key: cord-1013575-u9vgycib authors: Volkmann, Ariane; Williamson, Anna-Lise; Weidenthaler, Heinz; Meyer, Thomas P.H.; Robertson, James S.; Excler, Jean-Louis; Condit, Richard C.; Evans, Eric; Smith, Emily R.; Kim, Denny; Chen, Robert T. title: The Brighton Collaboration standardized template for collection of key information for risk/benefit assessment of a Modified Vaccinia Ankara (MVA) vaccine platform date: 2020-10-17 journal: Vaccine DOI: 10.1016/j.vaccine.2020.08.050 sha: b5aba3cfcc74f9043faf56ed6ca652fde32e9a68 doc_id: 1013575 cord_uid: u9vgycib The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. The Modified Vaccinia Ankara (MVA) vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features specifically of the MVA-BN vector system, followed by a template with details on the safety and characteristics of an MVA-BN based vaccine against Zaire ebolavirus and other filovirus strains. The MVA-BN-Filo vaccine is based on a live, highly attenuated poxviral vector incapable of replicating in human cells and encodes glycoproteins of Ebola virus Zaire, Sudan virus and Marburg virus and the nucleoprotein of the Thai Forest virus. This vaccine has been approved in the European Union in July 2020 as part of a heterologous Ebola vaccination regimen. The MVA-BN vector is attenuated following over 500 serial passages in eggs, showing restricted host tropism and incompetence to replicate in human cells. MVA has six major deletions and other mutations of genes outside these deletions, which all contribute to the replication deficiency in human and other mammalian cells. Attenuation of MVA-BN was demonstrated by safe administration in immunocompromised mice and non-human primates. In multiple clinical trials with the MVA-BN backbone, more than 7800 participants have been vaccinated, demonstrating a safety profile consistent with other licensed, modern vaccines. MVA-BN has been approved as smallpox vaccine in Europe and Canada in 2013, and as smallpox and monkeypox vaccine in the US in 2019. No signal for inflammatory cardiac disorders was identified throughout the MVA-BN development program. This is in sharp contrast to the older, replicating vaccinia smallpox vaccines, which have a known risk for myocarditis and/or pericarditis in up to 1 in 200 vaccinees. MVA-BN-Filo as part of a heterologous Ebola vaccination regimen (Ad26.ZEBOV/MVA-BN-Filo) has undergone clinical testing including Phase III in West Africa and is currently in use in large scale vaccination studies in Central African countries. This paper provides a comprehensive picture of the MVA-BN vector, which has reached regulatory approvals, both as MVA-BN backbone for smallpox/monkeypox, as well as for the MVA-BN-Filo construct as part of an Ebola vaccination regimen, and therefore aims to provide solutions to prevent disease from high-consequence human pathogens. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. The Modified Vaccinia Ankara (MVA) vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features specifically of the MVA-BN vector system, followed by a template with details on the safety and characteristics of an MVA-BN based vaccine against Zaire ebolavirus and other filovirus strains. The MVA-BN-Filo vaccine is based on a live, highly attenuated poxviral vector incapable of replicating in human cells and encodes glycoproteins of Ebola virus Zaire, Sudan virus and Marburg virus and the nucleoprotein of the Thai Forest virus. This vaccine has been approved in the European Union in July 2020 as part of a heterologous Ebola vaccination regimen. The MVA-BN vector is attenuated following over 500 serial passages in eggs, showing restricted host tropism and incompetence to replicate in human cells. MVA has six major deletions and other mutations of genes outside these deletions, which all contribute to the replication deficiency in human and other mammalian cells. Attenuation of MVA-BN was demonstrated by safe administration in immunocompromised mice and non-human primates. In multiple clinical trials with the MVA-BN backbone, more than 7800 participants have been vaccinated, demonstrating a safety profile consistent with other licensed, modern vaccines. MVA-BN has been approved as smallpox vaccine in Europe and Canada in 2013, and as smallpox and monkeypox vaccine in the US in 2019. No signal for inflammatory cardiac disorders was identified throughout the MVA-BN development program. This is in sharp contrast to the older, replicating vaccinia smallpox vaccines, which have a known risk for myocarditis and/or pericarditis in up to 1 in 200 vaccinees. MVA-BN-Filo as part of a heterologous Ebola vaccination regimen (Ad26.ZEBOV/MVA-BN-Filo) has undergone clinical testing including Phase III in West Africa and is currently in use in large scale vaccination studies in Central African countries. This paper provides a comprehensive picture of the MVA-BN vector, which has reached regulatory approvals, both as MVA-BN backbone for smallpox/monkeypox, as well as for the MVA-BN-Filo construct as part of an Ebola vaccination regimen, and therefore aims to provide solutions to prevent disease from high-consequence human pathogens. Ó 2020 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). The Brighton Collaboration (www.brightoncollaboration.org) was launched in 2000 to improve the science of vaccine safety [1] . The Brighton Collaboration formed the Viral Vector Vaccines Safety Working Group (V3SWG) in October 2008 to improve our ability to anticipate potential safety issues and meaningfully assess or interpret safety data, thereby facilitating greater public acceptance when a viral vector vaccine is licensed [2] . The V3SWG has developed a standardized template describing the key characteristics of a novel viral vaccine vector to facilitate the scientific discourse among key stakeholders and increase the transparency and comparability of information. This introduction and the ''specific instructions" provide definitions and additional guidance for completing the template (V2.0) that follows. Viral vector vaccines are laboratory-generated, chimeric viruses that are based upon replicating or non-replicating virus vectors into which have been spliced genes encoding antigenic proteins for a target pathogen. Consideration of safety issues associated with viral vector vaccines requires a clear understanding of the agents used for construction of the vaccine. These include (1) the wild type virus from which the vector is derived, referred to in the template as ''wild type virus"; (2) the vector itself before incorporation of the foreign antigen, referred to in the template as ''viral vector"; and (3) the final recombinant viral vector vaccine, referred to in the template as ''vaccine". Wild type viruses used as vectors may originate from human or non-human hosts and may have low or high pathogenic potential in humans regardless of species of origin. Viral vectors can originate from attenuated human vaccines, from attenuated human viruses, from human viruses with low pathogenic potential, from animal viruses with low human pathogenic potential, and from vectors (for the expression of proteins) which are then adapted as a viral vector (such as DNA plasmids or baculovirus vector vaccines) to be used as a vaccine in humans or animals. Thus, viral vectors usually, but not always, have properties in a human host that differ from wild type virus from which they were derived. Incorporation of a target antigen into a viral vector to create a vaccine may alter the properties of the vector such that the vaccine may have properties that differ from the vector. The Brighton Collaboration Vaccine Vector template is designed to describe vectors into which transgenes may be incorporated to create vaccines. However, pursuant to understanding completely the safety aspects of a given vector, consideration is given to the wild type virus from which the vector is derived ( The world was declared to be free of smallpox in May 1980 and this was because of global vaccination with vaccinia virus (VACV) based vaccines [3] . Although these vaccines were very successful at preventing smallpox there were serious adverse events indicating the need for a less virulent vaccine [4, 5] . Due to these often-severe post-vaccination complications associated with Vaccinia viruses, there were several attempts to generate a more attenuated, safe smallpox vaccine. Modified vaccinia Ankara (MVA) originates from the dermal Vaccinia Virus Ankara strain (Chorioallantois Vaccinia Virus Ankara, CVA) that was maintained in the Vaccination Institute Ankara for many years and used as the basis for vaccination of humans. During the period of 1960 to 1974, Prof. Anton Mayr and his colleagues (University of Munich, Germany, Institute for Microbiology and Infectious Diseases of Animals) succeeded in attenuating CVA by over 570 continuous passages in primary CEF (chicken embryo fibroblast) cells. A reduced virulence of CVA was reported from passage 371 on CEF cells [6] . From passage 516, the attenuated CVA virus was renamed MVA to discriminate it from other attenuated Vaccinia virus strains [7] [8] [9] . In clinical trials with MVA, the pock lesions associated with vaccinia virus vaccination are not seen [9] . This attenuated MVA vaccine was used in more than 120,000 vaccinees for priming prior to administration of a conventional smallpox vaccine in a two-step protocol used in the 1970s in Europe [5, 8, 10] . In the last decades, multiple recombinant MVA vectors have been tested as vaccine candidates against various pathogens, such as human immunodeficiency viruses, Mycobacterium tuberculosis, Plasmodium falciparum or Middle East Respiratory Syndrome virus [8, 11] . MVA-BN, that is derived from the MVA strain developed in Prof. Anton Mayr's laboratory, is a further attenuated MVA strain, which has lost its ability to replicate in most mammalian cell types, including human cell lines and is safe in severely immune compromised animals [12, 13] . The hallmark of MVA-BN is the fact that it does not productively replicate in the human keratinocyte cell line HaCat, the human cervix adenocarcinoma cell line HeLa, the human embryo kidney cell line 293 (HEK293), and the human bone osteosarcoma cell line 143B [12, 14] . However, like other MVA strains, MVA-BN effectively infects mammalian cells. Infection of mammalian cells results in transcription of the viral genes, but no MVA-BN virus is released from the cells due to a genetic block in the viral assembly and egress. The infected cells eventually undergo apoptosis (programmed cell death) [15] [16] [17] . There are several deletions and other mutations in MVA that account for the change in host-range of the virus. Six major deletions mainly account for a reduction in the size of the original vaccinia genome from 204.5 kb to 178 kb for the MVA strain [14, 18] . Sequencing of the genome revealed that these deletions included immune evasion genes, host interactive protein genes and some structural proteins [19] . Due to the lack of replication competence in many mammalian cells including human cells, MVA-BN can be safely administered to immunocompromised humans. This safety feature has also been confirmed in severely immunocompromised animals [12, 13] . MVA-BN is now a licensed smallpox vaccine (since 2013 in EU and Canada and since 2019 in the US, where it is also licensed as MVA-BN was shown to be more attenuated compared to two other MVA isolates, namely MVA-572 and MVA-I721, and even fails to replicate in immune compromised animals [10, 12] . Despite its high attenuation and reduced virulence, MVA-BN has been shown to elicit both humoral and cellular immune responses to Vaccinia virus and foreign genes cloned into the MVA-BN genome [31] [32] [33] [34] [35] [36] . MVA is a potent inducer of type I interferon (IFN) in human cells. MVA expresses a soluble interleukin-1 receptor, which has been implicated as an antivirulence factor for certain Poxviruses. MVA does not express soluble receptors for IFN-c, IFN-a/-b, tumor necrosis factor and CC chemokines [37] . Neurovirulence assessment of vaccinia virus based smallpox vaccines demonstrated the inability of MVA to replicate in suckling mouse brains following intracranial inoculation of 10-100 plaque forming units (pfu) of virus, while all other vaccinia virus strains tested (Dryvax Ò , Lister, Copenhagen, IHD-J, WR) replicated to peak titers of 10 7 to 10 8 pfu per gram tissue. Moreover, none of the doses of MVA tested, i.e. up to 10 5 pfu administered intracranially, induced death in the suckling mice. In contrast, mortality induced by Dryvax Ò started at a dose of 10 pfu; Lister, Copenhagen, IHD-J and WR induced death in some mice at 1 pfu and injection of 10 3 pfu was 100% lethal confirming neurovirulence reported for these strains [38] . MVA-BN replicates extensively and rapidly in CEF cells and also in certain other avian cell lines. (replicating or non-replicating) MVA-BN is a non-replicating vector in humans. VACV is the virus used for the replicating smallpox vaccine that was utilized during eradication and now ACAM2000. [39] 3.2 What is the natural host for the wild type virus? The original host is unknown, but VACV can replicate in a range of animals including primates, rodents, lagomorphs and ungulates as well as humans. The origin of the VACV is debated and there is some evidence that it originated from a horse poxvirus which was able to infect cows. In Brazil and in India VACV is endemic in animals, with occasional transmission to humans, and is thought to originate from smallpox vaccine campaigns. [ [39] [40] [41] [42] [43] 3.3. How is the wild type virus normally transmitted? The typical manifestation of the wildtype virus infection are vesiculopustular lesions or dermal vesicles (pox lesions). These lesions contain infectious virus particles. Transmission can occur by close contact with infected area. There is no evidence that VACV is transmitted via airborne infection. There is evidence that shedding of VACV from the vaccination lesion of healthy primary vaccinees occurs from about the third day to the end of the third week after vaccination. There are rare reports of transmission of VACV. [44] 3.4. Does the wild type virus establish a latent or persistent infection? No, the infections are acute [45] 3.5. Does the wild type virus replicate in the nucleus? No. Poxviruses replicate in the cytoplasm. [46] (continued on next page) [ 51, 52] Reference for myocarditis and/or pericarditis frequency: [53] In immunocompromised humans Can be fatal and so vaccination with VACV is contraindicated. Applicable for the replicating vacciniabased smallpox vaccines [54, 55] In human neonates, infants, children Children <12 months of age have an increased rate of the complications listed above for healthy human host [55, 56] During pregnancy and in the unborn in humans Poxviruses make excellent vaccine delivery vehicles since their genomes allow large insertions of foreign DNA [20, 21] . Conventionally, foreign genes are inserted into poxviruses by homologous recombination into non-essential genes or into intergenic regions [22] . The genes are under the control of a poxvirus promoter and may have a reporter gene or selection marker to aid selection of recombinants [23] [24] [25] [26] . The foreign genes are usually modified to remove the poxvirus early transcription termination signals (TTTTTNT) [27] and must be devoid of introns. Recently a Horsepox virus genome has been made by chemical synthesis and rescued by coinfection with Shope fibroma virus [28] demonstrating that this strategy can potentially be used in the future to synthesize other poxviruses. One of the most successful poxvirus vectored vaccines is the VACV vectored rabies vaccine distributed in oral baits for foxes, which has almost completely eradicated terrestrial rabies in parts of Europe [29, 30] . Host restricted poxviruses, such as the canary poxvirus, ALVAC, have been registered as commercial vaccine vectors for a number of veterinary diseases including equine influenza, canine distemper, rabies, feline leukemia and West-Nile fever [31] . This publication presents the properties of MVA-BN as a vaccine vector and specifically focuses on MVA-BN-Filo as a component of an Ebola vaccine two-dose regimen (Ad26.ZEBOV/MVA-BN-Filo) which was granted Marketing Authorization by the European Commission on July 1, 2020 [32] (Table 1 ). The findings, opinions, conclusions, and assertions contained in this consensus document are those of the individual members of the Working Group. They do not necessarily represent the official positions of any participant's organization (e.g., government, university, or corporations) and should not be construed to represent any Agency determination or policy. The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: The authors Ariane Volkmann, Heinz Weidenthaler, and Thomas Meyer are employees of Bavarian Nordic. The Brighton Collaboration: addressing the need for standardized case definitions of adverse events following immunization (AEFI). Vaccine The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG). Vaccine Smallpox: emergence, global spread, and eradication Reactions to small pox vaccine in naive and previously-vaccinated individuals IMVAMUNE: modified vaccinia Ankara strain as an attenuated smallpox vaccine Comparative experimental works on cow pox virus vaccines Changes in the vaccinia virus through continuing passages in chick embryo fibroblast cultures Modified vaccinia virus ankara: history, value in basic research, and current perspectives for vaccine development Intracutaneous smallpox vaccination with a weak pathogenic vaccinia virus Experiences with immunization against orthopox viruses of humans and animals using vaccine strain MVA Modified vaccinia virus ankara (MVA) as production platform for vaccines against influenza and other viral respiratory diseases Modified vaccinia Ankara strains with identical coding sequences actually represent complex mixtures of viruses that determine the biological properties of each strain Safety of modified vaccinia virus Ankara (MVA) in immune-suppressed macaques Introduction of the six major genomic deletions of modified vaccinia virus Ankara (MVA) into the parental vaccinia virus is not sufficient to reproduce an MVA-like phenotype in cell culture and in mice Recombinant MVA vaccines: dispelling the myths Host range and cytopathogenicity of the highly attenuated MVA strain of vaccinia virus: propagation and generation of recombinant viruses in a nonhuman mammalian cell line Mapping of deletions in the genome of the highly attenuated vaccinia virus MVA and their influence on virulence Genomic sequence of chorioallantois vaccinia virus Ankara, the ancestor of modified vaccinia virus Ankara The complete genomic sequence of the modified vaccinia Ankara strain: comparison with other orthopoxviruses Poxvirus-based vectors as vaccine candidates Poxviruses as vaccine vectors Generation of Recombinant Vaccinia Viruses Stable expression of the vaccinia virus K1L gene in rabbit cells complements the host range defect of a vaccinia virus mutant Engineering recombinant poxviruses using a compact GFP-blasticidin resistance fusion gene for selection Construction, characterization, and immunogenicity of a multigene modified vaccinia Ankara (MVA) vaccine based on HIV type 1 subtype C Generation of recombinant Vaccinia viruses Removal of cryptic poxvirus transcription termination signals from the human immunodeficiency virus type 1 envelope gene enhances expression and immunogenicity of a recombinant vaccinia virus Vaccine xxx (xxxx) xxx Construction of an infectious horsepox virus vaccine from chemically synthesized DNA fragments Largescale eradication of rabies using recombinant vaccinia-rabies vaccine Oral vaccination of wildlife against rabies: opportunities and challenges in prevention and control Development and registration of recombinant veterinary vaccines. The example of the canarypox vector platform Safety and immunogenicity of novel adenovirus Type 26-and modified vaccinia ankara-vectored Ebola vaccines: a randomized clinical trial Immunogenicity and safety of three consecutive production lots of the non replicating smallpox vaccine MVA: A randomised, double blind, placebo controlled phase III trial Phase 3 efficacy trial of modified vaccinia ankara as a vaccine against smallpox Safety and immunogenicity of novel modified vaccinia ankara-vectored RSV vaccine: a randomized phase I clinical trial Therapeutic vaccination of HIV-1-infected patients on HAART with a recombinant HIV-1 nef-expressing MVA: safety, immunogenicity and influence on viral load during treatment interruption Phase I study of a Poxviral TRICOM-based vaccine directed against the transcription factor Brachyury Therapeutic vaccination with MVA-HIV-1 nef elicits Nef-specific T-helper cell responses in chronically HIV-1 infected individuals Modified vaccinia virus Ankara undergoes limited replication in human cells and lacks several immunomodulatory proteins: implications for use as a human vaccine A mouse-based assay for the preclinical neurovirulence assessment of vaccinia virus-based smallpox vaccines Poxviruses and the evolution of host range and virulence Genomic analysis, phenotype, and virulence of the historical brazilian smallpox vaccine strain IOC: implications for the origins and evolutionary relationships of Vaccinia virus An emergent poxvirus from humans and cattle in Rio de Janeiro State: Cantagalo virus may derive from Brazilian smallpox vaccine Vaccinia virus natural infections in Brazil: the good, the bad, and the ugly Bovine Vaccinia: insights into the disease in cattle Transmission of vaccinia virus and rationale for measures for prevention Poxvirus pathogenesis Poxvirus DNA replication MVA as a vector for vaccines against HIV-1 Biosafety aspects of modified vaccinia virus Ankara (MVA)-based vectors used for gene therapy or vaccination Environmental risk assessment of clinical trials involving modified vaccinia virus Ankara (MVA)-based vectors Hazard characterization of modified vaccinia virus ankara vector: what are the knowledge gaps? Adverse reactions after smallpox vaccination Economics of cardiac adverse events after smallpox vaccination: lessons from the 2003 US Vaccination Program A prospective study of the incidence of myocarditis/pericarditis and new onset cardiac symptoms following smallpox and influenza vaccination Disseminated vaccinia in a military recruit with human immunodeficiency virus (HIV) disease Clinical guidance for smallpox vaccine use in a postevent vaccination program Complications of smallpox vaccination Smallpox vaccination reactions, prophylaxis, and therapy of complications ACAM2000 TM Prescribing Information Smallpox vaccine: contraindications, administration, and adverse reactions Membrane fusion during poxvirus entry Smallpox vaccines: targets of protective immunity Centers for Disease C, Prevention. Smallpox vaccination and adverse reactions. Guidance for clinicians Use of vaccinia virus smallpox vaccine in laboratory and health care personnel at risk for occupational exposure to orthopoxviruses -recommendations of the Advisory Committee on Immunization Practices (ACIP) Spread of vaccinia virus through shaving during military training Repair of a previously uncharacterized second host-range gene contributes to full replication of modified vaccinia virus Ankara (MVA) in human cells Safety, immunogenicity, and surrogate markers of clinical efficacy for modified vaccinia Ankara as a smallpox vaccine in HIV-infected subjects Safety and immunogenicity of modified vaccinia ankara-bavarian nordic smallpox vaccine in vaccinia-naive and experienced human immunodeficiency virusinfected individuals: an open-label, controlled clinical Phase II Trial A randomized phase II trial to compare safety and immunogenicity of the MVA-BN smallpox vaccine at various doses in adults with a history of AIDS A Phase 1 dose escalation trial of BN-CV301, a recombinant poxviral vaccine targeting MUC1 and CEA with costimulatory molecules Phase I trial of a modified vaccinia ankara priming vaccine followed by a fowlpox virus boosting vaccine modified to express brachyury and costimulatory molecules in advanced solid tumors Safety and immunogenicity of modified vaccinia Ankara as a smallpox vaccine in people with atopic dermatitis A multicenter, open-label, controlled Phase II study to evaluate safety and immunogenicity of MVA smallpox vaccine (IMVAMUNE) in 18-40 year old subjects with diagnosed atopic dermatitis Safety and immunogenicity of modified vaccinia Ankara in hematopoietic stem cell transplant recipients: a randomized, controlled trial A comparative Phase I study of combination, homologous subtype-C DNA, MVA, and Env gp140 protein/adjuvant HIV vaccines in two immunization regimes Immunogenicity of a novel Clade B HIV-1 vaccine combination: results of phase 1 randomized placebo controlled trial of an HIV-1 GM-CSFexpressing DNA prime with a modified vaccinia Ankara vaccine boost in healthy HIV-1 uninfected adults Safety and vaccine-induced HIV-1 immune responses in healthy volunteers following a late MVA-B boost 4 years after the last immunization Safety and immunogenicity of PENNVAX-G DNA prime administered by biojector 2000 or CELLECTRA electroporation device with modified Vaccinia Ankara-CMDR boost Subtype C gp140 vaccine boosts immune responses primed by the South African AIDS vaccine initiative DNA-C2 and MVA-C HIV vaccines after more than a 2-Year Gap A randomized, double-blind, placebo-controlled Phase II trial investigating the safety and immunogenicity of modified Vaccinia Ankara smallpox vaccine (MVA-BN(R)) in 56-80-year-old subjects Vaccine xxx (xxxx) xxx Boosting with subtype C CN54rgp140 protein adjuvanted with glucopyranosyl lipid adjuvant after priming with HIV-DNA and HIV-MVA is safe and enhances immune responses: a Phase I trial Safety and immunogenicity of IMVAMUNE, a promising candidate as a third generation smallpox vaccine Clinical and immunologic responses to multiple doses of IMVAMUNE (Modified Vaccinia Ankara) followed by Dryvax challenge A randomized, double-blind, dose-finding Phase II study to evaluate immunogenicity and safety of the third generation smallpox vaccine candidate IMVAMUNE Safety and immunogenicity of IMVAMUNE(R) smallpox vaccine using different strategies for a post event scenario Phase II randomized, double-blinded comparison of a single high dose (5x10(8) TCID50) of modified vaccinia Ankara compared to a standard dose (1x10(8) TCID50) in healthy vaccinia-naive individuals Comparison of lyophilized versus liquid modified vaccinia Ankara (MVA) formulations and subcutaneous versus intradermal routes of administration in healthy vaccinia-naive subjects Cardiac safety of Modified Vaccinia Ankara for vaccination against smallpox in a young, healthy study population Safety and immunogenicity of a modified vaccinia Ankara vaccine using three immunization schedules and two modes of delivery: a randomized clinical non-inferiority trial Intergenic region 3 of modified vaccinia ankara is a functional site for insert gene expression and allows for potent antigen-specific immune responses Construction and biological characteristic for the recombinant modified vaccinia virus ankara co-expressing modified GP5 and M protein of porcine reproductive and respiratory syndrome virus Genetic stability of recombinant MVA-BN A monovalent chimpanzee adenovirus Ebola vaccine boosted with MVA Safety and immunogenicity of a 2-dose heterologous vaccination regimen with Ad26.ZEBOV and MVA-BN-Filo Ebola vaccines: 12-month data from a Phase 1 randomized clinical trial in Uganda and Tanzania Safety and immunogenicity of a 2-dose heterologous vaccine regimen with Ad26.ZEBOV and MVA-BN-Filo Ebola vaccines: 12-month data from a phase 1 randomized clinical trial in A two-dose heterologous prime-boost vaccine regimen eliciting sustained immune responses to Ebola Zaire could support a preventive strategy for future outbreaks A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates Correction: A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates Broad antibody and cellular immune response from a Phase 2 clinical trial with a novel multivalent poxvirus based RSV vaccine Recombinant modified vaccinia virus Ankara expressing a soluble form of glycoprotein B causes durable immunity and neutralizing antibodies against multiple strains of human cytomegalovirus Environmental resistance, disinfection, and sterilization of poxviruses Rapid protection in a monkeypox model by a single injection of a replicationdeficient vaccinia virus Modified vaccinia virus Ankara protects macaques against respiratory challenge with monkeypox virus Survival of lethal poxvirus infection in mice depends on TLR9, and therapeutic vaccination provides protection Immunogenicity and protection after vaccination with a modified Vaccinia virus Ankara-vectored yellow fever vaccine in the hamster model Protective efficacy of monovalent and trivalent recombinant MVA-based vaccines against three encephalitic alphaviruses Poxvirus-based active immunotherapy synergizes with CTLA-4 blockade to increase survival in a murine tumor model by improving the magnitude and quality of cytotoxic T cells Evaluation of the immunogenicity and impact on the latent HIV-1 reservoir of a conserved region vaccine, MVA.HIVconsv, in antiretroviral therapy-treated subjects A phase I trial of preventive HIV vaccination with heterologous poxviral-vectors containing matching HIV-1 inserts in healthy HIV-uninfected subjects First field efficacy trial of the ChAd63 MVA ME-TRAP vectored malaria vaccine candidate in 5-17 months old infants and children Safety and efficacy of novel malaria vaccine regimens of RTS, S/AS01B alone, or with concomitant ChAd63-MVA-vectored vaccines expressing ME ChAdOx1 and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice Smallpox and Monkeypox Vaccine, Live, Nonreplicating) Prescribing Information Vaccinating against monkeypox in the Democratic Republic of the Congo Smallpox vaccination and its consequences: first experiences with the highly attenuated smallpox vaccine ''MVA"