6,472
Views
44
CrossRef citations to date
0
Altmetric
Letter

Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens

ORCID Icon, ORCID Icon &
Pages 2269-2285 | Received 09 Apr 2019, Accepted 19 Jul 2019, Published online: 05 Sep 2019

ABSTRACT

The devastating Ebola virus (EBOV) outbreak in West Africa in 2013–2016 has flagged the need for the timely development of vaccines for high-threat pathogens. To be better prepared for new epidemics, the WHO has compiled a list of priority pathogens that are likely to cause future outbreaks and for which R&D efforts are, therefore, paramount (R&D Blueprint: https://www.who.int/blueprint/priority-diseases/en/). To this end, the detailed characterization of vaccine platforms is needed. The vesicular stomatitis virus (VSV) has been established as a robust vaccine vector backbone for infectious diseases for well over a decade. The recent clinical trials testing the vaccine candidate VSV-EBOV against EBOV disease now have added a substantial amount of clinical data and suggest VSV to be an ideal vaccine vector candidate for outbreak pathogens. In this review, we discuss insights gained from the clinical VSV-EBOV vaccine trials as well as from animal studies investigating vaccine candidates for Blueprint pathogens.

This article is part of the following collections:
Vaccines for Emerging Diseases

Introduction

Emerging infectious diseases are a major threat to public health. Ebola virus (EBOV) and other high-threat pathogens have recently been declared one of the top ten health threats of 2019 by the World Health Organization (WHO).Citation1 Out of the five Public Health Emergencies of International Concern (PHEIC) that WHO has declared to date, four have been caused by emerging or re-emerging viruses. The last few years, humanity experienced novel and increasingly frequent outbreaks for which timely and efficient countermeasures were lacking. Between 2013 and 2016, more than 11,000 people lost their lives to the dramatic West African Ebola virus disease epidemic. In 2015, the Middle East Respiratory Syndrome coronavirus (MERS-CoV) demonstrated to the world how rapidly a virus can spread into new regions due to travel, when a 68-year-old Korean man returning from the Middle East was infected by MERS-CoV and became the index case for a chain of secondary mainly nosocomial transmissions cumulating in 138 MERS-CoV infections, including 36 deaths.Citation2 The 2016 Zika virus outbreak in the Americas and its resulting cluster of associated neurological disorders and neonatal malformations,Citation3 also was declared a PHEIC, to name just a few recent outbreaks that emphasize the urgency to create strategies to contain future outbreaks fast and effectively. In the future, socioeconomic factors including an increased global human population and lifespan, urbanization, intensified global travel and mobility and the effects of climate change will further ripen conditions for the emergence and rapid spread of viruses between and among populations. Vaccines are the most effective way to prevent and control this viral spread; however, few vaccine candidates for emerging infections exist. As a result of this phenomenon of supply vacuum, novel viral outbreaks such as the EBOV crisis found the global medical community ill-prepared, and the deployment of a preventive vaccine was delayed due to the lack of clinical-stage vaccine candidates.

In response, WHO initiated a Blueprint list of priority diseases to accelerate Research & Development efforts for pathogens that bear a particularly high potential to cause epidemics while no or limited specific countermeasures are available and may, therefore, cause future public health emergencies.Citation4 WHO’s R&D blueprint, composed and regularly updated by an international committee of experts, identifies and defines priority pathogens on the basis of a number of criteria weighted by their relative importance. Major prioritization criteria focus on whether and how the pathogen is transmitted to humans, the extent of medical countermeasures available, and the severity and case-fatality rate of the corresponding disease. Other factors such as potential societal impacts and the evolutionary potential of the pathogen are also taken into account.Citation5 The current Blueprint, reviewed in February 2018, identified as present priority diseases Crimean-Congo Hemorrhagic Fever (CCHF), Ebola Viral Disease (EVD) and Marburg Viral Disease (MVD), Lassa Fever, MERS and Severe Acute Respiratory Syndrome (SARS), Nipah and henipaviral diseases, Rift Valley Fever (RVF), Zika disease as well as “disease X”, a – yet unknown – disease.Citation6

In this context, the detailed characterization and assessment of vaccine platforms capable of being applied to swiftly develop vaccines against a variety of pathogens are of utmost importance. The ideal vaccine candidate for outbreak scenarios should be able to be manufactured in a rapid and scalable fashion, be safe, induce both strong cellular and humoral immune responses and rapidly confer long-term immunity after a single immunization. Viral vector vaccines can express a variety of heterologous antigens and, therefore, represent ideal vaccine platforms for developing novel vaccine candidates. As such, vaccine candidates based on the Rhabdovirdae family have shown to feature many of these properties.

Vesicular stomatitis virus (VSV) is a negative-stranded RNA virus and a member of the Rhabdoviridae family, consisting of a simple genome organization encoding five structural proteins: the nucleoprotein (N), phosphoprotein (P), matrix (M), glycoprotein (G) and the RNA-dependent RNA polymerase (L).Citation7 During the last decade, the recombinant VSV (rVSV) platform has been tested for multiple emerging and neglected viral diseases as well as for therapeutic cancer vaccines in animal studies.Citation8

Based on the successful establishment of a method to generate VSV from DNA by Rose et al., effective VSV cloning strategies have been implementedCitation9 and multiple studies revealed the capacity of VSV to express foreign antigens and thus its potential to be used as vaccine candidates.Citation10Citation13 Benefits of the rVSV platform include  (reviewed inCitation14,Citation15):

  1. fast production of high titers and its propagation in almost all mammalian cells;

  2. lack of reassortment and corresponding potential to undergo genetic shift in vivo;

  3. inability of the vector’s viral RNA to integrate into the host genome;

  4. simple genetic modification with the possibility to accommodate one or multiple antigenic inserts;

  5. the low seroprevalence in the human population;

  6. mild pathogenicity in humans;

  7. induction of humoral and cellular immune responses.

A commonly used VSV vaccine design strategy utilizes an rVSV vector lacking VSV-G – rVSVΔG – which is modified to encode the glycoprotein (GP) of the pathogen of interest in replacement of VSV-G, expressing the foreign GP on the viral membrane.Citation15 Besides being an efficient and stable way to insert antigens of interest, this approach holds two major advantages. It serves as an attenuation factor, as the pathogenicity of wild-type VSV has largely been attributed to VSV-G. An exchange of glycoproteins may affect cell-tropism and, therefore, additionally, attenuate the vaccine candidate.Citation14,Citation16,Citation17 Toxicity and pathogenicity are thereby significantly decreased leading to a more benign safety profile. Simultaneously, antibody responses to wild-type VSV are mostly directed against VSV-G in humans and, therefore, eliminating VSV-G is an efficient way to significantly reduce preexisting vector-specific immunity.Citation18 It, however, needs to be considered that rVSV is a replication competent vector and that vulnerable populations including immunocompromised individuals, pregnant women, infants and the elderly may be at higher risk for adverse events.

The present review now endeavors to summarize pre-clinical and clinical data of VSV vaccine candidates against emerging infectious diseases. We here specifically focus on those viruses listed by WHO as prioritized pathogens in the R&D Blueprint, and for which VSV vector vaccine candidates have been described: Ebola, Marburg, Lassa, Nipah, Zika, and CCHF viruses as well as MERS- and SARS-coronaviruses. We place a particular emphasis on the Ebola vaccine candidate VSV-EBOV, as this vaccine is currently the most advanced candidate of the rVSV platform and the only one that has entered human clinical trials besides an rVSV-vectored HIV vaccine candidateCitation19 and an oncolytic vaccine (NCT02923466, both in phase I).

Ebola virus

Ebola virus (EBOV), in the genus of Ebolavirus and the family of Filoviridae, is one of the six ebolavirus species, Sudan ebolavirus (SUDV), Taï Forest ebolavirus (TAFV, previously Côte d’Ivoire ebolavirus), Reston ebolavirus (RESTV), Bundibugyo ebolavirus (BDBV),Citation20 and the newly discovered Bombali virus (BOMV).Citation21 EBOV is the etiological agent of EVD, which can result in hemorrhagic fever and multi-organ failure. The high case-fatality rate and potential threat posed by EBOV not only as an emerging virus but also as a potential bioterrorism agent, led the first research efforts on vaccine candidates against EBOV to be initiated more than a decade ago. However, when the West African EVD epidemic was declared a PHEIC by WHO on August 8th, 2014,Citation22 swift development of a preventive vaccine became an absolute priority. In light of this development, today’s most clinically advanced rVSV-vectored vaccine candidate – and the most advanced EBOV vaccine candidate to date – has entered clinical trials at an unprecedented pace. The vaccine has been known under multiple names, such as rVSVΔG-ZEBOV-GP, rVSV-ZEBOV, VSV-EBOV, and V920. For reason of uniformity, VSV-EBOV is used to reference this vaccine throughout this review.

VSV-EBOV is derived from a chimeric VSV Indiana strain, in which VSV G is replaced by the transmembrane GP of Ebola virus (formerly Zaire ebolavirus (ZEBOV), now designated EBOV),Citation20 derived from the Kikwit strain. VSV-EBOV was first described by a group led by Heinz Feldmann and Ute Stroeher at the National Microbiology Laboratory of Canada’s Public Health agency. While the objective of the study was to assess EBOV GP pathogenicity in mice using VSV as a vector to express EBOV GP, the researchers found that inoculated mice did not develop EVD, and did, in fact, not show any symptoms after challenge with a mouse-adapted EBOV strain.Citation17,Citation23 In a following non-human primate (NHP) study, vaccinated animals were completely protected from homologous challenge,Citation24 and subsequent studies in NHPs provided further evidence for the tolerability and efficacy of the vaccine, administered via several routes of administration – orally, intranasally or intramuscularly, and also when receiving a high-dose aerosol EBOV challenge.Citation25,Citation26 Furthermore, in mice, even doses of as low as two plaque-forming units resulted in protection.Citation27 To date, VSV-EBOV has been described to be immunogenic and to elicit high titers of binding and neutralizing antibodies over a wide dose range in small and large animal models as well as in humans (reviewed in Monath et al.).Citation16

In 2016, the results of phase I clinical trials generated by the VEBCON (VSV-EBOV consortium with four independent, yet harmonized investigator-initiated trials in Switzerland, Germany, Gabon and Kenya) provided a first safety and immunogenicity assessment of VSV-EBOV in humans.Citation28 Results of four more phase I trials in the United States and Canada were reported simultaneously (Regules et al., NEJM 2016) or shortly thereafter.Citation29Citation31 To date, a total of 17 phase I-III clinical trials have been completed or are ongoing (summarized in and ).Citation16,Citation32 VSV-EBOV is furthermore the only vaccine that is being deployed during the 10th EBOV outbreak in the Democratic Republic of the Congo (DRC). This outbreak now constitutes the worst EBOV outbreak in the history of the country and the second-worst outbreak globally, and has consequently been recognized as a PHEIC on July 17th, 2019. The recent developments emphasize the importance of a detailed evaluation of the ample pre-clinical, clinical and field data on VSV-EBOV gathered to date in order to ultimately expedite licensure and facilitate vaccine supply.

Table 1. VSV-based EBOV vaccine clinical phase I trials

Table 2. VSV-based EBOV vaccine clinical phase II, III and observational trials

Clinical dose-finding and vaccination strategy

In clinical trials, various dose levels have been assessed, ranging from 3 × 103 plaque-forming units (PFU) to 1 × 108 PFU.Citation32 Considering both risk and benefit profiles evaluated in human dose-finding phase I trials, a dose of 2 × 107 PFU was determined as the preferred doseCitation30 and was consecutively used in phase II and III trialsCitation33Citation37 as well as in WHO’s vaccination strategy during subsequent EBOV outbreaks. Due to the high demand for vaccine doses in the growing DRC outbreak and, consecutively, to a relative vaccine shortage, WHO’s Strategic Advisory Group of Experts (SAGE) on Immunization has now, however, revised this recommendation for the current outbreak. Potency data from the currently deployed vaccine lots show that a 2-fold dosage reduction would on average still approximate a concentration of 2 × 107 PFU, and, therefore, a 50% reduced vaccine dose is recommended to be given to high-risk contacts (including health-care workers as well as contacts and contacts of confirmed EVD cases). In addition, a vaccine with a 5-fold dose reduction is recommended to be utilized more widely (i.e. in broader populations in which EVD cases occurred), potentially preventing tertiary EVD cases more rapidly.Citation38 Efficacy data for this dosage, however, need to yet be generated.

In all clinical trials, VSV-EBOV was administered intramuscularly and primarily as a single-shot regimen – a regimen that had previously been shown to be protective soon after administration in NHP.Citation15,Citation24Citation26 These tested single-shot regimens are indispensable in outbreak situations, especially in volatile regions with insufficient infrastructure where follow-up vaccinations are often not possible. Yet, in comparison, prime-boost strategies utilizing VSV-EBOV may be beneficial when durability of the vaccine response needs to be expanded. First data have been gathered in a phase I trial, where a subgroup of 30 individuals received a prime-boost regimen (days 0 and 28) of VSV-EBOV at three different dose levels. The vaccinees developed higher antibody (Ab) and virus neutralization titers compared to participants who only received a single-shot; yet these observations were no longer statistically significant six months after initial vaccination.Citation29 Prime-boost regimens are further evaluated in ongoing studies: The PREVAC study is presently assessing a prime only as well as a prime-boost regimen (at days 0 and 56) of VSV-EBOV in comparison to other vaccine strategies in an estimated 4500 volunteers, including children from 1 year of age, with a follow-up time of 12 months. Another ongoing study, the PREPARE study investigates immune response durability and vaccine efficacy is investigated in persons at occupational risk of EBOV exposure, such as health-care workers and laboratory personnel, across a three-year timeframe. Here, participants are either vaccinated with a single shot of VSV-EBOV or receive a booster immunization 18 months after initial vaccination. In addition, participants whose antibody levels decrease beneath a predefined threshold will also be offered a booster vaccination.

Safety data

As studies published to date differ widely across variables such as size, study population characteristics, dosage, etc., a comprehensive concluding safety assessment has not been made. Nonetheless, VSV-EBOV has been described as safe and overall well tolerated, while also being reactogenic in the clinical studies published to date. The adverse events documented were generally mild to moderate and transient. Importantly, there were no vaccine-related serious adverse events (SAE), other than one case of fever and two allergic reactions (one of them when the vaccine was co-administered with amoxicillin) – all which were resolved without sequelae – in now over 18,000 individuals who have received the vaccine in clinical trials.Citation16,Citation33 Besides local reactogenicity, which was reported in up to 90–100% of the volunteers,Citation28,Citation29 the most frequently reported adverse events were systemic solicited reactions such as headaches, which were reported in 21–71%, fever in 10–50%, and fatigue in 12–50% of vaccinees in all published phase II/III studies.Citation33-Citation37 An initially unexpected adverse event constituted the emergence of arthritis cases that tested PCR-positive for rVSV in the phase I trial in Geneva, Switzerland. The study was halted when 11/51 volunteers developed oligoarthritis after having received 1 × 107 PFU or 5 × 107 PFU of VSV-EBOV and was ultimately resumed with a reduced dose of 3 × 105 PFU. Arthritis occurred on median 11 days after vaccination and lasted for 8 days. The development of arthritis did, however, not correlate with the dosage, as 13/51 individuals who had received the reduced dose likewise developed arthritis. In the reduced dose cohort, the risk of arthritis furthermore correlated with increasing age.Citation39 It was also observed that its development correlated with higher vaccine efficacy.Citation40 Three affected participants also developed rVSV-PCR positive skin lesions, with infectious rVSV recovered from one participant. After the study halt, simultaneously running phase I studies of VSV-EBOV included arthralgia/arthritis as a solicited adverse event, but while transient arthralgia was likewise reported, the high frequency of arthritis cases was not observed to the same extent.Citation28Citation31,Citation41 Finally, arthritis frequency was assessed in a large placebo-controlled phase III trial. Here, the incidence of arthritis or joint swelling was 4.9% in a study population of 1050 individuals. Its onset and duration was comparable to the observations in the Geneva trial, and again, age was associated with a higher incidence, which was more pronounced in the lower dose cohort (here: 2 × 107 PFU vs. 1 × 108 PFU).Citation35 The first emerging data from a small subset of 90 surveyed individuals vaccinated during the current EVD outbreak in the DRC seem to mirror the safety assessment obtained in these clinical trials, and while adverse events were frequent, arthralgia and rash were reported in only 7% and 5%, respectively.Citation42 Importantly, vaccine satisfaction was reported to be high, which is a very encouraging finding, as misconceptions about the vaccine and EVD constitute a serious challenge for health-care workers to administer vaccinations and care in the current outbreak setting.

Besides the assessment of adverse events, early clinical studies also focused on evaluating potential viral shedding by vaccinated individuals via testing of saliva and urine. In the NHP model, viral shedding was previously not observed.Citation24 In humans, rVSV viremia could be detected early after vaccination with resolution 3–5 days p.v.; however, viral shedding was extremely rare and presented in individuals with very high viremia.Citation31 Notably, children (6–12 years of age) and, less pronouncedly, adolescents were found to have higher viremia and shedding frequency than adults when vaccinated with 2 × 107 PFU of VSV-EBOV, raising the question whether lower vaccine doses might be required in pediatric populations.Citation41

Vaccine assessment in vulnerable populations

It is important to note that vaccine safety was primarily assessed in healthy adults. Pregnant women and children are, however, disproportionally affected by EVD and safety data from these populations are scarce but essential.

Until now, a small number of children have been included in clinical trials. Despite the observed aforementioned higher viremia in a total of 40 pediatric individuals by Agnandji and colleagues, the vaccine had a comparable safety and immunogenicity profile to that of adults receiving the same dose.Citation41 A subsequent phase III trial as well as vaccine usage under expanded access showed an acceptable safety profile in over 500 vaccinated children aged 6–17.Citation33,Citation43 Unpublished and ongoing trials are now also including children aged 1 year and above (Médecins Sans Frontières (MSF) (NCT03161366) and PREVAC trials) and will hopefully provide more data on safety and efficacy of VSV-EBOV in the pediatric population.

To date, pregnant women have been excluded from VSV-EBOV vaccine trials due to the safety concerns of administering a live-attenuated vaccine to this vulnerable population. At the same time, mortality rates are exorbitantly high with a lethality of around 90% in EVD-affected pregnant women and roughly 100% for the unborn children. Of those pregnant women who have inadvertently received the vaccine, no case outcomes have been published to date. However, 43 women reported pregnancies within 2 months of receiving the vaccine in a controlled phase II/III trial. Reassuringly, in this cohort pregnancy losses did not occur more often than in the control cohort and no congenital anomalies were observed.Citation37 The policy that pregnant women are not being offered the possibility of receiving VSV-EBOV has been a topic of repeated debate and controversy.Citation44,Citation45 MSF among others, have strongly argued in favor of inclusion of pregnant women, given the enormously high probability of death once infected. In view of the above, the Congolese Ministry of Health, backed by WHO, has recently announced a change of this policy, offering pregnant and lactating women as well as children under the age of 12 months who are contacts of identified EVD cases the vaccine as of June 15th Citation46,Citation47 which is not only vital for the protection of this population, but is also an opportunity to generate data on vaccine safety and efficacy in this group and will be of great value in future outbreaks.

As EVD outbreaks occur in a population with high HIV incidence rates, it is all the more important to evaluate how safe the vaccine is in immunocompromised individuals. VSV-EBOV was reported to be tolerable in severely immunocompromised NOD-SCID miceCitation48 and was consecutively tested in a simian-human immunodeficiency (SHIV) model. SHIV infected macaques did not develop overt symptoms such as fever after vaccination with 1 × 107 PFU of VSV-EBOV, a dosage previously shown to be protective in NHPs. The immunogenicity of the vaccine was limited, and EBOV-GP-specific antibodies could not be detected prior to challenge. When challenged with a lethal dose of EBOV 31 days post-vaccination (p.v.), 4/6 of vaccinated animals showed clinical signs of illness and two of them ultimately died. These two animals had been most affected by SHIV as they displayed the highest SHIV viremia and had the lowest CD4+ T cell counts in the group. Also, unlike the survivors, they did not show VSV viremia after vaccination.Citation49 This study underlines the need for a further assessment of VSV-EBOV in immunocompromised individuals and probably for adapting vaccine dosage and schedule. One phase II trial of VSV-EBOV to date has included a small subgroup of 22 HIV-positive individuals: no SAE was reported in HIV-positive participants within one month of vaccination, but immunogenicity as measured by antibody responses was found to be lower as compared to HIV-negative individuals.Citation34 To generate data in a larger cohort, the ongoing ACHIV phase II trial is specifically examining the safety and immunogenicity of VSV-EBOV in an estimated 200 HIV-positive individuals.

Efficacy of VSV-EBOV and utilization during outbreaks

VSV-EBOV contains the GP of the EBOV Kikwit strain, yet, in an NHP model, it showed cross-protection against the heterologous West African outbreak strain of EBOV Makona.Citation50 The subsequent phase III trial “Ebola Ça Suffit” is the only completed clinical trial reporting human efficacy data of VSV-EBOV to date – and, as such, is the only trial reporting efficacy data of any EVD vaccine at the time of this review. The study was conducted towards the end of the West African epidemic and has yielded highly encouraging results. The trial utilized a ring-vaccination strategy: a cluster of contacts and contacts-of-contacts around a case of EVD was identified and randomized to receive either immediate or delayed vaccination after 21 days, the duration of one incubation period reported for EBOV, and EVD cases from day 10 p.v. were assessed.Citation33,Citation51 As the study reported a vaccine efficacy of 100%, randomization was lifted and all ring contacts were offered the vaccine, now also including children. None of the 3796 immediately vaccinated individuals developed EVD from 10 days post vaccination (p.v.), while EVD occurred in 16/2041 contacts of the delayed vaccination cohort plus in seven never vaccinated contacts. The perfect efficacy rate had initially been questioned and spurred a controversial debate since the trial design was unblinded and, as the number of EVD cases had already decreased, only few EVD cases were reported in the delayed vaccination cohort. The authors’, however, rejected the notion that trial results were biased due to the open trial design.Citation52,Citation53

On the basis of these efficacy data, VSV-EBOV has received Priority Medicine (PRIME) designation by the European Medical Association (EMA) as well as Breakthrough Therapy designation by the Food and Drug Administration (FDA), which enables a faster licensure process. The submission of a rolling licensure application to the FDA has been initiated by the manufacturer Merck in November 2018.Citation54 Owing to its PRIME and Breakthrough Therapy designation, VSV-EBOV has been pre-approved for emergency use during outbreaks by SAGE as part of its Expanded Access framework (also called compassionate use program), while pending licensure.Citation55 Under the framework, WHO provides the vaccine to consenting at-risk individuals, health-care workers as well as ring contacts outside of clinical studies.Citation55,Citation56 In a new flare-up of EVD in Guinea in March 2016, after the end of the West African epidemic was declared, the framework was utilized to vaccinate over 1500 individuals including children from the age of 6, and no EVD cases were reported in the vaccinated population.Citation57 Another 3481 people were vaccinated during the 9th EBOV outbreak in the DRC in May and June 2018.Citation58 During the current 10th EBOV outbreak in the DRC, which was declared on August 1st, 2018 and has claimed the lives of 2592 humans as of July 21st, 2019, VSV-EBOV has been administered a total of 171,052 times since the initiation of vaccination seven days later.Citation59

In addition to WHO, MSF is also providing the vaccine using the ring vaccination strategy and has assessed vaccine efficacy in a clinical trial including 500 individuals from the age of one. The trial was reportedly completed on November 30, 2018 (clinicaltrials.gov, NCT03161366) and the results are awaited with great anticipation. Meanwhile, WHO recently reported highly encouraging preliminary results of the ring vaccination regimen applied during the recent DRC EBOV outbreaks including a total of 93,965 vaccinated individuals and reported an estimated vaccine efficacy of 97.5%, thus strongly supporting the findings of the phase III trial.Citation60

Stability of the vaccine

In the context of the deployment of VSV-EBOV in the field setting, the storage of the vaccine, which must be kept at a temperature of −70°C maximum, remains a challenge. The establishment and maintenance of cold chains in often rural surroundings with suboptimal infrastructure has been difficult.Citation61 To date, stability data remain scarce, yet one study found that the vaccine stayed stable with comparable viral titers for 7 days when stored at +4°C.Citation62 In a guinea-pig challenge model, vaccination with VSV-EBOV conferred protection from disease even when stored in various suboptimal conditions over 7 days (4°C, room temperature, 32°C, or three cycles of freeze-thawing from −80°C to room temperature), and only the vaccine kept at 32°C showed a significant reduction in virus titer.Citation63 The data indicate that short-term interruptions of the cold chain may be tolerated without affecting vaccine efficacy.

Correlates of protection and duration of immune responses

Understanding the correlates of protection in EVD remains a challenge. There is still no biomarker that associates with EBOV vaccine efficacy. The identification of EBOV-associated correlates of protection is challenging as markers can only be defined when EBOV incidence is sufficiently high such as during outbreaks. While the “Ebola Ça Suffit!” trial provided strong evidence for high protection of VSV-EBOV, blood samples to analyze immunogenicity and protection markers could not be generated during this trial.

First insights into possible correlates of protection have been provided by assessing immune responses of survivor and fatal cases from EBOV-infected individuals following the West African EVD epidemic. One study demonstrated that fatal cases were associated with an increased expression of the exhaustion markers CTLA-4 and PD-1 on CD4+ and CD8+ T cells.Citation64 A transcriptomic analysis of blood RNA from EBOV-infected patients showed elevated expression levels of genes associated with interferons and acute phase signaling pathways.Citation65 Lower levels of inflammation correlated with virus clearance and robust EBOV-specific T-cell responses. Although these novel data provide insight into innate and adaptive immune responses, their contribution to protection in humans is still not defined. Notably, correlates of protection may differ between natural EVD infection and vaccine-induced immune responses.

Studies comparing immune responses in EVD survivors are currently ongoing (César Muñoz-Fontela, Florian Klein personal communication).

Animal models can be crucial to decipher immune response mechanisms important for protection. In the past, guinea pig as well as non-human-primate EBOV models were used to unravel protective mechanisms. Andrea Marzi and colleagues comprehensively analyzed EBOV challenges in cynomolgus macaques after vaccination. Depletion methods, in which CD4+ and CD8+ T cells as well as B-cells were blocked, revealed a critical role for EBOV GP-specific antibodies.Citation66 The pivotal role of VSV-EBOV-induced innate immunity in conferring early antiviral control and generating effective humoral immune responses was demonstrated in an NHP challenge model. Transcriptional whole blood profiling from vaccinated animals challenged at different timepoints (28, 21, 14, 7 and 3 days p.v.) uncovered that genes associated with antiviral innate responses were upregulated at early timepoints, and were delayed in an animal that did not survive challenge at day 3 p.v.Citation50,Citation67

The collection of samples obtained during the West African epidemic and the ongoing outbreak in the DRC together with numerous individuals vaccinated with VSV-EBOV can now generate a highly valuable body of information to fill critical knowledge gaps regarding mechanisms crucial for immune protection. A first meta-analysis has now been published by Gross and colleagues summarizing antibody responses of all clinical trials in which VSV-EBOV was tested.Citation68 All studies observed induction of EBOV GP-specific antibody responses following immunization with VSV. Antibody responses developed at 14 days p.v., peaked around day 28 and remained detectable for at least 2 years.Citation28,Citation29,Citation40 Besides evaluating humoral responses, clinical trials also evaluated all other arms of immune responses. One study focused on T follicular helper (TFH) cells and observed a correlation with antibody titers.Citation69 Cellular immune responses were observed in the cohort receiving a dose of 2 × 107 PFU. Here, EBOV GP specific T-cells secreted mainly CD107a and TNFα, while lower dose groups showed antigen-specific T-cell responses.Citation70 Recent advances in our understanding of the innate immune system and the use of systems biology approaches are beginning to reveal the fundamental mechanisms by which the innate immune system orchestrates protective immune responses to vaccination. We investigated innate immune responses following VSV-EBOV immunization and our data revealed an innate immune signature that correlated with antibody responses in the Hamburg VSV-EBOV vaccine trial.Citation71 In addition, changes in expression levels of circulating miRNAs have been associated with increased EBOV GP-specific antibody titers. Furthermore, innate immune signatures were assessed in vaccinees from Europe and Africa, revealing an early induction at day 1 p.v. of chemokines and cytokines, associated with monocyte functionality.Citation72

Comprehensive evaluations of immunogenicity following VSV-EBOV vaccination are still ongoing. Systems vaccinology approaches in which clinical, immunological, transcriptomic and metabolomic data from clinical vaccine studies can now be integrated and analyzed with data from survivors will be instrumental to further identify EBOV vaccine correlates of protection.

Experience in use as post-exposure prophylaxis

VSV-EBOV is intended for use as a prophylactic vaccine; however, its implementation as a post-exposure prophylaxis is intriguing, as a swift identification and vaccination of EVD-affected individuals – if effective – would contribute to a timely containment of future outbreaks. In NHPs, VSV-EBOV was fully protective when given 7 days before challenge and ameliorated disease when given 3 days pre-exposure.Citation50 In mice, vaccination with VSV-EBOV 24 h post-infection (p.i.) conferred complete protection, while guinea pigs only showed partial protection from challenge when vaccinated 24 h pre- to 24 h post-exposure. Lastly, in NHP, 50% protection was conferred in animals vaccinated 20–30-min post-exposure.Citation73 In line with this finding, a separate study described an overall 50% protection from EVD when NHP were vaccinated either 1 h or 24 h p.i. or when vaccinated twice, at 1 h and 24 h p.i. with half the vaccine dose, respectively. Surprisingly, however, post-exposure prophylaxis with an rVSV-vectored vaccine against Marburg virus conferred similar protection against EVD, and the authors argued that VSV-driven innate immune activation as well as filovirus-specific immune responses may play a role in protection from EVD.Citation74

Of note, compared to the disease course in humans, the onset of clinical disease is faster and its manifestation more severe in the NHP model, with a lethality of close to 100%. When NHPs were challenged with Sudan ebolavirus (SUDV), which leads to lethal disease in NHP later than does EBOV, 100% of NHP were protected when vaccinated with rVSV-SUDV 20–30 min p.i.Citation75 The efficacy of post-exposure prophylaxis may thus well be higher in humans than in the NHP model.Citation76

In humans, VSV-EBOV was first used as an emergency post-exposure vaccine in 2011, long before entering clinical trials, and has been used in a total of seven individuals with occupational exposure to EBOV. Here, the vaccine was administered 24 h to 3 days post-exposure and none of the individuals developed EVD.Citation77Citation79 It is of course unknown whether, and to what extent, post-exposure prophylaxis prevented the development of EVD, but albeit that these case reports are anecdotal in nature, they are encouraging and provide the basis for current EVD post-exposure recommendations.Citation76

Vector-specific immunity

Although pre-existing anti-VSV immunity is rarely found in humans,Citation8 anti-vector immunity after vaccination with a VSV vector vaccine might impair vaccine efficacy of future VSV-based vaccines and prime-boost regimens. While data investigating pre-existing immunity remain scarce, we recently demonstrated that vaccination with VSV-EBOV induced both vector-specific humoral and cellular immune responses.Citation80 The VSV-specific antibody responses generated in humans in this study following vaccination with VSV-EBOV were non-neutralizing and correlated with EBOV GP-specific antibody responses.Citation80 With regard to the potential simultaneous use of VSV-EBOV and VSV-based Lassa virus (LASV) vaccines in West Africa, Marzi et al. vaccinated cynomolgus macaques with VSV-EBOV 90 days after having received a protective dose of the Lassa vaccine VSVΔG/LASVGPC. Despite having significant anti-VSV antibody titers before vaccination with VSV-EBOV, the vaccination conferred full protection from EBOV challenge, suggesting that a repeated vaccination with VSV-vectored vaccines may be efficacious.Citation81

Future perspectives for VSV-based Ebola vaccines

While clinical trials of VSV-EBOV are still ongoing, efforts to create more attenuated second-generation rVSV-EBOV vaccines are already underway with the goal to reduce reactogenicity while maintaining immunogenicity. In one approach, VSV GP was substituted with an EBOV GP exhibiting the mutation F88A which impairs cell entry. VSV*ΔG(EBOV-GPF88A) induced immunogenicity in guinea pigs comparable to vaccination with rVSV expressing propagation-competent EBOV GP. However, the vaccine construct was found to be genetically unstable and quickly reverted back to a more infectious variant when passaged. Further mutations may, therefore, have to be introduced in order to maintain propagation restriction.Citation82 N4CT1-EBOVGP1 is another attenuated rVSV-EBOV vaccine candidate. The rVSV vector is attenuated by combining a translocation of the N protein from position 1 to 4 (N4) with a truncation of the VSV-G cytoplasmatic tail (CT), and expresses the EBOV GP from the EBOV-Mayinga strain from the first position of the rVSV genome.Citation83 The vector has previously been tested as an HIV vaccine candidate in phase I clinical trials and was well tolerated.Citation19,Citation84 N4CT1-EBOVGP1 vaccinated mice developed both humoral and cellular immune responses and guinea pigs and macaques were completely protected from EBOV challenge, mounting antibody responses in the latter by day 14 p.v.Citation83 Importantly, vaccinated primates were also protected against heterologous challenge with EBOV-Makona.Citation85 N4CT1-EBOVGP1 is licensed by Profectus BioSciences, a biotech company that has specialized in vaccine development and uses the attenuated rVSV vector in a number of vaccine candidates. N4CT1-EBOVGP1 is now being evaluated in a phase I dose-escalation trial (NCT02718469).

Marburg virus

Marburg hemorrhagic fever, caused by Marburg virus (MARV) of the genus Marburgvirus, is characterized by its high case-fatality rate. Also belonging to the family of Filoviridae, MARV shares a lot of characteristics with EBOV. However, historically less frequent outbreaks with fewer affected individuals have occurred.Citation86 It both poses a great potential public health threat as an emerging virus and is listed as a category A bioterrorism agent by the Center for Disease Control (CDC), making vaccine development a priority. Despite decades of research, no licensed vaccines or therapies are available to date. However, ample preclinical data on rVSV-vectored MARV vaccine candidates have been generated – often alongside the EBOV vaccine candidates, both as a pre-exposure prophylactic as well as a post-exposure vaccine (reviewed inCitation15,Citation87,Citation88).

Analogous to its EBOV counterpart, rVSV-MARV vaccine candidates utilize the immunogenic MARV GP and have shown tolerability as well as efficacy in various animal models, including the NHP model. As a preventive vaccine, VSV∆G/MARVGP, which uses GP strain Musoke as the expressed antigen, was shown to completely protect cynomolgus macaques 28 days p.v. from lethal MARV challenge, and even when challenged almost 4 months after initial vaccination and with a different MARV strain (Popp).Citation24

A subsequent study further demonstrated that the vaccine was capable to confer cross-protection when NHP were challenged with the more virulent strain Angola, as well as the genetically distant Ravn strain with an amino acid deviation of around 20%. In this study, a dose of 2 × 107 PFU was administered; high anti-MARV IgG titers developed and rVSV viremia was detected around 3 days p.v., closely mirroring the observations made for VSV-EBOV. The primates did not develop any signs of clinical illness or abnormalities in hematology or blood chemistry values.Citation89

Peri- and post-exposure prophylaxis efficacy have been evaluated in animal models. A vaccination 20–30 min p.i. fully protected rhesus macaques from homologous challenge with >10,000 LD50 doses of MARV, and administration 24 h and 48 h post-exposure protected 5/6 and 2/6 macaques, respectively, suggesting efficacy – albeit limited – as a post-exposure treatment, even at high exposure doses.Citation90,Citation91 In contrast, when two vaccine candidates expressing MARV-Angola GP were tested for post-exposure efficacy 20–30 min after homologous MARV challenge, survival rates were only 25–75% depending on the challenge dosage and attenuation of the vector used, probably due to the higher pathogenicity as well as shorter incubation period of the MARV-Angola variant, which is of high clinical importance, as the MARV-Angola strain has been responsible for the worst outbreak of Marburg hemorrhagic fever so far.Citation92 Notably, all surviving animals had mounted anti-MARV specific IgG antibody responses by day 10, indicating that post-exposure treatment may confer protection by delaying disease progression long enough for the infected subject to develop sufficient immunity.Citation91,Citation92

The durability of post-vaccination immunity was furthermore assessed by monthly measurements of anti-MARV-GP antibodies in initially VSV∆G/MARVGP vaccinated macaques. All animals developed strong humoral responses that remained overall stable over the course of 13 months. When the macaques were then challenged with homologous MARV-Musoke, no animal developed clinical signs of illness, demonstrating efficacy of the vaccine for at least a year.Citation93 Taken together with data from other studies, the strong protective capacity of rVSV-MARV seems to be primarily mediated by a strong humoral immune response.Citation24,Citation89,Citation92,Citation94

Combination of rVSV-vectored filovirus vaccines

While the largest EVD outbreaks to date have been caused by EBOV, EVD outbreaks caused by other ebolavirus species have also been described, and MARV and EBOV outbreaks occur in overlapping geographic regions. Cross-protection of VSV-EBOV and -MARV against heterologous strains has been demonstrated as outlined above;Citation50,Citation89 however, limited or no cross-protection might exist between the more distantly related species and genera.Citation24,Citation95,Citation96 Therefore, a combination of rVSV-vectored filovirus vaccines would be desirable and has been evaluated.

After being vaccinated with a multi-component vaccine consisting of rVSV-ΔG-EBOV-, -MARV- and -SUDV-GP, macaques survived challenge with either of the respective species or TAFV. Monkeys challenged with MARV, EBOV or TAFV did not show clinical signs of disease, while those challenged with SUDV did. The authors argued that this observation could be due to slower replication kinetics of rVSVΔG-SUDV or the differential affinity of GP to antigen-presenting cells. They, therefore, tested a two-stage vaccination administering rVSVΔG-SUDV two weeks prior to vaccination with rVSVΔG-EBOV and -MARV, which resulted in complete protection from challenge with no clinical evidence of infection both when challenged with SUDV and subsequently back challenged with MARV.Citation97 In another study, the rVSV-N4CT1 vector expressing either EBOV, SUDV or MARV GP from the first position of the rVSV genome was administered to macaques as a trivalent vaccine and none of the animals succumbed to the disease after challenge with any of the pathogens. However, a subset of vaccinated macaques did develop mild signs of disease as well as low levels of filovirus viremia, especially after challenge with EBOV, when this attenuated vector was used.Citation98 The same vector is currently employed in efforts to develop a multivalent vaccine candidate against MARV, SUDV, EBOV and additionally Lassa virus.Citation99

Lassa virus

The arenavirus LASV is one of the primary causal agents of hemorrhagic fevers worldwide with more than 300,000 estimated infections recorded annually in West Africa, where it is endemic.Citation100 It is a rodent-transmitted disease and, while disease courses are highly diverse depending on LASV clade, morbidity is significant.Citation100 Since January 2019, Nigeria has been facing an ongoing outbreak of Lassa fever with 526 confirmed cases of LASV infections and 121 deaths (case-fatality rate 23%, Nigeria Center for Disease Control (NCDC) as of March 31, 2019).Citation101 Due to its widespread geographic distribution, high incidence and the lack of a preventive vaccine or approved therapies, LASV has been identified as an emerging virus for which R&D efforts are given very high priority by multiple international professional organizations, including the Coalition for Epidemic Preparedness Innovations (CEPI) and WHO.Citation102

Analogous to rVSV vectors expressing MARV and EBOV GP, Drs. Feldmann and Stroeher had described a LASV Glycoprotein C (GPC) expressing rVSV, VSVΔG/LASVGPC (clade IV, isolate Josiah), with no detectable pathogenicity in mice.Citation17 As the NHP model is the most relevant animal model for LASV infection, efficacy of a vaccine dose of 2 × 107 PFU was assessed in macaques (n = 4).Citation103 VSVΔG/LASVGPC was well tolerated and no viral shedding occurred after vaccination. Hematology and blood chemistry analysis found a slight thrombocyte decrease as well as a slight elevation of ALT in 3/4 animals. 100% of the animals remained asymptomatic after lethal LASV challenge 28 days after vaccination in contrast to controls, which succumbed to infection, and high LASV-specific antibody titers as well as moderate T-cell responses could be detected. Immunity was not sterile and animals developed viremia that was cleared by day 10.Citation103 Viremia was, however, not observed in consecutive experiments.Citation81,Citation104 Another vaccine construct with VSV expressing LASV nucleoprotein (NP) meanwhile only showed limited protection and was not pursued further.Citation104

The genetic heterogeneity observed between LASV clades raises concerns on whether VSVΔG/LASVGPC can act as a universal vaccine and this question was examined in subsequent studies. In inbred guinea pigs as well as NHPs, vaccination with VSVΔG/LASVGPC resulted in full protection against homologous challenge as well as against challenge with heterologous LASV clade IV isolates 28 days p.v. Again, challenge of inbred guinea pigs with the clade I isolate Pinneo, one of the most genetically divergent isolates compared to Josiah, did not result in clinical disease, while it has to be noted that animals vaccinated with an irrelevant control vector did develop symptomatic disease, but were able to clear infection.Citation104 Further work to elucidate Lassa cross-clade immunity is currently ongoing.Citation105

A better understanding of the durability of VSVΔG/LASVGPC is needed to estimate its potential benefit as a vaccine candidate. Stein and colleagues, therefore, sought to examine how early and how long VSVΔG/LASVGPC can confer protection. In a guinea pig-adapted (GPA) LASV model, animals vaccinated with 1 × 106 PFU VSVΔG/LASVGPC were challenged at a timepoint between 7 and 355 days p.v. As early as 7 days p.v. the vaccine provided full protection from clinical disease; however, immunity was not sterile. Antibody responses peaked on day 51 and were sustained until the final sampling date one year later. When challenged 355 p.v., the guinea pigs did develop signs of disease, but 71% of animals were still protected from death.Citation106

In recent developments, CEPI furthermore awarded considerable research funds to develop, manufacture and ultimately stockpile a Lassa virus vaccine based on the above mentioned rVSV-N4CT1 platform.Citation107

While further research including additional studies in NHPs is warranted, the data suggest that VSVΔG/LASVGPC may be a universal vaccine candidate that can confer early and lasting immunity.

Crimean-Congo hemorrhagic fever virus (CCHFV)

Based on the promising results obtained from the studies with rVSV vaccine candidates expressing filo- and arenavirus GP, a replication-competent rVSV expressing glycoprotein precursor (GPC) of CCHFV, likewise a hemorrhagic fever virus, has recently been developed and tested in an immunocompromised (STAT-1 knock-out) mouse model. In this study, a prime-only and a prime-boost regimen (days 0 and 7) were compared to mock vaccination, and immunogenicity, as well as protection from challenge with CCHFV 35 days after prime vaccination were assessed. Both groups receiving either one or two injections developed CCHFV-GP binding as well as virus neutralizing antibodies. The prime-boost group showed milder signs of infection and 100% of animals in both prime-only and prime-boost groups survived the challenge, while none of the mock-vaccinated did.Citation108

Nipah virus

The Henipavirus Nipah virus (NiV) is a zoonotic pathogen that can cause severe pulmonary and neurologic disease with a high case-fatality rate and has been responsible for almost yearly outbreaks on the Indian subcontinent. The NiV vaccine portfolio currently under development includes rVSV-vectored Nipah vaccines, which utilize rVSV expressing two different structural proteins as antigens, the NiV fusion (F) and attachment (G) glycoproteins – two surface proteins that together are required for viral cell entry. The rVSV vector expressing either antigen has been described in two different models, one with the insertion of either protein after VSV-G (VSV-G/F(NiV)), the other in place of the VSV glycoprotein (rVSV∆G-G/F(NiV)). In mice, intranasal (i.n.) administration of rVSV-G(NiV) and/or -F(NiV) induced neutralizing Ab (nAb) formation, and highest nAb formation was observed when rVSV-G(NiV) alone or in combination with rVSV-F(NiV) was given. The rVSV∆G vector was evaluated using i.m. vaccination and likewise induced nAb, especially when rVSV∆G-G- and -F(NiV) were used in combination.Citation109 Of note, the combined administration of rVSV∆G-G/F(NiV) was found to be lethal when given intranasally in 5/8 postnatal mice in a later study, while the individual administration of either vaccine candidate did not cause any symptoms, even when directly injected into the brain.Citation110

Vaccination with rVSV∆G-G- or -F(NiV) was demonstrated to be fully protective against NiV challenge in a Syrian hamster model, where none of the animals developed disease.Citation111 Similarly, the vector VSV-EBOV, complemented with NiV F or G protein downstream of EBOV-GP, was assessed in the hamster model and conferred the same level of protection from disease.Citation112 Importantly, while the vaccine was administered 28 days prior to challenge in this study, a follow-up study additionally demonstrated peri-exposure protection. Full protection was achieved until one day before vaccination and decreased to 17% until one day after challenge, while administration three days after challenge did not show protection.Citation113 Interestingly, similarly to observations made by Marzi et al. in an EBOV post-exposure vaccine model,Citation74 an rVSV vector vaccine control also mediated partial protection when given one day pre-challenge.Citation113 Antibody generation was associated with protectionCitation113 and a serum-transfer experiment confirmed that protection was antibody-dependent.Citation112 A simultaneously assessed rVSV-∆G-EBOV vector expressing NiV nucleocapsid protein (N) was shown to be inferior to the F and G variants.Citation112 The more immunogenic rVSV-∆G-EBOV-G(NiV) was subsequently demonstrated to be completely protective in African Green Monkeys, described as the most appropriate animal model for NiV infection.Citation114,Citation115

MERS- and SARS-coronaviruses

SARS as well as MERS constitute severe pulmonary infections that are caused by SARS- and MERS- coronaviruses (CoV), respectively. A severe outbreak of SARS-CoV, which originated in China in 2002, spread swiftly and ultimately affected over 8000 humans within a few months.Citation116 While the virus has been contained, its potential to reemerge and cause new outbreaks remains high, and the rVSV platform has been employed to create SARS-CoV vaccine candidates for such a future scenario. VSV-S expresses the immunogenic spike protein (S) of SARS-CoV between the G and the L gene of VSV. In a mouse model, vaccination with VSV-S induced higher levels of neutralizing antibodies than immunization with SARS-CoV and conferred protection as measured by low or undetectable SARS-CoV viral loads in the respiratory tract, even when challenged 4 months p.v. Using antisera from VSV-S vaccinated mice in naïve mice, it could further be shown that the protective effect was mediated by antibodies.Citation117 This vaccine candidate was then attenuated by replacing VSV G with SARS-CoV S, resulting in VSVΔG-S. Immunogenicity of the replication-defective vector was surprisingly greater than the immunogenicity of VSV-S when applied i.m., an effect the authors argued that could be due to a higher expression of SARS-CoV S protein in VSVΔG-S, among others. In an analogous manner, VSVΔG was later used to create a MERS-CoV vaccine by expressing the MERS-CoV spike protein (S), and VSVΔG-MERS displayed immunogenicity in rhesus monkeys.Citation118 To date, no human clinical trials have been reported for VSV-based CoV vaccines and the WHO roadmap for MERS-CoV research and product development does not currently include rVSV-vectored vaccines in the port- folio of vectored vaccines.Citation119

Zika virus

rVSV vectors have also been employed in designing vaccine candidates against Zika virus (ZIKV), a flavivirus closely related to Dengue, West-Nile, Japanese encephalitis and Yellow Fever viruses, which has been associated with severe neurological disorders such as microcephaly in newborns and Guillan-Barré Syndrome. In 2015–2017, the spread of ZIKV infections received attention when it rapidly disseminated from Brazil throughout the Americas, where no autochthonous case of ZIKV infection had been described before 2015.Citation120,Citation121

rVSV vaccine vector expressing diverse ZIKV antigens have been described in proof-of-concept studies. In 2017, an rVSV vectors that entails a mutated M protein as an attenuation factor (VSVm) and expresses either Zika envelope (ZENV) alone or ZENV containing the precursor to membrane (prM) proteins (ZprME) was described. The constructs remained replication competent. In mice, vaccination did not trigger any overt pathology and VSVm-ZprME vaccination did induce neutralizing antibody titers.Citation122 As ZIKV infection does not lead to disease in adult mice, challenge studies were performed in offspring of vaccinated mothers and, therefore, statements about the efficacy of the vaccine were limited.

In another approach, the attenuated VSV-EBOV was used as a vector to express ZprME or prM and soluble envelope proteins as antigens (ZprMsolE) and efficacy was tested in type I interferon receptor deficient mice (IFNAR/). The ZIKV vaccine candidates did not result in pathogenicity in mice, however, unexpectedly vaccination with VSV-EBOV control vector led to severe clinical symptoms in the immunocompromised mice, thus the VSV-EBOV-based ZIKV vaccine constructs appear to be more attenuated than the original VSV-EBOV in this study. Mice challenged with ZIKV 28 days p.v. were fully protected from disease. Antigenicity of ZprME seemed to be superior to ZprMsolE and 50% and 100% protection was achieved when mice were vaccinated with rVSV-ZprME 1 and 3 days pre-challenge, respectively. Interestingly, vaccination with the vaccines was sufficient to also protect outbred CD1 mice from EBOV challenge.Citation123

In a study assessing multiple ZIKV antigens in a VSV vector, which was attenuated via a point mutation in the VSV L protein, combining the ZIKV prM and E proteins with the nonstructural NS1 protein showed robust humoral and cellular responses in immunocompetent mice, as well as protection from disease in IFNAR/ mice.Citation124

Furthermore, a VSV vector expressing the ZIKV capsid protein was also shown to be immunogenic with dominating cellular immune responses and reduced viral replication in the spinal cord and brain tissues of immunocompetent mice.Citation125 No human clinical trials using VSV-based ZIKV vaccine candidates have been initiated to date.

Conclusion

The rVSV vaccine platform exhibits many advantages as a vaccine vector backbone for the expression of viral antigens and has been tested as a vaccine candidate against a multitude of emerging infections. As a result of the recent clinical phase I-III trials of VSV-EBOV, we have now gained a significant amount of data in humans, which encouragingly suggest a favorable safety profile of the vaccine candidate as well as swift protection after a single immunization. VSV can readily be attenuated while remaining immunogenic. While the comprehensive details on VSV vaccine-mediated protective immunity have yet to be elucidated, the data generated to date support the premise that protection appears to be primarily inferred by antibody responses. However, cellular immunity is simultaneously induced. In addition, the fact that recombinants can be manufactured quickly with diverse antigens further adds to its value as a vaccine platform for outbreak pathogens. While we have focused here on WHO blueprint priority pathogens, a variety of VSV-based vaccine candidates have been described for other emerging or reemerging pathogens, including enterovirus 71,Citation126 Chikungunya,Citation127,Citation128 West-Nile,Citation129 Dengue,Citation130 Severe fever with thrombocytopenia syndrome,Citation131 and Andes viruses.Citation132Citation134 Animal studies of VSV-based vector vaccines against MARV, LASV, NiV, ZIKV, and MERS- and SARS-CoV described above have yielded promising results and the progression of rVSV vaccine candidates to clinical trials may add to the portfolio of effective outbreak vaccines in the foreseeable future.

Disclosure of potential conflicts of interest

The authors declare no conflict of interest.

Acknowledgments

We would like to thank Katherine M. Ball, JD, for her kind and helpful suggestions to edit this manuscript’s English writing style.

Additional information

Funding

This work has been supported by the German Center for Infection Research (DZIF, Clinical Leave Stipend for AF).

References

  • WHO. Ten threats to global health in 2019 [Internet]. [accessed 2019 Apr 3]. https://www.who.int/emergencies/ten-threats-to-global-health-in-2019
  • Oh M, Park WB, Park S-W, Choe PG, Bang JH, Song K-H, Kim ES, Bin KH, Kim NJ. Middle East respiratory syndrome: what we learned from the 2015 outbreak in the Republic of Korea. Korean J Intern Med. 2018;33:233–46. doi:10.3904/kjim.2018.031.
  • WHO. WHO Director-General summarizes the outcome of the emergency committee regarding clusters of microcephaly and Guillain-Barré syndrome [Internet]. [accessed 2019 Apr 3]. https://www.who.int/en/news-room/detail/01-02-2016-who-director-general-summarizes-the-outcome-of-the-emergency-committee-regarding-clusters-of-microcephaly-and-guillain-barré-syndrome
  • WHO. WHO publishes list of top emerging diseases likely to cause major epidemics [Internet]. 2015 [accessed 2019 Mar 24]. https://www.who.int/medicines/ebola-treatment/WHO-list-of-top-emerging-diseases/en/
  • WHO. Methodology for prioritizing severe emerging diseases for research and development background [Internet]. 2017 [accessed 2019 Jun 27]. http://www.who.int/csr/research-and-development/meeting-report-prioritization.pdf?ua=1
  • WHO. WHO research and development blueprint 2018 annual review of diseases prioritized under the research and development blueprint informal consultation [internet]. [accessed 2019 Jun 28]. http://www.who.int/csr/research-and-development/documents/prioritizing_diseases_progress/en/
  • Rose J, Schubert M. Rhabdovirus genomes and their Products. In: RR Wagner, editor. The rhabdoviruses. Boston (MA): Springer US; 1987. p. 129–66.
  • Zemp F, Rajwani J, Mahoney DJ. Rhabdoviruses as vaccine platforms for infectious disease and cancer. Biotechnol Genet Eng Rev. 2018;34:122–38. doi:10.1080/02648725.2018.1474320.
  • Lawson ND, Stillmans EA, Whitt MA, Rose JK. Recombinant vesicular stomatitis viruses from DNA. PNAS. 1995;92:4477–81. doi:10.1073/pnas.92.10.4477.
  • Roberts A, Buonocore L, Price R, Forman J, Rose JK, Roberts A, Kretzschmar E, Perkins AS, Forman J, Price R, et al. Attenuated vesicular stomatitis viruses as vaccine vectors. J Virol. 1999;73:3723–32.
  • Clarke DK, Nasar F, Lee M, Johnson JE, Wright K, Calderon P, Guo M, Natuk R, Cooper D, Hendry RM, et al. Synergistic attenuation of vesicular stomatitis virus by combination of specific G Gene truncations and N Gene translocations. J Virol. 2007;81:2056. doi:10.1128/JVI.01911-06.
  • Rose NF, Marx PA, Luckay A, Nixon DF, Moretto WJ, Donahoe SM, Montefiori D, Roberts A, Buonocore L, Rose JK. An effective AIDS vaccine based on live attenuated vesicular stomatitis virus recombinants. Cell. 2001;106:539–49. doi:10.1016/S0092-8674(01)00482-2.
  • Mcgettigan JP, Sarma S, Orenstein JM, Pomerantz RJ, Schnell MJ. Expression and immunogenicity of human immunodeficiency virus type 1 gag expressed by a replication-competent rhabdovirus-based vaccine vector. J Virol. 2001;75:8724–32. doi:10.1128/JVI.75.18.8724-8732.2001.
  • Clarke DK, Michael Hendry R, Singh V, Rose JK, Seligman SJ, Klug B, Kochhar S, Mac LM, Carbery B, Chen RT. Live virus vaccines based on a vesicular stomatitis virus (VSV) backbone: standardized template with key considerations for a risk/benefit assessment. Vaccine. 2016;34:6597–609. doi:10.1016/j.vaccine.2016.10.045.
  • Geisbert TW, Feldmann H. Recombinant vesicular stomatitis virus-based vaccines against Ebola and marburg virus infections. J Infect Dis. 2011;204:S1075–1081. doi:10.1093/infdis/jir349.
  • Monath TP, Fast PE, Modjarrad K, Clarke DK, Martin BK, Fusco J, Nichols R, Heppner DG, Simon JK, 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. doi:10.1016/j.jvacx.2019.100009.
  • Garbutt M, Liebscher R, Wahl-Jensen V, Jones S, Möller P, Wagner R, Volchkov V, Klenk H-D, Feldmann H, Ströher U. Properties of replication-competent vesicular stomatitis virus vectors expressing glycoproteins of filoviruses and arenaviruses. J Virol. 2004;78:5458–65. doi:10.1128/JVI.78.10.5458-5465.2004.
  • Rose NF, Roberts A, Buonocore L, Rose JK. Glycoprotein exchange vectors based on vesicular stomatitis virus allow effective boosting and generation of neutralizing antibodies to a primary isolate of human immunodeficiency virus type 1. J Virol. 2000;74:10903–10. doi:10.1128/JVI.74.23.10903-10910.2000.
  • Fuchs JD, Frank I, Elizaga ML, Allen M, Frahm N, Kochar N, Li S, Edupuganti S, Kalams SA, Tomaras GD, et al. First-in-human evaluation of the safety and immunogenicity of a recombinant vesicular stomatitis virus human immunodeficiency virus-1 gag vaccine (HVTN 090). Open Forum Infect Dis. 2015;2:ofv082. doi:10.1093/ofid/ofv082.
  • Kuhn JH, Becker S, Ebihara H, Geisbert TW, Johnson KM, Kawaoka Y, Lipkin WI, Negredo AI, Netesov SV, Nichol ST, et al. Proposal for a revised taxonomy of the family Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol. 2010;155:2083–103. doi:10.1007/s00705-010-0677-1.
  • Goldstein T, Anthony SJ, Gbakima A, Bird BH, Bangura J, Tremeau-Bravard A, Belaganahalli MN, Wells HL, Dhanota JK, Liang E, et al. The discovery of Bombali virus adds further support for bats as hosts of ebolaviruses. Nat Microbiol. 2018;3:1084–89. doi:10.1038/s41564-018-0227-2.
  • WHO. WHO | statement on the 1st meeting of the IHR emergency committee on the 2014 Ebola outbreak in West Africa [Internet]. WHO2014. [accesed 2019 Mar 6]. https://www.who.int/mediacentre/news/statements/2014/ebola-20140808/en/
  • Plummer FA, Jones SM. The story of Canada’s Ebola vaccine. CMAJ. 2017;189:E1326–7. doi:10.1503/cmaj.170704.
  • Jones SM, Feldmann H, Ströher U, Geisbert JB, Fernando L, Grolla A, Klenk H-D, Sullivan NJ, Volchkov VE, Fritz EA, et al. Live attenuated recombinant vaccine protects nonhuman primates against Ebola and Marburg viruses. Nat Med. 2005;11:786–90. doi:10.1038/nm1258.
  • Qiu X, Fernando L, Alimonti JB, Leno Melito P, Feldmann F, Dick D, Ströher U, Feldmann H, Jones SM. Mucosal immunization of cynomolgus macaques with the VSVDG/ZEBOVGP vaccine stimulates strong Ebola GP-specific immune responses. PLoS One. 2009;4:e5547. doi:10.1371/journal.pone.0005547.
  • Geisbert TW, Daddario-Dicaprio KM, Geisbert JB, Reed DS, Feldmann F, Grolla A, Ströher U, Fritz EA, Hensley LE, Jones SM, et al. Vesicular stomatitis virus-based vaccines protect nonhuman primates against aerosol challenge with Ebola and Marburg viruses. Vaccine. 2012;26:6894–900. doi:10.1016/j.vaccine.2008.09.082.
  • Jones SM, Ströher U, Fernando L, Qiu X, Alimonti J, Melito P, Bray M, Klenk H-D, Feldmann H, Strö U, et al. Assessment of a vesicular stomatitis virus-based vaccine by use of the mouse model of Ebola virus hemorrhagic fever. J Infect Dis. 2007;196:404–12. doi:10.1086/520591.
  • Agnandji ST, Huttner A, Zinser ME, Njuguna P, Dahlke C, Fernandes JF, Yerly S, Dayer J-A, Kraehling V, Kasonta R, et al. Phase 1 trials of rVSV Ebola vaccine in Africa and Europe. N Engl J Med. 2016;374:1647–60. doi:10.1056/NEJMoa1502924.
  • Regules JA, Beigel JH, Paolino KM, Voell J, Castellano AR, Hu Z, Muñoz P, Moon JE, Ruck RC, Bennett JW, et al. A recombinant vesicular stomatitis virus Ebola vaccine. N Engl J Med. 2017;376:330–41. doi:10.1056/NEJMoa1414216.
  • ElSherif MS, Brown C, Mackinnon-Cameron D, Li L, Racine T, Alimonti J, Rudge TL, Sabourin C, Silvera P, Hooper JW, et al. Assessing the safety and immunogenicity of recombinant vesicular stomatitis virus Ebola vaccine in healthy adults: A randomized clinical trial. CMAJ. 2017;189:819–27. doi:10.1503/cmaj.171389.
  • Heppner DG, Kemp TL, Martin BK, Ramsey WJ, Nichols R, Dasen EJ, Link CJ, Das R, Xu ZJ, Sheldon EA, et al. Safety and immunogenicity of the rVSV∆G-ZEBOV-GP Ebola virus vaccine candidate in healthy adults: a phase 1b randomised, multicentre, double-blind, placebo-controlled, dose-response study. Lancet Infect Dis. 2017;17:854–66. doi:10.1016/S1473-3099(17)30242-6.
  • Coller B-AG, Blue J, Das R, Dubey S, Finelli L, Gupta S, Helmond F, Grant-Klein RJ, Liu K, Simon J, et al. Clinical development of a recombinant Ebola vaccine in the midst of an unprecedented epidemic. Vaccine. 2017;35:4465–69. doi:10.1016/j.vaccine.2017.05.097.
  • Henao-Restrepo AM, Camacho A, Longini IM, Watson CH, Edmunds J, Egger M, Carroll MW, Dean NE, 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;389:505–18. doi:10.1016/S0140-6736(16)32621-6.
  • Kennedy SB, Bolay F, Kieh M, Grandits G, Badio M, Ballou R, Eckes R, Feinberg M, Follmann D, Grund B, et al. Phase 2 placebo-controlled trial of two vaccines to prevent Ebola in Liberia. N Engl J Med. 2017;377:1438–47. doi:10.1056/NEJMoa1614067.
  • Halperin SA, Arribas JR, Rupp R, Andrews CP, Chu L, Das R, Simon JK, Onorato MT, 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;215:1789–98. doi:10.1093/infdis/jix189.
  • Juan-Giner A, Tchaton M, Jemmy J-P, Soumah A, Boum Y, Faga EM, Cisse M, Grais RF. Safety of the rVSV ZEBOV vaccine against Ebola Zaire among frontline workers in Guinea. Vaccine. 2018;S0264-410X:31246–5.
  • Samai M, Seward JF, Goldstein ST, Mahon BE, Lisk DR, Widdowson M-A, Jalloh MI, Schrag SJ, Idriss A, Carter RJ, et al. The sierra leone trial to introduce a vaccine against Ebola: an evaluation of rVSV∆G-ZEBOV-GP vaccine tolerability and safety during the West Africa Ebola outbreak. J Infect Dis. 2018;6:217.
  • WHO. Strategic Advisory Group of Experts (SAGE) on immunization interim recommendations on vaccination against Ebola Virus Disease (EVD) [internet]. 2019 [accessed 2019 Jun 28]. www.who.int/immunization/sage/meetings/2019/april/SAGE_April_2019_meeting_summary.pdf?ua=1,
  • Huttner A, Dayer J-A, Yerly S, Combescure C, Auderset F, Desmeules J, Eickmann M, Finckh A, Goncalves AR, Hooper JW, et al. The effect of dose on the safety and immunogenicity of the VSV Ebola candidate vaccine: a randomised double-blind, placebo-controlled phase 1/2 trial. Lancet Infect Dis. 2015;15:1156–66. doi:10.1016/S1473-3099(15)00154-1.
  • Huttner A, Agnandji ST, Combescure C, Fernandes JF, Bache EB, Kabwende L, Ndungu FM, Brosnahan J, Monath TP, Lemaître B, et al. Determinants of antibody persistence across doses and continents after single-dose rVSV-ZEBOV vaccination for Ebola virus disease: an observational cohort study. Lancet Infect Dis. 2018;18:738–48. doi:10.1016/S1473-3099(18)30165-8.
  • Agnandji ST, Fernandes JF, Bache EB, Mba RMO, Brosnahan JS, Kabwende L, Pitzinger P, Staarink P, Massinga-Loembe M, Krähling V, et al. Safety and immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine in adults and children in Lambaréné, Gabon: A phase I randomised trial. PLoS Med. 2017;14:e1002402. doi:10.1371/journal.pmed.1002230.
  • Kasereka MC, Sawatzky J, Hawkes MT. Ebola epidemic in war-torn democratic republic of congo, 2018: acceptability and patient satisfaction of the recombinant vesicular stomatitis virus - Zaire Ebolavirus vaccine. Vaccine. 2019;37:2174–78. doi:10.1016/j.vaccine.2019.03.004.
  • Gsell P-S, Camacho A, Kucharski AJ, Watson CH, Bagayoko A, Nadlaou SD, Dean NE, Diallo A, Diallo A, Honora DA, et al. Ring vaccination with rVSV-ZEBOV under expanded access in response to an outbreak of Ebola virus disease in Guinea, 2016: an operational and vaccine safety report. Lancet Infect Dis. 2017;17:1276–84. doi:10.1016/S1473-3099(17)30242-6.
  • The PREVENT Working Group. Pregnant women & vaccines against emerging epidemic threats [Internet]. Prev Work Gr. [accessed 2019 Mar 17]. https://www.who.int/immunization/sage/meetings/2018/october/1_PREVENT_Recs_Excerpts_for_SAGE.PDF.
  • Caluwaerts S. Nubia’s mother: being pregnant in the time of experimental vaccines and therapeutics for Ebola. Reprod Health. 2017;14:157. doi:10.1186/s12978-017-0429-8.
  • Branswell H. Ebola vaccine will be given to pregnant women, marking reversal in policy [internet]. [accssed 2019 Mar 23]. https://www.statnews.com/2019/02/20/ebola-pregnancy-reversal/
  • RDC. EBOLA RDC - Evolution de la riposte contre l’épidémie d’Ebola dans les provinces du Nord Kivu et de l’Ituri au Samedi 15 juin 2019 [Internet]. 2019 [accessed 2019 Jun 28]. https://mailchi.mp/sante.gouv.cd/ebola_kivu_15juin19?e=77c16511ad
  • Jones SM, Ströher U, Fernando L, Qiu X, Alimonti J, Melito P, Bray M, Klenk H-D, Feldmann H. Assessment of a vesicular stomatitis virus-based vaccine by use of the mouse model of Ebola virus hemorrhagic fever. J Infect Dis. 2007;196:S404–412. doi:10.1086/520591.
  • Geisbert TW, Daddario-Dicaprio KM, Lewis MG, Geisbert JB, Grolla A. vesicular stomatitis virus-based Ebola vaccine is well-tolerated and protects immunocompromised nonhuman primates. PLoS Pathog. 2008;4:1000225. doi:10.1371/journal.ppat.1000225.
  • Marzi A, Robertson SJ, Haddock E, Feldmann F, Hanley PW, Scott DP, Strong JE, Kobinger G, Best SM, Feldmann H. VSV-EBOV rapidly protects macaques against infection with the 2014/15 Ebola virus outbreak strain. Science. 2015;349:739–42. doi:10.1126/science.aab3920.
  • Maria Henao-Restrepo A, Longini IM, Egger M, Dean NE, John Edmunds W, Camacho A, Carroll MW, Doumbia M, Draguez B, Duraff Our S, et al. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. Lancet. 2015;386:857–66. doi:10.1016/S0140-6736(15)61137-0.
  • Metzger WG, Vivas-Martínez S. Questionable efficacy of the rVSV-ZEBOV Ebola vaccine. Lancet. 2018;391:1021. doi:10.1016/S0140-6736(18)30302-7.
  • Longini IM, Røttingen J-A, Kieny MP, Edmunds WJ, Henao-Restrepo AM. Questionable efficacy of the rVSV-ZEBOV Ebola vaccine – authors’ reply. Lancet. 2018;391:1021–22. doi:10.1016/S0140-6736(18)30302-7.
  • Merck. Merck begins rolling submission of licensure application for V920 (rVSV∆G-ZEBOV-GP) to U.S. Food and drug administration [internet]. 2018 [accessed 2019 Mar 16]. https://investors.merck.com/news/press-release-details/2018/Merck-Begins-Rolling-Submission-of-Licensure-Application-for-V920-rVSVG-ZEBOV-GP-to-US-Food-and-Drug-Administration/default.aspx
  • Marzi A, Mire CE. Current Ebola virus vaccine progress. BioDrugs. 2019;33:9–14. doi:10.1007/s40259-018-0329-7.
  • Haug CJ. Keeping your cool — doing Ebola research during an emergency. NEJM. 2018;378:2353–55. doi:10.1056/NEJMc1711583.
  • Gsell P-S, Camacho A, Kucharski AJ, Watson CH, Bagayoko A, Danmadji Nadlaou S, Dean NE, Diallo A, Diallo A, Honora DA, et al. Ring vaccination with rVSV-ZEBOV under expanded access in response to an outbreak of Ebola virus disease in Guinea, 2016: an operational and vaccine safety report. Lancet Infect Dis. 2017;17:1276–84. doi:10.1016/S1473-3099(17)30242-6.
  • WHO. Ebola Virus Disease Democratic Republic of the Congo External Situation Report 17 [Internet]. 2018 [accessed 2019 Apr 9]. http://www.who.int/ebola/historical-outbreaks-drc/en/.
  • Ministère de la Santé RD du C. EBOLA RDC - Evolution de la riposte contre l’épidémie d’Ebola dans les provinces du Nord Kivu et de l’Ituri au Lundi 22 juillet 2019 [Internet]. 2019 [accessed 2019 Jul 30]. https://us13.campaign-archive.com/?u=89e5755d2cca4840b1af93176&id=1af4c6a868
  • WHO. Preliminary results on the efficacy of rVSV-ZEBOV-GP Ebola vaccine using the ring vaccination strategy in the control of an Ebola outbreak in the Democratic Republic of the Congo: an example of integration of research into epidemic response. [Internet]. 2019 [accessed 2019 Jun 14]. https://www.who.int/csr/resources/publications/ebola/ebola-ring-vaccination-results-12-april-2019.pdf
  • Widdowson M-A, Schrag SJ, Carter RJ, Carr W, Legardy-Williams J, Gibson L, Lisk DR, Jalloh MI, Bash-Taqi DA, Kargbo SAS, et al. Implementing an Ebola vaccine study-sierra leone background to trial conception. MMWR. 2016;65:98–106. doi:10.15585/mmwr.su6503a14.
  • Arnemo M, Sofie S, Watle V, Merete Schoultz K, Vainio K, Norheim G, Moorthy V, Fast P, Røttingen J-A, Gjøen T. Stability of a vesicular stomatitis virus-vectored Ebola vaccine. J Infect Dis. 2016;213:930–33. doi:10.1093/infdis/jiv532.
  • Stein DR, Sroga P, Warner BM, Deschambault Y, Poliquin G, Safronetz D. Evaluating temperature sensitivity of vesicular stomatitis virus–based vaccines. Emerg Infect Dis. 2019 [accessed 2019 Jul 26];25:1563–66. http://wwwnc.cdc.gov/eid/article/25/8/19-0281_article.htm.
  • Ruibal P, Oestereich L, Lüdtke A, Becker-Ziaja B, Wozniak DM, Kerber R, Korva M, Cabeza-Cabrerizo M, Bore JA, Koundouno FR, et al. Unique human immune signature of Ebola virus disease in Guinea. Nature. 2016;533:100–04. doi:10.1038/nature17949.
  • Liu X, Speranza E, Muñoz-Fontela C, Haldenby S, Rickett NY, Garcia-Dorival I, Fang Y, Hall Y, Zekeng E-G, Lüdtke A, et al. Transcriptomic signatures differentiate survival from fatal outcomes in humans infected with Ebola virus. Genome Biol. 2017;18:4. doi:10.1186/s13059-016-1137-3.
  • Marzi A, Engelmann F, Feldmann F, Haberthur K, Shupert WL, Brining D, Scott DP, Geisbert TW, Kawaoka Y, Katze MG, et al. Antibodies are necessary for rVSV/ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhuman primates. PNAS. 2013;110:1893–98. doi:10.1073/pnas.1209591110.
  • Menicucci AR, Jankeel A, Feldmann H, Marzi A, Messaoudi I. Antiviral innate responses induced by VSV-EBOV vaccination contribute to rapid protection. MBio. 2019;10. doi:10.1128/mBio.00669-19.
  • Gross L, Lhomme E, Pasin C, Richert L, Thiebaut R. Ebola vaccine development: systematic review of pre-clinical and clinical studies, and meta-analysis of determinants of antibody response variability after vaccination. Int J Infect Dis. 2018;74:83–96. doi:10.1016/j.ijid.2018.06.022.
  • Farooq F, Beck K, Paolino KM, Phillips R, Waters NC, Regules JA, Bergmann-Leitner ES. Circulating follicular T helper cells and cytokine profile in humans following vaccination with the rVSV-ZEBOV Ebola vaccine. Sci Rep. 2016;6:1–9. doi:10.1038/srep27944.
  • Dahlke C, Kasonta R, Lunemann S, Krähling V, Zinser ME, Biedenkopf N, Fehling SK, Ly ML, Rechtien A, Stubbe HC, et al. Dose-dependent T-cell dynamics and cytokine cascade following rVSV-ZEBOV immunization. EBioMedicine. 2017;19:107–18. doi:10.1016/j.ebiom.2017.03.045.
  • Rechtien A, Richert L, Lorenzo H, Martrus G, Hejblum B, Dahlke C, Kasonta R, Zinser M, Stubbe H, Matschl U, et al. Systems vaccinology identifies an early innate immune signature as a correlate of antibody responses to the Ebola vaccine rVSV-ZEBOV. Cell Rep. 2017;20:2251–61. doi:10.1016/j.celrep.2017.08.023.
  • Huttner A, Combescure C, Grillet S, Haks MC, Quinten E, Modoux C, Todagbe Agnandji S, Brosnahan J, Dayer J-A, Harandi AM, et al. A dose-dependent plasma signature of the safety and immunogenicity of the rVSV-Ebola vaccine in Europe and Africa. Sci Transl Med. 2017;9. doi:10.1126/scitranslmed.aaj1701.
  • Feldmann H, Jones SM, Daddario-Dicaprio KM, Geisbert JB, Strö Her U, Grolla A, Bray M, Fritz EA, Fernando L, Feldmann F, et al. Effective post-exposure treatment of Ebola infection. PLoS Pathog. 2007;3:e2. doi:10.1371/journal.ppat.0030002.
  • Marzi A, Hanley PW, Haddock E, Martellaro C, Kobinger G, Feldmann H. Efficacy of vesicular stomatitis virus-Ebola virus postexposure treatment in rhesus macaques infected with Ebola virus makona. J Infect Dis. 2016;214:S360–6. doi:10.1093/infdis/jiw389.
  • Geisbert TW, Daddario-Dicaprio KM, Williams KJN, Geisbert JB, Leung A, Feldmann F, Hensley LE, Feldmann H, Jones SM. Recombinant vesicular stomatitis virus vector mediates postexposure protection against sudan Ebola hemorrhagic fever in nonhuman primates. J Virol. 2008;82:5664–68. doi:10.1128/JVI.01931-07.
  • Fischer WA, Vetter P, Bausch DG, Burgess T, Davey RT, Fowler R, Hayden FG, Jahrling PB, Kalil AC, Mayers DL, et al. Ebola virus disease: an update on post-exposure prophylaxis. Rev Lancet Infect Dis. 2018;18:e183–92. doi:10.1016/S1473-3099(17)30677-1.
  • Günther S, Feldmann H, Geisbert TW, Hensley LE, Rollin PE, Nichol ST, Ströher U, Artsob H, Peters CJ, Ksiazek TG, et al. Management of accidental exposure to Ebola virus in the biosafety level 4 laboratory, Hamburg, Germany. J Infect Dis. 2011;204(Suppl):S785–90. doi:10.1093/infdis/jir298.
  • Lai L, Davey R, Beck A, Xu Y, Suffredini AF, Palmore T, Kabbani S, Rogers S, Kobinger G, Alimonti J, et al. Emergency postexposure vaccination with vesicular stomatitis virus-vectored Ebola vaccine after needlestick. JAMA. 2015;313:1249–55. doi:10.1001/jama.2015.1995.
  • Wong KK, Davey RT, Hewlett AL, Kraft CS, Mehta AK, Mulligan MJ, Beck A, Dorman W, Kratochvil CJ, Lai L, et al. Use of postexposure prophylaxis after occupational exposure to Zaire ebolavirus. Clin Infect Dis. 2016;63:376–79. doi:10.1093/cid/ciw256.
  • Poetsch JH, Dahlke C, Zinser ME, Kasonta R, Lunemann S, Rechtien A, Ly ML, Stubbe HC, Krähling V, Biedenkopf N, et al. Detectable Vesicular Stomatitis Virus (VSV)–specific humoral and cellular immune responses following VSV–Ebola virus vaccination in humans. J Infect Dis. 2018;219:556–61. doi:10.1093/infdis/jiy565.
  • Marzi A, Feldmann F, Geisbert TW, Feldmann H, Safronetz D. Vesicular stomatitis virus–based vaccines against lassa and Ebola viruses. Emerg Infect Dis. 2015;21:305–07. doi:10.3201/eid2101.140256.
  • Locher S, Schweneker M, Hausmann J, Zimmer G. Immunogenicity of propagation-restricted vesicular stomatitis virus encoding Ebola virus glycoprotein in guinea pigs. J Gen Virol. 2018;99:866–79. doi:10.1099/jgv.0.001031.
  • Matassov D, Marzi A, Latham T, Xu R, Ota-Setlik A, Feldmann F, Geisbert JB, Mire CE, Hamm S, Nowak B, et al. Vaccination with a highly attenuated recombinant vesicular stomatitis virus vector protects against challenge with a lethal dose of Ebola virus. J Infect Dis. 2015;212:443–51. doi:10.1093/infdis/jiv316.
  • Elizaga IDML, Li SS, Kochar NK, Wilson GJ, Allen IDMA, Van Tieu HN, Frank I, Sobieszczyk ME, Cohen KW, Sanchez B, et al. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial. PLoS One. 2018;13:e0202753. doi:10.1371/journal.pone.0202753.
  • Mire CE, Matassov D, Geisbert JB, Latham TE, Agans KN, Xu R, Ota-Setlik A, Egan MA, Fenton KA, Clarke DK, et al. Single-dose attenuated Vesiculovax vaccines protect primates against Ebola Makona virus. Nature. 2015;520:688–94. doi:10.1038/nature14428.
  • WHO | Marburg virus disease. WHO [Internet] 2017 [accessed 2019 Apr 9]. http://origin.who.int/mediacentre/factsheets/fs_marburg/en/
  • Reynolds P, Ebola MA. Marburg virus vaccines. Virus Genes. 2017;53:501–15. doi:10.1007/s11262-017-1455-x.
  • Cross RW, Mire CE, Feldmann H, Geisbert TW. Post-exposure treatments for Ebola and Marburg virus infections. Nat Rev Drug Discov. 2018;17:413–34. doi:10.1038/nrd.2017.251.
  • Daddario-Dicaprio KM, Geisbert TW, Geisbert JB, Ströher U, Hensley LE, Grolla A, Fritz EA, Feldmann F, Feldmann H, Jones SM. Cross-protection against Marburg virus strains by using a live, attenuated recombinant vaccine. J Virol. 2006;80:9659–66. doi:10.1128/JVI.00959-06.
  • Daddario-Dicaprio KM, Geisbert TW, Ströher U, Geisbert JB, Grolla A, Fritz EA, Fernando L, Kagan E, Jahrling PB, Hensley LE, et al. Postexposure protection against Marburg haemorrhagic fever with recombinant vesicular stomatitis virus vectors in non-human primates: an efficacy assessment. Lancet. 2006;367:1399–404. doi:10.1016/S0140-6736(06)68546-2.
  • Geisbert TW, Hensley LE, Geisbert JB, Leung A, Johnson JC, Grolla A, Feldmann H. Postexposure treatment of Marburg virus infection. Emerg Infect Dis. 2010;16:1119–22.
  • Woolsey C, Geisbert JB, Matassov D, Agans KN, Borisevich V, Cross RW, Deer DJ, Fenton KA, Eldridge JH, Mire CE, et al. Postexposure efficacy of recombinant vesicular stomatitis virus vectors against high and low doses of marburg virus variant angola in nonhuman primates. J Infect Dis. 2018;218:582–87. doi:10.1093/infdis/jiy293.
  • Mire CE, Geisbert JB, Agans KN, Satterfield BA, Versteeg KM. Durability of a vesicular stomatitis virus-based Marburg virus vaccine in nonhuman primates. PLoS One. 2014;9:e94355. doi:10.1371/journal.pone.0094355.
  • Nachbagauer R, Connor J, Marzi A, Menicucci AR, Engelmann F, Callison J, Horne EJ, Feldmann F, Jankeel A, Feldmann H, et al. Protection against marburg virus using a recombinant VSV-vaccine depends on T and B cell activation. Front Immunol. 2019;9. doi:10.3389/fimmu.2018.03122.
  • Falzarano D, Feldmann F, Grolla A, Leung A, Ebihara H, Strong JE, Marzi A, Takada A, Jones S, Gren J, et al. Single immunization with a monovalent vesicular stomatitis virus-based vaccine protects nonhuman primates against heterologous challenge with bundibugyo ebolavirus. J Infect Dis. 2011;204:S1082–S1089. doi:10.1093/infdis/jir350.
  • Mire CE, Geisbert JB, Marzi A, Agans KN, Feldmann H, Geisbert TW. Vesicular stomatitis virus-based vaccines protect nonhuman primates against bundibugyo ebolavirus. PLoS Negl Trop Dis. 2013;7:e2600. doi:10.1371/journal.pntd.0002600.
  • TW G, JB G, Leung A, KM D-D, LE H, Grolla A, Feldmann H. Single-injection vaccine protects nonhuman primates against infection with marburg virus and three species of Ebola virus. J Virol. 2009;83:7296–304. doi:10.1128/JVI.02516-08.
  • Matassov D, Mire CE, Latham T, Geisbert JB, Xu R, Ota-Setlik A, Agans KN, Kobs DJ, Wendling MQS, Burnaugh A, et al. Single-dose trivalent vesiculovax vaccine protects macaques from lethal ebolavirus and marburgvirus challenge. J Virol. 2018;92:e01190–17. doi:10.1128/JVI.01190-17.
  • Profectus Biosciences. Profectus biosciences awarded NIH contract for up to $22.25 million to develop multi-component vaccine for Ebola, Marburg, and Lassa viruses [Internet]. 2017 [accessed 2019 Mar 5]. www.profectusbiosciences.com
  • Warner BM, Safronetz D, Stein DR. Current research for a vaccine against Lassa hemorrhagic fever virus. Drug Des Devel Ther. 2018;12:2519–27. doi:10.2147/DDDT.S147276.
  • Nigeria Center for Disease Control (NCDC). 2019 LASSA Fever Outbreak Situation report week 13 [Internet]. [accessed 2019 Apr 9]. https://ncdc.gov.ng/diseases/sitreps/?cat=5&name=An update of Lassa fever outbreak in Nigeria
  • Plotkin SA. Vaccines for epidemic infections and the role of CEPI. Hum Vaccin Immunother. 2017;13:2755–62. doi:10.1080/21645515.2017.1306615.
  • Geisbert TW, Jones S, Fritz EA, Shurtleff AC, Geisbert JB, Liebscher R, Grolla A, Strö Her U, Fernando L, Daddario KM, et al. Development of a New Vaccine for the Prevention of Lassa Fever. PLoS Med. 2005;2:e183. doi:10.1371/journal.pmed.0020183.
  • Safronetz D, Mire C, Rosenke K, Feldmann F, Haddock E, Geisbert T, Feldmann H. A recombinant vesicular stomatitis virus-based lassa fever vaccine protects guinea pigs and macaques against challenge with geographically and genetically distinct Lassa viruses. PLoS Negl Trop Dis. 2015;9:e0003736. doi:10.1371/journal.pntd.0003736.
  • Kafetzopoulou LE, Pullan ST, Lemey P, Suchard MA, Ehichioya DU, Pahlmann M, Thielebein A, Hinzmann J, Oestereich L, Wozniak DM, et al. Metagenomic sequencing at the epicenter of the Nigeria 2018 Lassa fever outbreak. Science. 2019;363:74–77. doi:10.1126/science.aau9343.
  • Stein DR, Warner BM, Soule G, Tierney K, Frost KL, Booth S, Safronetz D. A recombinant vesicular stomatitis-based Lassa fever vaccine elicits rapid and long-term protection from lethal Lassa virus infection in guinea pigs. NPJ Vaccines. 2019;4. doi:10.1038/s41541-019-0104-x.
  • Profectus Biosciences. CEPI awards contract worth up to $36 million to profectus biosciences and emergent biosolutions to develop lassa virus vaccine [Internet]. 2018 [accessed 2019 Mar 5]. http://www.profectusbiosciences.com/news/releases.html
  • Rodriguez SE, Cross RW, Fenton KA, Bente DA, Mire CE, Geisbert TW. Vesicular stomatitis virus-based vaccine protects mice against crimean-congo hemorrhagic fever. Sci Rep. 2019;9. doi:10.1038/s41598-019-44210-6.
  • Chattopadhyay A, Rose JK. Complementing defective viruses that express separate paramyxovirus glycoproteins provide a new vaccine vector approach. J Virol. 2011;85:2004–11. doi:10.1128/JVI.01852-10.
  • Van Den Pol AN, Mao G, Chattopadhyay A, Rose JK, Davis JN. Chikungunya, influenza, Nipah, and semliki forest chimeric viruses with vesicular stomatitis virus: actions in the brain. J Virol. 2017;91:e02154–16. doi:10.1128/JVI.02154-16.
  • Lo MK, Bird BH, Chattopadhyay A, Drew CP, Martin BE, Coleman JD, Rose JK, Nichol ST, Spiropoulou CF. Single-dose replication-defective VSV-based Nipah virus vaccines provide protection from lethal challenge in Syrian hamsters. Antiviral Res. 2014;101:26–29. doi:10.1016/j.antiviral.2013.10.012.
  • Debuysscher BL, Scott D, Marzi A, Prescott J, Feldmann H. Single-dose live-attenuated Nipah virus vaccines confer complete protection by eliciting antibodies directed against surface glycoproteins. Vaccine. 2014;32:2637–44. doi:10.1016/j.vaccine.2014.02.087.
  • Debuysscher BL, Scott D, Thomas T, Feldmann H, Prescott J. Peri-exposure protection against Nipah virus disease using a single-dose recombinant vesicular stomatitis virus-based vaccine. NPJ Vaccines. 2016;1:16002. doi:10.1038/npjvaccines.2016.2.
  • Prescott J, Debuysscher BL, Feldmann F, Gardner DJ, Haddock E, Martellaro C, Scott D, Feldmann H. Single-dose live-attenuated vesicular stomatitis virus-based vaccine protects African green monkeys from Nipah Virus disease. Vaccine. 2015;33:2823–29. doi:10.1016/j.vaccine.2015.03.089.
  • Geisbert TW, Daddario-Dicaprio KM, Hickey AC, Smith MA, Chan Y-P. Development of an acute and highly pathogenic nonhuman primate model of nipah virus infection. PLoS One. 2010;5:10690. doi:10.1371/journal.pone.0010690.
  • Kapadia SU, Simon ID, Rose JK. SARS vaccine based on a replication-defective recombinant vesicular stomatitis virus is more potent than one based on a replication-competent vector. Virology. 2008;376:165–72. doi:10.1016/j.virol.2008.03.002.
  • Kapadia SU, Rose JK, Lamirande E, Vogel L, Subbarao K, Roberts A. Long-term protection from SARS coronavirus infection conferred by a single immunization with an attenuated VSV-based vaccine. Virology. 2005;340:174–82. doi:10.1016/j.virol.2005.06.007.
  • Liu R, Wang J, Shao Y, Wang X, Zhang H, Shuai L, Ge J, Wen Z, Bu Z. A recombinant VSV-vectored MERS-CoV vaccine induces neutralizing antibody and T cell responses in rhesus monkeys after single dose immunization. Antiviral Res. 2018;150:30–38. doi:10.1016/j.antiviral.2017.12.007.
  • Modjarrad K, Moorthy VS, Ben Embarek P, Van Kerkhove M, Kim J, Kieny M-P A roadmap for MERS-CoV research and product development: report from a World Health Organization consultation. Nat Med 2016; 22:701–05. doi:10.1038/nm.4131
  • Zanluca C, Campos Andrade de Melo V, Luiza Pamplona Mosimann A, Igor Viana Dos Santos G, Nunes Duarte Dos Santos C, Luz K. First report of autochthonous transmission of Zika virus in Brazil. Mem Inst Oswaldo Cruz Rio Janeiro 2015; 110:569–72. doi:10.1590/0074-02760150192
  • Gatherer D, Kohl A. Zika virus: a previously slow pandemic spreads rapidly through the Americas. J Gen Virol. 2016;97:269–73. doi:10.1099/jgv.0.000381.
  • Betancourt D, De Queiroz NMGP, Xia T, Ahn J, Barber GN. cutting Edge: innate immune augmenting vesicular stomatitis virus expressing zika virus proteins confers protective immunity. J Immunol. 2017;198:3023–28. doi:10.4049/jimmunol.1602180.
  • Emanuel J, Callison J, Dowd KA, Pierson TC, Feldmann H, Marzi A. A VSV-based Zika virus vaccine protects mice from lethal challenge. Sci Rep. 2018;8:11043. doi:10.1038/s41598-018-29401-x.
  • Li A, Yu J, Lu M, Ma Y, Attia Z, Shan C, Xue M, Liang X, Craig K, Makadiya N, et al. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nat Commun. 2018;9:1–17.
  • Shi X, Hu J, Guo J, Wu C, Xiong S, Dong C. A vesicular stomatitis virus-based vaccine carrying zika virus capsid protein protects mice from viral infection. Virol Sin. 2019;34:106–10. doi:10.1007/s12250-019-00083-7.
  • Yan Q, Wu L, Chen L, Qin Y, Pan Z, Chen M. Vesicular stomatitis virus-based vaccines expressing EV71 virus-like particles elicit strong immune responses and protect newborn mice from lethal challenges. Vaccine. 2016;34:4196–204. doi:10.1016/j.vaccine.2016.10.045.
  • Chattopadhyay A, Wang E, Seymour R, Weaver SC, Rose JK. A chimeric vesiculo/alphavirus is an effective alphavirus vaccine. J Virol. 2013;87:395–402. doi:10.1128/JVI.01860-12.
  • Chattopadhyay A, Aguilar PV, Bopp NE, Yarovinsky TO, Weaver SC, Rose JK. A recombinant virus vaccine that protects against both Chikungunya and Zika virus infections. Vaccine. 2018;36:3894–900. doi:10.1016/j.vaccine.2018.05.095.
  • Iyer AV, Pahar B, Boudreaux MJ, Wakamatsu N, Roy AF, Chouljenko VN, Baghian A, Apetrei C, Marx PA, Kousoulas KG. Recombinant vesicular stomatitis virus-based west Nile vaccine elicits strong humoral and cellular immune responses and protects mice against lethal challenge with the virulent west Nile virus strain LSU-AR01. Vaccine. 2009;27:893–903. doi:10.1016/j.vaccine.2008.11.087.
  • Lauretti F, Chattopadhyay A, Freitas De Oliveira França R, Castro-Jorge L, Rose J, Da Fonseca BAL. Recombinant vesicular stomatitis virus-based dengue-2 vaccine candidate induces humoral response and protects mice against lethal infection. Hum Vaccin Immunother. 2016;12:2327–33. doi:10.1080/21645515.2016.1208328.
  • Dong F, Li D, Wen D, Li S, Zhao C, Qi Y, Jangra RK, Wu C, Xia D, Zhang X, et al. Single dose of a rVSV-based vaccine elicits complete protection against severe fever with thrombocytopenia syndrome virus. NPJ Vaccines. 2019 Jan 25;4:5. eCollection 2019. doi:10.1038/s41541-018-0096-y.
  • Prescott J, Debuysscher BL, Brown KS, Feldmann H. Long-term single-dose efficacy of a vesicular stomatitis virus-based andes virus Vaccine in syrian hamsters. Viruses. 2014;6:516–23. doi:10.3390/v6020516.
  • Tsuda Y, Safronetz D, Brown K, Lacasse R, Marzi A, Ebihara H, Feldmann H. Protective efficacy of a bivalent recombinant vesicular stomatitis virus vaccine in the syrian hamster model of lethal ebola virus infection. J Infect Dis. 2011;204:S1090–1097. doi:10.1093/infdis/jir379.
  • Brown KS, Safronetz D, Marzi A, Ebihara H, Feldmann H. Vesicular stomatitis virus-based vaccine protects hamsters against lethal challenge with Andes virus. J Virol. 2011;85:12781–91. doi:10.1128/JVI.00794-11.