vac Vaccine against COVID-19 (RESIST). Available online: https://clinicaltrials.gov/ct2/show/NCT04530396?term=NCT04530396&draw=2&rank=1 (accessed on 17 December 2020).
  63. BBC. Coronavirus: Putin says vaccine has been approved for use. BBC News, 11 August 2020. [Google Scholar]
  64. Callaway, E. Russia’s fast-track coronavirus vaccine draws outrage over safety. Nature 2020, 584, 334–335. [Google Scholar] [CrossRef] [PubMed]
  65. Mahase, E. Covid-19: Russia approves vaccine without large scale testing or published results. BMJ 2020, 370, m3205. [Google Scholar] [CrossRef] [PubMed]
  66. Grzegorczyk, M. Belarus begins rollout of Russian Covid-19 vaccine. Emerging Europe, 29 December 2020. [Google Scholar]
  67. LiveMint. First 300,000 doses of Sputnik V vaccine arrive in Argentina to fight Covid-19. LiveMint, 24 December 2020. [Google Scholar]
  68. SouthernTimes. Algeria first in Africa to approve Sputnik V vaccine. The Southern Times, 11 January 2021. [Google Scholar]
  69. Wadhwa, T. Bolivia approves emergency use of Russia’s Sputnik V vaccine. People Dispatch, 7 January 2021. [Google Scholar]
  70. RFERL. Serbia Starts Inoculations With Russia’s Sputnik-V Vaccine. Radio Free Europe Radio Liberty, 6 January 2021. [Google Scholar]
  71. Reuters. Palestinians approve Russian COVID-19 vaccine for use in self-rule areas. Reuters, 11 January 2021. [Google Scholar]
  72. Reuters. Russia Completes Clinical Trials of Second Potential COVID-19 Vaccine: RIA. Available online: https://www.reuters.com/article/uk-health-coronavirus-russia-vaccine-vec/russia-completes-clinical-trials-of-second-potential-covid-19-vaccine-ria-idUSKBN26L13B (accessed on 17 December 2020).
  73. Ryzhikov, A.; Ryzhikov, Е.; Bogryantseva, M.; Usova, S.; Danilenko, E.; Nechaeva, E.; Pyankov, O.; Pyankova, O.; Gudymo, A.; Bodnev, S. A single blind, placebo-controlled randomized study of the safety, reactogenicity and immunogenicity of the “EpiVacCorona” Vaccine for the prevention of COVID-19, in volunteers aged 18–60 years (phase I–II). Russ. J. Infect. Immun. 2021, 11, 283–296. [Google Scholar] [CrossRef]
  74. ClinicalTrial. Study of the Tolerability, Safety, Immunogenicity and Preventive Efficacy of the EpiVacCorona Vaccine for the Prevention of COVID-19. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04780035?term=vaccine&cond=Covid19&draw=2 (accessed on 6 July 2021).
  75. Bureau, H. Russia Approves 2nd Covid-19 Vaccine ‘EpiVacCorona’ after Sputnik V. Available online: https://www.healthwire.co/russia-approves-2nd-covid-19-vaccine-epivaccorona-after-sputnik-v/ (accessed on 17 December 2020).
  76. ClinicalTrial. Study of the Safety, Reactogenicity and Immunogenicity of “EpiVacCorona” Vaccine for the Prevention of COVID-19 (EpiVacCorona). Available online: https://clinicaltrials.gov/ct2/show/NCT04527575?term=NCT04527575&draw=2&rank=1 (accessed on 17 December 2020).
  77. Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z. Development of an inactivated vaccine candidate for SARS-CoV-2. Science 2020, 369, 77–81. [Google Scholar] [CrossRef] [PubMed]
  78. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [Green Version]
  79. Liu, L.; Liu, W.; Zheng, Y.; Jiang, X.; Kou, G.; Ding, J.; Wang, Q.; Huang, Q.; Ding, Y.; Ni, W. A preliminary study on serological assay for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 238 admitted hospital patients. Microbes Infect. 2020, 22, 206–211. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, Y.; Zeng, G.; Pan, H.; Li, C.; Hu, Y.; Chu, K.; Han, W.; Chen, Z.; Tang, R.; Yin, W. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: A randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet 2020. [Google Scholar] [CrossRef]
  81. Wu, Z.; Hu, Y.; Xu, M.; Chen, Z.; Yang, W.; Jiang, Z.; Li, M.; Jin, H.; Cui, G.; Chen, P. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine (CoronaVac) in healthy adults aged 60 years and older: A randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect. Dis. 2021, 21, 803–812. [Google Scholar] [CrossRef]
  82. Reuters. Sinovac’s Coronavirus Vaccine Candidate Approved for Emergency Use in China—Source. Available online: https://www.reuters.com/article/us-health-coronavirus-china-vaccines/sinovacs-coronavirus-vaccine-candidate-approved-for-emergency-use-in-china-source-idUSKBN25O0Z3 (accessed on 17 December 2020).
  83. ClinicalTrial. Clinical Trial of Efficacy and Safety of Sinovac’s Adsorbed COVID-19 (Inactivated) Vaccine in Healthcare Professionals (PROFISCOV). Available online: https://clinicaltrials.gov/ct2/show/NCT04456595?term=NCT04456595&draw=2&rank=1 (accessed on 17 December 2020).
  84. ClinicalTrial. Clinical Trial for SARS-CoV-2 Vaccine (COVID-19). Available online: https://clinicaltrials.gov/ct2/show/NCT04582344?term=NCT04582344&draw=2&rank=1 (accessed on 17 December 2020).
  85. ClinicalTrial. Efficacy, Safety and Immunogenicity Study of SARS-CoV-2 Inactivated Vaccine (COVID-19). Available online: https://clinicaltrials.gov/ct2/show/NCT04508075?term=NCT04508075&draw=2&rank=1 (accessed on 17 December 2020).
  86. ModernaTX, Inc. Phase 3, Randomized, Stratified, Observer-Blind, Placebo-Controlled Study to Evaluate the Efficacy, Safety, and Immunogenicity of mRNA-1273 SARS-CoV-2 Vaccine in Adults Aged 18 Years and Older. ClinicalTrials.gov, 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04470427 (accessed on 17 December 2020).
  87. Hoffmann, M.; Kleine-Weber, H.; Pöhlmann, S. A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells. Mol. Cell 2020, 78, 779–784. [Google Scholar] [CrossRef] [PubMed]
  88. Toelzer, C.; Gupta, K.; Yadav, S.K.; Borucu, U.; Davidson, A.D.; Williamson, M.K.; Shoemark, D.K.; Garzoni, F.; Staufer, O.; Milligan, R. Free fatty acid binding pocket in the locked structure of SARS-CoV-2 spike protein. Science 2020, 370, 725–730. [Google Scholar] [CrossRef] [PubMed]
  89. Salazar, E.; Kuchipudi, S.V.; Christensen, P.A.; Eagar, T.; Yi, X.; Zhao, P.; Jin, Z.; Long, S.W.; Olsen, R.J.; Chen, J. Convalescent plasma anti–SARS-CoV-2 spike protein ectodomain and receptor-binding domain IgG correlate with virus neutralization. J. Clin. Investig. 2020, 130, 6728–6738. [Google Scholar] [CrossRef]
  90. Chen, X.; Li, R.; Pan, Z.; Qian, C.; Yang, Y.; You, R.; Zhao, J.; Liu, P.; Gao, L.; Li, Z. Human monoclonal antibodies block the binding of SARS-CoV-2 spike protein to angiotensin converting enzyme 2 receptor. Cell. Mol. Immunol. 2020, 17, 647–649. [Google Scholar] [CrossRef] [PubMed]
  91. Corbett, K.S.; Edwards, D.K.; Leist, S.R.; Abiona, O.M.; Boyoglu-Barnum, S.; Gillespie, R.A.; Himansu, S.; Schäfer, A.; Ziwawo, C.T.; DiPiazza, A.T.; et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 2020, 586, 567–571. [Google Scholar] [CrossRef]
  92. Corbett, K.S.; Flynn, B.; Foulds, K.E.; Francica, J.R.; Boyoglu-Barnum, S.; Werner, A.P.; Flach, B.; O’Connell, S.; Bock, K.W.; Minai, M. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N. Engl. J. Med. 2020, 383, 1544–1555. [Google Scholar] [CrossRef] [PubMed]
  93. Jackson, L.A.; Anderson, E.J.; Rouphael, N.G.; Roberts, P.C.; Makhene, M.; Coler, R.N.; McCullough, M.P.; Chappell, J.D.; Denison, M.R.; Stevens, L.J. An mRNA vaccine against SARS-CoV-2—Preliminary report. N. Engl. J. Med. 2020, 383, 1920–1931. [Google Scholar] [CrossRef] [PubMed]
  94. Anderson, E.J.; Rouphael, N.G.; Widge, A.T.; Jackson, L.A.; Roberts, P.C.; Makhene, M.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; Pruijssers, A.J. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N. Engl. J. Med. 2020, 383, 2427–2438. [Google Scholar] [CrossRef] [PubMed]
  95. Latestly. COVID-19 Vaccine EpiVacCorona Creates Immunity 1 Month After Inoculation, Says Russia’s Vector. Yahoo News, 27 November 2020. [Google Scholar]
  96. ClinicalTrial. Dose-Confirmation Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of mRNA-1273 COVID-19 Vaccine in Adults Aged 18 Years and Older. Available online: https://clinicaltrials.gov/ct2/show/NCT04405076?term=NCT04405076&draw=2&rank=1 (accessed on 17 December 2020).
  97. ClinicalTrial. A Study to Evaluate Efficacy, Safety, and Immunogenicity of mRNA-1273 Vaccine in Adults Aged 18 Years and Older to Prevent COVID-19. Available online: https://clinicaltrials.gov/ct2/show/NCT04470427?term=NCT04470427&draw=2&rank=1 (accessed on 17 December 2020).
  98. 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. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef] [PubMed]
  99. FDA. Coronavirus (COVID-19) Update: FDA Announces Advisory Committee Meeting to Discuss Second COVID-19 Vaccine Candidate. FDA News Release, 30 November 2020. [Google Scholar]
  100. EMA. EMA receives application for conditional marketing authorisation of Moderna COVID-19 vaccine. News, 1 December 2020. [Google Scholar]
  101. Reuters. UK starts real-time review of Moderna’s COVID-19 vaccine candidate. Reuters, 28 October 2020. [Google Scholar]
  102. Reuters. Switzerland starts ‘rolling review’ of Moderna COVID-19 vaccine. Reuters, 13 November 2020. [Google Scholar]
  103. ABC. US Food and Drug Administration authorises Moderna’s COVID-19 vaccine for emergency use. ABC News, 19 December 2020. [Google Scholar]
  104. HealthCanada (Ed.) Health Canada Authorizes Moderna COVID-19 Vaccine; Government of Canada: Ottawa, ON, Canada, 2020.
  105. EMA. EMA Recommends COVID-19 Vaccine Moderna for Authorisation in the EU; European Medicines Agency: Amsterdam, The Netherlands, 2021. [Google Scholar]
  106. Hussey, C. Israeli Ministry of Health Authorizes COVID-19 Vaccine Moderna for Use in Israel; Moderna: Cambridge, MA, USA, 2021. [Google Scholar]
  107. Hussey, C. Swissmedic Authorizes COVID-19 Vaccine Moderna for Use in Switzerland. Businesswire, 12 January 2021. [Google Scholar]
  108. Hussey, C. United Kingdom Medicines and Healthcare Products Regulatory Agency Authorizes Use of COVID-19 Vaccine Moderna; Moderna: Cambridge, MA, USA, 2021. [Google Scholar]
  109. Van Doremalen, N.; Lambe, T.; Spencer, A.; Belij-Rammerstorfer, S.; Purushotham, J.N.; Port, J.R.; Avanzato, V.A.; Bushmaker, T.; Flaxman, A.; Ulaszewska, M.; et al. ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature 2020, 586, 578–582. [Google Scholar] [CrossRef] [PubMed]
  110. Folegatti, P.M.; Ewer, K.J.; Aley, P.K.; Angus, B.; Becker, S.; Belij-Rammerstorfer, S.; Bellamy, D.; Bibi, S.; Bittaye, M.; Clutterbuck, E.A. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: A preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020, 396, 467–478. [Google Scholar] [CrossRef]
  111. ABCNews. Oxford says patient illness which forced pause to AstraZeneca coronavirus clinical trial ‘unlikely’ related to vaccine. ABCNews, 17 September 2020. [Google Scholar]
  112. Schwartz, L. Brazil Confirms Death of Volunteer in COVID-19 Vaccine Trial. MedScape, 21 October 2020. [Google Scholar]
  113. AstraZeneca. AZD1222 Clinical Trials Now Resumed Globally; AstraZeneca: Cambridge, MA, USA, 2020. [Google Scholar]
  114. Ramasamy, M.N.; Minassian, A.M.; Ewer, K.J.; Flaxman, A.L.; Folegatti, P.M.; Owens, D.R.; Voysey, M.; Aley, P.K.; Angus, B.; Babbage, G. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): A single-blind, randomised, controlled, phase 2/3 trial. Lancet 2020, 396, 1979–1993. [Google Scholar] [CrossRef]
  115. Duffy, K. AstraZeneca Admits the Best Results in Its COVID-19 Vaccine Study Came from a Dosing Error, and Experts Are Raising New Questions about the Shot; Business Insider: Sydney, Australia, 2020. [Google Scholar]
  116. MHRA (Ed.) Conditions of Authorisation for COVID-19 Vaccine AstraZeneca Agency; Government of United Kingdom: London, UK, 2021.
  117. PharmaLetter. Argentina Second to Back AstraZeneca’s COVID-19 Vaccine; The PharmaLetter: London, UK, 2020. [Google Scholar]
  118. VOA. Mexico Grants Emergency Approval to AstraZeneca-Oxford Coronavirus Vaccine. VOA News, 5 January 2021. [Google Scholar]
  119. AstraZeneca. AstraZeneca’s COVID-19 Vaccine Authorised in Five Other Countries; AstraZeneca: Cambridge, MA, USA, 2021. [Google Scholar]
  120. Voysey, M.; Clemens, S.A.C.; Madhi, S.A.; Weckx, L.Y.; Folegatti, P.M.; Aley, P.K.; Angus, B.; Baillie, V.L.; Barnabas, S.L.; Bhorat, Q.E. 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. [Google Scholar] [CrossRef]
  121. Tiede, A.; Sachs, U.J.; Czwalinna, A.; Werwitzke, S.; Bikker, R.; Krauss, J.K.; Donnerstag, F.; Weißenborn, K.; Höglinger, G.; Maasoumy, B. Prothrombotic immune thrombocytopenia after COVID-19 vaccine. Blood 2021. [Google Scholar] [CrossRef]
  122. Sah, R.; Shrestha, S.; Mehta, R.; Sah, S.K.; Raaban, A.R.; Dharma, K.; Rodriguez-Morales, A.J. AZD1222 (Covishield) vaccination for COVID-19: Experiences, challenges and solutions in Nepal. Travel Med. Infect. Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
  123. Román, G.C.; Gracia, F.; Torres, A.; Palacios, A.; Gracia, K.; Harris, D. Acute transverse myelitis (ATM): Clinical review of 43 patients with COVID-19-associated ATM and 3 post-vaccination ATM serious adverse events with the ChAdOx1 nCoV-19 vaccine (AZD1222). Front. Immunol. 2021, 12, 879. [Google Scholar] [CrossRef] [PubMed]
  124. Terpos, E.; Politou, M.; Ntanasis-Stathopoulos, I.; Karalis, V.; Merkouri, E.; Fotiou, D.; Gavriatopoulou, M.; Malandrakis, P.; Kastritis, E.; Trougakos, I.P. High Prevalence of Anti-PF4 Antibodies Following ChAdOx1 nCov-19 (AZD1222) Vaccination Even in the Absence of Thrombotic Events. Vaccines 2021, 9, 712. [Google Scholar] [CrossRef]
  125. BharatBiotech. COVAXIN—India’s First indigenous COVID-19 Vaccine. Available online: https://www.bharatbiotech.com/covaxin.html (accessed on 15 January 2021).
  126. Mohandas, S.; Yadav, P.D.; Shete, A.; Abraham, P.; Mohan, K.; Sapkal, G.; Mote, C.; Nyayanit, D.; Gupta, N.; Srinivas, V. Immunogenicity and protective efficacy of BBV152: A whole virion inactivated SARS CoV-2 vaccine in the Syrian hamster model. Res. Sq. 2020, 1, 1–25. [Google Scholar] [CrossRef]
  127. Yadav, P.; Ella, R.; Kumar, S.; Patil, D.; Mohandas, S.; Shete, A.; Bhati, G.; Sapkal, G.; Kaushal, H.; Patil, S. Remarkable immunogenicity and protective efficacy of BBV152, an inactivated SARS-CoV-2 vaccine in rhesus macaques. Res. Sq. 2020, 1, 1–17. [Google Scholar] [CrossRef]
  128. Ella, R.; Vadrevu, K.M.; Jogdand, H.; Prasad, S.; Reddy, S.; Sarangi, V.; Ganneru, B.; Sapkal, G.; Yadav, P.; Abraham, P.; et al. A Phase 1: Safety and Immunogenicity Trial of an Inactivated SARS-CoV-2 Vaccine-BBV152. medRxiv 2020, 1–34. [Google Scholar] [CrossRef]
  129. Ella, R.; Reddy, S.; Jogdand, H.; Sarangi, V.; Ganneru, B.; Prasad, S.; Das, D.; Raju, D.; Praturi, U.; Sapkal, G.; et al. Safety and immunogenicity clinical trial of an inactivated SARS-CoV-2 vaccine, BBV152 (a phase 2, double-blind, randomised controlled trial) and the persistence of immune responses from a phase 1 follow-up report. medRxiv 2020, 1–34. [Google Scholar] [CrossRef]
  130. BBCNews. Coronavirus: India approves vaccines from Bharat Biotech and Oxford/AstraZeneca. BBC News, 3 January 2021. [Google Scholar]
  131. 24GlobeNews. Bharat Biotech: Indian well being specialists voice their doubts, dismay on the hurried vaccine approval course of. 24 Globe News, 12 January 2021. [Google Scholar]
  132. FirstPost. Health experts voice doubts, concerns about hurried approval process for Bharat Biotech’s Covaxin. FirstPost, 12 January 2021. [Google Scholar]
  133. Frayer, L. India Prepares for Massive Vaccine Drive, But Some Fear It’s Moving Too Quickly. National Public Radio, 13 January 2021. [Google Scholar]
  134. Janssen. Clinical Trial COVID-19 Vaccine Candidate Underway; Janssen: Beerse, Belgium, 2021. [Google Scholar]
  135. WHO. Background Document on the Janssen Ad26.COV2.S (COVID-19) Vaccine; World Health Organization: Italy, Rome, 2021; Volume 1, p. 54. [Google Scholar]
  136. He, X.; Chandrashekar, A.; Zahn, R.; Wegmann, F.; Yu, J.; Mercado, N.B.; McMahan, K.; Martinot, A.J.; Piedra-Mora, C.; Beecy, S. Low-dose Ad26. COV2. S protection against SARS-CoV-2 challenge in rhesus macaques. Cell 2021. [Google Scholar] [CrossRef]
  137. Solforosi, L.; Kuipers, H.; Huber, S.K.R.; van der Lubbe, J.E.; Dekking, L.; Czapska-Casey, D.N.; Gil, A.I.; Baert, M.; Drijver, J.; Vaneman, J. Immunogenicity of one-and two-dose regimens of the Ad26. COV2. S COVID-19 vaccine candidate in adult and aged rhesus macaques. BioRxiv 2020. [Google Scholar] [CrossRef]
  138. Roozendaal, R.; Solforosi, L.; Stieh, D.; Serroyen, J.; Straetemans, R.; Wegmann, F.; Huber, S.K.R.; van der Lubbe, J.E.; Hendriks, J.; le Gars, M. SARS-CoV-2 binding and neutralizing antibody levels after vaccination with Ad26. COV2. S predict durable protection in rhesus macaques. bioRxiv 2021. [Google Scholar] [CrossRef]
  139. Van der Lubbe, J.E.; Huber, S.K.R.; Vijayan, A.; Dekking, L.; van Huizen, E.; Vreugdenhil, J.; Choi, Y.; Baert, M.R.; Feddes-de Boer, K.; Gil, A.I. Ad26. COV2. S-elicited immunity protects against G614 spike variant SARS-CoV-2 infection in Syrian hamsters and does not enhance respiratory disease in challenged animals with breakthrough infection after sub-optimal vaccine dosing. bioRxiv 2021. [Google Scholar] [CrossRef]
  140. Sadoff, J.; Le Gars, M.; Shukarev, G.; Heerwegh, D.; Truyers, C.; de Groot, A.M.; Stoop, J.; Tete, S.; Van Damme, W.; Leroux-Roels, I. Interim results of a phase 1–2a trial of Ad26. COV2. S Covid-19 vaccine. N. Engl. J. Med. 2021, 384, 1824–1835. [Google Scholar] [CrossRef] [PubMed]
  141. Sadoff, J.; Gray, G.; Vandebosch, A.; Cárdenas, V.; Shukarev, G.; Grinsztejn, B.; Goepfert, P.A.; Truyers, C.; Fennema, H.; Spiessens, B. Safety and efficacy of single-dose Ad26. COV2. S vaccine against Covid-19. N. Engl. J. Med. 2021, 384, 2187–2201. [Google Scholar] [CrossRef] [PubMed]
  142. FDA. FDA Issues Emergency Use Authorization for Third COVID-19 Vaccine. Available online: https://www.fda.gov/news-events/press-announcements/fda-issues-emergency-use-authorization-third-covid-19-vaccine (accessed on 17 December 2020).
  143. EMA. EMA Recommends COVID-19 Vaccine Janssen for Authorisation in the EU; European Medicines Agency: Amsterdam, The Netherlands, 2021. [Google Scholar]
  144. MHRA. Regulatory Approval of COVID-19 Vaccine Janssen; Medicines and Healthcare products Regulatory Agency: London, UK, 2021. [Google Scholar]
  145. Muir, K.-L.; Kallam, A.; Koepsell, S.A.; Gundabolu, K. Thrombotic thrombocytopenia after Ad26. COV2. S vaccination. N. Engl. J. Med. 2021, 384, 1964–1965. [Google Scholar] [CrossRef] [PubMed]
  146. Sadoff, J.; Davis, K.; Douoguih, M. Thrombotic thrombocytopenia after Ad26. COV2. S vaccination—response from the manufacturer. N. Engl. J. Med. 2021, 384, 1965–1966. [Google Scholar] [CrossRef] [PubMed]
  147. See, I.; Su, J.R.; Lale, A.; Woo, E.J.; Guh, A.Y.; Shimabukuro, T.T.; Streiff, M.B.; Rao, A.K.; Wheeler, A.P.; Beavers, S.F. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26. COV2. S vaccination, March 2 to April 21, 2021. JAMA 2021. [Google Scholar] [CrossRef] [PubMed]
  148. WHO (Ed.) Background Document on the Inactivated COVID-19 Vaccine BIBP Developed by China National Biotec Group (CNBG); World Health Organization: Rome, Italy, 2021; p. 23. [Google Scholar]
  149. Wang, H.; Zhang, Y.; Huang, B.; Deng, W.; Quan, Y.; Wang, W.; Xu, W.; Zhao, Y.; Li, N.; Zhang, J. Development of an inactivated vaccine candidate, BBIBP-CorV, with potent protection against SARS-CoV-2. Cell 2020, 182, 713–721. [Google Scholar] [CrossRef]
  150. Xia, S.; Zhang, Y.; Wang, Y.; Wang, H.; Yang, Y.; Gao, G.F.; Tan, W.; Wu, G.; Xu, M.; Lou, Z. Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBIBP-CorV: A randomised, double-blind, placebo-controlled, phase 1/2 trial. Lancet Infect. Dis. 2021, 21, 39–51. [Google Scholar] [CrossRef]
  151. Taylor, A. WHO grants emergency use authorization for Chinese-made Sinopharm coronavirus vaccine. The Washington Post, 2021; 1. [Google Scholar]
  152. WHO. WHO Lists Additional COVID-19 Vaccine for Emergency use and Issues Interim Policy Recommendations; World Health Organization: Rome, Italy, 2021. [Google Scholar]
  153. WHO (Ed.) Tracking SARS-CoV-2 Variants; World Health Organization: Rome, Italy, 2021; Volume 1, p. 33. [Google Scholar]
  154. Zhou, B.; Thao, T.T.N.; Hoffmann, D.; Taddeo, A.; Ebert, N.; Labroussaa, F.; Pohlmann, A.; King, J.; Steiner, S.; Kelly, J.N. SARS-CoV-2 spike D614G change enhances replication and transmission. Nature 2021, 592, 122–127. [Google Scholar] [CrossRef]
  155. Volz, E.; Hill, V.; McCrone, J.T.; Price, A.; Jorgensen, D.; O’Toole, Á.; Southgate, J.; Johnson, R.; Jackson, B.; Nascimento, F.F. Evaluating the effects of SARS-CoV-2 spike mutation D614G on transmissibility and pathogenicity. Cell 2021, 184, 64–75. [Google Scholar] [CrossRef]
  156. Korber, B.; Fischer, W.M.; Gnanakaran, S.; Yoon, H.; Theiler, J.; Abfalterer, W.; Hengartner, N.; Giorgi, E.E.; Bhattacharya, T.; Foley, B. Tracking changes in SARS-CoV-2 spike: Evidence that D614G increases infectivity of the COVID-19 virus. Cell 2020, 182, 812–827. [Google Scholar] [CrossRef] [PubMed]
  157. Deng, X.; Garcia-Knight, M.A.; Khalid, M.M.; Servellita, V.; Wang, C.; Morris, M.K.; Sotomayor-González, A.; Glasner, D.R.; Reyes, K.R.; Gliwa, A.S. Transmission, infectivity, and antibody neutralization of an emerging SARS-CoV-2 variant in California carrying a L452R spike protein mutation. MedRxiv 2021. [Google Scholar] [CrossRef]
  158. Wang, P.; Nair, M.S.; Liu, L.; Iketani, S.; Luo, Y.; Guo, Y.; Wang, M.; Yu, J.; Zhang, B.; Kwong, P.D. Antibody resistance of SARS-CoV-2 variants B. 1.351 and B. 1.1. 7. Nature 2021, 593, 130–135. [Google Scholar] [CrossRef]
  159. Edara, V.V.; Floyd, K.; Lai, L.; Gardner, M.; Hudson, W.; Piantadosi, A.; Waggoner, J.; Babiker, A.; Ahmed, R.; Xie, X. Infection and mRNA-1273 vaccine antibodies neutralize SARS-CoV-2 UK variant. medRxiv 2021. [Google Scholar] [CrossRef]
  160. Wang, P.; Casner, R.G.; Nair, M.S.; Wang, M.; Yu, J.; Cerutti, G.; Liu, L.; Kwong, P.D.; Huang, Y.; Shapiro, L. Increased resistance of SARS-CoV-2 variant P. 1 to antibody neutralization. Cell Host Microbe 2021, 29, 747–751.e744. [Google Scholar] [CrossRef]
  161. Garcia-Beltran, W.F.; Lam, E.C.; Denis, K.S.; Nitido, A.D.; Garcia, Z.H.; Hauser, B.M.; Feldman, J.; Pavlovic, M.N.; Gregory, D.J.; Poznansky, M.C. Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell 2021, 184, 2372–2383. [Google Scholar] [CrossRef]
  162. Edara, V.-V.; Lai, L.; Sahoo, M.; Floyd, K.; Sibai, M.; Solis, D.; Flowers, M.W.; Hussaini, L.; Ciric, C.R.; Bechnack, S. Infection and vaccine-induced neutralizing antibody responses to the SARS-CoV-2 B. 1.617. 1 variant. bioRxiv 2021. [Google Scholar] [CrossRef]
  163. PHE. Investigation of Novel SARS-CoV-2 Variant: Variant of Concern 202012/01, Technical Briefing 3; Public Health England: London, UK, 2020. [Google Scholar]
  164. Mwenda, M.; Saasa, N.; Sinyange, N.; Busby, G.; Chipimo, P.J.; Hendry, J.; Kapona, O.; Yingst, S.; Hines, J.Z.; Minchella, P. Detection of B. 1.351 SARS-CoV-2 Variant Strain—Zambia.MMWR Morb. Mortal. Wkly Rep. 2021, 70, 280–282. [Google Scholar] [CrossRef] [PubMed]
  165. Hirotsu, Y.; Omata, M. Discovery of a SARS-CoV-2 variant from the P. 1 lineage harboring K417T/E484K/N501Y mutations in Kofu, Japan. J. Infect. 2021, 82, 276–316. [Google Scholar] [CrossRef]
  166. Annavajhala, M.K.; Mohri, H.; Zucker, J.E.; Sheng, Z.; Wang, P.; Gomez-Simmonds, A.; Ho, D.D.; Uhlemann, A.-C. A novel SARS-CoV-2 variant of concern, B. 1.526, identified in New York. medRxiv 2021. [Google Scholar] [CrossRef]
  167. Hadfield, J.; Megill, C.; Bell, S.M.; Huddleston, J.; Potter, B.; Callender, C.; Sagulenko, P.; Bedford, T.; Neher, R.A. Nextstrain: Real-time tracking of pathogen evolution. Bioinformatics 2018, 34, 4121–4123. [Google Scholar] [CrossRef] [PubMed]
  168. Bernal, J.L.; Andrews, N.; Gower, C.; Gallagher, E.; Simmons, R.; Thelwall, S.; Tessier, E.; Groves, N.; Dabrera, G.; Myers, R. Effectiveness of COVID-19 vaccines against the B. 1.617. 2 variant. medRxiv 2021. [Google Scholar] [CrossRef]
  169. Abu-Raddad, L.J.; Chemaitelly, H.; Butt, A.A. Effectiveness of the BNT162b2 Covid-19 Vaccine against the B. 1.1. 7 and B. 1.351 Variants. N. Engl. J. Med. 2021, 385, 187–189. [Google Scholar] [CrossRef] [PubMed]
  170. Huang, B.; Dai, L.; Wang, H.; Hu, Z.; Tan, W.; Gao, G.F.; Yang, X. Neutralization of SARS-CoV-2 VOC 501Y. V2 by human antisera elicited by both inactivated BBIBP-CorV and recombinant dimeric RBD ZF2001 vaccines. BioRxiv 2021. [Google Scholar] [CrossRef]
  171. Wu, K.; Werner, A.P.; Koch, M.; Choi, A.; Narayanan, E.; Stewart-Jones, G.B.; Colpitts, T.; Bennett, H.; Boyoglu-Barnum, S.; Shi, W. Serum neutralizing activity elicited by mRNA-1273 vaccine. N. Engl. J. Med. 2021, 384, 1468–1470. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The phylogeny tree of SARS-CoV-2 variant strains. The figure depicts all discovered variant strains obtained from 3916 genome samples between December 2019 till July 2021 and their prevalence across the globe. The real-time data to prepare this phylogeny tree was obtained from [167].
Figure 1. The phylogeny tree of SARS-CoV-2 variant strains. The figure depicts all discovered variant strains obtained from 3916 genome samples between December 2019 till July 2021 and their prevalence across the globe. The real-time data to prepare this phylogeny tree was obtained from [167].
Viruses 13 01397 g001
Table 1. Current COVID-19 vaccine candidates.
Table 1. Current COVID-19 vaccine candidates.
Vaccine TypeCandidate VaccineDevelopersTrial PhaseNumber of Virologically Confirmed Symptomatic Cases of COVID-19Efficacy Compared to Placebo for the Prevention of SARS-CoV-2Efficacy of Vaccine Against Severe and non-Severe COVID-19Efficacy: Seroconversion RatesAssess Humoral ImmunogenicitySafety and Immunogenicity of a Booster Dose
Inactivated virusCoronaVacSinovac Research and Development Co., Ltd4XXXXXX
Inactivated virusVero cellSinopharm + China National Biotec Group Co + Wuhan Institute of Biological Products3XX XXX
Inactivated virusBBIBP-CorVSinopharm + China National Biotec Group Co + Beijing Institute of Biological Products4XXXXXX
Viral vector (Non-replicating)AZD1222AstraZeneca + University of Oxford4X XXXX
Viral vector (Non-replicating)Recombinant novel coronavirus vaccine (Adenovirus type 5 vector)CanSino Biological Inc./Beijing Institute of Biotechnology4XXXXX
Viral vector (Non-replicating)Gam-COVID-VacGamaleya Research Institute; Health Ministry of the Russian Federation3X XXX
Viral vector (Non-replicating)Ad26.COV2.SJanssen Pharmaceutical4XXXXXX
Protein subunitNVX-CoV2373Novavax3XXXXXX
RNA based vaccinemRNA-1273Moderna + National Institute of Allergy and Infectious Diseases (NIAID)4XXXXXX
RNA based vaccineBNT162b2 (Comirnaty)Pfizer/BioNTech + Fosun Pharma4XX XXX
Protein subunitCHO CellAnhui Zhifei Longcom Biopharmaceutical + Institute of Microbiology, Chinese Academy of Sciences3
RNA based vaccineCVnCoV VaccineCureVac AG3XXXXXX
Inactivated virusSARS-CoV-2 vaccine (vero cells)Institute of Medical Biology + Chinese Academy of Medical Sciences3XX XXX
Inactivated virusQazCovid-in®Research Institute for Biological Safety Problems, Rep of Kazakhstan3XX XXX
DNA based vaccineINO-4800Inovio Pharmaceuticals + International Vaccine Institute + Advaccine (Suzhou) Biopharmaceutical Co., Ltd2/3XXXXXX
DNA based vaccineAG0301-COVID19AnGes + Takara Bio + Osaka University2/3XX XXX
DNA based vaccinenCov vaccineZydus Cadila3XX XXX
Inactivated virusCovaxinBharat Biotech International Limited3XXXXXX
Inactivated virusInactivated SARS-CoV-2 vaccine (Vero cell)Shenzhen Kangtai Biological Products Co., Ltd.3 XXXXX
Protein subunitEpiVacCoronaFederal Budgetary Research Institution State Research Center of Virology and Biotechnology “Vector”3XXXXXX
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chong, W.C.; Chellappan, D.K.; Shukla, S.D.; Peterson, G.M.; Patel, R.P.; Jha, N.K.; Eri, R.D.; Dua, K.; Tambuwala, M.M.; Shastri, M.D. An Appraisal of the Current Scenario in Vaccine Research for COVID-19. Viruses 2021, 13, 1397. https://0-doi-org.brum.beds.ac.uk/10.3390/v13071397

AMA Style

Chong WC, Chellappan DK, Shukla SD, Peterson GM, Patel RP, Jha NK, Eri RD, Dua K, Tambuwala MM, Shastri MD. An Appraisal of the Current Scenario in Vaccine Research for COVID-19. Viruses. 2021; 13(7):1397. https://0-doi-org.brum.beds.ac.uk/10.3390/v13071397

Chicago/Turabian Style

Chong, Wai Chin, Dinesh K. Chellappan, Shakti D. Shukla, Gregory M. Peterson, Rahul P. Patel, Niraj Kumar Jha, Rajaraman D. Eri, Kamal Dua, Murtaza M. Tambuwala, and Madhur D. Shastri. 2021. "An Appraisal of the Current Scenario in Vaccine Research for COVID-19" Viruses 13, no. 7: 1397. https://0-doi-org.brum.beds.ac.uk/10.3390/v13071397

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop