ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Review

Development of vaccines for SARS-CoV-2

[version 1; peer review: 2 approved]
PUBLISHED 17 Aug 2020
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

Abstract

COVID-19 emerged in late 2019 and has rapidly spread through many countries globally. The causative SARS-CoV-2 virus was not known until recently, and there is little or no natural immunity in human populations. There is an urgent need for vaccines and drugs to combat this new pandemic. In just a few months, huge efforts and resources by government, academia, and industry have been thrown into the race to develop a vaccine. This brief review summarizes and discusses the array of technologies being applied to vaccine development, highlighting the strengths and weaknesses of the various approaches.

Keywords

COVID-19, Vaccines, SARS-CoV-2

Introduction

The first quarter of 2020 has been plagued by the emergence of a novel coronavirus disease, COVID-19 or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 was first identified in Wuhan, China, and has since spread globally at an alarming rate, with over 200 countries and territories being infected1,2. In March 2020, SARS-CoV-2 was declared a global pandemic by the World Health Organization (WHO)3, and as of 18 July 2020, there are over 14 million confirmed cases with over 594,000 deaths worldwide. These figures are expected to rise (https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports)2. With the emergence of SARS-CoV-2, there are currently over 2,400 listed studies on the National Institute of Health database (https://clinicaltrials.gov/ct2/results?cond=COVID-19) aimed at identifying a solution to this pandemic. The development of a much-needed vaccine against SARS-CoV-2 is of utmost importance. To date, there are more than 100 vaccine candidates being developed by industry and academic institutions using a wide range of technologies including live attenuated, viral vectored, DNA/RNA-based, protein-based, and inactivated vaccines46. At the time of writing, 19 vaccines are recorded as being in clinical trials (Table 1)47. This article highlights various technologies employed in vaccine development for COVID-19, potential hurdles, and recent advances.

Table 1. Vaccine platforms and their current status.

Technology
employed
DeveloperType of vaccine candidateCurrent stage in
clinical evaluation
Non-
replicating
viral vector
University of Oxford/AstraZenecaChAdOx1-SPhase III
CanSino Biologics Inc and Beijing Institute of BiotechnologyAdenovirus type 5 vectorPhase II
Shenzhen Geno-Immune Medical InstituteLentivirus modification
Gamaleya Research InstituteAdeno-basedPhase I
GeoVax/BravoVaxModified Vaccinia Ankara (MVA)-encoded virus-like particle (VLP)Pre-clinical
Stabilitech Biopharma LtdOral Ad5 S
Janssen Pharmaceutical CompaniesAd26 (alone or witd MVA boost)
AltimmuneAdenovirus-based NasoVAX expressing SARS-CoV-2 Spike protein
GreffexAd5 S (GREVAX™ platform)
VaxartOral vaccine platform
DZIF – German Center for Infection ResearchMVA-S encoded
IDIBAPS-Hospital Clinic, SpainMVA-S
GreffexAd5 S (GREVAX™ platform)
Centro Nacional de Biotecnología (CNB-CSIC), SpainMVA expressing structural proteins
Reitdera/LEUKOCARE/UnivercellsReplication defective Simian adenovirus (gRAd) encoding SARS-CoV-2 S
Valo tderapeutics LtdAdenovirus-based + HLA-matched peptides
National Center for Genetic Engineering and Biotechnology
(BIOTEC)/GPO, Thailand
Inactivated flu-based SARS-CoV2 vaccine + adjuvant
University of ManitobaDendritic cell-based vaccine
University of Georgia/University of IowaParainfluenza virus 5 (PIV5)-based vaccine expressing tde Spike protein
Bharat Biotech/Thomas Jefferson UniversityRecombinant deactivated rabies virus containing S1
Massachusetts Eye and Ear/Massachusetts General Hospital/AveXisAdeno-associated virus vector (AAVCOVID)
ImmunityBio, Inc. and NantKwest, Inc.[E1-, E2b-, E3-] hAd5-COVID19-Spike/nucleocapsid
National Research Centre, EgyptInfluenza A H1N1 vector
RNAModerna/NIAIDLipid nanoparticle (LNP)-encapsulated mRNAPhase II
BioNTech/Fosun Pharma/PfizerThree LNP-mRNAsPhase I/II
CureVacmRNAPhase I
People's Liberation Army (PLA) Academy of Military Sciences/
Walvax Biotechnology
Imperial College LondonLNP-nCoVsaRNA
University of Tokyo/Daiichi-SankyoLNP-encapsulated mRNAPre-clinical
Fudan University/Shanghai JiaoTong University/RNACure
Biopharma
LNP-encapsulated mRNA cocktail encoding VLP/receptor-binding
domain (RBD)
China CDC/Tongji University/SterminamRNA
Arcturus Therapeutics/Duke-NUS Medical School
FBRI SRC VB VECTOR, Rospotrebnadzor, Koltsovo
Greenlight Biosciences
IDIBAPS-Hospital Clinic, Spain
BIOCADLiposome-encapsulated mRNA
Centro Nacional de Biotecnología (CNB-CSIC), SpainReplicating defective SARS-CoV-2-derived RNAs
Translate Bio/Sanofi PasteurLNP-mRNA
CanSino Biologics/Precision NanoSystems
Chula Vaccine Research Center/University of Pennsylvania
RNAimmune, Inc.Several mRNA candidates
eTheRNAmRNA in an intranasal delivery system
DNAINOVIO PharmaceuticalsDNA plasmid vaccine with electroporation devicePhase I/II
Cadila Healthcare LimitedDNA plasmid vaccinePhase I/II (not yet
recruiting)
Genexine ConsortiumDNA vaccine (GX-19)Phase I
Takis Biotech/Applied DNA Sciences/EvviVaxDNAPre-clinical
BioNet-AsiaDNA vaccine
Entos Pharmaceuticals
Mediphage Bioceuticals/University of WaterloomsDNA vaccine
Zydus CadilaDNA plasmid vaccine
Karolinska Institute/Cobra Biologics (OPENCORONA Project)DNA with electroporation
Chula Vaccine Research Center
Osaka University/AnGes/Takara BioDNA plasmid vaccine
Immunomic Therapeutics, Inc./EpiVax, Inc./PharmaJet, Inc.Plasmid DNA, needle-free delivery
SymvivobacTRL-Spike
Scancell/University of Nottingham/ Nottingham Trent UniversityDNA plasmid vaccine RBD and N
National Research Centre, EgyptDNA plasmid vaccine S, S1, S2, RBD, and N
InactivatedSinovacFormaldehyde-inactivated + alumPhase III (not yet
recruiting)
Beijing Institute of Biological Products/SinopharmInactivatedPhase I/II
Wuhan Institute of Biological Products/Sinopharm
Institute of Medical Biology, Chinese Academy of Medical SciencesPhase I
Osaka University/BIKEN/NIBIOHNUnknownPre-clinical
Sinovac/DynavaxInactivated + CpG 1018
Valneva/Dynavax
National Research Centre, EgyptInactivated whole virus
Beijing Minhai Biotechnology Co., Ltd.Inactivated
Research Institute for Biological Safety Problems, Rep of Kazakhstan
Live
attenuated
virus
Codagenix/Serum Institute of IndiaDeoptimized live attenuated vaccinesPre-clinical
Indian Immunologicals Limited/Griffith UniversityCodon de-optimization of live attenuated vaccine
UMC Utrecht/Radboud UniversityRecombinant BCG (rBCG) technology
Protein
subunit
NovavaxFull-length recombinant SARS CoV-2 glycoprotein nanoparticle
vaccine adjuvanted with Matrix (M)
Phase I/II
Clover Biopharmaceuticals Inc./GSK/DynavaxNative like trimeric subunit Spike protein vaccinePhase I
Anhui Zhifei Longcom Biopharmaceutical/Institute of Microbiology,
Chinese Academy of Sciences
Adjuvanted recombinant protein (RBD-dimer)
Vaxine Pty Ltd/MedytoxRecombinant Spike protein with Advax™ adjuvant
ExpreS2ion BiotechnologiesDrosophila S2 insect cell expression system VLPsPre-clinical
Osaka University/BIKEN/National Institutes of Biomedical
Innovation, Japan
VLP recombinant protein + adjuvant
Chulalongkorn University/GPO, ThailandRBD protein fused with Fc of immunoglobulin G + adjuvant
AdaptVac (PREVENT-nCoV consortium)Capsid-like particle
Helix Biogen Consult, Ogbomoso, and Trinity Immonoefficient
Laboratory, Ogbomoso, Oyo State, Nigeria
Subunit
WRAIR/USAMRIIDS protein
AJ Vaccines
EpiVax/University of Georgia
National Institute of Infectious Diseases, JapanS protein + adjuvant
Sanofi Pasteur/GSKS protein (baculovirus production)
University of VirginiaS subunit intranasal liposomal formulation with GLA/3M052 adjuvants
ImmunoPrecise/LiteVax BVSpike-based (epitope screening)
Vaxil BioPeptide
Flow Pharma Inc
IMV IncPeptide antigens formulated in lipid nanoparticle formulation
Generex/EpiVaxIi-Key peptide
EpiVaxProtein subunit EPV-CoV-19
National Research Centre, EgyptProtein subunit S, N, M and S1 protein
Heat Biologics/University of Miamigp-96 backbone
University of Queensland/GSK/DynavaxMolecular clamp stabilized Spike protein
Baylor College of MedicineS1 or RBD protein
iBio/CC-PharmingSubunit protein, plant produced
VIDO-InterVac, University of SaskatchewanAdjuvanted microsphere peptide
LakePharma, Inc.Nanoparticle vaccine
Baiya Phytopharm/Chula Vaccine Research CenterPlant-based subunit
(RBD-Fc + adjuvant)
Biological E LtdAdjuvanted protein subunit (RBD)
University of SaskatchewanAdjuvanted microsphere peptide
University of PittsburghMicroneedle arrays S1 subunit
Saint-Petersburg Scientific Research Institute of Vaccines and
Serums
Recombinant protein, nanoparticles (based on S-protein and other
epitopes)
Innovax/Xiamen University/GSKCOVID-19 XWG-03 truncated S (Spike) proteins
OncoGenSynthetic long peptide vaccine candidate for S and M proteins
MIGAL Galilee Research InstituteOral Escherichia coli-based protein expression system of S and N proteins
Lomonosov Moscow State UniversityStructurally modified spherical particles of the tobacco mosaic virus
(TMV)
University of AlbertaSpike-based
AnyGo TechnologyRecombinant S1-Fc fusion protein
Yisheng BiopharmaRecombinant protein
VabiotechRecombinant S protein in IC-BEVS
Applied Biotechnology Institute, Inc.Orally delivered, heat-stable subunit
Medigen Vaccine Biologics Corporation/NIAID/DynavaxS-2P protein + CpG 1018
National University of San Martin and CONICET, ArgentinaProtein subunit
MOGAM Institute for Biomedical Research, GC Pharma
Axon Neuroscience SEPeptides derived from Spike protein
Intravacc/EpiVaxOuter membrane vesicle (OMV) subunit
OMV peptide
Neovii/Tel Aviv UniversityRBD-based
Kentucky Bioprocessing, Inc
Quadram InstituteOMV-based vaccine
BiOMViS Srl/University of Trento
FBRI SRC VB VECTOR, Rospotrebnadzor, KoltsovoPeptide vaccine
Subunit vaccine
Virus-like particlesMedicago Inc./Université LavalPlant-derived VLPPre-clinical
Saiba GmbHVLP based on RBD displayed on VLPs
Navarrabiomed, Oncoimmunology groupVLPs, lentivirus and baculovirus vehicles
VBI Vaccines Inc.Enveloped VLP (eVLP)
Mahidol University/The Government Pharmaceutical Organization
(GPO)/Siriraj Hospital
VLP + adjuvant
IrsiCaixa AIDS Research Institute/IRTA-CReSA/Barcelona
Supercomputing Centre/Grifols
S protein integrated in HIV VLPs
Imophoron Ltd and Bristol University Max Planck CentreADDomer™ multiepitope display
Doherty InstituteUnknown
OSIVAXVLP
University of Sao PauloVLPs/whole virus
ARTES BiotechnologyeVLP
Replicating
viral vector
Shenzhen Geno-Immune Medical InstituteMinigenes engineered based on multiple viral genes, using an
efficient lentiviral vector system (NHP/TYF) to express viral proteins
and immune modulatory genes
Phase I
Cadila Healthcare LimitedMeasles vectorPre-clinical
Institute Pasteur/Themis/University of Pittsburg Center for Vaccine
Research
FBRI SRC VB VECTOR, Rospotrebnadzor, Koltsovo
DZIF – German Center for Infection Research/CanVirex AGMeasles virus (S, N targets)
Tonix Pharmaceuticals/Southern ResearchHorsepox vector expressing S protein
University of Hong KongInfluenza vector expressing RBD
IAVI/MerckReplication-competent vesicular stomatitis virus (VSV) chimeric virus
technology (VSVΔG) delivering the SARS-CoV-2 Spike (S) glycoprotein
BIOCAD and IEMLive viral vectored vaccine based on attenuated influenza virus
backbone (intranasal)
Lancaster University, UKAvian paramyxovirus vector (APMV)
FBRI SRC VB VECTOR, Rospotrebnadzor, KoltsovoRecombinant vaccine based on influenza A virus for the prevention of
COVID-19 (intranasal) VSV vector
University of Western OntarioVSV-S
Israel Institute for Biological Research/Weizmann Institute of
Science
Fundação Oswaldo Cruz and Instituto ButantanAttenuated influenza expressing an antigenic portion of the Spike
protein
Intravacc/Wageningen Bioveterinary Research/Utrecht UniversityNewcastle disease virus vector (NDV-SARS-CoV-2/Spike)
UW–Madison/FluGen/Bharat BiotechM2-deficient single replication (M2SR) influenza vector
UnknownTulane UniversityUnknownPre-clinical

The table was adapted from5,6,21.

DNA/RNA-based platforms

DNA- and RNA-based platforms present the greatest potential for speed of production since culturing and fermentation are not required for their synthesis. The potential of this approach is showcased by Moderna, whose vaccine candidate advanced to clinical testing just 2 months after sequence identification7. There are many other advantages associated with DNA-based vaccines. Notably, they are renowned for their safety profile since the vectors employed are non-replicating and encode and express only the target antigen. The vectors therefore are unable to revert to a disease-causing form, which is a risk with viral vectors. Another key advantage with DNA-based vaccination is the absence of vector-specific immunity, which allows these products to be utilized in prime and boost regimens with multiple products intended for the same patient8. RNA-based vaccines are also a promising alternative owing to their potential for low-cost manufacture and good safety profile in animal studies911. However, both DNA- and RNA-based vaccines have their own set of challenges. Both vaccines could suffer from the drawback of having low immunogenicity, as DNA vaccines could potentially integrate into the human genome, while there are concerns about the stability of RNA vaccines12. As there are currently no approved DNA or RNA vaccines for medical use in humans12,13, the question about low immunogenicity has not yet been resolved (Table 2). The lack of currently approved DNA or RNA vaccines provides considerable regulatory uncertainty, and their progression through the regulatory process will almost certainly take significantly longer than for more conventional vaccine technologies. Currently, there is one vaccine in phase II clinical trials, three in phase I/II clinical trials, and four in phase I clinical trials for SARS-CoV-2 based on the DNA/RNA platform. Inovio Pharmaceuticals is developing a SARS-CoV-2 vaccine, INO-4800, using a DNA-based platform, with phase I clinical trials being undertaken in conjunction with pre-clinical studies in order to hasten development14. The International Vaccine Institute (IVI) has announced that the Coalition for Epidemic Preparedness Innovations (CEPI) has awarded $6.9 million of funding to INOVIO to work with IVI and the Korea National Institute of Health (KNIH) for a phase I/II clinical trial of INO-4800 in South Korea. The trial will be conducted in parallel with INOVIO’s phase I study, which is currently ongoing with 40 healthy adults receiving the vaccine candidate and will eventually be expanded to older adults15. An RNA vaccine candidate, mRNA-1273, is being developed by Moderna in collaboration with NIAID. The Biomedical Advanced Research and Development Authority (BARDA), a division of the Office of the Assistant Secretary for Preparedness and Response (ASPR) within the US Department of Health and Human Services (HHS), has agreed to cooperate to hasten the development of mRNA-1273 by pledging up to $483 million in funding16. Healthy adults aged 18 to 55 were recruited for phase I trials. Two injections of mRNA-1273 were given 28 days apart at a dose of 25 μg, 100 μg, or 250 μg. Dire adverse events were absent, and no trial halting rules were met, although one participant in the 25 μg group withdrew because of an unsolicited adverse event, transient urticaria, which was postulated to be linked to the first vaccination. Live virus neutralization capable of reducing SARS-CoV-2 infectivity by 80% or more was detected at day 43 post-vaccination in all participants. Of the three doses evaluated, the 100 μg dose was optimal based on its capacity to trigger high neutralization responses and Th1-skewed CD4 T cell responses. The reactogenicity profile at 100 μg was better than for the other two doses. The safety, reactogenicity, and immunogenicity of mRNA-1273 are currently being evaluated in phase II trials using two vaccinations given 28 days apart. A total of 600 participants, 300 aged 18–54 and 300 aged 55 and above, have been assigned to three groups: placebo, 50 μg of vaccine, or 100 μg of vaccine at both vaccinations17. A phase III trial of mRNA-1273 is expected to begin in July with an estimated 30,000 participants. With the expected final dose of 100 μg, Moderna is confident to scale up manufacturing to approximately 500 million to 1 billion doses per year18.

Table 2. Pros and cons of different vaccine formulations and examples of licensed vaccines.

Vaccine platformsProsConsExamples of licensed vaccines
targeted for humans
RNA•   Potential low-cost
manufacturing
•   Ease of manufacturing
•   Good safety profile
•   May have low immunogenicity
due to instability
•   May require multiple doses
-
DNA•   Potential low-cost
manufacturing
•   Ease of manufacturing
•   Good safety profile
•   Good stability
•   Does not induce anti-vector
immunity
•   Potential integration to human
genome
•   Low immunogenicity
-
Virus vectors
(replicating/non-
replicating viral vectors
and virus-like particles)
•   High-efficiency gene
transduction
•   High specific delivery of genes
to target cells
•   Induction of robust immune
responses
•   Increased cellular immunity
•   Low titer production
•   May induce anti-vector
immunity
•   Generation of replication-
competent virus, which can
induce tumorigenesis
•   JYNNEOS (Smallpox/
Monkeypox)
•   ACAM2000 (Smallpox)
•   Adenovirus type 4 and type 7
vaccine, live, oral (febrile acute
respiratory)
Inactivated•   Good safety profile
•   Can be used in
immunocompromised patients
•   Requires booster doses
•   Low production titer
•   Poliovax (Polio)
•   Flucelvax Quadrivalent
(Influenza)
•   Ixiaro (Japanese Encephalitis)
•   Imovax (Rabies)
Live attenuated virus•   High potency
•   Triggers long-lasting immunity
•   Low-cost manufacturing
•   Possible regression to virulence
strain
•   Limited use in
immunocompromised patients
•   ERVEBO (Ebola virus)
•   MMR II (Measles, Mumps, and
Rubella)
•   BCG vaccine (Tuberculosis)
Protein subunit•   Can be used in
immunocompromised patients
•   Good safety profile
•   Low immunogenicity
•   Conjugation could lead to
batch-wise variation
•   PedvaxHIB
(Haemophilusinfluenzae type b)
•   Engerix-B (Hepatitis B)
•   Recombivax HB (Hepatitis B)

This table was adapted from811,20,2228.

Virus vectors

Virus-based vectors are powerful tools for vaccination. Their effectiveness stems from their ability to infect cells, which allows them to be highly efficient, specific, and able to trigger robust immune responses. Despite their advantages, viral vectors have several disadvantages (Table 2). For example, the use of vaccinia virus and adenovirus may lead to immunity against the vector, which will reduce the efficacy of the vaccine. The use of retrovirus and lentivirus, on the other hand, may lead to the risk of tumorigenesis in patients, which results from the integration of viral long terminal repeats into proto-oncogenes19. Additionally, certain viral vectors such as adeno-associated virus may not be cost effective owing to their low titer production20. Ultimately, viral vectors still present themselves as a valid choice for vaccine development against SARS-CoV-2, as illustrated by their successful use in the eradication of smallpox20. Examples of such vaccines currently under development are ChAdOx1-S, Ad5-nCoV, aAPC, and LV-SMENP-DC. The latter two vaccines, developed by Shenzhen Geno-Immune Medical Institute, are currently in phase I (aAPC) and phase II (LV-SMENP-DC) clinical studies. The Ad5-nCoV vaccine pioneered by CanSino Biologics is the first SARS-CoV-2 vaccine to reach phase II clinical trials5. Phase I clinical studies for Ad5-nCoV were conducted between March 16 and March 27. No serious adverse events within 28 days of vaccination were reported in vaccine recipients. Specific antibodies, including neutralizing antibodies, increased significantly at day 14 and peaked 28 days post-vaccination. Specific T-cell response peaked at day 14 post-vaccination29. A phase II study is currently underway with 500 participants registered in three different groups: 250 participants will receive the vaccine, 125 participants will receive a lower vaccine dose, and 125 participants will receive placebo. The immune response will be tested at 0, 14, and 28 days and 6 months after vaccination30. ChAdOx1-S, developed by the University of Oxford in partnership with AstraZeneca, is the first vaccine candidate to reach phase III clinical trials1,5. A study has reported that a single dose of the vaccine is able to elicit a strong immune response in rhesus macaques31. Additionally, a trial study conducted in pigs demonstrated that ChAdOx is able to elicit a greater antibody response when given a booster shot, suggesting that a two-dose approach may give better protection in humans against SARS-CoV-232. However, it should be noted that nose swabs and oropharynx and mediastinal lymph node testing revealed viral gRNA in both vaccinated and control animal groups at 3 and 5 days post-inoculation. Interestingly, viral genome was detected in cervical lymph node in the vaccinated group but not in the control group. Detection of virus in these tissues indicates the vaccine does not provide complete protection against SARS-CoV-2 infection. Viral RNA (gRNA and sgRNA) load in lung tissue was also shown to vary dramatically between individual animals but was determined to be significantly lower in the vaccinated group than in the control group. Another troubling result to note from this study is that despite observing neutralizing antibodies in vaccinated animals, the titers reported were extremely low31. Generally, neutralizing antibodies elicited by effective vaccines can be diluted more than a thousand-fold and still maintain their effectiveness33. However, the results reported by Oxford show that the serum could be diluted only 4- to 40-fold before losing its neutralizing activity31,33. ChAdOx1-S was well tolerated in humans, with a trial of over 320 vaccinated individuals showing no strong adverse effects34. The phase III clinical trials of this vaccine will involve 8,000 individuals in the United Kingdom, 5,000 individuals in Brazil, and 2,000 individuals in South Africa35. AstraZeneca has reached an agreement with Europe’s Inclusive Vaccines Alliance (IVA) to supply up to 400 million doses of ChAdOx1-S starting by the end of 2020 at no profit. The total manufacturing capacity of this vaccine currently stands at 2 billion doses34.

Inactivated vaccines

Inactivated vaccines have been successfully employed over the past 70 years and are widely used today36. Inactivated vaccines are produced using bacteria or viruses by deactivating them with heat, chemicals, or radiation. These processes terminate the pathogen’s ability to replicate, leading to them being more stable and having higher safety profiles. These attributes allow for their use in immunocompromised individuals37. However, the characteristics that contribute to their strengths are also weaknesses. As the pathogens are inactivated, these vaccines generally stimulate a much weaker immune response than live vaccines and require several doses for effective immunity to be established. The immune response to an inactivated vaccine is also typically humoral. Antibody titers against the targeted antigen will diminish with time, leading to the need for booster shots22,36 (Table 2). Regardless, inactivated vaccines are effective agents that have prevented countless deaths due to various infections in humans, notably against wild poliovirus 2, which has not been detected since 1999 and was declared eradicated in September 2015 by WHO38. There are currently nine inactivated vaccine candidates for SARS-CoV-2 listed by WHO. Of these, one vaccine candidate—PiCoVacc, developed by Sinovac—is listed as ready to commence phase III clinical trials and is in the midst of preparing to recruit participants. It is very promising to note that Sinovac Biotech has demonstrated protection by PiCoVacc against SARS-CoV-2 in monkeys. Monkeys were immunized three times with two different doses (3 or 6 μg per dose) of PiCoVacc at day 0, 7, and 14 before virus challenge at week 3. It was demonstrated that monkeys vaccinated with PiCoVacc produced anti-SARS-CoV-2 neutralizing antibody titers similar to those of recovered patients. This study also indicated that PiCoVacc is safe, as there was no infection enhancement or immunopathological exacerbation observed in vaccinated monkeys. PiCoVacc is currently in phase I human clinical trials as stated above. The company plans to initiate phase II and III clinical trials with PiCoVacc by the end of this year39. There are two other inactivated vaccine candidates being developed by the Beijing Institute of Biological Products and the Wuhan Institute of Biological Products, respectively, both of which are currently in phase I/II clinical trials. Both institutes are collaborating with Sinopharm for the development of these vaccines.

Live attenuated virus

Another promising approach in vaccine design is the use of live attenuated virus (LAV), developed by codon deoptimization. This technology has proven itself to be cost effective for large-scale manufacturing and has a smooth regulatory approval pathway, as it has demonstrated high efficacy and potency in both in vitro and in vivo experiments against different respiratory viruses in mice, human cells, and non-human primates, providing lasting immunity with just a single dose2325. LAV have a simple production process using well-established Vero cells, which are currently the most widely accepted cell line by regulatory authorities for vaccine development, since they have been in use for vaccine manufacturing for nearly 40 years. LAVs are also proven to grow and infect microcarrier beads in large-scale fermenters up to 6,000 L and in serum free media with zero loss in production output40. Existing manufacturing infrastructure can be easily utilized because of its cost effectiveness for large-scale manufacturing41. LAV has certain drawbacks, notably the existing possibility of reversion to virulence, which has strong safety implications, especially in immunocompromised patients26 (Table 2). Nonetheless, codon deoptimization technology in vaccine design has promise against SARS-CoV-2. It is a tried and tested new engineering technology, and designs such as introducing point mutations have proved that the drawbacks can be overcome with relative ease42. One example of its recent use is in a vaccine against Ebola virus, where an estimated efficacy of 97.5% was recorded from a preliminary analysis of 90,000 individuals who were exposed for 10 days or more to Ebola virus after vaccination43. Codon deoptimization vaccine candidates have virtually no risk of reversion to virulence because of the large number of substitutions that are made in the coding sequence. This is a crucial safety feature of vaccines developed using codon deoptimization. Vaccines developed against SARS-CoV-2 using LAV technology are all currently in pre-clinical development. For example, Indian Immunologicals Limited is currently working together with Griffith University to develop a vaccine using codon deoptimization as a strategy against SARS-CoV-2. The vaccine candidate is expected to provide a long-lasting immunity against SARS-CoV-2 following a single vaccination and is also expected to provide cross-protection against other coronaviruses such as MERS and SARS-CoV-144.

Protein subunit

Protein subunit vaccines are a popular choice for the design of a SARS-CoV-2 vaccine owing to their strong safety profile, which is particularly advantageous for immunocompromised patients. They are less likely to cause complications in vaccinated individuals, as the antigenic components employed in protein subunit vaccines are purified and do not involve the use of infectious viruses45. However, as with other vaccine platforms, protein subunit vaccines come with their own set of challenges, with the most prominent one being their lack of efficacy. A subunit vaccine presents an antigen to the immune system without the involvement of viral particles using a specific, isolated protein of the pathogen. In the likely scenario where the isolated proteins are denatured, they will become associated with other antibodies different from what was initially targeted, leading to the lack of efficacy46 (Table 2). Nonetheless, novel designs and delivery can overcome this limitation and demonstrate enhanced efficacy, e.g. against Zika virus in animal studies and against malaria in in vivo studies47,48. The success of protein subunit vaccines is highlighted by the hepatitis B vaccine, and we have now controlled these diseases to the point of virtual elimination49,50. Currently, there are many vaccines under pre-clinical studies using this platform. For example, Danish company Expression Biotechnologies has announced that it was awarded an EU horizon 2020 grant amounting to €2.7 million for the development of a SARS-CoV-2 vaccine candidate. The company aims to conduct phase I/IIa clinical trials within 12 months after preliminary studies51. One other notable example of a vaccine candidate designed on the basis of this platform is developed by Novavax, which is currently in phase I/II clinical trials. There are three other protein subunit vaccine candidates currently in phase I clinical trials5.

Outlook

SARS-CoV-2 has given rise to an unprecedented economic, health, and societal challenge globally. The key to tackle this pandemic is the development of safe and effective vaccines. Monumental energy has been poured into vaccine R&D; however, there are many challenges that need to be overcome before a successful marketable vaccine becomes available. Over 100 vaccine candidates are in pre-clinical trials, with 19 in clinical trials and many others in early development5,6. Vaccine developers are turning a blind eye to industrial benchmarks for traditional vaccine development, as speed is considered a major priority in developing a successful vaccine against SARS-CoV-2. For example, clinical trials are being conducted simultaneously instead of sequentially using adaptive designs that are optimized for speed. Researchers, manufacturers, funders, and governing parties are targeting vaccines to be available by 2021, impelling proposals to move forward with reduced numbers of study participants and cut short safety follow-up in clinical studies. A successful COVID-19 vaccine will be administered on a global scale; thus, its safety profile must be solid. Therefore, approaches for generating a COVID-19 vaccine must not compromise safety aspects and will require careful evaluation of effectiveness and safety at each step to avoid major setbacks. There are also unresolved obstacles in ensuring global access to a developed COVID-19 vaccine. For example, during the early days of the HIV and H1N1 outbreaks, even after the successful development of a vaccine, it was difficult to achieve global distribution. Similarly, for COVID-19, a vaccine approved for marketing does not necessarily lead to easy accessibility. Manufacturing capability must be enhanced so that the production of vaccine doses can be magnified while ensuring vaccine affordability for the global community. Coordinated efforts among vaccine developers, funders, and manufacturers are essential to ensure that successful vaccine candidates can be manufactured in adequate numbers and distributed evenly across the globe.

Another important issue is whether COVID-19 vaccines will sensitize humans to a phenomenon known as antibody-dependent enhancement (ADE). Although antibodies are generally protective, rare cases have been documented in which pathogen-specific antibodies enhance disease—a phenomenon known as ADE. ADE is most prominently associated with dengue virus52. ADE has also been observed for both MERS and SARS-CoV-1 despite evidence pointing towards the fact that coronavirus diseases in humans lack the clinical, epidemiological, biological, or pathological attributes of ADE exemplified by the dengue viruses. Therefore, ADE warrants full consideration in the safety profiling of potential vaccine candidates against SARS-CoV-2 to avoid this phenomenon being observed, as SARS-CoV-2 shares a high sequence identity with SARS-CoV-153,54.

Safety aspects of the COVID-19 vaccines under development are of the utmost importance. Thorough clinical trials need to be carried out prior to marketing of the vaccine. Recent reports in India on the possibility of launching a vaccine in record time without allowing sufficient time for clinical trials are disturbing. Although COVID-19 vaccine development is of the utmost global importance, it is crucial that we do not cut corners and that vaccines are approved only after having passed through rigorous clinical trials.

Concluding remarks

There are an extraordinary number of COVID-19 vaccines currently under development around the world. Highly optimistic timeframes of 12–18 months to get to the distribution of approved vaccines are being touted. Vaccines typically take many years or even decades to develop, e.g. the smallpox vaccine took 25 years to develop for human use, the HPV vaccine 15 years, the rotavirus vaccine 15 years, and the influenza vaccine 28 years. The COVID-19 vaccine initiatives aspire to have vaccines ready for distribution by 2021. This is a truly remarkable and unprecedented timeframe for vaccine development. The majority of vaccines won’t progress beyond pre-clinical or early clinical testing, but, of course, we need only one effective COVID-19 vaccine to halt this pandemic. Fewer than 10% of drugs or vaccines that enter clinical trials ever progress to become available for human use. For example, 30% fail at phase I trials. Of those that progress to phase II, 69% fail and then a further 42% fail at phase III. Of those products that successfully pass through phase III trials, a further 15% do not gain regulatory approval. Because of the drastically shorter time being allotted for the pre-clinical development of COVID-19 vaccine candidates, it is conceivable that a higher proportion of these candidates may fail during clinical testing. In addition to vaccine development, a number of resource-intensive activities will need to be undertaken in parallel, particularly the construction of manufacturing facilities capable of producing at a global scale. We are hopeful the unmatched resources being directed into the international vaccine R&D effort will lead to a successful vaccine capable of stopping this new pandemic.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 17 Aug 2020
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Ng WH, Liu X and Mahalingam S. Development of vaccines for SARS-CoV-2 [version 1; peer review: 2 approved] F1000Research 2020, 9(Faculty Rev):991 (https://doi.org/10.12688/f1000research.25998.1)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 1
VERSION 1
PUBLISHED 17 Aug 2020
Views
1
Cite
Reviewer Report 17 Aug 2020
Gavin Koh, Diseases of the Developing World, GlaxoSmithKline, Uxbridge, UB11 1BT, UK 
Approved
VIEWS 1
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Koh G. Reviewer Report For: Development of vaccines for SARS-CoV-2 [version 1; peer review: 2 approved]. F1000Research 2020, 9(Faculty Rev):991 (https://doi.org/10.5256/f1000research.28692.r69047)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Views
2
Cite
Reviewer Report 17 Aug 2020
Shinji Makino, Department of Microbiology and Immunology, Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA 
Approved
VIEWS 2
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Makino S. Reviewer Report For: Development of vaccines for SARS-CoV-2 [version 1; peer review: 2 approved]. F1000Research 2020, 9(Faculty Rev):991 (https://doi.org/10.5256/f1000research.28692.r69046)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 17 Aug 2020
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.