Next Article in Journal
COVID-19 Vaccination and Rates of Infections, Hospitalizations, ICU Admissions, and Deaths in the European Economic Area during Autumn 2021 Wave of SARS-CoV-2
Next Article in Special Issue
Protective CD8+ T Cell Response Induced by Modified Vaccinia Virus Ankara Delivering Ebola Virus Nucleoprotein
Previous Article in Journal
VSV-Based Vaccines Reduce Virus Shedding and Viral Load in Hamsters Infected with SARS-CoV-2 Variants of Concern
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Utilization of Viral Vector Vaccines in Preparing for Future Pandemics

US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, Washington, DC 20201, USA
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(3), 436; https://doi.org/10.3390/vaccines10030436
Submission received: 22 February 2022 / Revised: 9 March 2022 / Accepted: 10 March 2022 / Published: 12 March 2022
(This article belongs to the Special Issue A Modern Take on Replicating Viral Vaccines)

Abstract

:
As the global response to COVID-19 continues, government stakeholders and private partners must keep an eye on the future for the next emerging viral threat with pandemic potential. Many of the virus families considered to be among these threats currently cause sporadic outbreaks of unpredictable size and timing. This represents a major challenge in terms of both obtaining sufficient funding to develop vaccines, and the ability to evaluate clinical efficacy in the field. However, this also presents an opportunity in which vaccines, along with robust diagnostics and contact tracing, can be utilized to respond to outbreaks as they occur, and limit the potential for further spread of the disease in question. While mRNA-based vaccines have proven, during the COVID-19 response, to be an effective and safe solution in terms of providing a rapid response to vaccine development, virus vector-based vaccines represent a class of vaccines that can offer key advantages in certain performance characteristics with regard to viruses of pandemic potential. Here, we will discuss some of the key pros and cons of viral vector vaccines in the context of preparing for future pandemics.

1. Introduction

As of the end of January 2022, there have been over 373 million diagnosed cases of COVID-19, with more than 5.65 million deaths worldwide [1]. These numbers have unfortunately underscored key gaps in preparedness for pandemic threats. As the global response to COVID-19 continues, public and private partners must also keep an eye on the future to improve our collective preparedness posture for future pandemics.
There are several viral threats identified by the World Health Organization (WHO) as representing major public health risks due to their potential to cause epidemics and pandemics. These include severe acute respiratory syndrome (SARS) coronavirus (CoV) 2, or SARS-CoV-2, Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, Crimean-Congo hemorrhagic fever virus, Ebola viruses, Marburg virus, Lassa virus, Nipah virus, Hendra virus, Rift Valley fever virus, Zika virus, and “disease X” [2]. Similar lists of priority viral threats have been published by GAVI and CEPI on their respective websites, which include these threats, plus Chikungunya virus and monkeypox virus [3,4]. While COVID-19 has had an obvious global impact both in terms of lives lost and economic disruption, there is a collective threat of viral pathogens with pandemic potential, representing viruses that could be particularly problematic due to their ability to rapidly spread and cause severe disease, morbidity, and mortality (Table 1). In some cases, the endemic nature of these diseases, whether in human or animal populations, adds to the risk of epidemic or pandemic disease.
Diagnostics and disease surveillance will play a critical role in stopping outbreaks early; however, there are key roles that vaccines can play in reducing disease burden in the context of outbreaks. While there is an understandable push to leverage “platform” technologies that could represent rapid plug-and-play solutions as part of a vaccine response, the most critical aspect is that the right vaccine technology is utilized for the threat in question. In some cases, such as Ebola and Lassa viruses, this may mean viral vector vaccines due to the ability to provide rapid onset to protection with a single dose. Historically, Ebola outbreaks have been sporadic and unpredictable in terms of size and geographic location [24]. As a result, the now-licensed Ebola vaccine has been utilized in response to outbreaks by vaccinating those at risk of exposure, since efficacy was demonstrated in 2015 [25,26]. The clinical operations associated with Ebola vaccine responses were made possible by a vaccine with high efficacy and a rapid onset of protection, two key performance parameters associated with the use of a live virus-vector vaccine in this case [26]. For Lassa virus, human and animal studies indicate that convalescence is largely mediated by the cellular response. Antibody titers are highest only after infected individuals have already recovered from disease [27]. Since cellular response plays a dominant role during resolution of natural infection, vaccine development for Lassa virus has included a focus on generating a potent CD8+ T cell response [27]. Because viral vectors are live, attenuated viruses, they generally elicit a robust cellular response, and will be an important platform option for viruses where this is a key to optimal protective immunity.
With the goal of pandemic preparedness in mind, it will remain critical that we find the right vaccine for each respective threat. For diseases of which outbreaks will continue to be sporadic, and geographically isolated at the start, there will be time to intervene with vaccines prior to the event becoming an epidemic or pandemic. Some of the advantages offered by vaccines utilizing virus vectors may provide opportunities to respond in a reactive setting if targeted vaccination campaigns can be used to slow the spread of disease. A recent review by Vrba et al. highlighted the breadth of viruses that have been used to express heterologous antigens as a means for delivery as a vaccine [28]. In this review, we will focus on those virus vectors that have proceeded to clinical development, and how these vectors could be applied to emerging viral threats of pandemic potential.

2. Target Product Profile

Preparedness for the threats noted above would be best served by developing a pipeline of candidate vaccines with key performance parameters in mind at the outset. Viral vector vaccines have positive aspects for several of the parameters noted in Table 2.

2.1. Safety and Efficacy

Several viral vector-based vaccines are approved or authorized for use, based on a favorable balance between the benefits of vaccine effectiveness, the seriousness of the threat, and a comparably favorable safety profile. The approved vesicular stomatitis virus-vector Ebola vaccine showed early indications of arthralgia and arthritis events during clinical development [26,29]. While these early findings altered safety monitoring plans, ultimately, these events occurred at a low frequency, and were resolved relatively quickly [29]. Being a live virus vaccine, the thorough characterization of biodistribution and viral shedding was also monitored [30].
Recent findings of rare thrombolytic events with two different adenovirus-vector COVID-19 vaccines underscore the importance of tailoring safety monitoring plans during clinical development, and continued safety monitoring post-authorization [31]. The rare yet serious adverse events associated with adenovirus vectors have resulted in several regulatory authorities amending their respective guidance on usage [32]. However, given the significant threat of COVID-19, the benefits of these vaccines, and the rarity of thrombolytic events, the vaccines remain authorized for use. If the benefit-risk profile warrants continued development, safety monitoring can be fine-tuned to address key questions associated with the vector in question.
A high level of efficacy can be critical with diseases with high morbidity and mortality rates, which may require a relatively reactogenic vaccine. Smallpox is a good example of a disease for which vaccines with higher levels of reactogenicity were acceptable given the high case fatality rates (~30%) and significant morbidity in survivors (e.g., blindness and skin scarring) [33]. Analyses from the United States vaccination efforts throughout the 1960s showed an adverse event profile that included progressive vaccinia, eczema vaccinatum, postvaccinial encephalitis, and generalized vaccinia at rates from 1.5 to 241 cases per million vaccinations [34]. As the global burden of smallpox disease declined over the 20th century, safer vaccines that use vaccinia vectors, but include extensive cell culture passaging to attenuate the vaccine, have been pursued [35].

2.2. Clinical Operations

Efficacy following a single dose can be a huge benefit when a rapid response is required, or when tracking individuals for subsequent doses presents a challenge. Specifically, when clinical operations may entail use in a ring vaccination setting, rapid onset to protection with a single dose is crucial to the ability to slow an outbreak. When combined with disease surveillance, a vaccine that elicits rapid protection against disease can be utilized to protect those likely to have been exposed or be exposed, such as contacts of index cases and healthcare workers. Protection with a single dose also confers advantages around the total number of doses needed, which can be a critical consideration in resource-constrained settings, not only because of the number of doses, but also the infrastructure to track needs for additional doses.
Cold-chain requirements are also important to consider, although responses to Ebola and SARS-CoV-2 have demonstrated that the distribution and administration of a vaccine requiring storage at −70 °C are feasible [36,37]. That said, a vaccine that can be stored long-term at refrigerated or room temperatures offers clear logistical advantages in storage, both in terms of central inventory, and at the point of use. Viral vector vaccines can be developed with such storage and shelf-life considerations in mind. Viral vectors are amenable to techniques such as lyophilization to improve storage and shelf-life considerations [38], although this can also introduce complications in the field with diluent requirements.

2.3. Research and Development

While not inherently a key vaccine characteristic, development speed will be a critical aspect in how quickly we can meet key performance parameters. Generally speaking, when considering the three major vaccine classes (nucleic acid-based, viral vector, and recombinant protein constructs) likely to be pursued, nucleic acid-based vaccines likely will be the quickest into clinical development, followed by virus vector vaccines, and then protein subunit-based vaccines.
Part of the speed associated with vaccine development will lie in whether the production platform has been applied to other pathogens. For example, the rapid progress of COVID-19 vaccines utilizing mRNA and adenovirus vector platforms was enabled by past research and development efforts using these same platforms for other pathogens [39]. For viral vector vaccines, experience in applying reverse genetics technologies to generate constructs and the conduct of nonclinical studies to understand toxicity and immunogenicity profiles associated with the vector provides a foundation on which candidate development can quickly progress. Examples of viral vectors with such experience will be discussed in the next section.

3. Virus Vectors in Development

3.1. Adenovirus Vectors

The utility of adenoviruses as vaccine vectors became clear when the earliest attempts at using them for gene therapy were hindered by the robust induction of innate and adaptive immune responses [40]. These findings, along with their broad cellular tropism, high gene expression, and manufacturing ease, have made adenoviruses one of the most widely used vaccine platforms. Adenoviruses are typically attenuated for use as vaccine vectors by deleting the E1/E3 genomic locus, which is replaced with a transgene encoding the target pathogen antigen. E1 deletion renders the virus non-replicating and E3 deletion removes proteins that would otherwise help the virus evade immune detection in infected cells. E1/E3 deleted adenovirus vectors can accommodate transgene inserts of up to 7.5–8 kb in size [40]. Table 3 highlights the use of various vectors against a range of infectious diseases.
Human adenovirus 5 (Ad5) was originally the most widely used adenovirus vector for vaccine development. Ad5 vector COVID-19 vaccines are approved as single shot vaccines in China (Ad5-nCoV), with an estimated 57.5% vaccine efficacy against symptomatic COVID-19 [51], and as part of a heterologous boost to an Ad26 prime in Russia (Sputnik V) [80].
The early clinical development of Ad5-based vaccines found that high seroprevalence for Ad5 within the human population reduced antigen-specific immunity for Ad5 vector HIV vaccines [81]. In response, the development of adenovirus vector vaccines has shifted to focus primarily on either human adenoviruses with low seroprevalence (e.g., human serotypes 26 (Ad26) and 35 (Ad35)) or primate adenoviruses [82,83].
Ad26 emerged as an attractive alternate to address the pre-existing immunity issues with Ad5, as it has low human seroprevalence, and is more immunogenic compared to other alternate adenovirus vectors [84]. Prior to the COVID-19 pandemic, Ad26 vector vaccines had been evaluated in several clinical trials for various pathogens, and were determined to have an acceptable safety profile, and induce strong and durable humoral and cellular immune responses [85]. The first approved Ad26 vector vaccine was authorized under exceptional circumstances by the European Medicines Agency (EMA) in 2020 for Ebola virus when combined with a heterologous boost of a modified vaccinia Ankara (MVA) vector-based vaccine known as Zabdeno and Mvabea [43]. This heterologous prime/boost regimen was well-tolerated and generated robust antibody responses in clinical trials [85].
An Ad26 vector vaccine for SARS-CoV-2 (Ad26.COV2.S) is one of three vaccines authorized by the US Food and Drug Administration (FDA) for COVID-19. In a Phase 3 trial, a single shot of the Janssen Ad26.COV2.S vaccine demonstrated an efficacy of 66.9% against moderate to severe–critical COVID-19, with an onset 14 days after vaccine administration [86]. In an analysis of Ad26.COV2.S immunogenicity against COVID-19 variants of concern, while some reduction in neutralizing antibodies was seen for variants versus the prototype SARS-CoV-2 strain, the CD8+ and CD4+ T cell responses remained comparable against all strains [87]. The induction of a CD8+ T cell response may prove to be important for controlling infections with COVID-19 variants if they can escape neutralization by vaccine- or infection-generated antibodies targeting the original strain.
Ad35 is another human adenovirus alternate to Ad5 that has lower seroprevalence in humans and is immunogenic, although less so than Ad26 [84,88]. While there are no clinical candidates for the viruses of interest discussed here that use Ad35, the AdCLD-CoV19 candidate uses an Ad5 vector in which the native fiber knob protein used for virus-cell attachment is replaced by the Ad35 counterpart [72]. This design hopes to take advantage of the high immunogenic potential of Ad5, while avoiding pre-existing immunity issues.
ChAdOx1 is a serotype Y25 chimpanzee adenovirus vector with deletion of the E1/E3 loci and additional modifications that replace regions of the native E4 loci with that of Ad5 to increase virus yields [89]. The use of a chimpanzee adenovirus vector circumvents the issues with pre-existing immunity to human Ad5, which is confirmed by studies that show a very low seroprevalence of neutralizing antibodies for Y25 in humans [89]. ChAdOx1 is in use for several clinical stage vaccine candidates for the viruses of interest (Table 3).
Following demonstration that a single dose of a ChAdOx1 MERS-CoV vaccine generated protective immunity in non-human primates (NHP), the candidate completed Phase 1 human trials, where it proved to be well tolerated, and elicited humoral and cellular responses [61,90]. These data informed the selection of this platform for the development of AZD1222 to target SARS-CoV-2. AZD1222 is well tolerated as a two-dose primary series, and in Phase 3 trials, demonstrated an overall estimated efficacy of 74.0% at 15 or more days after the second dose [91]. This vaccine received conditional marketing authorization from the EMA in early 2021 [60]. Similar to Ad26.COV2.S, in addition to generating neutralizing antibodies, AZD1222 also elicited spike antigen-specific CD4+ and CD8+ T cells [92].
Chimpanzee adenovirus type 3 (cAd3) was identified as one of the most immunogenic primate adenovirus vectors, based on early screens in animal models [83]. In clinical trials, a cAd3-based Ebola virus vaccine was well tolerated and able to induce humoral and cellular responses, either alone [93], or as a priming immunization for a MVA-BN-Filo boost [94]. cAd3 is also used in the clinical development of monovalent vaccines for two other filoviruses, Sudan virus and Marburg virus, as well as a bivalent vaccine for Ebola virus and Sudan virus [66,67,68,69].
Additional primate adenovirus vectors in clinical development include the chimpanzee adenoviruses serotype 68 (ChAd68) and serotype 36 (Ad36), as well as the gorilla adenovirus serotype 32 (GRAd32), which are all in Phase 1 trials for SARS-CoV-2 (Table 3). The Ad36 vector SARS-CoV-2 vaccine is being assessed in a trial as an intranasal vaccine, which previously generated both systemic and sterilizing mucosal immunity in small animal models [71,95].
Single cycle adenovirus vectors (SC Ad) do not produce viral progeny, but are different from all the above-mentioned non-replicating vectors, in that they can go through a single round of gene amplification. This is achieved by retaining the genes required to amplify the inserted antigen cassette, but deleting the late viral genes that are involved in viral assembly [96]. The result is a vector with a safety profile similar to other non-replicating adenovirus vectors, but with increased amplification of the antigen target gene, higher production of the antigen target protein, and increased immunogenicity [96]. There is currently a human SC Ad serotype 6 vector (SC Ad6) in clinical development for SARS-CoV-2 [70]. Ad6 has lower seroprevalence in humans than Ad5 and has proven to be immunogenic in preclinical animal studies [97].

3.2. Modified Vaccinia Ankara (MVA)

MVA was originally attenuated through the serial tissue culture passage of the vaccinia virus strain Ankara. This resulted in genome deletions that restricted the host range of MVA, thus rendering the vector non-replicating in human cells, and removed immunomodulatory genes [98]. In turn, MVA has a long history as a safe and effective vaccine, as is evidenced by the recent approval of MVA for smallpox and monkeypox, and its prior use in the later stages of the World Health Organization’s (WHO) smallpox eradication campaign [35].
As an orthopoxvirus, MVA has a particularly large genome that can accommodate the insertion of transgenes of up to 25–30 kB in size [28]. This has made MVA an attractive vector to use in multivalent vaccines that target several pathogens at a time. The multivalent MVA Filovirus vaccine (MVA-BN Filo or Mvabea) has a transgene insert encoding for the surface glycoproteins (GP) of Ebola virus, Sudan virus, and Marburg virus, as well as for the nucleoprotein of Tai Forest virus [99]. MVA-BN Filo received conditional marketing authorization from the EMA in early 2021 as the boosting immunization in a heterologous regimen with adenovirus serotype 26 expressing Ebola virus GP (Zabdeno) [43]. This regimen was well tolerated in clinical trials and generated humoral and cellular responses that persist for up to one year after the boost [100].
Since vaccination for smallpox largely stopped following worldwide eradication in 1980, pre-existing immunity that could impact MVA vector immunogenicity is currently limited to adults over ~40 years old [101]. In animal studies to determine the effect of pre-existing immunity on MVA vector-generated immunity, some reduction in immunogenicity was observed, but this did not impact overall vaccine efficacy [102]. The MVA vector is in use for several clinical stage vaccine candidates for viruses of interest (Table 3).

3.3. Paramyxovirus Vectors

Several paramyxoviruses are used as vaccine vectors, including measles virus (MeV), parainfluenza viruses, and Newcastle disease virus (NDV). These vectors can accommodate transgene inserts of 4.5–6 kB, and easily incorporate the membrane GPs of other RNA viruses into their envelope [103,104]. The paramyxovirus vectors discussed here all infect via the intranasal route and are capable of inducing both systemic and local mucosal immune memory [104].
MeV was pursued as a vector based on the success of the live attenuated vaccine for measles, which has a long history as a safe and effective vaccine that generates durable immune memory responses [103]. A large proportion of the human population is seropositive for MeV due to its inclusion in routine childhood immunizations; however, based on animal and human studies, this does not appear to affect the immunogenicity of the MeV vector [105].
The MeV vector vaccines discussed here are based on the MeV Schwarz strain, which was attenuated through serial passage, but retained its ability to replicate and produce viral progeny [106]. A single shot of an MeV vector Lassa virus vaccine (MV-LASV) generated humoral and cellular responses in an NHP study, and was protective when animals were infected, either one month or one year after vaccination [107]. MV-LASV and a MeV vector based Zika virus candidate have completed Phase 1 clinical trials (Table 3).
Parainfluenza virus (PIV) vectors in clinical development include those that utilize human parainfluenza 3 (HPIV3) and parainfluenza virus 5 (PIV5), which, like all PIVs, require a genome length in a multiple of six nucleotides for efficient replication [104].
HPIV3 is a replicating vector that is used as the basis of an Ebola virus vaccine (HPIV3-EbovZ GP) that has completed Phase 1 clinical trials [48]. A unique element of HPIV3-EbovZ GP is that the vaccine was administered intranasally in an NHP study and elicited systemic and local respiratory T cell responses that protected animals from lethal Ebola virus infection [108]. This may be beneficial, as Ebola virus is transmitted via direct contact with infectious body fluids, which may include inoculation via oral or respiratory mucosa [19]. As HPIV3 is a common pediatric respiratory pathogen, the issue of pre-existing immunity is avoided by replacing the native surface proteins of HPIV3 with the Ebola GP [110].
PIV5 is a replicating vector that is currently being assessed in a Phase 1 SARS-CoV-2 trial as an intranasally administered vaccine [49]. PIV5 is a canine pathogen that does not cause disease in humans, but seropositivity is possible if an individual is in regular contact with dogs [111]. However, pre-existing immunity is not a significant concern due to swapping the native PIV surface proteins with that of the target virus; furthermore, a PIV5 vector influenza vaccine proved immunogenic in PIV5 vaccinated canines [111].
NDV is an avian virus that is naturally attenuated for humans, as a key immunomodulatory protein, V protein, is species-specific [112]. NDV is being assessed as a vector for a SARS-CoV-2 vaccine (NDV-HXP-S) that is currently in Phase 1/2 clinical trials using an inactivated form of NDV-HXP-S administered intramuscularly, with or without the Toll-like Receptor 9 agonist adjuvant, CpG 1018 [79]. An interim report of one of the trials found that the vaccine has an acceptable safety profile and is a potent immunogen [113]. A benefit of this vaccine is that it uses egg-based manufacturing, which could make it suitable for large-scale production if existing influenza vaccine manufacturing capacity is leveraged [114].

3.4. Vesicular Stomatitis Virus (VSV)

VSV is an attenuated, replicating rhabdovirus vaccine vector that can accommodate antigen inserts of up to ~6 kB in size [28]. For the viruses of interest, the vaccine constructs replace the native VSV surface GP with that of the target virus (Table 3). This serves the purpose of providing the antigenic target, but also attenuates the virus, as GP is a major virulence factor of VSV [115]. The VSV vector is also sometimes further attenuated through the down-regulation of VSV nucleoprotein and the truncation of the native GP cytoplasmic tail [116]. Replacement of the native VSV GP also reduces any potential effects of pre-existing immunity, which is already limited as VSV infection in humans is extremely rare [115].
A VSV vector Ebola virus vaccine (rVSVΔG-ZEBOV-GP) was licensed as ERVEBO by the FDA in 2019 as a single dose vaccine [43]. The effectiveness of this vaccine was established in Phase 3 clinical trials, where robust vaccine efficacy was observed within ten days following the administration of a single dose [26,117]. Vaccine generated anti-GP antibodies are correlated with protection and can persist for at least 24 months [118,119]. A smaller clinical assessment showed that the vaccine also elicits a cellular response [120]. rVSVΔG-ZEBOV-GP has a favorable safety profile and, while some instances of arthralgia and arthritis were reported, they were rare and self-limiting [121]. A SARS-CoV VSV vaccine is currently in a Phase 2b/3 trial (Table 3).
Another rhabdovirus, rabies virus (RABV), is also used as a vaccine vector and similarly can be pseudotyped with the GP of a different target virus, fully replacing the native RABV GP. While there are no RABV vector vaccine candidates in clinical development, the preclinical testing of inactivated vector vaccines for Ebola virus, Sudan virus, Marburg virus, and Lassa virus found the candidates to be immunogenic, either alone or when administered in combination as a multivalent vaccine [122].

4. Conclusions and Future Perspectives

Viral vector vaccines have important positive aspects that could be useful in an outbreak, epidemic, or pandemic response. As noted above, such vaccines have played a major role in recent Ebola and COVID-19 responses. These vaccines offer the potential for high levels of efficacy with a single dose, as well as manageable operational logistics. Viral vector vaccines are now in development for several other threats of pandemic potential; however, only a handful of different vectors are being actively pursued for these threats. Understanding the likely safety profile of vectors will inform the selection and development of lead candidates for the next outbreaks or pandemics. Additional investments in a more diverse array of viral vectors are warranted as such vaccines may provide additional tools for future outbreak and pandemic responses. Key factors such as levels of baseline seropositivity against the vector, safety profiles, and development as replication-defective versus live virus vectors should be taken into account when prioritizing likely candidates.
As public and private stakeholders begin to pivot from the COVID-19 response to focus on preparedness for future pandemics, a concerted effort will be required to build a more robust pipeline of vaccines in clinical development, with virus vectors being an important component of that pipeline, in combination with mRNA and protein subunit vaccines. The vaccine community, as a whole, must continue to evaluate lead vectors while investigating the potential for other viral vectors to be progressed into clinical development, and ultimately, to licensed products.

Author Contributions

Conceptualization, D.N.W.; formal analysis, K.A.H.; investigation, K.A.H., K.M.B. and D.N.W.; writing—original draft preparation, K.A.H. and D.N.W.; writing—review and editing, K.A.H., K.M.B. and D.N.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The opinions presented herein are those of the authors and do not represent the views of the United States Department of Health and Human Services.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 31 January 2022).
  2. World Health Organization. Prioritizing Diseases for Research and Development in Emergency Contexts. Available online: https://www.who.int/activities/prioritizing-diseases-for-research-and-development-in-emergency-contexts (accessed on 28 December 2021).
  3. Coalition for Epidemic Preparedness Innovations. Priority Diseases. Available online: https://cepi.net/research_dev/priority-diseases/ (accessed on 2 December 2021).
  4. Gavi. 10 Infectious Diseases That Could Be the Next Pandemic. Available online: https://www.gavi.org/vaccineswork/10-infectious-diseases-could-be-next-pandemic (accessed on 28 December 2021).
  5. Ioannidis, J.P.A. Infection fatality rate of COVID-19 inferred from seroprevalence data. Bull World Health Organ. 2021, 99, 19–33f. [Google Scholar] [CrossRef] [PubMed]
  6. De Wit, E.; van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol 2016, 14, 523–534. [Google Scholar] [CrossRef] [PubMed]
  7. Li, Y.D.; Chi, W.Y.; Su, J.H.; Ferrall, L.; Hung, C.F.; Wu, T.C. Coronavirus vaccine development: From SARS and MERS to COVID-19. J. Biomed. Sci. 2020, 27, 104. [Google Scholar] [CrossRef] [PubMed]
  8. Memish, Z.A.; Perlman, S.; Van Kerkhove, M.D.; Zumla, A. Middle East respiratory syndrome. Lancet 2020, 395, 1063–1077. [Google Scholar] [CrossRef]
  9. Forna, A.; Nouvellet, P.; Dorigatti, I.; Donnelly, C.A. Case Fatality Ratio Estimates for the 2013-2016 West African Ebola Epidemic: Application of Boosted Regression Trees for Imputation. Clin. Infect. Dis. 2020, 70, 2476–2483. [Google Scholar] [CrossRef] [PubMed]
  10. Agua-Agum, J.; Allegranzi, B.; Ariyarajah, A.; Aylward, R.; Blake, I.M.; Barboza, P.; Bausch, D.; Brennan, R.J.; Clement, P.; Coffey, P.; et al. After Ebola in West Africa--Unpredictable Risks, Preventable Epidemics. N. Engl. J. Med. 2016, 375, 587–596. [Google Scholar] [CrossRef] [PubMed]
  11. Lefebvre, A.; Fiet, C.; Belpois-Duchamp, C.; Tiv, M.; Astruc, K.; Aho Glélé, L.S. Case fatality rates of Ebola virus diseases: A meta-analysis of World Health Organization data. Med. Mal. Infect. 2014, 44, 412–416. [Google Scholar] [CrossRef] [PubMed]
  12. Nyakarahuka, L.; Kankya, C.; Krontveit, R.; Mayer, B.; Mwiine, F.N.; Lutwama, J.; Skjerve, E. How severe and prevalent are Ebola and Marburg viruses? A systematic review and meta-analysis of the case fatality rates and seroprevalence. BMC Infect. Dis. 2016, 16, 708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Richmond, J.K.; Baglole, D.J. Lassa fever: Epidemiology, clinical features, and social consequences. BMJ 2003, 327, 1271–1275. [Google Scholar] [CrossRef]
  14. Nigeria Centre for Disease Control. Lassa fever Situation Report Epi Week 52: 21–27 December 2020. Available online: https://ncdc.gov.ng/themes/common/files/sitreps/e9cf6841a83cdfdef7d73200813b9126.pdf (accessed on 14 January 2022).
  15. Kenmoe, S.; Demanou, M.; Bigna, J.J.; Nde Kengne, C.; Fatawou Modiyinji, A.; Simo, F.B.N.; Eyangoh, S.; Sadeuh-Mba, S.A.; Njouom, R. Case fatality rate and risk factors for Nipah virus encephalitis: A systematic review and meta-analysis. J. Clin. Virol. 2019, 117, 19–26. [Google Scholar] [CrossRef]
  16. Soman Pillai, V.; Krishna, G.; Valiya Veettil, M. Nipah Virus: Past Outbreaks and Future Containment. Viruses 2020, 12, 465. [Google Scholar] [CrossRef] [Green Version]
  17. Gómez Román, R.; Tornieporth, N.; Cherian, N.G.; Shurtleff, A.C.; L’Azou Jackson, M.; Yeskey, D.; Hacker, A.; Mungai, E.; Le, T.T. Medical countermeasures against henipaviruses: A review and public health perspective. Lancet Infect. Dis. 2022, 22, e13–e27. [Google Scholar] [CrossRef]
  18. Halpin, K.; Rota, P. A Review of Hendra Virus and Nipah Virus Infections in Man and Other Animals. Zoonoses Infect. Affect. Hum. Anim. 2014, 22, 997–1012. [Google Scholar] [CrossRef]
  19. Nasirian, H. New aspects about Crimean-Congo hemorrhagic fever (CCHF) cases and associated fatality trends: A global systematic review and meta-analysis. Comp. Immunol. Microbiol. Infect. Dis. 2020, 69, 101429. [Google Scholar] [CrossRef] [PubMed]
  20. Javelle, E.; Lesueur, A.; Pommier de Santi, V.; de Laval, F.; Lefebvre, T.; Holweck, G.; Durand, G.A.; Leparc-Goffart, I.; Texier, G.; Simon, F. The challenging management of Rift Valley Fever in humans: Literature review of the clinical disease and algorithm proposal. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 4. [Google Scholar] [CrossRef] [PubMed]
  21. Ahmed, A.; Ali, Y.; Elduma, A.; Eldigail, M.H.; Mhmoud, R.A.; Mohamed, N.S.; Ksiazek, T.G.; Dietrich, I.; Weaver, S.C. Unique Outbreak of Rift Valley Fever in Sudan, 2019. Emerg. Infect. Dis. 2020, 26, 3030–3033. [Google Scholar] [CrossRef] [PubMed]
  22. Costa, M.C.N.; Cardim, L.L.; Teixeira, M.G.; Barreto, M.L.; Carvalho-Sauer, R.d.C.O.d.; Barreto, F.R.; Carvalho, M.S.I.; Oliveira, W.K.; França, G.V.A.; Carmo, E.H.; et al. Case Fatality Rate Related to Microcephaly Congenital Zika Syndrome and Associated Factors: A Nationwide Retrospective Study in Brazil. Viruses 2020, 12, 1228. [Google Scholar] [CrossRef] [PubMed]
  23. Ikejezie, J.; Shapiro, C.N.; Kim, J.; Chiu, M.; Almiron, M.; Ugarte, C.; Espinal, M.A.; Aldighieri, S. Zika Virus Transmission—Region of the Americas, 15 May 2015–15 December 2016. MMWR Morb. Mortal. Wkly. Rep. 2017, 66, 329–334. [Google Scholar] [CrossRef] [Green Version]
  24. Jacob, S.T.; Crozier, I.; Fischer, W.A., 2nd; Hewlett, A.; Kraft, C.S.; Vega, M.A.; Soka, M.J.; Wahl, V.; Griffiths, A.; Bollinger, L.; et al. Ebola virus disease. Nat. Rev. Dis. Primers 2020, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  25. Inungu, J.; Iheduru-Anderson, K.; Odio, O.J. Recurrent Ebolavirus disease in the Democratic Republic of Congo: Update and challenges. AIMS Public Health 2019, 6, 502–513. [Google Scholar] [CrossRef]
  26. Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: Final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef] [Green Version]
  27. Purushotham, J.; Lambe, T.; Gilbert, S.C. Vaccine platforms for the prevention of Lassa fever. Immunol. Lett. 2019, 215, 1–11. [Google Scholar] [CrossRef]
  28. Vrba, S.M.; Kirk, N.M.; Brisse, M.E.; Liang, Y.; Ly, H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines 2020, 8, 680. [Google Scholar] [CrossRef] [PubMed]
  29. Halperin, S.A.; Arribas, J.R.; Rupp, R.; Andrews, C.P.; Chu, L.; Das, R.; Simon, J.K.; Onorato, M.T.; Liu, K.; Martin, J.; et al. Six-Month Safety Data of Recombinant Vesicular Stomatitis Virus-Zaire Ebola Virus Envelope Glycoprotein Vaccine in a Phase 3 Double-Blind, Placebo-Controlled Randomized Study in Healthy Adults. J. Infect. Dis. 2017, 215, 1789–1798. [Google Scholar] [CrossRef]
  30. Wolf, J.; Jannat, R.; Dubey, S.; Troth, S.; Onorato, M.T.; Coller, B.A.; Hanson, M.E.; Simon, J.K. Development of Pandemic Vaccines: ERVEBO Case Study. Vaccines 2021, 9, 190. [Google Scholar] [CrossRef] [PubMed]
  31. Kelton, J.G.; Arnold, D.M.; Nazy, I. Lessons from vaccine-induced immune thrombotic thrombocytopenia. Nat. Rev. Immunol. 2021, 21, 753–755. [Google Scholar] [CrossRef]
  32. U.S. Centers for Disease Control and Prevention. Interim Clinical Considerations for Use of COVID-19 Vaccines Currently Approved or Authorized in the United States. Available online: https://www.cdc.gov/vaccines/covid-19/clinical-considerations/covid-19-vaccines-us.html?CDC_AA_refVal=https%3A%2F%2Fwww.cdc.gov%2Fvaccines%2Fcovid-19%2Finfo-by-product%2Fclinical-considerations.html (accessed on 12 February 2022).
  33. Simonsen, K.A.; Snowden, J. Smallpox; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  34. Belongia, E.A.; Naleway, A.L. Smallpox vaccine: The good, the bad, and the ugly. Clin. Med. Res. 2003, 1, 87–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Meyer, H.; Ehmann, R.; Smith, G.L. Smallpox in the Post-Eradication Era. Viruses 2020, 12, 138. [Google Scholar] [CrossRef] [Green Version]
  36. Jusu, M.O.; Glauser, G.; Seward, J.F.; Bawoh, M.; Tempel, J.; Friend, M.; Littlefield, D.; Lahai, M.; Jalloh, H.M.; Sesay, A.B.; et al. Rapid Establishment of a Cold Chain Capacity of −60 °C or Colder for the STRIVE Ebola Vaccine Trial During the Ebola Outbreak in Sierra Leone. J. Infect. Dis. 2018, 217, S48–S55. [Google Scholar] [CrossRef] [PubMed]
  37. Ortiz, J.R.; Robertson, J.; Hsu, J.S.; Yu, S.L.; Driscoll, A.J.; Williams, S.R.; Chen, W.H.; Fitzpatrick, M.C.; Sow, S.; Biellik, R.J.; et al. The potential effects of deploying SARS-Cov-2 vaccines on cold storage capacity and immunization workload in countries of the WHO African Region. Vaccine 2021, 39, 2165–2176. [Google Scholar] [CrossRef] [PubMed]
  38. Crommelin, D.J.A.; Volkin, D.B.; Hoogendoorn, K.H.; Lubiniecki, A.S.; Jiskoot, W. The Science is There: Key Considerations for Stabilizing Viral Vector-Based Covid-19 Vaccines. J. Pharm. Sci. 2021, 110, 627–634. [Google Scholar] [CrossRef] [PubMed]
  39. Wherry, E.J.; Jaffee, E.M.; Warren, N.; D’Souza, G.; Ribas, A.; Force, A.C.; Task, C. How Did We Get a COVID-19 Vaccine in Less Than 1 Year? Clin. Cancer Res. 2021, 27, 2136–2138. [Google Scholar] [CrossRef] [PubMed]
  40. Tatsis, N.; Ertl, H.C. Adenoviruses as vaccine vectors. Mol. Ther. 2004, 10, 616–629. [Google Scholar] [CrossRef] [PubMed]
  41. NeuroRx, I. Phase 2b/3 Trial of VSV-ΔG SARS-CoV-2 Vaccine (BRILIFE) against Approved Comparator Vaccine. Available online: https://ClinicalTrials.gov/show/NCT04990466 (accessed on 14 December 2021).
  42. Gamaleya Research Institute of Epidemiology and Microbiology. Study of Safety and Immunogenicity of BVRS-GamVac-Combi. Available online: https://ClinicalTrials.gov/show/NCT04128059 (accessed on 14 December 2021).
  43. Tomori, O.; Kolawole, M.O. Ebola virus disease: Current vaccine solutions. Curr. Opin. Immunol. 2021, 71, 27–33. [Google Scholar] [CrossRef] [PubMed]
  44. International AIDS Vaccine Initiative. A Clinical Trial to Evaluate the Safety and Immunogenicity of rVSV∆G-LASV-GPC Vaccine in Adults in Good General Heath. Available online: https://ClinicalTrials.gov/show/NCT04794218 (accessed on 14 December 2021).
  45. Themis Bioscience GmbH. A Trial to Evaluate the Optimal Dose of MV-LASV (V182-001). Available online: https://ClinicalTrials.gov/show/NCT04055454 (accessed on 14 December 2021).
  46. Themis Bioscience GmbH. Zika-Vaccine Dose Finding Study Regarding Safety, Immunogenicity and Tolerability (V186-001). Available online: https://ClinicalTrials.gov/show/NCT02996890 (accessed on 14 December 2021).
  47. Themis Bioscience GmbH. Safety and Immunogenicity of a Novel Vaccine Formulation MV-ZIKA-RSP (V187-001). Available online: https://ClinicalTrials.gov/show/NCT04033068 (accessed on 14 December 2021).
  48. National Institute of Allergy and Infectious Diseases. Evaluating the Live-Attenuated Human Parainfluenza Virus Type 3 Vectored Vaccine Candidate Expressing Ebolavirus Zaire Glycoprotein as the Sole Envelope Glycoprotein. Available online: https://ClinicalTrials.gov/show/NCT03462004 (accessed on 14 December 2021).
  49. CyanVac LLC. Phase 1 Study of Intranasal PIV5-vectored COVID-19 Vaccine Expressing SARS-CoV-2 Spike Protein in Healthy Adults. Available online: https://ClinicalTrials.gov/show/NCT04954287 (accessed on 14 December 2021).
  50. International Clinical Trials Registry Platform. A Phase III Clinical Trial of Influenza Virus Vector COVID- 19 Vaccine for Intranasal Spray (DelNS1-2019-nCoV-RBD-OPT1). Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2100051391 (accessed on 14 December 2021).
  51. Halperin, S.A.; Ye, L.; MacKinnon-Cameron, D.; Smith, B.; Cahn, P.E.; Ruiz-Palacios, G.M.; Ikram, A.; Lanas, F.; Lourdes Guerrero, M.; Muñoz Navarro, S.R.; et al. Final efficacy analysis, interim safety analysis, and immunogenicity of a single dose of recombinant novel coronavirus vaccine (adenovirus type 5 vector) in adults 18 years and older: An international, multicentre, randomised, double-blinded, placebo-controlled phase 3 trial. Lancet 2022, 399, 237–248. [Google Scholar] [CrossRef] [PubMed]
  52. A Ph 2 Trial with an Oral Tableted COVID-19 Vaccine. Available online: https://ClinicalTrials.gov/show/NCT05067933 (accessed on 14 December 2021).
  53. COVID-19 Vaccination Using a 2nd Generation (E1/E2B/E3-Deleted) Adenoviral-COVID-19 in Normal Healthy Volunteers. Available online: https://ClinicalTrials.gov/show/NCT04591717 (accessed on 14 December 2021).
  54. McMaster University. Phase 1 Trial of ChAd68 and Ad5 Adenovirus COVID-19 Vaccines Delivered by Aerosol. Available online: https://ClinicalTrials.gov/show/NCT05094609 (accessed on 14 December 2021).
  55. Gamaleya Research Institute of Epidemiology and Microbiology. Study of Safety and Immunogenicity of BVRS-GamVac. Available online: https://ClinicalTrials.gov/show/NCT04130594 (accessed on 14 December 2021).
  56. U.S. Food & Drug Administration. Janssen COVID-19 Vaccine. Available online: https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/janssen-covid-19-vaccine (accessed on 14 December 2021).
  57. European Medicines Agency. COVID-19 Vaccine Janssen. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/covid-19-vaccine-janssen (accessed on 14 December 2021).
  58. Nogrady, B. Mounting evidence suggests Sputnik COVID vaccine is safe and effective. Nature News 2021, 595, 339–340. [Google Scholar] [CrossRef] [PubMed]
  59. Janssen Vaccines & Prevention, B.V. A Study to Evaluate the Safety, Reactogenicity and Immunogenicity of Ad26.ZIKV.001 in Healthy Adult Volunteers. Available online: https://ClinicalTrials.gov/show/NCT03356561 (accessed on 14 December 2021).
  60. European Medicines Agency. Vaxzevria (previously COVID-19 Vaccine AstraZeneca). Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/vaxzevria-previously-covid-19-vaccine-astrazeneca (accessed on 14 December 2021).
  61. Bosaeed, M.; Balkhy, H.H.; Almaziad, S.; Aljami, H.A.; Alhatmi, H.; Alanazi, H.; Alahmadi, M.; Jawhary, A.; Alenazi, M.W.; Almasoud, A.; et al. Safety and immunogenicity of ChAdOx1 MERS vaccine candidate in healthy Middle Eastern adults (MERS002): An open-label, non-randomised, dose-escalation, phase 1b trial. Lancet Microbe. 2022, 3, e11–e20. [Google Scholar] [CrossRef]
  62. University of Oxford. A Study of a New Vaccine against Two Types of Ebola. Available online: https://ClinicalTrials.gov/show/NCT05079750 (accessed on 14 December 2021).
  63. University of Oxford. Safety and Immunogenicity of a Candidate RVFV Vaccine (RVF001). Available online: https://ClinicalTrials.gov/show/NCT04754776 (accessed on 14 December 2021).
  64. University of Oxford. Research Study to Assess New Chikungunya and Zika Vaccines in Healthy Adults in Mexico. Available online: https://ClinicalTrials.gov/show/NCT04440774 (accessed on 14 December 2021).
  65. GlaxoSmithKline. A Study to Evaluate the Safety and Immunogenicity of a Candidate Ebola Vaccine in Adults. Available online: https://ClinicalTrials.gov/show/NCT02485301 (accessed on 14 December 2021).
  66. National Institute of Allergy and Infectious Diseases. cAd3-Marburg Vaccine in Healthy Adults. Available online: https://ClinicalTrials.gov/show/NCT03475056 (accessed on 14 December 2021).
  67. Albert, B.; Sabin Vaccine Institute. Evaluation of Safety, Tolerability and Immune Responses of Ebola-S and Marburg Vaccines in Healthy Adults. Available online: https://clinicaltrials.gov/ct2/show/NCT04723602 (accessed on 14 December 2021).
  68. National Institute of Allergy and Infectious Diseases. Ebola Sudan Chimpanzee Adenovirus Vector Vaccine in Healthy Adults. Available online: https://ClinicalTrials.gov/show/NCT04041570 (accessed on 14 December 2021).
  69. National Institute of Allergy and Infectious Diseases. Safety, Tolerability, and Immunogenicity of the Ebola Chimpanzee Adenovirus Vector Vaccine (cAd3-EBO), VRC-EBOADC069-00-VP, in Healthy Adults. Available online: https://ClinicalTrials.gov/show/NCT02231866 (accessed on 14 December 2021).
  70. Tetherex Pharmaceuticals Corporation. A Phase 1, First-In-Human Study of the Investigational COVID-19 Vaccine SC-Ad6-1 in Healthy Volunteers. Available online: https://ClinicalTrials.gov/show/NCT04839042 (accessed on 14 December 2021).
  71. Bharat Biotech International Limited. Safety and Immunogenicity of an Intranasal SARS-CoV-2 Vaccine (BBV154) for COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04751682 (accessed on 14 December 2021).
  72. Cellid Co., Ltd. Safety and Immunogenicity Study of AdCLD-CoV19: A COVID-19 Preventive Vaccine in Healthy Volunteers. Available online: https://ClinicalTrials.gov/show/NCT04666012 (accessed on 14 December 2021).
  73. ReiThera Srl. Study of GRAd-COV2 for the Prevention of COVID-19 in Adults. Available online: https://ClinicalTrials.gov/show/NCT04791423 (accessed on 14 December 2021).
  74. Biocad. Clinical Study of the Safety and Immunogenicity of a Recombinant Viral Vector AAV5 (Adeno-Associated Virus Type 5)-RBD (Receptor Binding Domain)-S Vaccine for the Prevention of Coronavirus Infection (COVID-19). Available online: https://ClinicalTrials.gov/show/NCT05037188 (accessed on 14 December 2021).
  75. Universitätsklinikum Hamburg-Eppendorf. Safety and Immunogenicity of the Candidate Vaccine MVA-SARS-2-S and a Booster Vaccination with a Licensed Vaccine against COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04569383 (accessed on 14 December 2021).
  76. Universitätsklinikum Hamburg-Eppendorf. Safety, Tolerability and Immunogenicity of the Candidate Vaccine MVA-SARS-2-ST against COVID-19. Available online: https://ClinicalTrials.gov/show/NCT04895449 (accessed on 14 December 2021).
  77. City of Hope Medical Center. SARS-CoV-2 Vaccine (COH04S1) Versus Emergency Use Authorization SARS-COV-2 Vaccine for the Treatment of COVID-19 in Patients with Blood Cancer. Available online: https://ClinicalTrials.gov/show/NCT04977024 (accessed on 14 December 2021).
  78. Universitätsklinikum Hamburg-Eppendorf. Safety, Tolerability and Immunogenicity of Vaccine Candidate MVA-MERS-S. Available online: https://ClinicalTrials.gov/show/NCT03615911 (accessed on 14 December 2021).
  79. Institute of Vaccines and Medical Biologicals Vietnam. A Phase 1/2 Safety and Immunogenicity Trial of COVID-19 Vaccine COVIVAC. Available online: https://ClinicalTrials.gov/show/NCT04830800 (accessed on 14 December 2021).
  80. Cazzola, M.; Rogliani, P.; Mazzeo, F.; Matera, M.G. Controversy surrounding the Sputnik V vaccine. Respir. Med. 2021, 187, 106569. [Google Scholar] [CrossRef] [PubMed]
  81. Zak, D.E.; Andersen-Nissen, E.; Peterson, E.R.; Sato, A.; Hamilton, M.K.; Borgerding, J.; Krishnamurty, A.T.; Chang, J.T.; Adams, D.J.; Hensley, T.R.; et al. Merck Ad5/HIV induces broad innate immune activation that predicts CD8+ T-cell responses but is attenuated by preexisting Ad5 immunity. Proc. Natl. Acad. Sci. USA 2012, 109, 3503–3512. [Google Scholar] [CrossRef] [Green Version]
  82. Teigler, J.E.; Iampietro, M.J.; Barouch, D.H. Vaccination with adenovirus serotypes 35, 26, and 48 elicits higher levels of innate cytokine responses than adenovirus serotype 5 in rhesus monkeys. J. Virol. 2012, 86, 9590–9598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Colloca, S.; Folgori, A.; Ammendola, V.; Capone, S.; Cirillo, A.; Siani, L.; Naddeo, M.; Grazioli, F.; Esposito, M.L.; Ambrosio, M.; et al. Generation and screening of a large collection of novel simian Adenovirus allows the identification of vaccine vectors inducing potent cellular immunity in humans. Sci. Transl. Med. 2012, 4, 115ra2. [Google Scholar] [CrossRef] [Green Version]
  84. Abbink, P.; Lemckert, A.A.; Ewald, B.A.; Lynch, D.M.; Denholtz, M.; Smits, S.; Holterman, L.; Damen, I.; Vogels, R.; Thorner, A.R.; et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J. Virol. 2007, 81, 4654–4663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Custers, J.; Kim, D.; Leyssen, M.; Gurwith, M.; Tomaka, F.; Robertson, J.; Heijnen, E.; Condit, R.; Shukarev, G.; Heerwegh, D.; et al. Vaccines based on replication incompetent Ad26 viral vectors: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2021, 39, 3081–3101. [Google Scholar] [CrossRef]
  86. Sadoff, J.; Gray, G.; Vandebosch, A.; Cárdenas, V.; Shukarev, G.; Grinsztejn, B.; Goepfert, P.A.; Truyers, C.; Fennema, H.; Spiessens, B.; et al. Safety and Efficacy of Single-Dose Ad26.COV2.S Vaccine against Covid-19. N. Engl. J. Med. 2021, 384, 2187–2201. [Google Scholar] [CrossRef] [PubMed]
  87. Alter, G.; Yu, J.; Liu, J.; Chandrashekar, A.; Borducchi, E.N.; Tostanoski, L.H.; McMahan, K.; Jacob-Dolan, C.; Martinez, D.R.; Chang, A.; et al. Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans. Nature 2021, 596, 268–272. [Google Scholar] [CrossRef]
  88. Geisbert, T.W.; Bailey, M.; Hensley, L.; Asiedu, C.; Geisbert, J.; Stanley, D.; Honko, A.; Johnson, J.; Mulangu, S.; Pau, M.G.; et al. Recombinant adenovirus serotype 26 (Ad26) and Ad35 vaccine vectors bypass immunity to Ad5 and protect nonhuman primates against ebolavirus challenge. J. Virol. 2011, 85, 4222–4233. [Google Scholar] [CrossRef] [Green Version]
  89. Dicks, M.D.; Spencer, A.J.; Edwards, N.J.; Wadell, G.; Bojang, K.; Gilbert, S.C.; Hill, A.V.; Cottingham, M.G. A novel chimpanzee adenovirus vector with low human seroprevalence: Improved systems for vector derivation and comparative immunogenicity. PLoS One 2012, 7, e40385. [Google Scholar] [CrossRef] [Green Version]
  90. Van Doremalen, N.; Haddock, E.; Feldmann, F.; Meade-White, K.; Bushmaker, T.; Fischer, R.J.; Okumura, A.; Hanley, P.W.; Saturday, G.; Edwards, N.J.; et al. A single dose of ChAdOx1 MERS provides protective immunity in rhesus macaques. Sci. Adv. 2020, 6, eaba8399. [Google Scholar] [CrossRef]
  91. Falsey, A.R.; Sobieszczyk, M.E.; Hirsch, I.; Sproule, S.; Robb, M.L.; Corey, L.; Neuzil, K.M.; Hahn, W.; Hunt, J.; Mulligan, M.J.; et al. Phase 3 Safety and Efficacy of AZD1222 (ChAdOx1 nCoV-19) Covid-19 Vaccine. N. Engl. J. Med. 2021, 385, 2348–2360. [Google Scholar] [CrossRef]
  92. Swanson, P.A.; Padilla, M.; Hoyland, W.; McGlinchey, K.; Fields, P.A.; Bibi, S.; Faust, S.N.; McDermott, A.B.; Lambe, T.; Pollard, A.J.; et al. AZD1222/ChAdOx1 nCoV-19 vaccination induces a polyfunctional spike protein-specific TH1 response with a diverse TCR repertoire. Sci. Transl. Med. 2021, 13, eabj7211. [Google Scholar] [CrossRef] [PubMed]
  93. Tapia, M.D.; Sow, S.O.; Ndiaye, B.P.; Mbaye, K.D.; Thiongane, A.; Ndour, C.T.; Mboup, S.; Ake, J.A.; Keshinro, B.; Akintunde, G.A.; et al. Safety, reactogenicity, and immunogenicity of a chimpanzee adenovirus vectored Ebola vaccine in adults in Africa: A randomised, observer-blind, placebo-controlled, phase 2 trial. Lancet Infect. Dis. 2020, 20, 707–718. [Google Scholar] [CrossRef]
  94. Woolsey, C.; Geisbert, T.W. Current state of Ebola virus vaccines: A snapshot. PLoS Pathog. 2021, 17, e1010078. [Google Scholar] [CrossRef] [PubMed]
  95. Bricker, T.L.; Darling, T.L.; Hassan, A.O.; Harastani, H.H.; Soung, A.; Jiang, X.; Dai, Y.-N.; Zhao, H.; Adams, L.J.; Holtzman, M.J.; et al. A single intranasal or intramuscular immunization with chimpanzee adenovirus-vectored SARS-CoV-2 vaccine protects against pneumonia in hamsters. Cell Rep. 2021, 36, 109400. [Google Scholar] [CrossRef] [PubMed]
  96. Barry, M. Single-cycle adenovirus vectors in the current vaccine landscape. Expert Rev. Vaccines 2018, 17, 163–173. [Google Scholar] [CrossRef] [PubMed]
  97. Anguiano-Zarate, S.S.; Matchett, W.E.; Nehete, P.N.; Sastry, J.K.; Marzi, A.; Barry, M.A. A Replicating Single-Cycle Adenovirus Vaccine Against Ebola Virus. J. Infect. Dis. 2018, 218, 1883–1889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Kaynarcalidan, O.; Moreno Mascaraque, S.; Drexler, I. Vaccinia Virus: From Crude Smallpox Vaccines to Elaborate Viral Vector Vaccine Design. Biomedicines 2021, 9, 1780. [Google Scholar] [CrossRef] [PubMed]
  99. Callendret, B.; Vellinga, J.; Wunderlich, K.; Rodriguez, A.; Steigerwald, R.; Dirmeier, U.; Cheminay, C.; Volkmann, A.; Brasel, T.; Carrion, R.; et al. A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates. PLoS One 2018, 13, e0192312. [Google Scholar] [CrossRef] [Green Version]
  100. Pollard, A.J.; Launay, O.; Lelievre, J.D.; Lacabaratz, C.; Grande, S.; Goldstein, N.; Robinson, C.; Gaddah, A.; Bockstal, V.; Wiedemann, A.; et al. Safety and immunogenicity of a two-dose heterologous Ad26.ZEBOV and MVA-BN-Filo Ebola vaccine regimen in adults in Europe (EBOVAC2): A randomised, observer-blind, participant-blind, placebo-controlled, phase 2 trial. Lancet Infect. Dis. 2021, 21, 493–506. [Google Scholar] [CrossRef]
  101. Taub, D.D.; Ershler, W.B.; Janowski, M.; Artz, A.; Key, M.L.; McKelvey, J.; Muller, D.; Moss, B.; Ferrucci, L.; Duffey, P.L.; et al. Immunity from smallpox vaccine persists for decades: A longitudinal study. Am. J. Med. 2008, 121, 1058–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Altenburg, A.F.; van Trierum, S.E.; de Bruin, E.; de Meulder, D.; van de Sandt, C.E.; van der Klis, F.R.M.; Fouchier, R.A.M.; Koopmans, M.P.G.; Rimmelzwaan, G.F.; de Vries, R.D. Effects of pre-existing orthopoxvirus-specific immunity on the performance of Modified Vaccinia virus Ankara-based influenza vaccines. Sci. Rep. 2018, 8, 6474. [Google Scholar] [CrossRef] [PubMed]
  103. Zuniga, A.; Wang, Z.; Liniger, M.; Hangartner, L.; Caballero, M.; Pavlovic, J.; Wild, P.; Viret, J.F.; Glueck, R.; Billeter, M.A.; et al. Attenuated measles virus as a vaccine vector. Vaccine 2007, 25, 2974–2983. [Google Scholar] [CrossRef] [PubMed]
  104. Samal, S.K. Paramyxoviruses as Vaccine Vectors. In Viral Vectors in Veterinary Vaccine Development; Springer: Cham, Switzerland, 2020; pp. 113–139. [Google Scholar]
  105. Reisinger, E.C.; Tschismarov, R.; Beubler, E.; Wiedermann, U.; Firbas, C.; Loebermann, M.; Pfeiffer, A.; Muellner, M.; Tauber, E.; Ramsauer, K. Immunogenicity, safety, and tolerability of the measles-vectored chikungunya virus vaccine MV-CHIK: A double-blind, randomised, placebo-controlled and active-controlled phase 2 trial. Lancet 2019, 392, 2718–2727. [Google Scholar] [CrossRef]
  106. Frantz, P.N.; Teeravechyan, S.; Tangy, F. Measles-derived vaccines to prevent emerging viral diseases. Microbes Infect. 2018, 20, 493–500. [Google Scholar] [CrossRef] [PubMed]
  107. Mateo, M.; Reynard, S.; Journeaux, A.; Germain, C.; Hortion, J.; Carnec, X.; Picard, C.; Baillet, N.; Borges-Cardoso, V.; Merabet, O.; et al. A single-shot Lassa vaccine induces long-term immunity and protects cynomolgus monkeys against heterologous strains. Sci. Transl. Med. 2021, 13, eabf6348. [Google Scholar] [CrossRef] [PubMed]
  108. Meyer, M.; Garron, T.; Lubaki, N.M.; Mire, C.E.; Fenton, K.A.; Klages, C.; Olinger, G.G.; Geisbert, T.W.; Collins, P.L.; Bukreyev, A. Aerosolized Ebola vaccine protects primates and elicits lung-resident T cell responses. J. Clin. Investig. 2015, 125, 3241–3255. [Google Scholar] [CrossRef] [Green Version]
  109. Osterholm, M.T.; Moore, K.A.; Kelley, N.S.; Brosseau, L.M.; Wong, G.; Murphy, F.A.; Peters, C.J.; LeDuc, J.W.; Russell, P.K.; Van Herp, M.; et al. Transmission of Ebola viruses: What we know and what we do not know. mBio 2015, 6, e00137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Bukreyev, A.; Marzi, A.; Feldmann, F.; Zhang, L.; Yang, L.; Ward, J.M.; Dorward, D.W.; Pickles, R.J.; Murphy, B.R.; Feldmann, H.; et al. Chimeric human parainfluenza virus bearing the Ebola virus glycoprotein as the sole surface protein is immunogenic and highly protective against Ebola virus challenge. Virology 2009, 383, 348–361. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, Z.; Xu, P.; Salyards, G.W.; Harvey, S.B.; Rada, B.; Fu, Z.F.; He, B. Evaluating a parainfluenza virus 5-based vaccine in a host with pre-existing immunity against parainfluenza virus 5. PLoS One 2012, 7, e50144. [Google Scholar] [CrossRef]
  112. Park, M.S.; Shaw, M.L.; Muñoz-Jordan, J.; Cros, J.F.; Nakaya, T.; Bouvier, N.; Palese, P.; García-Sastre, A.; Basler, C.F. Newcastle disease virus (NDV)-based assay demonstrates interferon-antagonist activity for the NDV V protein and the Nipah virus V, W, and C proteins. J. Virol. 2003, 77, 1501–1511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Pitisuttithum, P.; Luvira, V.; Lawpoolsri, S.; Muangnoicharoen, S.; Kamolratanakul, S.; Sivakorn, C.; Narakorn, P.; Surichan, S.; Prangpratanporn, S.; Puksuriwong, S.; et al. Safety and Immunogenicity of an Inactivated Recombinant Newcastle Disease Virus Vaccine Expressing SARS-CoV-2 Spike: Interim Results of a Randomised, Placebo-Controlled, Phase 1/2 Trial. medRxiv 2021. [CrossRef]
  114. Sun, W.; Liu, Y.; Amanat, F.; Gonzalez-Dominguez, I.; McCroskery, S.; Slamanig, S.; Coughlan, L.; Rosado, V.; Lemus, N.; Jangra, S.; et al. A Newcastle disease virus-vector expressing a prefusion-stabilized spike protein of SARS-CoV-2 induces protective immune responses against prototype virus and variants of concern in mice and hamsters. BioRxiv 2021. [CrossRef]
  115. Fathi, A.; Dahlke, C.; Addo, M.M. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum. Vaccin. Immunother. 2019, 15, 2269–2285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Cooper, D.; Wright, K.J.; Calderon, P.C.; Guo, M.; Nasar, F.; Johnson, J.E.; Coleman, J.W.; Lee, M.; Kotash, C.; Yurgelonis, I.; et al. Attenuation of recombinant vesicular stomatitis virus-human immunodeficiency virus type 1 vaccine vectors by gene translocations and g gene truncation reduces neurovirulence and enhances immunogenicity in mice. J. Virol. 2008, 82, 207–219. [Google Scholar] [CrossRef] [Green Version]
  117. Coller, B.G.; Blue, J.; Das, R.; Dubey, S.; Finelli, L.; Gupta, S.; Helmond, F.; Grant-Klein, R.J.; Liu, K.; Simon, J.; et al. Clinical development of a recombinant Ebola vaccine in the midst of an unprecedented epidemic. Vaccine 2017, 35, 4465–4469. [Google Scholar] [CrossRef] [PubMed]
  118. Grais, R.F.; Kennedy, S.B.; Mahon, B.E.; Dubey, S.A.; Grant-Klein, R.J.; Liu, K.; Hartzel, J.; Coller, B.-A.; Welebob, C.; Hanson, M.E.; et al. Estimation of the correlates of protection of the rVSVΔG-ZEBOV-GP Zaire ebolavirus vaccine: A post-hoc analysis of data from phase 2/3 clinical trials. Lancet Microbe 2021, 2, E70–E78. [Google Scholar] [CrossRef]
  119. Halperin, S.A.; Das, R.; Onorato, M.T.; Liu, K.; Martin, J.; Grant-Klein, R.J.; Nichols, R.; Coller, B.A.; Helmond, F.A.; Simon, J.K.; et al. Immunogenicity, Lot Consistency, and Extended Safety of rVSVΔG-ZEBOV-GP Vaccine: A Phase 3 Randomized, Double-Blind, Placebo-Controlled Study in Healthy Adults. J. Infect. Dis. 2019, 220, 1127–1135. [Google Scholar] [CrossRef]
  120. Dahlke, C.; Kasonta, R.; Lunemann, S.; Krähling, V.; Zinser, M.E.; Biedenkopf, N.; Fehling, S.K.; Ly, M.L.; Rechtien, A.; Stubbe, H.C.; et al. Dose-dependent T-cell Dynamics and Cytokine Cascade Following rVSV-ZEBOV Immunization. EBioMedicine 2017, 19, 107–118. [Google Scholar] [CrossRef] [Green Version]
  121. Monath, T.P.; Fast, P.E.; Modjarrad, K.; Clarke, D.K.; Martin, B.K.; Fusco, J.; Nichols, R.; Heppner, D.G.; Simon, J.K.; Dubey, S.; et al. rVSVΔG-ZEBOV-GP (also designated V920) recombinant vesicular stomatitis virus pseudotyped with Ebola Zaire Glycoprotein: Standardized template with key considerations for a risk/benefit assessment. Vaccine X 2019, 1, 100009. [Google Scholar] [CrossRef]
  122. Kurup, D.; Fisher, C.R.; Scher, G.; Yankowski, C.; Testa, A.; Keshwara, R.; Abreu-Mota, T.; Lambert, R.; Ferguson, M.; Rinaldi, W.; et al. Tetravalent Rabies-Vectored Filovirus and Lassa Fever Vaccine Induces Long-term Immunity in Nonhuman Primates. J. Infect. Dis. 2021, 224, 995–1004. [Google Scholar] [CrossRef]
Table 1. Characteristics of select viruses of pandemic potential.
Table 1. Characteristics of select viruses of pandemic potential.
VirusFatality RateDisease/
Symptoms
Largest OutbreakLicensed
Vaccine
SARS-CoV-20.27% [5]Respiratory illness - acute respiratory distress syndrome (ARDS)COVID-19 pandemic (as of 31 January 2022) [1]
Est. Cases: 373,229,380
Est. Deaths: 5,658,702
Licensed: Yes
SARS-CoV10% [6]Respiratory illness - ARDS2002–2004 SARS pandemic [6]
Est. Cases: 8096
Est. Deaths: 774
Licensed: No
(Clinical stage candidates [7])
MERS-CoV34.3% [8]Respiratory illness - ARDS2012–2019 sporadic outbreaks [8]
Est. Cases: 2499
Est. Deaths: 858
Licensed: No
Ebola virus50%
(25–90% range) [9]
Hemorrhagic fever2013–2016 West Africa epidemic [10]
Est. Cases: 28,616
Est. Deaths: 11,310
Licensed: Yes
Sudan virus55% [11]Hemorrhagic fever2000–2001 Uganda outbreak [11]
Est. Cases: 425
Est. Deaths: 224
Licensed: No
Marburg
virus
53.8% [12]Hemorrhagic fever2004–2005 Angola outbreak [12]
Est. Cases: 252
Est. Deaths: 227
Licensed: No
Lassa virus1–2% [13]Hemorrhagic feverEndemic in West Africa
2020 Nigeria cumulative: [14]
Est. cases: 6732
Est deaths: 244
Licensed: No
Nipah virus61.0% [15]Respiratory symptoms
Encephalitis
1998–1999 Malaysia outbreak [16]
Est. Cases: 265
Est. Deaths: 105
Licensed: No
(Clinical stage candidate [17])
Hendra
virus
57% [18]Respiratory symptoms
Encephalitis
Totality of human cases (1994–2008) [17]
Est. Cases: 7
Est. Deaths: 4
Licensed: No
(Clinical stage candidate for NiV cross-protective for HeV in NHP [16])
Crimean-Congo
hemorrhagic fever virus
26.5% [19]Hemorrhagic feverTurkey, 2002–2009 period [19]
Est. Cases: 4431
Est. Deaths: 222
Licensed: No
(Clinical stage candidate)
Rift Valley fever virus0.5 to 2% [20]Respiratory symptoms
Hemorrhagic fever
Encephalitis
2019 Sudan outbreak [21]
Est. Cases: 1129
Est. Deaths: 123
Licensed: No
Zika virusNon-fatal/
10% for CZS [22]
Fever, arthralgia, maculopapular rash
Congenital Zika syndrome (CZS
2015–2016 Zika epidemic (Region of the Americas)
Est. Cases: 707,133 [23]
Est. Deaths due to CZS (Brazil): 603/6059 CZS cases [22]
Licensed: No
Table 2. Target product profile considerations.
Table 2. Target product profile considerations.
CriteriaObjective
Indication for useFor active immunization of persons considered at-risk of exposure; reactive use in response to outbreaks may be preferrable
Target populationAll adults and pediatrics down to 6 months of age
Safety/ReactogenicitySafety and reactogenicity that provide a favorable benefit-risk profile in context with vaccine efficacy; ideally only mild, transient vaccination-related adverse events (AE) and no vaccine-related serious AEs (SAEs)
Efficacy (clinical)Greater than 90% efficacy in preventing infection or disease in healthy adults (70% minimum).
If demonstration of clinical efficacy is not feasible, pre-clinical immunogenicity and efficacy in a standardized and relevant animal model together with clinical immunogenicity may be considered.
Efficacy (nonclinical based on Animal Rule)Demonstration of protection in relevant animal models in line with FDA Animal Rule guidance
Onset to protectionRapid onset to protection within two weeks after first dose
Duration of protectionPrimary series confers long-lasting protection of 1 year or more, and can be maintained by booster doses
Dosing regimenSingle-dose primary series
Route of administrationInjectable (IM, ID, or SC) using standard volumes suitable for a single injection, but oral or other needle-free approaches would be preferred.
Storage temperature Room temperature > 2–8 °C > −20 °C
Table 3. Approved and clinical-stage viral vector vaccine candidates for viruses of interest.
Table 3. Approved and clinical-stage viral vector vaccine candidates for viruses of interest.
ReplicationVectorTargetVaccineStatus
ReplicatingVSVSARS-CoV-2VSV-ΔG SARS-CoV-2Phase 2/3 active [41]
MERS-CoV-2BVRS-GamVac-Combi
(Ad5 & VSV)
Phase 1/2 active [42]
EBOVrVSVΔG-ZEBOV-GPApproved [43]
GamEvac-Combi & GamEvac-Lyo
(Ad5 & VSV)
Licensed (Russia) [43]
LASVrVSV∆G-LASV-GPCPhase 1 active [44]
MeVLASVMV-LASVPhase 1 complete [45]
ZIKVMV-ZIKA & MV-ZIKA-RSPPhase 1 complete [46,47]
HPIV3EBOVHPIV3-EbovZ GPPhase 1 complete [48]
PIV5SARS-CoV-2CVXGA1-001Phase 1 active [49]
LAIVSARS-CoV-2DelNS1-2019-nCoV-RBD-OPT1Phase 3 active [50]
Non-replicatingAd5SARS-CoV-2Ad5-nCoVAuthorized (China) [51]
VXA-Cov2-1Phase 2 active [52]
hAd5-S-Fusion+N-ETSDPhase 1/2 active [53]
Ad5-triCoV/MacPhase 1 active [54]
MERS-CoVMERS BVRS-GamVacPhase 1/2 active [55]
MERS BVRS-GamVac-Combi
(Ad5 & VSV)
Phase 1/2 active [42]
EBOVAd5-EBOVLicensed (China) [43]
GamEvac-Combi & Lyo
(Ad5 & VSV)
Licensed (Russia) [43]
Ad26SARS-CoV-2Ad26.COV2.S Authorized [56,57]
Sputnik LightApproved (Russia) [58]
EBOVAd26.ZEBOV-GP
(in Zabdeno/Mvabea vaccine)
Authorized [43]
ZIKVAd26.ZIKV.001Phase 1 complete [59]
ChAdOx1SARS-CoV-2ChAdOx1-SAuthorized [60]
MERS-CoVChAdOx1 MERSPhase 1 complete [61]
EBOV/SUDVChAdOx1 biEBOVPhase 1 active [62]
RVFVChAdOx1 RVFPhase 1 active [63]
ZIKVChAdOx1 ZikaPhase 1 active [64]
CAd3EBOVChAd3-EBO-ZPhase 2 complete [65]
MARVcAd3-MARVPhase 1 complete [66,67]
SUDVcAd3-EBO SPhase 1 complete [67]
EBOV/SUDVcAd3-EBOPhase 1 complete [68]
ChAd68SARS-CoV-2ChAdV68-SPhase 1 active [69]
Ad6 single cycleSARS-CoV-2SC-Ad6-1Phase 1 active [70]
Ad36SARS-CoV-2BBV154Phase 1 active [71]
Ad5/35SARS-CoV-2AdCLD-CoV19Phase 1/2 active [72]
GRAd32SARS-CoV-2GRAd-COV2Phase 2/3 active [73]
AAV5SARS-CoV-2AAV5-RBD-SPhase 1/2 active [74]
MVASARS-CoV-2MVA-SARS-2-SPhase 1 complete [75]
MVA-SARS2-STPhase 1 active [76]
COH04S1Phase 2 active [77]
MERS-CoV-2MVA-MERS-SPhase 1 complete [78]
Filovirus
multivalent
MVA-BN-Filo
(in Zabdeno/Mvabea vaccine)
Authorized [43]
NDVSARS-CoV-2NDV-HXP-S +/− CpG 1018Phase 1/2 active [79]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hofmeyer, K.A.; Bianchi, K.M.; Wolfe, D.N. Utilization of Viral Vector Vaccines in Preparing for Future Pandemics. Vaccines 2022, 10, 436. https://doi.org/10.3390/vaccines10030436

AMA Style

Hofmeyer KA, Bianchi KM, Wolfe DN. Utilization of Viral Vector Vaccines in Preparing for Future Pandemics. Vaccines. 2022; 10(3):436. https://doi.org/10.3390/vaccines10030436

Chicago/Turabian Style

Hofmeyer, Kimberly A., Katherine M. Bianchi, and Daniel N. Wolfe. 2022. "Utilization of Viral Vector Vaccines in Preparing for Future Pandemics" Vaccines 10, no. 3: 436. https://doi.org/10.3390/vaccines10030436

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

Article Metrics

Back to TopTop