2,790
Views
3
CrossRef citations to date
0
Altmetric
Review

Leveraging on the genomics and immunopathology of SARS-CoV-2 for vaccines development: prospects and challenges

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, , , ORCID Icon, , , , , , , ORCID Icon, , ORCID Icon, , ORCID Icon & ORCID Icon show all
Pages 620-637 | Received 21 May 2020, Accepted 11 Aug 2020, Published online: 16 Sep 2020

ABSTRACT

The incidence and case-fatality rates (CFRs) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, the etiological agent for Coronavirus Disease 2019 (COVID-19), have been rising unabated. Even though the entire world has been implementing infection prevention and control measures, the pandemic continues to spread. It has been widely accepted that preventive vaccination strategies are the public health measures for countering this pandemic. This study critically reviews the latest scientific advancement in genomics, replication pattern, pathogenesis, and immunopathology of SARS-CoV-2 infection and how these concepts could be used in the development of vaccines. We also offer a detailed discussion on the anticipated potency, efficacy, safety, and pharmaco-economic issues that are and will be associated with candidate COVID-19 vaccines.

Introduction

Severe Acute Respiratory Syndrome Coronavirus −2 (SARS-CoV-2) is the third highly pathogenic coronavirus that has affected the human race in the 21st century. As the incidence and case fatality rate (CFR) of SARS-CoV-2 infection, the etiological agent of Coronavirus Disease 2019 (COVID-19), continues to rise across more than 210 countries and territories, several preventive and control measures have been adopted to halt the spread of SARS-CoV-2 and minimize COVID-19 associated deaths. Essentially, all countries in the world have had SARS-CoV-2 infections, with over 30 million infected and about one million COVID-19-related deaths.Citation1 Several observers have attributed the low reported incidence rate of COVID-19 in Sub-Saharan Africa to under-diagnosis, probably due to inadequate molecular diagnostic capacity.Citation2 Although it may seem that both European and American countries have the worst CFRs associated with COVID-19, the relatively low CFR reported in Africa is not a true reflection of the reality. This is because the majority of African countries have limited or weak epidemiological system that can account for accurate COVID-19 data. This consequently reflects the grossly under-reported COVID-19 cases and associated mortality in Africa .Citation2

Besides the apparent scenarios of underreporting of COVID-19 data in Africa, another possible reason for this variation may be due to the differences in the proportions of elderly population in these continents, as age is a significant risk factor for developing symptomatic and severe form of COVID-19 .Citation3 High COVID-19 fatality rates have been observed in patients of ˃ 60 years, especially in countries with high average life expectancy .Citation4 Average life expectancy in most African countries ranges from 56.75 to 69.90 and 58.78 to 73.68 years for males and females, respectively .Citation5 In other continents, higher life expectancy is obtainable in America with 70.65 to 76.49 and 75.56 to 81.41 years for males and females, respectively; in Asia with 68.50 to 75.17 and 71.05 to 79.89 years for males and females, respectively; in Europe with 69.37 to 79.68 and 78.56 to 84.45 years for males and females, respectively .Citation5 This implies that countries in the Europe, America, and Asia with higher life expectancy have a relatively higher proportion of the elderly population compared to most African countries. Consequently, it will be expected that these countries with a comparatively higher proportion of elderly citizens to have a higher rate of COVID-19-related mortality due to the vulnerability of the elderly individuals to severe COVID-19 .Citation3

Specifically, age alongside other risk factors such as metabolic, immunologic, respiratory, and oncogenic disorders are determinants of the severity of SARS-CoV-2 infection .Citation6 Aside from these, the genetic makeup of SARS-CoV-2 and host immune response are other vital determinants that contribute to SARS-CoV-2 virulence .Citation7 Hence, understanding these features are crucial to predict future transmission dynamics of SARS-CoV-2 infection, immune protection against re-infection, antiviral, and vaccines development .Citation8

There are ongoing scientific investigations, which seek to elucidate the structural basis of SARS-CoV-2 pathology and attempt to predict its transmission dynamics .Citation9 Mathematical models have been used to determine the transmission rate of SARS-CoV-2. The transmission rate can be referred to as the R0, a measure of the transmissibility of SARS-CoV-2. R0 predicts how many people an infected person can transmit SARS-CoV-2 in a population with no prior immunity to the pathogen. Generally, the higher the R0, the more contagious the pathogen. An R0 of < 1 means that the outbreak dies out, while R0 > 1 means the infection will continue to spread .Citation9

Genetic analysis suggests that SARS-CoV-2 is related to SARS-CoV-1 .Citation10 Early mathematical modeling reports revealed R0 value range of 2.0 to 3.9 for SARS-CoV-2 as compared to the R0 values for SARS-CoV-1 (1.7-1.9) and MERS (<1) .Citation11-16 This explains why SARS-CoV-2 is more contagious than SARS-CoV-1 and MERS-CoV. Unlike the R0, the effective reproduction number (RE) of SARS-CoV-2 is influenced by the level of an individual’s immunity and behavioral changes in the absence of vaccines, anti-viral agents or public health interventions .Citation9

Currently, the only available means of controlling the spread of SARS-CoV-2 is through consistent adherence to physical distancing, personal hygiene, strict quarantine with monitored self-isolation of infected persons, and case contacts. Despite all these measures, the world continues to experience a sustained rise in new cases of SARS-CoV-2. Hence, most scientists arguably believe that vaccination will be the best way to overcome COVID-19 pandemic .Citation9,Citation16 For now, there is no available preventive vaccine, although, many have been approved for clinical trials. Furthermore, Remdesivir, a licensed coronavirus-specific antiviral drug produced by Gilead is now available in developed countries. However, it’s too expensive for use in developing countries .Citation16 For a COVID-19 vaccination program to be successfully accepted and implemented in every nation, it must be safe, potent, accessible, affordable, and equity in its distribution must be ensured. Hence, this study critically reviewed the latest scientific advancement in genomics, replication pattern, pathogenesis and immunopathology of SARS-CoV-2 infection and how these concepts could be used in the development of vaccine models. Also, we proffer a detailed narrative on the anticipated potency, efficacy, safety, and pharmaco-economic issues that are and will be associated with candidate COVID-19 vaccines.

Genomic Organization of SARS-CoV-2

Coronaviruses are enveloped viruses possessing positive-sense RNA genome that ranges between 26 and 32 kb in size .Citation17 They can be categorized into four genera, namely Alpha, Beta, Gamma, and Delta Coronaviruses .Citation18 SARS-CoV-2 belongs to the Beta genera of coronaviruses. It has a genome of about 29.9 kb in size and contains 15 genes, including the S gene which codes for the S-protein found on the surface of the envelope .Citation19 The RNA genome of SARS-CoV-2 encodes a large non-structural polyprotein (ORF1 a/b) () .Citation18 The first Open Reading Frame (ORF1 a/b), which is about two-third of the RNA genome, is then translated into ppla and pplab polyproteins .Citation19 These polyproteins are cleaved by proteolytic enzymes to produce 16 non-structural proteins (NSPs), which form the viral replicase transcriptase complex .Citation18 The NSPs of SARS-CoV-2 functions by forming double-membrane vesicles where viral transcription and replication occur using membranes originating from the rough endoplasmic reticulum.Citation18 The remaining one-third of the RNA genome, ORF 3A, ORF 6, ORF 7, ORF 8, ORF 9 code for four structural proteins and five accessory proteins .Citation20,Citation21 The four structural proteins which play significant roles in SARS-CoV-2 virion assembly and infection consist of the surface spike glycoprotein (S), the membrane protein (M), an envelope protein (E), and the nucleocapsid protein (N) Citation20,Citation21 (). The spike surface glycoprotein (S) which is the primary inducer of the immune response, primarily mediates invasion of the host cell by binding to a receptor referred to as angiotensin-converting enzyme 2 (ACE-2) which is located on the surface of the host cell membrane .Citation22 Apart from ACE2, CD147 has also been described as another route of viral entry .Citation23

Figure 1. Genomic organization of SARS-CoV-2

UTR = Untranslated Region: ORF = Open Reading frame: M = Membrane protein: N = Nucleocapsid: E = Envelop protein: S= Spike protein
Figure 1. Genomic organization of SARS-CoV-2

Indeed, RNA viruses, including SARS-CoV-2, have high mutation rates which are significantly correlated with enhanced virulence and evolvability .Citation24 At proteomic level, amino acid substitutions have been reported in NSP2, NSP3 and S protein .Citation21

Various researchers have investigated mutation hotspots on the genomic sequence of SARS-CoV-2. Pachetti et al evaluated 220 complete genome sequences from four geographic areas, including Asia, Oceania, Europe, North America. Mutations were reportedly found on sequences belonging ORF1ab (1397 nsp2, 2891 nsp3, 14408 RdRp, 17746 and 17857 NSP143, 18060 NSP14), S (23403 spike protein) and ORF9a (28881, nucleocapsid phosphoprotein) .Citation25 It was also observed that genomes from Europe and North America experienced the highest mutation frequency with a distinct mutation hotspot pattern when compared to other geographic areas .Citation25 In another comparative study by Yin Citation26 on 558 SARS-CoV-2 genome sequences from 33 countries and territories. They used single nucleotide polymorphisms that recognized mutations on the SARS-CoV-2 genomes due to the error-prone RNA-dependent RNA polymerase to reveal major mutations occurring in the S protein, RNA polymerase, RNA primase, and nucleoprotein which are critical proteins in the virus genome .Citation26

The first common single nucleotide polymorphism mutation in the viral genome was reportedly observed in the leader sequence (241C > U), this mutation coevolved with three other vital mutations (3037C > U, 14408C > U, 23403A > G) and consequentially caused amino acid mutations in NSP3, RNA primase, and spike glycoprotein, respectively .Citation26 Genetic analyses have also revealed that there are two major prevalent types: the Single Nucleotide Polymorphism (SNP) mutation in the nsp8 (28144 U > C) which resulted from mutation of amino acid leucine (L) to serine (S) that is responsible for the difference in the two prevalent types of the SARS-CoV-2 virus, the S type (8782C > U) and the L type (28144U > C) .Citation26,Citation27

Besides these, SNP mutation (23403A > G) also occurred in the genes encoding for spike protein (S protein: D614G). However, the function of this mutation is still under extensively study, although it could be significant in the affinity and binding strength of the virus to the ACE-2 receptor. Besides, it has been speculated that D614G strains could be more virulent, thus increasing the infectiousness of SARS-CoV-2 and severity of infected individuals, especially in places where this mutation is prominent .Citation28

Mutations in the RNA-dependent RNA polymerase (28144U > C, 28881G > A, 28882G > A, 28883G > C) were also reported. This could improve the fidelity of the virus concerning its lethality. Based on the findings of Yin et al, Citation26 the highest frequency of SNPs was concentrated in the S-glycoprotein (23403A > G) followed by NSP3 (3037C > U) and least in M-protein (27046C > U) .Citation26 Interestingly, another study suggested that NSP2 and NSP3 mutations play a significant role in virulence and differentiation mechanism of SARS-CoV-2 () .Citation29

Table 1. SARS-CoV-2 Genetic Mutations and their Implications on Vaccine development

Of interest, is the mutation in S-protein, which is significant in binding of the host cell (tropism) and also serves as a target of neutralizing antibodies. Mutation in the S-protein induces conformational changes, which probably led to the changes of the antigenicity of SARS-CoV-2 .Citation18 This has made scientists explore the possible differences between the host tropism and transmission rate of SARS-CoV-2. Also, genomic surveillance of SARS-CoV-2 has been used to determine the correlation of these mutations to virulence diversity and their implications on reinfection, immunity, and vaccines development .Citation34

SARS-CoV-2 mutation data might provide valuable insights for the development of efficient vaccines, antivirals and laboratory test assays and protocols. Efforts are being made all over the world to control the spread of COVID-19 and most importantly, to develop a potent vaccine. Understanding the genetic evolution and mutation course of SARS-CoV-2 would significantly aid in the development of efficient diagnostic assays, therapeutic drugs, and vaccines. As global efforts are being made to contain and decrease the incidence of COVID-19, it is essential to consider all the features of the circulating strains during the development of a polyvalent vaccine which would be worthy in the fight against the disease .Citation35

It is worthy of note that the sequences of 30,983 complete genomes from 80 countries situated in Africa, Asia, America, and Oceania, were analyzed by Alouane et al .Citation36 In comparison to the reference genome Wuhan-Hu-1 genome, the analysis revealed the occurrence of 3,206 variant sites with uniform distribution in all genome sequences analyzed .Citation36 A low frequency of mutations of about 182 mutations, with a prevalence of 5.67% was reportedly observed .Citation36 Nevertheless, hotspot mutations with a prevalence higher than 10% were observed on 14 different locations on the genome sequence, seven regions on ORF1ab gene (nsp2, nsp3, nsp6, nsp12, nsp13, nsp14 and nsp 15), 3 in the nucleocapsid protein and one each in the spike protein, RNA primase, ORF3a, and ORF8 respectively .Citation36 Also, 35 non-synonymous mutations (<1%) were identified in the receptor-binding domain (RBD) of the spike protein. In contrast, only four of the mutations could potentially enhance the binding affinity of the spike protein to the human ACE-2 receptor.

The corroborative findings of the phylogenetic analysis and mutation observations demonstrated that proteomic divergence of analyzed strains in comparison to the reference is insignificant. Therefore, the dawdling mutation rate in SARS-CoV-2 genomes makes the possibility of developing an effective global vaccine .Citation36

Pathology and Immunopathological Response to SARS-CoV-2 Infection

Despite the controversies about the origin of SARS-CoV-1, several studies were able to trace its source to a zoonotic origin where COVID-19 patients were exposed to an animal market in Wuhan; a where live animals are sold. Subsequently, efforts have been made to search for a reservoir and/or intermediate hosts of SARS-CoV-2 from which the infection might have spread to humans. Initially, two snake species were identified as the possible reservoir hosts of SARS-CoV-2. However, the only consistently identified SARS-CoV-2 reservoirs are mammals and bats .Citation37 Notably, genomic sequencing of some SARS-CoV-2 isolates showed 88% nucleotide homology with two bat-derived-SARS-like Coronaviruses .Citation38 Thus, indicating bats as the most likely reservoir of SARS-CoV-2 .Citation38

Although SARS-CoV-2 may affect many organs of the human body, the respiratory tract is the primary site of infection .Citation9 Following coughing, sneezing, or talking by an infected person, the virus in the respiratory droplets could come in contact with the mucous membrane (especially the mucosa of nasal cavity and pharynx) of a susceptible host leading to respiratory tract colonization and/or infection .Citation39

A complex interaction occurs between the receptor-binding domain (RBD) of the S-protein on the virus surface and ACE-2 receptor on the host cell leading to the internalization of the SARS-CoV-2 through clathrin-mediated endocytosis .Citation39,Citation40 These ACE-2 receptors are expressed on the cell surfaces of the respiratory tract, heart, gastrointestinal tract, kidneys and testicles .Citation41,Citation42 The virus moves from the upper respiratory tract to the lungs where it multiplies, translocate into the blood causing viremia, and induces its hematogenous spread to organs that express ACE-2 receptors on their cell surfaces. This host–virus interaction represents the beginning of SARS-CoV-2 replication which could ultimately lead to multi-organ disorders .Citation43-45

The pathogenesis of SARS-CoV-2 is believed to be closely similar to that of SARS-CoV .Citation46 These pathological changes include; diffuse alveolar damage, pneumocytes desquamation, pulmonary edema with hyaline membrane formation, and cytomegaly of pneumocytes .Citation42,Citation47 The incubation period of the SARS-CoV-2 is between 6 and 14 days and 5.2 days on average .Citation48 Infected persons commonly develop fever, non-productive cough, difficulty breathing, myalgia, confusion, headaches, rhinorrhea, and less common chest pain, diarrhoea, nausea and vomiting .Citation49 Blood glucose level could be elevated in diabetic patients due to the virus-induced release of stress hormones .Citation50

There is currently limited information on the innate immune status of patients diagnosed with SARS-CoV-2 infection. Some studies show an early increase in serum levels of pro-inflammatory cytokines in SARS-CoV-2 infection, indicating a possible cytokine storm-mediated syndrome .Citation51,Citation52 Active innate immune response to viral infection relies heavily on type – I interferon (IFN) responses and their downstream cascade, resulting in viral replication that eventually leads to adaptive immune response. SARS-CoV-1 and SARS-CoV-2 have similar entry receptor known as ACE-2 receptor that is expressed predominantly in small subsets of lung cells called type – 2 alveolar cells .Citation53 SARS-CoVs infects macrophages and T cells, a central feature of SARS-CoVs mediated pathogenesis. There are a limited number of macrophages in the lungs that express ACE-2 .Citation53 If ACE-2 is minimally expressed by the possible target immune cells, other receptors may be utilized, or other modes of cell entry such as antibody-dependent enhancement may be utilized .Citation9

Innate immune cells identify invasion of the virus to mount an antiviral response, often by pathogen-associated molecular patterns (PAMPs). For RNA viruses like coronavirus, PAMPs are recognized by either the endosomal RNA receptors – TLR3 and TLR7, or the cytosolic RNA sensor – RIG-I/MDA5, in the form of viral genomic RNA or their intermediates during viral replication like dsRNA. This recognition event triggers downstream signaling cascade, i.e., NF- κB and IRF3, followed by their nuclear translocation. These transcription factors induce type-I IFN expression and other pro-inflammatory cytokines in the nuclei, and these initial responses comprise the first-line defense against viral infection at the entry site .Citation54

Type-I IFN via IFNAR, in turn, activates the JAK-STAT pathway, where JAK1 and TYK2 kinases phosphorylate STAT1 and STAT2. STAT1/2 form an IRF9 complex and travel together into the nucleus to initiate transcription of IFN-stimulated genes (ISGs) under the influence of IFN-stimulated response element (ISRE) containing promoters .Citation54 The successful engagement of type-I IFN response should be able to suppress viral replication and dissemination at an early stage. Coronaviruses use several strategies to interfere with the signals that lead to Type-I IFN production and/or downstream IFNAR signals. This damping technique is closely correlated with the severity of the disease .Citation55 At the induction stage of Type-I IFN, SARS-CoV interferes directly or indirectly with downstream signalling of RNA sensors such as the ubiquitination and degradation of MAVS and TRAF3/6 RNA sensor adapter molecules and the inhibition of IRF3 nuclear translocation .Citation56

In severe or lethal cases of SARS-CoV infection, an increased influx of neutrophils and monocyte-macrophages is consistently observed .Citation57,Citation58 Dysregulated type-I IFN and inflammatory monocyte-macrophages are the leading cause of lethal pneumonia seen in a mouse model of SARS-CoV infection .Citation55 Infiltrated myeloid cells with excessive type-I IFN are, therefore, the primary cause of lung dysfunction that adversely affect the outcome of the infection. Delayed type-I IFN is thought to compromise new viral regulation upon SARS-CoV infection leading to the influx of hyperinflammatory neutrophils and monocyte-macrophages. The rise in these innate immune cells contributes to the worsening consequences for infected the person that manifested in lung immunopathology .Citation9 Similar scenario with varying degrees of immune involvement is expected in SARS-CoV-2 infection. Ironically, virus transmission is reported to occur even in people diagnosed with asymptomatic conditions. This may be an indication of the innate immune response being suppressed during the early phase of SARS-CoV-2 infection .Citation9

Based on the cumulative evidence from the previous infection with coronavirus, innate immune response plays a critical role in pro- or anti-inflammatory responses, this can be used as an approach for immune intervention. Later, successful viral replication results in type-I IFN hyper-production and the release of neutrophils and macrophages, which are the critical sources of pro-inflammatory cytokines. During COVID-19, with related changes in total neutrophils and lymphocytes, SARS-CoV-2 is likely to cause delayed type-I IFN and viral control loss in an early infection .Citation9 Individuals vulnerable to COVID-19 are those with underlying diseases, including diabetes, hypertension, and cardiovascular disease .Citation59

The immune response by type 1 helper T cell (Th1) typically plays a dominant role in adaptive immunity to viral infections. Cytokine microenvironment produced by antigen-presenting cells dictates the direction of T cell responses. Generally, helper T cells orchestrate the overall adaptive response. In contrast, cytotoxic T cells are necessary to destroy cells infected with viruses. Humoral immune response, particularly the development of neutralizing antibodies, plays a protective role by limiting delayed-phase infection and will prevent potential reinfection. In SARS-CoV, the structural proteins, S, N, M, and E proteins were extensively mapped for both T and B cell epitopes () .Citation59

Table 2. Immunogenic protective epitopes of Coronaviruses, their Sequences and Gene Location

SARS-CoV infection caused seroconversion as early as day four after disease onset and was discovered in most patients by day 14 .Citation80 A few serological descriptions of SARS-CoV-2 have been published .Citation81 In a preliminary analysis, one patient showed peak-specific Immunoglobulin M (IgM) on day nine after the onset of disease and then switched to IgG by week 2 .Citation81 Interestingly, sera from confirmed COVID-19 patients showed some cross-reactivity with SARS-CoV, but not other coronaviruses. The T-cell response has been investigated extensively in SARS-CoV infections.

Studies have shown that responses to CD8 + T cells were more frequent with greater magnitude than responses to CD4+ T cells .Citation82 Strong T-cell responses were significantly associated with higher neutralizing antibodies, with more serum Th2 cytokines (IL-4, IL-5, IL-10) in more severe infections .Citation83

Available clinical data strongly suggested that the response to Th1 is key to effective control of SARS-CoV, Citation83 and possibly also valid for SARS-CoV-2 in particular .Citation84 Cautiously so, CD8+ T cell response needs to be well regulated so as not to cause lung pathology. Since most epitopes found for SARS-CoV are focused on the viral structural proteins, comparing those epitopes associated with SARS-CoV with those of SARS-CoV-2 would be informative. If one can distinguish overlapping epitopes among these coronaviruses, using convalescent serum from recovered SARS patients would be useful for application in passive immunization .Citation9

Pharmaco-Economics of SARS-COV-2 Vaccination

The current global economic crisis and recession due to the COVID-19 pandemic have led to decreased budgetary allocations to the health sector. This results in tremendous consequences on healthcare systems and the health of human lives, most especially geriatric individuals. Recently, the pandemic is rapidly transmitting at the community level, and there is yet to be an effective means to prevent the disease. Vaccines are, no doubt, highly pharmacological and economical means of controlling infections and preventing death .Citation85 The development of effective and cost-efficient vaccines against SARS-CoV-2 is urgently needed to diminish the global COVID-19 pandemic. Despite the slow processes in the vaccines, they are still essential and indispensable. One of such reasons is that the COVID-19 pandemic is still evolving globally with an increase in confirmed cases and casualties, and the level of infection is yet to be attained. Furthermore, the duration of the impact of COVID-19 on global health may assume a seasonal pattern which can persist for an extended period .Citation86

Over 40 pharmaceutical companies and intellectual institutions across several countries have disclosed their initiatives through ongoing programs as well as the level of progress based on the development of vaccines targeted towards SARS-CoV-2 .Citation87 Several vaccines are currently undergoing design and preparation. At the same time, efforts are being made to commence efficacy evaluation for some other candidate vaccines in animals and phase one clinical trials. The measurement of the impact of vaccination is not only restricted to public health terms, but also based on economic values: decreasing healthcare spending, reducing lost of labor force, and contributing to the development of the society and the economy .Citation88 Several developed countries have taken into consideration the pivotal role of vaccination in their respective preventive healthcare programs. The economic benefits of vaccination reveal preventive healthcare as one of the best measures to identify efficiency and profits .Citation89

Vaccination is an essential part of public health; thus, there is an urgent need to reserve an adequate percentage of the budget to guarantee vaccine accessibility and affordability to the populace. The economic and pharmacological values of SARS-CoV-2 needs to be effectively communicated to the government, policymakers, healthcare workers and the populace to instill political will and public acceptance. There is also the need to evaluate further not just the effect of SARS-CoV-2 vaccination on health and medical expenses, but also its broader economic impact on society.

Private pharmaceutical companies, foundations, national governments and donor funding agencies are financial drivers for vaccine research, development, and production .Citation90 Pandemic products are high-risk investments for pharmaceutical industries, thereby requiring public funding from governments, industries, and individuals .Citation91 Processes from vaccine discovery to approval for use cost billions of dollars and several years to complete with an average success rate of 60% .Citation92,Citation93

For instance, the coalition for epidemic preparedness innovations (CEPI) was established in 2016 to oversee stages of vaccine research and development . Since the start of COVID-19 pandemic, CEPI has committed $23 million for vaccine development, the United States is channeling $3 billion for coronavirus vaccine research and development (R & D), and the Canadian government has made available $2.7 million for vaccine research .Citation94 In another vein, the Prime Minister of Canada promised to invest $192 million into the development and production of SARS-CoV-2 vaccine and therapeutic interventions. This will be achieved through a partnership with indigenous pharmaceutical companies and research institutes .Citation95

It was projected that vaccine R & D will cost between $200 million and $1.5 billion. It will take many years for the vaccine to pass through stages of clinical trials to ascertain its safety and effectiveness .Citation96 Is this in response to the extended timeline projection of the CEPI vaccine development, different vaccine candidates in different stages of R & D are expected to be available for use within one year. Using different technologies such as modified virus-like particles, messenger RNA (mRNA), nanoparticles and DNA-based technology, there are currently 26 candidate vaccines in clinical evaluation and 139 in preclinical evaluations pharmaceutical companies and research institutes globally .Citation97 Of these, there are two major vaccine candidates developed by Moderna Therapeutics and Inovio Pharmaceuticals, which are mRNA based and DNA-based vaccines, respectively .Citation97

There are commitments from several national governments and donor funding agencies to finance the SARS-CoV-2 vaccine, the anticipated profit of $30 billion in 2019 and expected turnover projection of $1.4 trillion by the end of 2020. Furthermore, key Biopharmaceutical companies have indeed committed between US$100 million to some billion US$ in research and development of COVID-19 vaccines .Citation98 For instance, Pfizer is starting a Phase 3 trial, Merck is well along in COVID-19 vaccine development. GSK-Sanofi have partnered, are well along in development, and have negotiated a US$2 billion contract with the US government for vaccine supply .Citation99

Recently, Moderna Therapeutics received $483 million from biomedical advanced research and development agency (BARDA) to develop the mRNA-based vaccine .Citation99 This vaccine may not be affordable for low- and middle-income countries (LMICs) due to high cost involved in different development stages Citation100 and little financial strength to acquire this vaccine, amongst other government priorities for their citizens. Equitably, however, vaccines should be made accessible and affordable to all countries because LMICs have the potential of becoming disease epicenter of current and future outbreaks .Citation81,Citation101 In this vein, part of the funds contributed by Bill and Melinda Gates targeted towards combating COVID-19 have been set aside for LMICs .Citation81

Currently, there is no global agency charged with financing vaccine production. However, global stakeholders are considering large-scale vaccine manufacturing and procurement .Citation81,Citation100

Overview of Ongoing Vaccine Development

Vaccine development is typically a long, arduous process sometimes extending from 10 to 15 years Citation102 before final approval. Newer technologies and improvement in molecular and genetic techniques coupled with previous experiences, have yielded several strategies and platforms for vaccine development .Citation103 These platforms include – live vaccines with vector viruses, inactivated vaccines with viral proteins and gene-based vaccines .Citation104 Most of the platforms and strategies are novel or based on existing developing platforms. Consequently, Ain Shams University, Egypt is planning to start Phase 3 of testing the Bacilli Calmette-Guerin BCG vaccine in Egyptian healthcare workers on the frontline of the pandemic .Citation104,Citation105 The rationale behind this hypothesis is in two ways. Firstly, BCG vaccination was shown to confer long-term changes to the innate immunity dubbed “trained immunity” and has been known to be protective against several viruses such as Vaccinia, Influenza A, Herpes Simplex Virus 2 and respiratory syncytial virus via macrophage-dependent epigenetic changes .Citation105-108 The heterologous immunomodulatory effect of BCG on nonrelated conditions provide a promising avenue for investigation in relation to the COVID-19 pandemic. Secondly, countries with a BCG vaccination policy experienced a reduction in both COVID-19 cases and deaths worldwide as exemplified by several epidemiological studies in the European hotspot with inadequate or no BCG coverage .Citation109 This suggests that BCG vaccination may be a protecting factor against COVID-19. However, being observational studies, these data should be interpreted with caution and should be considered only as hypothesis due to the significant differences in collecting data relating to COVID-19 between countries .Citation108

The WHO reported about 149 COVID-19 vaccine candidates in its updated continuously list, however, Le et al. Citation110 projected the number to be more as there are some not listed by the WHO. Of these, some vaccine candidates have proceeded into clinical trials, while others are in various stages of animal testing and will be planning to enter clinical trials (). In collaboration with several countries, CEPI is currently supporting and funding a good number of such trials and vaccine development with proposed support of up to $2 billion .Citation116,Citation117 Two inactivated vaccines are in phase 1 clinical trials .Citation110

Table 3. Overview of Some Ongoing COVID-19 Vaccine Trials

Sinovac Biotech reported on 23rd April 2020 – one of their many first COVID-19 vaccines in development conferred protection to rhesus macaques from infection by SARS-CoV-2. The scientists gave two separate doses of the candidate vaccine to eight rhesus macaque monkeys. About 3 weeks after the two separate doses of vaccination, the monkeys’ lungs were challenged with SARS-CoV-2 virus, and none developed an infection .Citation118 On 24th April 2020, a potential coronavirus vaccine – ChAdOx1 developed at the University of Oxford began clinical trial on people .Citation119

Need for Thorough Efficacy, Potency and Safety Evaluations of SARS-CoV-2 vaccine

It is a rule of thumb that any vaccine after its design and preparation, must undergo efficacy and safety evaluation as well as a validation of its quality standard before entering clinical trials. Generally, three phases of clinical trials are necessary to evaluate the safety, potency, and efficacy of any vaccine .Citation19 A few vaccine candidates for SARS-CoV-2 have shown efficacy in in-vitro studies, however not many have progressed to randomized animal or human trials, hence may have limited use in tackling COVID-19 infection. Bao et al Citation120 recently reported a demonstration of recovery by SARS-CoV-2 challenged rhesus macaques from prior infection with an associated immunity to re-challenge. Early on, it was not clear if COVID-19 patients are at risk of reinfection during the recovery stage. In the search for an answer, rhesus macaques were re-challenged by Bao et al Citation120 with SARS-CoV-2 while at an early recovery phase from the initial infection. During the infection, some of the clinical observations were loss of weight, interstitial pneumonia and systemic viral dissemination majorly in respiratory and gastrointestinal tracts. Gladly, after the monkeys were re-challenged with an identical SARS-CoV-2 strain, viral dissemination, clinical manifestations and histopathological changes were not detected and were absent. Possibly, the protection of rhesus macaques from the reinfection by SARS-CoV-2 might have been caused by a notably enhanced neutralizing antibody response. The outcome of this experiment depicted that primary SARS-CoV-2 infection protects from subsequent reinfection. Therein, a proof of principle for using vaccines to stimulate immunity in humans was demonstrated .Citation120

Because of the extensive sequence diversity of coronavirus, sadly, limited cross-protection offered by most vaccine candidates have remained a clog in the wheel of developing efficacious vaccines against human coronavirus infections in recent decades .Citation87 Several vaccines candidates have been studied for previous CoVs such as SARS-CoV and MERS-CoV .Citation87

In addition to scaling up novel researches, some of these previous options can also be further evaluated for their safety, efficacy, and potency in tackling SARS-CoV-2 .Citation87 This will increase the chances of coming up with an effective vaccine to stem the COVID-19 tide.

Moreover, there has also been the question of how long the vaccine-induced immunity for SARS-CoV-2 lasts. There still exist several grey areas. For instance, experience from studies of human coronavirus infections causing the common cold suggested that immunity may be short-lived probably just for about 3 months or less and individuals stand a chance of re-infection within a short while after initial exposure .Citation120 On the flip side, longitudinal studies of antibody-mediated immunity from recovered SARS patients suggests that immunity after infection may last for 1-2 years .Citation121 Furthermore, cellular immunity is also long-lasting with prompt elicitation of cytotoxic T-cell responses to challenge 1-year post-recovery .Citation121 Bearing in mind that SARS-CoV-2 is a novel virus, there is a need to follow up the persistence of neutralizing antibody levels over time. Animal models are pre-requisites for the evaluation of a vaccine efficacy at the preclinical level. It is worrisome that just a handful of suitable animal models are currently on the radar for SARS-CoV-2 vaccine, especially given the virus is a new pathogen. Good enough, a human ACE-2 transgenic mouse, as well as primate macaques, both, proved suitable models recently .Citation87

A recent study in a bid to elucidate the pathogenicity of SARS-CoV-2 used an hACE-2 transgenic mouse infected with the virus .Citation120 Weight loss, as well as virus replication in lung tissue, were observed in the hACE-2 transgenic mice model. Also observed was the typical histopathology of interstitial pneumonia. Additionally, the bronchial epithelial cells, alveolar epithelia, as well as alveolar macrophages all showed viral antigens. In view of the above, the mouse model may, therefore, facilitate therapeutic drugs and vaccine development against SARS-CoV-2 .Citation120 Furthermore, a report from the Institute of Zoology of the Chinese Academy of Sciences shows the observation of similar symptoms with human infections in primate macaques as well as viral load changes and computed tomography (CT) images of the lungs. As a result, the primate macaques’ model, which has undergone validation, is already being used in drug trials and functional evaluation .Citation122

Another significant limitation to SARS-CoV-2-related research is highly contagious and pathogenic nature of the organism. To bridge the gap caused by the scarcity of animal models in the evaluation of the efficacy of SARS-CoV-2 vaccines, virus-like particles (VLPs) can serve as alternative tools. These virus-like particles (VLPs) are structures of many proteins that resembles in its organization and conformation, authentic native viruses, except that they lack viral genome. Application of these VLPs in similar studies has resulted in safe and effective positive outcomes .Citation35 The search for a potent SARS-CoV-2 vaccine is one with multiple approaches. Except in the case of live attenuated vaccines and live vector vaccines, adjuvants are pre-requisite for boosting immune response in the development of other types of vaccine.

In order to avoid the potential undesired effects of coronavirus vaccination, attention has also been given to the use of adjuvants. Adjuvants are substances that modify and boosts the efficacy and immunogenicity of the vaccines .Citation35 A study revealed that by using a chemical adjuvant (a delta inulin-based polysaccharide), lung immunopathology previously observed in mice after SARS challenge experiments were no longer seen .Citation35 After that, there was a hypothesis that the adjuvant helped to avert an exacerbated Th2-polarized response after challenge causative of the adverse effects. Above, therefore, is a scenario where an adjuvant-enhanced vaccine-induced protection as well as helped to minimize adverse effects from coronavirus vaccination .Citation35

In order to fast track the development of a SARS-CoV-2 vaccine, however, there is a need to place premium on adjuvants that have been widely used in other vaccines that have gained market approval .Citation53 However, suitable adjuvants should be selected based on vaccine design. This is bearing in mind that adjuvants can regulate the type of immune response. Additionally, in order to have a more efficacious and effective vaccine, a combination of different types of adjuvants may be necessary. At the moment, GSK has offered to make her established vaccine adjuvant platform technology available to facilitate the development of an effective vaccine against SARS-CoV-2. In addition, GSK has made this adjuvant available to Sanofi as well in their ongoing development program. To this end, agreements have been reached with the University of Queensland, Australia and Clover Biopharmaceutical, Inc. Citation87

Safety is an essential factor that must be prioritized in the course of any vaccine development with SARS-CoV-2 vaccine not being an exemption. There exists a dearth of information on the safety of vaccines against SARS-CoV-2. Currently, the S protein has proven to be the right candidate antigen for SARS-CoV-2 vaccine development. Sadly, apart from receptor binding and membrane fusion, the S protein has also shown other biological activities .Citation121 There have been increasing concerns about the induction of antigen-dependent enhancement (ADE) and other adverse effects derived from vaccination or natural re-exposure. ADE is a situation that sets in when non-neutralizing antibodies against proteins of a virus facilitate virus entry to host cells, as well as enhancing virus infectivity .Citation87 ADE has been already seen in cats vaccinated against a species-specific coronavirus (feline infectious peritonitis coronavirus) .Citation35

In the case of SARS-CoV-2, many studies have associated full-length S protein with severe liver damage, enhanced infection otherwise known as ADE and chances of viral spread within the central nervous system (CNS) .Citation19 These adverse effects are linked to S protein-specific antibodies .Citation87 Currently, there is no clarity on the particular domains and critical amino acids in S protein of SARS-CoV-2 that elicit the lung damage, and this will pose safety issues should the full-length S protein be used as an antigen .Citation87

There have been attempts to solve the S glycoprotein-induction of ADE by using truncated versions of the same protein. Researchers believed that the use of the receptor-binding domain (RBD) or the S1 subunit of the S glycoprotein could lead to the induction of neutralizing antibodies while avoiding ADE. The concept is to focus the induction of antibodies to relevant S regions for efficient virus neutralization and, by doing so, to avert the induction of potential non-neutralizing antibodies targeting other regions of the S protein. At the moment, there is seemingly a paucity of direct evidence that justifies this phenomenon .Citation35 More fundamental researches are needed to understand better the structure and function of the S protein will be of help in considering the selection of mutant residues in antigen design.

Prospects and Possible Challenges Associated With SARS-CoV-2 Vaccine Strategies

Owing to the SARS-CoV-2 pandemic, it is now of importance to explore the science of preventive and therapeutic vaccine as a tool to ameliorate the prevailing condition. It is worthy of note that various strategies have been employed in achieving this goal.

Most candidate vaccines are predicated on the induction of serum virus-neutralizing (VN) antibodies and systemic cell-mediated immune responses in the animal models as indicators of protection, but the correlates of immunity to COVID-19 in humans are unknown .Citation122

The pathogenesis of COVID-19 in humans is unclear as more research is underway, and as such vaccine strategies may need to be altered if the virus infects both the respiratory and intestinal tracts (pneumoenteric, like Bovine CoV) and is also shed in faeces. Oro-nasal vaccine prime and parenteral spike (S) vaccine booster may be optimal to prevent both enteric and respiratory infections as faecal and nasal shedding used for some animal CoV vaccines .Citation123

Several types of COVID-19 vaccines are proposed and are in various stages of development Citation124 with emphasis on criteria for safety and efficacy and duration of immunity. However, vaccines for pandemics also required rapid and speedy development with high production capacity .Citation125

Despite the novel platforms, SARS-CoV-2 vaccine development poses challenges to researchers. Although the virus’s S-protein is a promising immunogen for protection, optimizing antigen design is critical to ensure optimal immune response. Scientists and researchers are left in the middle of thought over the best approach. For instance, targeting the full-length protein or only the receptor-binding domain .Citation126 More so, correlates of protection may be inferred from experience with SARS and MERS vaccines, but they are not yet established. As with naturally acquired infection, the potential duration of immunity is unknown; similarly, whether single-dose vaccines will confer immunity is uncertain .Citation126

A recent study by Kuldeep et al Citation127 revealed the possibility of developing a universal CoV vaccine based on the similarity in T-cell epitopes of SARS and MERS-CoV that confirmed the potential for cross-reactivity among CoVs. It was noted that SARS-CoV-2 shares high genetic similarity with the SARS-CoV such that vaccines developed for SARS-CoV may exhibit cross-reactivity with SARS-CoV-2. However, comparative evaluation performed on full-length S=protein sequences of SARS-CoV-2 and SARS-CoV identified that the most variable residues were located in the S1 subunit of S protein, the critical CoV vaccine target. These findings suggest that the specific neutralizing antibodies that are effective against the SARS-CoV might not be effective against the SARS-CoV-2 .Citation127

Vaccine development is a lengthy and expensive process with high attrition, and it typically takes multiple candidates and many years to produce a licensed vaccine .Citation82 Consequent upon this fact, vaccine developers typically follow a linear algorithm, with multiple pauses for data analysis or manufacturing process checks. Vaccine development at pandemic state involves a fast start and many steps executed in parallel before confirming a successful outcome of another step, hence resulting in elevated financial risk .Citation92 Although classical inactive and attenuated vaccines are currently under investigation, challenges of speedy production of these virus vaccines in large quantities in cell culture under biosafety level-3 conditions abounds, and this limits their rapid deployment in the face of emerging pandemics .Citation128

Recombinant vector vaccines are in various stages of development for SARS or MERS, which might be a promising candidate for SARS-CoV-2 .Citation129 However, safety remains a significant concern for vaccines, therefore the need to investigate an adverse event or vaccine-induced immunopathology evident during candidate vaccine studies in animal models .Citation130 Eosinophil-related lung pathology was observed in mice vaccinated with formalin and ultraviolet-inactivated SARS vaccine, Citation124 or γ-irradiated inactivated MERS-CoV vaccine post-murine challenge. However, adding toll-like receptor agonists to an ultraviolet-inactivated SARS-CoV vaccine reduced the Th2-associated lung pathology .Citation131 In one ferret study, the MVA-S vaccine was associated with liver pathology, but this was not evident in other studies Citation131,Citation132

In tests of a SARS-CoV-2 S-protein candidate vaccine, antibody-dependent enhancement (ADE) of infection was reported post-challenge in hamsters, but not in mice using a spike (S) protein nanoparticle vaccine for MERS .Citation87 ADE has remained a long-term obstacle to the development of safe vaccines for feline infectious peritonitis, a systemic CoV infection of cats .Citation87 In feline infectious peritonitis-infected cats, ADE was triggered by antibody-mediated virus entry into macrophages via Ig Fc receptors .Citation53

The inconsistencies in these events among animal models necessitate an improved understanding of the biological basis for their occurrence and a better knowledge of human immunology to avoid similar reactions in humans because the animals used in the experiment shares the similar physiologic pattern with humans .Citation133 More so, experts generally agree that animal experiments and human clinical trials of candidate vaccines for COVID-19 should include a careful assessment of possible immune complications before releasing the vaccine to the public .Citation134

There are particular disadvantages of each vaccine strategy. Nucleic acid vaccines can be easier to design. However, DNA vaccines may not be as immunogenic enough to elicit host antibody defensive mechanism. In contrast, mRNA vaccines may be more unstable and challenging to predict their immunologic effect .Citation135 Additionally, viral vector vaccines and subunit vaccines generally exhibit higher safety profiles. They are more immunogenic, but they may have reduced efficacy due to pre-existing immunity to the vector, and subunit vaccines may be globally inaccessible due to its expensive cost of production .Citation136

The non-replicating adenovirus type 5 viral vector vaccine strategies by CanSino Biological, Inc., and Beijing Institute of Biotechnology is designed with a gene encoding the antigenic SARS-CoV-2 S=protein .Citation137 The viral vector delivers the spike protein gene into human cells, leading to the production of the protein that is designed to trigger an immune response. This robust strategy has scaled through preclinical trials. It has recently begun phase I clinical trials, with more than 100 participants aged 18 to 60 years. Despite this landmark of success, possible limitations exist. The type of adenovirus used in this candidate is a virus associated with the common cold, and, consequently, it is possible that people may already have been exposed to the viral vector, which could hinder human cell uptake of the viral vector or even lead to possible safety concerns if immunization triggers an inappropriate immune response .Citation138

The sequence for the vaccine strategy, called mRNA-1273, by Moderna, Inc. and National Institute of Allergy and Infectious Diseases (NIAID) was first identified in mid-January and contains genetic code for the SARS-CoV-2 spike protein such that on administration, the mRNA sequence prompts cells to start producing the antigenic SARS-CoV-2 S-protein, thus initiating an immune response. mRNA vaccines do not contain infectious material and are easier to develop and manufacture; this serves as its advantage .Citation139 Nevertheless, mRNA vaccine candidates, including those in development by Moderna, have not made it past phase III clinical trials .Citation140

DNA vaccine strategy called INO-4800 by Inovio Pharmaceuticals, was designed to be administered intradermally. Administration of this vaccine candidate requires the use of an electroporation device called CELLECTRA, which uses a small electrical current to make the human cells more permeable and thereby enables proper entry and incorporation of the DNA molecule into the cell .Citation141 This vaccine strategy is in its first clinical trial. It consists of plasmid DNA that, upon administration, prompts human cells to produce the antigenic SARS-CoV-2 S-protein .Citation142 While DNA vaccines carry certain advantages, including optimal development speeds and thermostability, past trials have shown that producing sufficient immunogenicity can be a challenge, thereby eliciting little immunological effect. Additionally, the administration can often require larger volumes of DNA vaccine compared to more traditional vaccine types, and it requires the use of an electroporation device, which can be inconvenient .Citation142,Citation143

Suitable Animal Models of COVID-19 Vaccine Candidates

Rhesus Macaques

Studies proved that humans share the same essential 12 amino acid residues present in ACE-2 with macaques .Citation143 Therefore, it has been chosen as a closer model system to humans, making it fit for COVID-19 vaccine testing. At a high SARS-CoV-2 viral load, macaques suffered pulmonary infiltrates when observed in radiographs .Citation144,Citation145 This matches with the milder symptoms seen in SARS-CoV-2 infected humans. Further, it was also observed that SARS-CoV-2 affected older macaques than the young ones, and this is also the same in human .Citation145 Consequently, rhesus macaques can be a promising animal model for efficacy and safety study of candidate COVID-19 vaccines.

Ferrets (Mustela putorius furo)

Ferrets are domesticated forms of European polecat that are commonly used in veterinary research. Ferrets cough, sneeze, and have similar lung morphology as humans .Citation146 These could support their use as an animal model for SARS-CoV-2. Vasan et al Citation147 have shown the susceptibility of ferrets to SARS-CoV-2. It is also interesting to note that SARS-CoV-2-infected ferrets could spread the virus with high transmission rates .Citation148 Importantly, since most of the countries are now under community transmission stage, ferrets may be a valuable animal model to study COVID-19 vaccines .Citation149

Mice

The use of mice as an animal model has economic benefits as it is cost-effective. However, mice have 11 of the 29 amino acids while for a rat, 13 of 29 amino acids differ in the binding region of ACE-2, respectively, when compared to humans; therefore, SARS-CoV-2 does not infect mice .Citation150 Perlman Laboratory developed humanized mouse models for SARS, where they incorporated human ACE-2 in mice .Citation151 Successful SARS-CoV-2 infection transgenic mice that express human ACE-2 were observed. The development of typical interstitial pneumonia with infiltration of significant macrophages and lymphocytes in the alveolar interstitium and the accumulation of macrophages in alveolar cavities were noted on histopathological examination .Citation152 This made it another choice animal model to study COVID-19 vaccines safety .Citation152

Syrian Hamster (also: Golden Hamster; Mesocricetus auratus)

Syrian hamster ACE2 shows maximum similarity to that of human, with differences in only 4 amino acids out of 29. This made this animal a model of choice for SARS-CoV-2 infection. Importantly, Syrian hamsters show similar immune responses as that of humans where IFN-gamma and pro-inflammatory cytokines are induced at day two post-infection. Consequently, there were a decline in type II interferon and IL6 and increase of TGF- β at day 7 post-infection .Citation150 Besides, Syrian hamsters also transmit infections to other non-infected Syrian hamsters .Citation150 It was also reported that Syrian hamsters showed similar SARS-CoV-2 pathogenesis and immune responses to humans. These make them suitable animal models for COVID-19 vaccine studies .Citation149

Conclusion

There are uncertainties on the extent and time of the decline of the COVID-19 pandemic. Hence, preventive vaccination measures remain the only hope to mitigate the spread of SARS-CoV-2 infections. The various SARS-CoV-2 vaccine development strategies show a promising future as much advances had been made in a minimal time frame. However, there is need for more research funding that will enable an extension of the ongoing clinical trials to all or most parts of the world, in order to ensure unified vaccine trails outcomes, regardless of racial and regional variation. It is highly recommended that comprehensive evaluation of the various vaccine platforms be considered in order to assure their potency, efficacy, safety, and global accessibility.

Authors’ Biographies

1.Idris Nasir Abdullahi: A Lecturer and Researcher with specialty in Medical Virology, subspecialty in Immunovirology, Virus Diagnostics and Epidemiology

2.Anthony Uchenna Emeribe: A lecturer, specialty in Chemical Pathology and Molecular Medicine

3.Hafeez Adekola Aderinsayo: A PhD student in Medical Microbiology

4.Sharafudeen Dahiru Abubakar: A Lecturer and specializes in Molecular Pathology

5.Amos Dangana: Chief Medical Laboratory Scientist

6.Halima Ali Shuwa: A PhD student in Immunology

7.Sunday Theophilus Nwoba: A PhD Student

8.Jelili Olaide Mustapha: A Medical Laboratory Technologist

9.Muyideen Titilope Haruna: A PhD Student

10.Kafayat Adepeju Olowookere: Medical Laboratory Scientist

11.Olawale Sunday Animasaun: A Resident of NFETLP and Medical Laboratory Scientist (Medical Microbiology Specialty)

12.Charles Egede Ugwu: Medical Laboratory Scientist

13.Solomon Oloche Onoja: Medical Laboratory Scientist

14.Abdullahi Sani Gadama: Medical Doctor, PhD (Immunology) Student

15.Musa Mohammed: Medical Laboratory Scientist and MSc (Immunology) Student

16.Isa Muhammad Daneji: A lecturer, Physician and Medical Microbiologist

17.Dele Ohinoyi Amadu: Chief Medical Laboratory Scientist

18.Peter Elisha Ghamba: Medical Laboratory Scientist

19.Nkechi Blessing Onukegbe: Medical Laboratory Scientist

20.Muhammad Sagir Shehu: Medical Laboratory Scientist

21.Chiladi Isomah: A lecturer, Medical Laboratory Scientist and Medical Microbiologist

22.Adamu Babayo: Medical Laboratory Scientist

23.Abdurrahman El-fulaty Ahmad: A Lecturer, Immunologist and Specializes in Immunology of Infectious Diseases

Disclosure of potential conflicts of interest

None declared.

Contributors’ Statement

INA, AUE and HAA conceptualized and planned the study. All the authors conducted a literature search, compilation of data and wrote the first draft manuscript. All authors read and approved the final manuscript for intellectual content before submission. INA is responsible for the overall content as guarantor.

Additional information

Funding

None received

References

  • Worldometers.info. Situation Update Worldwide; Dove, Delaware, USA. 2020; Available from: https://www.worldometers.info/coronavirus/#countries Accessed July 10th, 2020.
  • Adepoju P. Africa’s struggle with inadequate COVID-19 testing. The Lancet Microbe. 2020 May;1(1):e12. doi:10.1016/S2666-5247(20)30014-8.
  • Le Couteur DG, Anderson RM, Newman AB. COVID-19 through the lens of gerontology. J Gerontol A Biol Sci Med Sci. 2020. doi:10.1093/gerona/glaa077.
  • Natale F, Ghio D, Tarchi D et al. COVID-19 cases and case fatality rate by age. Knowledge for policy. 2020 May 4th; https://ec.europa.eu/knowledge4policy/sites/know4pol/files/jrc120420_covid_risk_and_age.pdf
  • World Data. Average life expectancy by country. Worlddata.info. 2020. Accessed 16 August 2020. https://www.worlddata.info/life-expectancy.php.
  • Yang K, Sheng Y, Huang C, Jin Y, Xiong N, Jiang K, Lu H, Liu J, Yang J, Dong Y, et al.. Clinical characteristics, outcomes, and risk factors for mortality in patients with cancer and COVID-19 in Hubei, China: a multicentre, retrospective, cohort study. Lancet Oncol. 2020;21(7):904–13. doi:10.1016/S1470-2045(20)30310-7.
  • Felsenstein S, Herbert JA, McNamara PS, Hedrich CM. COVID-19: Immunology and treatment options. Clin Immunol. 2020;215:108448. doi:10.1016/j.clim.2020.108448.
  • Mousavizadeh L, Ghasemi S. Genotype and phenotype of COVID-19: their roles in pathogenesis. J Microbiol Immunol Infect. 2020. doi:10.1016/j.jmii.2020.03.022.
  • Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38(1):1–9. doi:10.12932/AP-200220-0772.
  • Wang H, Li X, Li T, Zhang S, Wang L, Wu X, Liu J. The genetic sequence, origin, and diagnosis of SARS-CoV-2. Eur J Clin Microbiol Infect Dis. 2020 Apr 24;1–7. doi:10.1007/s10096-020-03899-4.
  • Giordano G, Blanchini F, Bruno R, Colaneri P, Di Filippo A, Di Matteo A, Colaneri M. Modelling the COVID-19 epidemic and implementation of population-wide interventions in Italy. Nat Med. 2020;26(6):855–60. doi:10.1038/s41591-020-0883-7.
  • D’Arienzo M, Coniglio A. Assessment of the SARS-CoV-2 basic reproduction number, R0, based on the early phase of COVID-19 outbreak in Italy. Biosaf Health. 2020;2(2):57–59. doi:10.1016/j.bsheal.2020.03.004.
  • Zhao S, Lin Q, Ran J, Musa SS, Yang G, Wang W, Lou Y, Gao D, Yang L, He D. Preliminary estimation of the basic reproduction number of novel coronavirus (2019-nCoV) in China, from 2019 to 2020: a data-driven analysis in the early phase of the outbreak. Int J. Inf Dis. 2020;92:214–17. doi:10.1016/j.ijid.2020.01.050.
  • Read J, Bridgen JR, Cummings DAT, Ho A, Jewell CP. Novel coronavirus 2019-nCoV: early estimation of epidemiological parameters and epidemic predictions. medRxiv. Preprint at medRxiv doi:10.1101/2020.01.23.20018549 2020.
  • Petrosillo N, Viceconte G, Ergonul O, Ippolito G, Petersen E. COVID-19, SARS and MERS: are they closely related?. Clin Microb Infect. 2020;26:729–34. doi:10.1016j.cmi.2020.03.026.
  • Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y, Ren R, Leung KSM, Lau EHY, Wong JY Early Transmission Dynamics in Wuhan, China, of Novel Coronavirus–Infected Pneumonia. N Engl J Med [Preprint]. 2020a; Retrieved from: doi:10.1056/NEJMoa2001316. Accessed February 16, 2020. 382 13 1199–207
  • Su S, Wong G, Shi W, Liu J, Lai A, Zhou J,Liu W, Bi Y, Gao GF. Epidemiology, genetic recombination and pathogenesis of coronaviruses. Trends Microbiol. 2016;7:490–502. doi:10.1016/j.tim.2016.03.003.
  • Li H, Liu S, Yu X, Tang S, Tang C. Coronavirus disease 2019 (COVID-19): current status and future perspectives. Int J Antimicrob Agents. 2020;55(5):105951.
  • Guo YR, Cao QD, Hong ZS, Tan YY, Chen SD, Jin HJ,Tan KS, Wang DY, Yan Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak—An update on the status. Mil Med Res. 2020;7(11):1–10. doi:10.1186/s40779-020-00240-0.
  • Ramaiah A, Arumugaswami V. Insights into cross-species evolution of novel human coronavirus 2019-nCoV and defining immune determinants for vaccine development. BioRxiv. Epub. 2020; doi: doi:10.1101/2020.01.29.925867.
  • Wu A, Peng Y, Huang B, Ding X, Wang X, Niu P,Meng J, Zhu Z, Zhang Z, Wang J, Sheng J, Quan L, Xia Z, Tan W, Cheng G, Jiang T. Genome composition and divergence of the novel coronavirus (2019-nCoV) originating in China. Cell Host Microbe. 2020. doi:10.1016/j.chom.2020.02.001.
  • Liu C, Zhou Q, Li Y, Garner L, Watkins S, Carter L, Smoot J, Gregg AC, Daniels AD, Jervey S, et al.. Research and Development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases. ACS Cent Sci. 2020;6(3):315–31. doi:10.1021/acscentsci.0c00272.
  • Wang K, Chen W, Zhou Y-S, Lian J-Q, Zhang Z, Du P, et al.. SARS-CoV-2 invades host cells via a novel route: CD147-spike protein. BioRxiv. 2020.
  • Duffy S. Why are RNA virus mutation rates so damn high?. PLoS Biol. 2018;16(8):e3000003. doi:10.1371/journal.pbio.3000003.
  • Pachetti M, Marini B, Benedetti F, Giudici F, Mauro E, Storici P, Masciovecchio C, Angeletti S, Ciccozzi M, Gallo RC, et al.. Emerging SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. J Transl Med. 2020;18(1). doi:10.1186/s12967-020-02344-6.
  • Yin C. Genotyping coronavirus SARS-CoV-2: methods and implications. Genomics. published online ahead of print. 2020;112(5):3588–96. Apr 27 doi:10.1016/j.ygeno.2020.04.016.
  • Tang X, Wu C, Li X, Song Y, Yao X, Wu X,Duan Y, Zhang H,Wang Y, Qian Z, Cui J, Lu J. On the origin and continuing evolution of SARS-CoV-2. Natl Sci Rev. 2020;7(6):1012–13. doi:10.1093/nsr/nwaa036.
  • Eaaswarkhanth M, Al-Madhoun A, Al-Mulla F. Could the D614G substitution in the SARS-CoV-2 spike (S) protein be associated with higher COVID-19 mortality?. Int J Infect Dis. 2020; 96:459–60. doi:10.1016/j.ijid.2020.05.071.
  • Angeletti S, Benvenuto D, Bianchi M, Giovanetti M, Pascarella S, Cicozzi M. COVID-2019: the role of the nsp2 and nsp3 in its pathogenesis. J Med Virol. 2020. Epub doi:10.1002/jmv.25719
  • Jin X, Xu K, Jiang P, Lian J, Hao S, Yao H, Jia H, Zhang Y, Zheng L, Zheng N, et al.. Virus strain from a mild COVID-19 patient in Hangzhou represents a new trend in SARS-CoV-2 evolution potentially related to Furin cleavage site. Emerg. Microb. Infect. 2020;9(1):1474–88. doi:10.1080/22221751.2020.1781551.
  • Holland LA, Kaelin EA, Maqsood R, Estifanos B, Wu LI, Varsani A, Halden RU, Hogue BG, Scotch M, Lim ES, et al.. An 81-Nucleotide Deletion in SARS-CoV-2 ORF7a identified from sentinel surveillance in Arizona (January to March 2020). J Virol. 2020;94(14):14. doi:10.1128/JVI.00711-20.
  • van Dorp L, Acman M, Richard D, Shaw LP, Ford CE, Ormond L, Owen CJ, Pang J, Tan CCS, Boshier FAT, et al.. Emergence of genomic diversity and recurrent mutations in SARS-CoV-2. Infect Genet Evol. 2020;83:104351. doi:10.1016/j.meegid.2020.104351.
  • Happi C, Ihekweazu C, Oluniyi P SARS-CoV-2 genomes from Nigeria reveal community transmission, multiple virus lineages and spike protein mutation associated with higher transmission and pathogenicity - novel 2019 coronavirus/Genome Reports - Virological. 2020. Accessed 7 August 2020. https://virological.org/t/sars-cov-2-genomes-from-nigeria-reveal-community-transmission-multiple-virus-lineages-and-spike-protein-mutation-associated-with-higher-transmission-and-pathogenicity/494
  • Abdullahi IN, Emeribe AU, Ajayi OA, Oderinde BS, Amadu DO, Osuji AI. Implications of SARS-CoV-2 genetic diversity and mutations on pathogenicity of COVID-19 and biomedical interventions. J Taibah Univ Med Sci. 2020. doi:10.1016/j.jtumed.2020.06.005.
  • Padron-Regalado E. Vaccines for SARS-CoV-2: lessons from other coronavirus strains. Infect Dis Ther. 2020;23:1–20.
  • Alouane T, Laamarti M, Essabbar A, Hakmi M, Bouricha E, Chemao-Elfihri MW, Kartti  S, Boumajdi N, Bendani H, Laamarti R, Ghrifi F, Allam L. Genomic Diversity And Hotspot Mutations In 30,983 SARS-Cov-2 Genomes: Moving Toward A Universal Vaccine for the “confined virus”?. bioRxiv. 2020. preprint. doi:10.1101/2020.06.20.163188.
  • Bassetti M, Vena A, Giacobbe DR. The novel Chinese coronavirus (2019-nCoV) infections: challenges for fighting the storm. Eur J Clin Invest. 2020:e13209. doi:10.1111/eci.13209.
  • Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, et al.. Genomic characterization and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395(10224):565–74. doi:10.1016/S0140-6736(20)30251-8.
  • Yang P, Wang X. COVID-19: a new challenge for human beings. Nature. 2020. doi:10.1038/s41423-020-0407-X.
  • Wang H, Yang P, Liu K, Guo F, Zhang G, Jiang C. SARS coronavirus entry into host cells through a novel clathrin- and caveolae-independent endocytic pathway. Cell Res. 2008;18(2):290–301. doi:10.1038/cr.2008.15.
  • Zou X, Chen K, Zou J, Han P, Han Z. The single cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different organs vulnerable to Wuhan 2019-nCoV infection. Front Med. 2020;14(2):185-192.
  • Jin Y, Yang H, Ji W, Wu W, Chen S, Zhang W, et al.. Virology, epidemiology, pathogenesis and control of COVID-19. Viruses. 2020;12(4):372. doi:https.//doi.10.3390/v12040372.
  • Harmer D, Gilbert M, Borman R, Clark KL. Quantitative mRNA expression profiling of ACE2, a novel homologue of angiotensin converting enzyme. FEBS Lett. 2002;532(1–2):107–10. doi:10.1016/S0014-5793(02)03640-2.
  • Letko M, Marzi A, Munster V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat Microbiol. 2020;5:562–69. doi:10.1038/s41564-020-0688-y.
  • Lin L, Lu L, Cao W, Li T. Hypothesis for potential pathogenesis of SARS-CoV-2 infection – a review of immune changes in patients with viral pneumonia. Emerg Microb infect. 2020;9(1):727–32. doi:10.1080/22221751.2020.1746199.
  • Li X, Geng M, Peng Y, Meng L, Lu S. Molecular immune pathogenesis and diagnosis of COVID-19. J Pharm Anal. 2020:1–17. doi:https.//doi.10.1016/j.jpha.2020.03.001.
  • Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, Tai Y, Bai C, Gao T, Song J, Xia P, Dong J, Zhao J, Wang FS. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Resp Med. 2020. doi:10.1016/S2213-2600(20)30076-X.
  • Rothan HA, Byrareddy SN. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J Autoimmun. 2020. doi:https.//doi.10.1016/j.jaut.2020.102433..
  • 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.
  • Wang A, Chao W, Xu Z, Gu J. Timely blood glucose management for the outbreak of 2019 novel coronavirus disease (COVID-19) is urgently needed. Diabetic Res Clin Pract. 2020;162.
  • Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, LIT LCW, HUI DSC, CHAN MHM, CHUNG SSC, et al.. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol. 2004;136(1):95–103. doi:10.1111/j.1365-2249.2004.02415.x.
  • Mahallawi WH, Khabour OF, Zhang Q, Makhdoum HM, Suliman BA. MERS-CoV infection in humans is associated with a pro-inflammatory Th1 and Th17 cytokine profile. Cytokine. 2018;104:8–13. doi:10.1016/j.cyto.2018.01.025.
  • Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, et al.. A novel coronavirus from patients with pneumonia in China, 2019. New Engl J Med. 2020;382(8):727–33. doi:10.1056/NEJMoa2001017.
  • de Wit E, van Doremalen N, Falzarano D, Munster VJ. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol. 2016;14(8):523–34. doi:10.1038/nrmicro.2016.81.
  • Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39(5):529–39. doi:10.1007/s00281-017-0629-x.
  • Kindler E, Thiel V, Weber F. Interaction of SARS and MERS coronaviruses with the antiviral interferon response. Adv Virus Res. 2016;96:219–43.
  • Perlman S, Dandekar AA. Immunopathogenesis of coronavirus infections: implications for SARS. Nat Rev Immunol. 2005;5(12):917–27. doi:10.1038/nri1732.
  • Zumla A, Hui DS, Perlman S. Middle East respiratory syndrome. Lancet. 2015;386(9997):995–1007. doi:10.1016/S0140-6736(15)60454-8.
  • Liu WJ, Zhao M, Liu K, Xu K, Wong G, Tan W, Gao GF. T-cell immunity of SARS-CoV: implications for vaccine development against MERS-CoV. Antiviral Res. 2017;137:82–92. doi:10.1016/j.antiviral.2016.11.006.
  • Tsao YP, Lin JY, Jan JT, Leng CH, Chu CC, Yang YC, Chen SL. HLAA*0201 T-cell epitopes in severe acute respiratory syndrome (SARS) coronavirus nucleocapsid and spike proteins. Biochem Biophys Res Commun. 2020 2006;344(1):63–71. doi:10.1016/j.bbrc.2006.03.152.
  • Lv Y, Ruan Z, Wang L, Ni B, Wu Y. Identification of a novel conserved HLA-A*0201-restricted epitope from the spike protein of SARS-CoV. BMC Immunol. 2009;10:61. doi:10.1186/1471-2172-10-61.
  • Wang BM, Chen HB, Jiang XD, Zhang MH, Wan T, Li N, Zhou XY, Wu YF, Yang F, Yu YZ, et al.. Identification of an HLA-A*0201-restricted CD8(þ) T-cell epitope SSp-1 of SARS-CoV spike protein. Blood. 2004;104:200e206. doi:10.1182/blood-2003-11-3979.
  • Kohyama S, Ohno S, Suda T, Taneichi M, Yokoyama S, Mori M, Kobayashi A, Hayashi H, Uchida T, Matsui M. Efficient induction of cytotoxic T lymphocytes specific for severe acute respiratory syndrome (SARS)-associated coronavirus by immunization with surface-linked liposomal peptides derived from a non-structural polyprotein 1a. Antivir Res. 2009;84:168e177. doi:10.1016/j.antiviral.2009.09.004.
  • Doytchinova I, Flower D. The HLA-A2-supermotif: a QSAR definition. Organ Biomol Chem. 2003;1:2648e2654.
  • Huang J, Cao Y, Du J, Bu X, Ma R, Wu C. Priming with SARS CoV S DNA and boosting with SARS CoV S epitopes specific for CD4+ and CD8+ T cells promote cellular immune responses. Vaccine. 2007;25:6981e6991. doi:10.1016/j.vaccine.2007.06.047.
  • Du L, Zhao G, Lin Y, Sui H, Chan C, Ma S, He Y, Jiang S, Wu C, Yuen KY, et al.. Intranasal vaccination of recombinant adeno-associated virus encoding receptor-binding domain of severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein induces strong mucosal immune responses and provides long-term protection against SARSCoV infection. J Immunol. 2008;180:948e956.
  • Zhi Y, Kobinger GP, Jordan H, Suchma K, Weiss SR, Shen H, Schumer G, Gao G, Boyer JL, Crystal RG, et al.. Identification of murine CD8 T cell epitopes in codon-optimized SARS-associated coronavirus spike protein. Virol. 2005;335:34e45. doi:10.1016/j.virol.2005.01.050.
  • Zhao J, Zhao J, Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. JVirol. 2010;84:9318e9325. doi:10.1128/JVI.01049-10.
  • Poh WP, Narasaraju T, Pereira NA, Zhong F, Phoon MC, Macary PA, Wong SH, Lu J, Koh DR, Chow VT. Characterization of cytotoxic Tlymphocyte epitopes and immune responses to SARS coronavirus spike DNA vaccine expressing the RGD-integrin-binding motif. J MedVirol. 2009;81:1131e1139.
  • Rivino L, Tan AT, Chia A, Kumaran EA, Grotenbreg GM, MacAry PA, Bertoletti A. Defining CD8+ T cell determinants during human viral infection in populations of Asian ethnicity. J Immunol. 2013;191:4010e4019. doi:10.4049/jimmunol.1301507.
  • Ohno S, Kohyama S, Taneichi M, Moriya O, Hayash H, Oda H, Mori M, Kobayashi A, Akatsuka T, Uchida T, et al.. Synthetic peptides coupled to the surface of liposomes effectively induce SARS coronavirusspecific cytotoxic T lymphocytes and viral clearance in HLA-A*0201 transgenic mice. Vaccine. 2009;27:3912e3920. doi:10.1016/j.vaccine.2009.04.001.
  • Oh HL, Chia A, Chang CX, Leong HN, Ling KL, Grotenbreg GM, Gehring AJ, Tan YJ, Bertoletti A. Engineering T cells specific for a dominant severe acute respiratory syndrome coronavirus CD8 T cell epitope. JVirol. 2011;85:10464e10471. doi:10.1128/JVI.05039-11.
  • Ng OW, Chia A, Tan AT, Jadi RS, Leong HN, Bertoletti A, Tan YJ. Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection. Vaccine. 2016;34:2008e2014. doi:10.1016/j.vaccine.2016.02.063.
  • Cheung YK, Cheng SC, Sin FW, Chan KT, Xie Y. Induction of T-cell response by a DNA vaccine encoding a novel HLA-A*0201 severe acute respiratory syndrome coronavirus epitope. Vaccine. 2007;25:6070e6077. doi:10.1016/j.vaccine.2007.05.025.
  • Liu J, Wu P, Gao F, Qi J, Kawana-Tachikawa A, Xie J, Vavricka CJ, Iwamoto A, Li T, Gao GF. Novel immunodominant peptide presentation strategy: a featured HLA-A*2402-restricted cytotoxic T-lymphocyte epitope stabilized by intrachain hydrogen bonds from severe acute respiratory syndrome coronavirus nucleocapsid protein. JVirol. 2010;84:11849e11857. doi:10.1128/JVI.01464-10.
  • Blicher T, Kastrup JS, Buus S, Gajhede M. High-resolution structure of HLA-A*1101 in complex with SARS nucleocapsid peptide. Acta Crystallogr D Biol Crystallogr. 2005;61:1031e1040.
  • Roder G, Kristensen O, Kastrup JS, Buus S, Gajhede M. Structure of a SARS coronavirus-derived peptide bound to the human major histocompatibility complex class I molecule HLA-B*1501. Acta Crystallogr Sect F Struct Biol Cryst Commun. 2008;64:459e462. doi:10.1107/S1744309108012396.
  • Zhao J, Zhao J, Mangalam AK, Channappanavar R, Fett C, Meyerholz DK, Agnihothram S, Baric RS, David CS, Perlman S. Airway memory CD4(þ) T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity. 2016;44:1379e1391.
  • Tully, Claire M. Novel bivalent viral-vectored vaccines induce potent humoral and cellular immune responses conferring protection against stringent influenza A virus challenge. J Immunol. 2017;199(4):1333–41. doi:10.4049/jimmunol.1600939.
  • Liu W, Fontanet A, Zhang PH, Zhan L, Xin ZT, Baril L, ang F, Lv H, Cao W. Two-year prospective study of the humoral immune response of patients with severe acute respiratory syndrome. J Infect Dis. 2006;193(6):792–95. doi:10.1086/500469.
  • Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, Si HR, Zhu Y, Li B, Huang CL, Chen HD, Chen J, Luo Y, Guo H, Jiang RD, Liu MQ, Chen Y, Shen XR, Wang X, Zheng XS, Zhao K, Chen QJ, Deng F, Liu LL, Yan B, Zhan FX, Wang YY, Xiao GF, Shi ZL. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020. [Preprint]. doi:10.1038/s41586-020-2012-7.
  • Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CRRawlings SA, Sutherland A, Premkumar L, Jadi RS, Marrama D, de Silva AM, Frazier A, Carlin AF, Greenbaum JA, Peters B, Krammer F, Smith DM, Crotty S, Sette A. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020 Jun 25;181(7):1489–501. e15. doi:10.1016/j.cell.2020.05.015
  • Li CK, Wu H, Yan H, Ma S, Wang L, Zhang M, Tang X, Temperton NJ, Weiss RA, Brenchley JM, et al.. T cell responses to whole SARS coronavirus in humans. J Immunol. 2008;181(8):5490–500. doi:10.4049/jimmunol.181.8.5490.
  • Kumar S, Nyodu R, Maurya VK, Saxena SK. Host immune response and immunobiology of human SARS-CoV-2 infection. Saxena S. editor. Coronavirus Disease 2019 (COVID-19). Medical Virology:. 2020. From pathogenesis to disease control; Gateway East, Singapore: Springer; 43-53.
  • Remy V, Largeron N, Quilici S, Carroll S. The economic value of vaccination: why prevention is wealth. Value Health J Int Soc Pharmacoecon Outcomes Res. 2014;17:A450. doi:10.1016/j.jval.2014.08.1211.
  • Neher RA, Dyrdak R, Druelle V, Hodcroft EB, Albert J. Potential impact of seasonal forcing on a SARS-CoV-2 pandemic. MedRxiv. 2020. doi:10.1101/2020.02.13.20022806.
  • Zhang J, Zeng H, Gu J, Li H, Zheng L, Zou Q. Progress and prospects on vaccine development against SARS-CoV-2. Vaccines. 2020;8(2):153. doi:10.3390/vaccines8020153..
  • Leroy O, Geels M, Korejwo J, Dodet B, Imbault N, Jungbluth S. Roadmap for the establishment of a European vaccine R&D infrastructure. Vaccine. 2014;32:7021–24. doi:10.1016/j.vaccine.2014.08.014.
  • Rémy V, Largeron N, Quilici S, Carroll S. The economic value of vaccination: why prevention is wealth. J Mark Access Health Policy. 2015. 3:10.3402/jmahp.v3.29414. Published 2015Aug:12. doi:10.3402/jmahp.v3.29414.
  • Sim S, Jit M, Constenla D, Peters D, Hutubessy RA. Scoping review of investment cases for vaccines and immunization programs. Value Health. 2019;22(8):942–52. doi:10.1016/j.jval.2019.04.002.
  • Gates B. Responding to Covid-19 — A once-in-a-century pandemic?. New Engl J Med. 2020;382(18):1677–79. doi:10.1056/NEJMp2003762.
  • Gouglas D, Thanh Le T, Henderson K, Kaloudis A, Danielsen T, Hammersland NC, Robinson JM, Heaton PM, Røttingen JA. Estimating the cost of vaccine development against epidemic infectious diseases: a cost minimization study. Lancet Glob Health. 2018;6(12):e1386–e1396. doi:10.1016/S2214-109X(18)30346-2.
  • CBC. 2020. The Economics of Finding A Vaccine For COVID-19 | CBC Radio. [online] Available at: <https://www.cbc.ca/radio/costofliving/dispatches-from-the-small-businesses-struggling-with-covid-19-and-the-economics-of-creating-a-vaccine-1.5503847/the-economics-of-finding-a-vaccine-for-covid-19-1.5505056> [ Accessed 19 April 2020]
  • Abedi M Canada To Spend $192M On Developing COVID-19 Vaccine. [online] Global News. 2020; Available at: <https://globalnews.ca/news/6717883/coronavirus-canada-vaccine-spending/> [ Accessed April 20, 2020].
  • Dunn A The Wuhan Coronavirus Has Now Claimed More Lives Than SARS. Top Scientists Told Us It Could Take Years and Cost $1 Billion To Make A Vaccine to Fight the Epidemic …. [online] Business Insider. 2020; https://www.businessinsider.com/wuhan-coronavirus-vaccine-could-take-years-timeline-and-cost-2020-2. Accessed April 20, 2020.
  • Pang J, Wang M, Ang I, Tan S, Lewis R, Chen J, Gutierrez RA, Gwee SXW, Chua PEY, Yang Q, et al.. Potential rapid diagnostics, vaccine and therapeutics for 2019 novel coronavirus (2019-nCoV): a systematic review. J Clin Med. 2020;9(3):623. doi:10.3390/jcm9030623.
  • World Health Organization. Draft landscape of COVID-19 candidate vaccines as at 31st July, 2020. https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines Accessed 10th August, 2020
  • Pagliusi S, Jarrett S, Hayman B, Kreysa U, Prasad SD, Reers M, Hong Thai P, Wu K, Zhang YT, Baek YO, et al.. Emerging manufacturers engagements in the COVID −19 vaccine research, development and supply. Vaccine. 2020;38(34):5418–542375. doi:10.1016/j.vaccine.2020.06.022. Yamey G, Schäferhoff M, Pate M, Chawla M, Ranson K, Zhao F, Hatchett R, Wilder R. Funding the development and manufacturing of COVID-19 vaccines: Background paper for the World Bank/CEPI financing COVID-19 vaccine development consultation on February 20, 2020. The Center for Policy Impact in Global Health. Duke Global Working Paper Series: number 20, March 2020. Available at: http://centerforpolicyimpact.org/ourwork/our-publications/funding-development-and-manufacturing-of-covid-19-vaccines/Roy M. Moderna Receives $483 Million BARDA Award For COVID-19 Vaccine Development. [online] Financial Post. 2020; https://business.financialpost.com/pmn/business-pmn/moderna-receives-483-million-barda-award-for-covid-19-vaccine-development. Accessed April 22, 2020.
  • Lurie N, Saville M, Hatchett R, Halton J. Developing Covid-19 Vaccines at Pandemic Speed. New Engl J Med. 2020;382(21):1969–73. doi:10.1056/NEJMp2005630.
  • Hotez PJ, Bottazzi ME. Developing a Low-Cost and Accessible COVID-19 Vaccine for Global Health. Preprints. PLoS Negl Trop Dis. 2020;14(7):e0008548. doi:10.1371/journal.pntd.0008548
  • Bregu M, Draper SJ, Hill AV, Greenwood BM. Accelerating vaccine development and deployment: report of a royal society satellite meeting. Philos Trans R Soc B:Biol Sci. 2011;366(1579):2841–49. doi:10.1098/rstb.2011.0100.
  • Wiedermann U, Garner-Spitzer E, Wagner A. “Primary vaccine failure to routine vaccines: why and what to do?”. Hum Vaccin Immunother. 2016;12(1):239–43. doi:10.1080/21645515.2015.1093263.
  • vfa. Vaccines to protect against Covid-19, the new coronavirus infection. Verband Forschender Arzneimittelhersteller. May 16, 2020. Accessed 20 July 2020. https://www.vfa.de/de/arzneimittel-forschung/woran-wir-forschen/impfstoffe-zum-schutz-vor-coronavirus-2019-ncov
  • Netea MG, Joosten LAB, Latz E, Mills KHG, Natoli G, Stunnenberg HG, ONeill LAJ, Xavier RJ. Trained immunity: a program of innate immune memory in health and disease. Science. 2016;352(6284):aaf1098. doi:10.1126/science.aaf1098.
  • Arts RJW, Moorlag SJCFM, Novakovic B, Li Y, Wang SY, Oosting M, Kumar V, Xavier RJ, Wijmenga C, Joosten LAB, Reusken CBEM, Benn CS, Aaby P, Koopmans MP, Stunnenberg HG, van Crevel R, Netea MG. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe. 2018;23(1):89–100. e5. doi:10.1016/j.chom.2017.12.010.
  • Moorlag S. Non-specific effects of BCG vaccine on viral infections. Clin Microbiol Infect. 2019;25:1473–78. doi:10.1016/j.cmi.2019.04.020.
  • O’Neill LAJ, Netea MG. BCG-induced trained immunity: can it offer protection against COVID-19?. Nat Rev Immunol. 2020;20:335–37. doi:10.1038/s41577-020-0337-y.
  • O’Connor E, Teh J, Kamat AM, Lawrentschuk N. Bacillus Calmette Guérin (BCG) vaccination use in the fight against COVID-19 - what’s old is new again?. Future Oncol. 2020;16(19):1323–25. doi:10.2217/fon-2020-0381.
  • Le TT, Andreadakis Z, Kumar A, Román RG, Tollefsen S, Saville M The COVID-19 vaccine development landscape [Internet]. Nat Rev Drug Discov. Nature Publishing Group. 2020. Accessed 20 July 2020. https://www.ncbi.nlm.nih.gov/pubmed/32273591.
  • CEPI. IVI, INOVIO, and KNIH to partner with CEPI in a Phase I/ II clinical trial of INOVIO’s COVID-19 DNA vaccine in South Korea. 2020a. Accessed 20 July 2020. https://cepi.net/news_cepi/ivi-inovio-and-knih-to-partner-with-cepi-in-a-phase-i-ii-clinical-trial-of-inovios-covid-19-dna-vaccine-in-south-korea/.
  • Gao Q, Bao L, Mao H, Wang L, Xu K, Yang M, Li Y, Zhu L, Wang N, Lv Z, et al.. Development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020;369(6499):77–81. doi:10.1126/science.abc1932.
  • Gendon IUZ. Advantages and disadvantages of inactivated and live influenza vaccine. Vopr Virusol. 2004;49:4.
  • Broderick KE, Humeau LM. Enhanced Delivery of DNA or RNA Vaccines by Electroporation. Methods Mol Biol. 2017;1499:193-200. doi:10.1007/978-1-4939-6481-9_12.
  • Vartak A, Sucheck SJ. Recent advances in subunit vaccine carriers. Vaccines. 2016;4(2):12. doi:10.3390/vaccines4020012.
  • Yao H, Lu X, Chen Q, Xu K, Chen Y, Cheng L, Liu F, Wu Z, Wu H, Jin C, Zheng M, Wu N, Jiang C, Li L. Patient-derived mutations impact pathogenicity of SARS-CoV-2. MedRxiv. 2020. doi:10.1101/2020.04.14.20060160.
  • CEPI. 2020b. Netherlands and Switzerland join the search for COVID-19 vaccines. Accessed 20 July 2020. https://cepi.net/news_cepi/netherlands-and-switzerland-join-the-search-for-covid-19-vaccines/.
  • Science. COVID-19 vaccine protects monkeys from new coronavirus, Chinese biotech reports. Health, Coronavirus. 2020. Accessed 20 July 2020. https://www.sciencemag.org/news/2020/04/covid-19-vaccine-protects-monkeys-new-coronavirus-chinese-biotech-reports.
  • University of Oxford Covid-19 vaccine: everything we know so far. Telegram. 2020. Accessed 20 July 2020.https://www.telegraph.co.uk/global-health/science-and-disease/oxford-universitycoronavirusvaccine-covid/.
  • Bao LL, Deng W, Huang B, Gao H, Ren L, Wei Q. Reinfection could not occur in SARS-CoV-2 infected rhesus macaques. bioRxiv. 2020. preprint doi:10.1101/2020.03.13.990226
  • Hui DS, Azhar E, Madani TA, Ntoumi F, Kock R, Dar O, Ippolito G, McHugh TD, Memish ZA, Drosten C. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health—The latest 2019 novel coronavirus outbreak in Wuhan, China. Int J Infect Dis. 2020;91:264–66. IJID Off. Publications of International Society of Infectious Diseases.
  • Amanat A, Krammer F. SARS-CoV-2 Vaccines: Status Report. Immun. 2020 March 007. [Epub ahead of print]. doi:https://dx.doi.10.1016%2Fj.immuni.2020.03.007.
  • Linda JS Vaccines for COVID-19: perspectives, prospects, and challenges based on candidate SARS, MERS, and animal coronavirus vaccines. 2020. Accessed 20 July 2020. https://www.emjreviews.com/allergy-immunology/article/vaccines-for-covid-19-perspectives-prospects-and-challenges-based-on-candidate-sars-mers-and-animal-coronavirus-vaccines/.
  • World Health Organization. Draft landscape of Covid-19 candidate vaccines. 2020a; https://www.who.int/who-documents-detail/draft-landscape-of-covid-19-candidate-vaccines. Accessed May 19, 2020.
  • Rauch S. New vaccine technologies to combat outbreak situations. Front Immunol. 2018;9:1963. doi:10.3389/fimmu.2018.01963.
  • Jaimes JA, André NM, Chappie JS, Millet JK, Whittaker GR. Phylogenetic Analysis and Structural Modeling of SARS-CoV-2 Spike Protein Reveals an Evolutionary Distinct and Proteolytically Sensitive Activation Loop. J Mol Biol. 2020;432(10):3309-3325. doi:10.1016/j.jmb.2020.04.009.
  • Dhama K, Sharun K, Tiwari R, Dadar M, Malik YS, Singh KP, Chaicumpa W. COVID-19, an emerging coronavirus infection: advances and prospects in designing and developing vaccines, immunotherapeutics, and therapeutics. Hum Vaccin Immunother. 2020;16(6):1232-1238. doi:10.1080/21645515.2020.1735227.
  • Jordan B. Vaccination against infectious bronchitis virus: a continuous challenge. Vet Microbiol. 2017;206:137–43. doi:10.1016/j.vetmic.2017.01.002.
  • Schindewolf C, Menachery VD. Middle east respiratory syndrome vaccine candidates: cautious optimism. Viruses. 2019;11(1):e74. doi:10.3390/v11010074.
  • Jin H, Xiao C, Chen Z, Kang Y, Ma Y, Zhu K, Xie Q, Tu Y, Yu Y, Wang B, et al.. Induction of Th1 type response by DNA vaccinations with N, M, and E genes against SARS-CoV in mice. Biochem Biophys Res Commun. 2005;328(4):979. doi:10.1016/j.bbrc.2005.01.048.
  • Shang W, Yang Y, Rao Y, Rao X. The outbreak of SARSCoV-2 pneumonia calls for viral vaccines. NPJ Vaccines. 2020;5(1):1–3. doi:10.1038/s41541-020-0170-0.
  • Lynne P Avoiding pitfalls in the pursuit of a COVID-19 vaccine. 2020; www.pnas.org/cgi/doi/10.1073/pnas.2005456117. Accessed April 18, 2020.
  • Laura G-R, Subbarao K Emerging Respiratory Viruses: Challenges and Vaccine Strategies. Laboratory of infectious diseases, national institute of allergy and infectious diseases, National institutes of health, Bethesda, Maryland 20892. 2006; https://cmr.asm.org/content/cmr/19/4/614.full.pdf. Accessed April 27, 2020.
  • Ricke D, Malone R. Medical countermeasures analysis of 2019-nCoV and vaccine risks for antibody-dependent enhancement (ADE). SSRN Electronic Journal. 2020. v1 doi:10.20944/preprints202003.0138
  • Coalition for Epidemic Preparedness Innovations (CEPI). CEPI collaborates with the Institut Pasteur in a consortium to develop COVID-19 vaccine. 2020c; https://cepi.net/news_cepi/cepi-collaborates-withthe-institut-pasteur-in-a-consortium-to-develop-covid-19-vaccine/. Accessed April 19, 2020.
  • Craven J COVID-19 vaccine tracker and regulatory focus. 2020;https://www. raps.org/news-and-articles/news-articles/2020/3/covid-19-vaccine-tracker. Accessed April 18, 2020.
  • CanSinoBIO. CanSinoBIO’s investigational vaccine against COVID-19 approved for phase 1 clinical trial in China. 2020; http://www.cansinotech.com/homes/article/show/56/153.html. Accessed April 18, 2020.
  • INOVIO. Inovio accelerates timeline for COVID-19 DNA vaccine INO-4800. 2020; http://ir.inovio.com/news-and-media/news/press-release-details/2020/Inovio-Accelerates-Timeline-for-COVID-19-DNA-VaccineINO-4800/default.aspx. Accessed April 18, 2020.
  • Kramps T, Elbers K. RNA vaccines: methods and protocols. 2017. New York, NY: Humana Press. doi:10.1007/978-1-4939-6481-9.
  • Cohen J. With record-setting speed, vaccine makers take their first shots at the new coronavirus. Science. March 31, 2020 2020. https://www.sciencemag.org/news/2020/03/recordsetting-speed-vaccine-makers-take-their-first-shots-newcoronavirus Accessed April 18, 2020
  • Garde D An updated guide to the coronavirus drugs and vaccines in development. 2020; https://www.statnews.com/2020/03/19/an-updated-guide-to-thecoronavirus-drugs-and-vaccines-in-development/. Accessed April 18, 2020.
  • Tregoning JS, Kinnear E. Using plasmids as DNA vaccines for infectious diseases. Microbiol Spectr. 2014;2(6):6. doi:10.1128/microbiolspec.PLAS-0028-2014.
  • Melin AD, Janiak MC, Marrone F, Arora PS, Higham JP. Comparative ACE2 variation and primate COVID-19 risk. 2020. bioRxiv. 2020. 04.09.034967 doi:10.1101/2020.04.09.034967
  • Munster VJ, Feldmann F, Williamson BN, van Doremalen N, Pérez-Pérez L, Schulz J, Meade-White K, Okumura A, Callison J, Brumbaugh B, Avanzato VA, Rosenke R, Hanley PW, Saturday G, Scott D, Fischer ER, de Wit E. Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature. 2020. doi:10.1038/s41586-020-2324-7.
  • Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, et al.. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science. 2020;eabb7314. doi:10.1126/science.abb7314.
  • Cameron MJ, Kelvin AA, Leon AJ, Cameron CM, Ran L, Xu L, Chu Y, Danesh A. Lack of innate interferon responses during SARS coronavirus infection in a vaccination and reinfection ferret model. PLoS One. 2020. doi:10.1371/journal.pone.0045842.
  • Callaway E. Labs rush to study coronavirus in transgenic animals - some are in short supply. Nature. 2020;579:183. doi:10.1038/d41586-020-00698-x.
  • Kim YI, Kim SG, Kim SE, Kim EH, Park SJ, Chang JH, Chang J-H, Kim EJ, Lee S, Casel MAB. Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe. 2020;27(5):704–709.e2. doi:10.1016/j.chom.2020.03.023.
  • Bijayeeta D, Hemal S, Goel S. Current global vaccine and drug efforts against COVID-19: pros and cons of bypassing animal trials. J Biosci. 2020;45:82. doi:10.1007/s12038-020-00053-2.
  • Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, Chan WM, Fan Z, Tsoi HW. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in golden Syrian hamster model: implications for disease pathogenesis and transmissibility. Clin Infect Dis. 2020. doi:10.1093/cid/ciaa325.
  • McCray PB, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L, Shi L, Netland J, Jia HP, Halabi C, Sigmund CD. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 2007;81(2):813–21. doi:10.1128/JVI.02012-06.
  • Bao LD, Huang B, Gao H, Liu J, Ren L, Wei Q, Yu P. The pathogenicity of SARS-CoV-2 in hACE2 transgenic Mice. Nature. 2020. doi:10.1038/s41586-020-2312.
  • Zhou M, Xu D, Li X, Li H, Shan M, Tang J, Wang M, Wang FS, Zhu X, Tao H, et al.. Screening and identification of severe acute respiratory syndrome-associated coronavirus-specific CTL epitopes. J Immunol. 2006;177(4):2138–45. doi:10.4049/jimmunol.177.4.2138..

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.