1
|
Williams A, Ravel J, Kaul R, Armstrong E, Huibner S, Holm JB. Temporal Dynamics of the Vaginal Microbiome and Host Immune Markers Before, During, and After Metronidazole Treatment for Bacterial Vaginosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643085. [PMID: 40161639 PMCID: PMC11952498 DOI: 10.1101/2025.03.13.643085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
This study analyzed metagenomic and immune marker profiles of seven individuals before, during, and after a 7-day course of metronidazole treatment for bacterial vaginosis (BV). Treatment reduced BV-associated bacteria and immune marker levels, with distinct early (days 1-4) and late (days 5-7) phases. Post-treatment, variability in microbial and immune marker profiles demonstrated a rapid resurgence of certain BV associated bacteria, highlighting the need for additional strategies like probiotics to maintain a healthy vaginal microbiome. The study found significant host and microbial influences on immune response variance, with IP-10 and sEcad highly correlated with the vaginal microbiome. The findings identify optimal timing for administering live biotherapeutics to restore D-lactic acid-producing Lactobacillus species dominance and underscore the complexity of BV infection and treatment response among different people.
Collapse
Affiliation(s)
- Amanda Williams
- Center for Advanced Microbiome Research and Innovation, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Center for Advanced Microbiome Research and Innovation, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rupert Kaul
- Department of Medicine, University of Toronto; Toronto, Canada
- Department of Medicine, University Health Network, Toronto, Canada
| | - Eric Armstrong
- Department of Medicine, University of Toronto; Toronto, Canada
| | - Sanja Huibner
- Department of Medicine, University of Toronto; Toronto, Canada
| | - Johanna B. Holm
- Center for Advanced Microbiome Research and Innovation, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Rasmussen SA, Kim J, Jamieson DJ. Vaccines in Pregnancy: An Update on Recommendations From CDC's Advisory Committee on Immunization Practices. Birth Defects Res 2025; 117:e2459. [PMID: 39996387 DOI: 10.1002/bdr2.2459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Vaccinations in pregnancy are an essential part of prenatal care and play a critical role in protecting both pregnant persons and their infants from certain infectious diseases. In the United States, recommendations for vaccines are made through a comprehensive review of currently available scientific literature, including clinical trials and post-marketing surveillance data, by the Advisory Committee on Immunization Practices (ACIP). The ACIP is an advisory committee to the US Centers for Disease Control and Prevention (CDC), comprised of medical and public health experts who develop evidence-based recommendations and guidelines for vaccinations, including for pregnant persons. The ACIP has several work groups that review scientific evidence on an ongoing basis, and full-committee public meetings are held at least three times a year. As more data regarding the safety and efficacy of vaccines in pregnancy become available, these recommendations continue to evolve. To develop these recommendations, the ACIP carefully considers the risks of exposure to infectious agents against the potential risks of vaccination. We review here current ACIP recommendations for vaccinations and their use in pregnant persons. Recommendations are divided into four categories: vaccines recommended during pregnancy, vaccines recommended during pregnancy under certain circumstances, vaccines not recommended or contraindicated during pregnancy, and vaccines without specific ACIP recommendations. To ensure optimal care during pregnancy, healthcare providers who care for pregnant persons need to be familiar with these recommendations.
Collapse
Affiliation(s)
- Sonja A Rasmussen
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiyoung Kim
- Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine, Hempstead, New York, USA
- Long Island Jewish Forest Hills Hospital, Northwell Health, New York City, New York, USA
| | | |
Collapse
|
3
|
Lamb ER, Criss AK. Terminal complement complexes with or without C9 potentiate antimicrobial activity against Neisseria gonorrhoeae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633325. [PMID: 39868146 PMCID: PMC11760736 DOI: 10.1101/2025.01.16.633325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The complement cascade is a front-line defense against pathogens. Complement activation generates the membrane attack complex (MAC), a 10-11 nm diameter pore formed by complement proteins C5b through C8 and polymerized C9. The MAC embeds within the outer membrane of Gram-negative bacteria and displays bactericidal activity. In the absence of C9, C5b-C8 complexes can form 2-4 nm pores on membranes, but their relevance to microbial control is poorly understood. Deficiencies in terminal complement components uniquely predispose individuals to infections by pathogenic Neisseria, including N. gonorrhoeae (Gc). Increasing antibiotic resistance in Gc makes new therapeutic strategies a priority. Here, we demonstrate that MAC formed by complement activity in human serum disrupts the Gc outer and inner membranes, potentiating the activity of antimicrobials against Gc and re-sensitizing multidrug resistant Gc to antibiotics. C9-depleted serum also disrupts Gc membranes and exerts antigonococcal activity, effects that are not reported in other Gram-negative bacteria. C5b-C8 complex formation potentiates Gc sensitivity to azithromycin but not lysozyme. These findings expand our mechanistic understanding of complement lytic activity, suggest a size limitation for terminal complement-mediated enhancement of antimicrobials against Gc, and suggest complement manipulation can be used to combat drug-resistant gonorrhea. Importance The complement cascade is a front-line arm of the innate immune system against pathogens. Complement activation results in membrane attack complex (MAC) pores forming on the outer membrane of Gram-negative bacteria, resulting in bacterial death. Individuals who cannot generate MAC are specifically susceptible to infection by pathogenic Neisseria species including N. gonorrhoeae (Gc). High rates of gonorrhea and its complications like infertility, and high-frequency resistance to multiple antibiotics, make it important to identify new approaches to combat Gc. Beyond direct anti-Gc activity, we found the MAC increases the ability of antibiotics and antimicrobial proteins to kill Gc and re-sensitizes multidrug-resistant bacteria to antibiotics. The most terminal component, C9, is needed to potentiate the anti-Gc activity of lysozyme, but azithromycin activity is potentiated regardless of C9. These findings highlight the unique effects of MAC on Gc and suggest novel translational avenues to combat drug-resistant gonorrhea.
Collapse
Affiliation(s)
- Evan R. Lamb
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alison K. Criss
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
4
|
Richards PT, Briseño JAA, Brunton BA, Maury W. In Vivo Investigation of Filovirus Glycoprotein-Mediated Infection in a BSL2 Setting. Methods Mol Biol 2025; 2877:183-198. [PMID: 39585622 PMCID: PMC11727417 DOI: 10.1007/978-1-0716-4256-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Highly pathogenic viruses in the Filoviridae family are causative agents of filovirus disease (FVD). Ebola virus (EBOV) is one such member and, of all filoviruses, represents the largest threat to global public health. The study of FVD has been hampered by the lack of tools to study filovirus infection outside maximum containment laboratories. Recombinant vesicular stomatitis virus (VSV) lacking its native glycoprotein and expressing a filovirus glycoprotein (VSV-filo GP) has improved our understanding of GP-mediated host-cell interactions as well as adaptive and humoral immune responses in in vitro and in vivo studies. Furthermore, mouse models suitable for these studies are readily available. Here, we describe multiple injection routes for investigating filovirus GP-mediated infection and pathogenesis using VSV-filo GP and interferon α/β receptor-deficient (Ifnar-/-) mice as models. These tools can be safely used outside maximum containment laboratories, are cost effective, and easy to manipulate.
Collapse
Affiliation(s)
- Paige T Richards
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | | | - Bethany A Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
5
|
Michalow J, Hall L, Rowley J, Anderson RL, Hayre Q, Chico RM, Edun O, Knight J, Kuchukhidze S, Majaya E, Reed DM, Stevens O, Walters MK, Peters RPH, Cori A, Boily MC, Imai-Eaton JW. Prevalence of chlamydia, gonorrhoea, and trichomoniasis among male and female general populations in sub-Saharan Africa from 2000-2024: A systematic review and meta-regression analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.16.24319070. [PMID: 39763549 PMCID: PMC11702751 DOI: 10.1101/2024.12.16.24319070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Background Sub-Saharan Africa (SSA) has the highest sexually transmitted infection (STI) prevalence globally, but information about trends and geographic variation is limited by sparse aetiologic studies, particularly among men. This systematic review assessed chlamydia, gonorrhoea, and trichomoniasis prevalence by sex, sub-region, and year, and estimated male-to-female prevalence ratios for SSA. Methods We searched Embase, MEDLINE, Global Health, PubMed, and African Index Medicus for studies measuring STI prevalence among general populations from January 1, 2000, to September 17, 2024. We adjusted observations for diagnostic test performance and used log-binomial mixed-effects meta-regressions to estimate prevalence trends and sex-prevalence ratios. Findings Of 5202 records identified, we included 211 studies from 28 countries. In 2020, estimated prevalence among 15-49-year-olds in SSA for chlamydia was 6.6% (95%CI: 5.0-8.2%, n=169 observations) among females and 4.7% (3.4-6.1%, n=33) among males, gonorrhoea was 2.4% (1.4-3.3%, n=171) and 1.7% (0.7-2.6%, n=31), and trichomoniasis was 6.8% (3.6-9.9%, n=188) and 1.7% (0.7-2.7%, n=19). Male-to-female ratio estimates were 0.61 (0.53-0.71) for chlamydia, 0.81 (0.61-1.09) for gonorrhoea, and 0.23 (0.18-0.28) for trichomoniasis. From 2010-2020, chlamydia prevalence increased by 34.5% (11.1-62.9%) in SSA, while gonorrhoea and trichomoniasis trends were not statistically significant. Chlamydia and gonorrhoea prevalence were highest in Southern and Eastern Africa, whereas trichomoniasis was similar across sub-regions. Interpretation SSA has a high, geographically varied STI burden, with increasing prevalence of chlamydia. Region-specific sex-prevalence ratios differed from existing global ratios and should be considered in future burden estimates. Enhanced sex-stratified surveillance is crucial to guide national programmes and reduce STI prevalence in SSA. Funding Gates Foundation, Imperial College London, NIH, UKRI.
Collapse
Affiliation(s)
- Julia Michalow
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Lauren Hall
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Jane Rowley
- Department of Global HIV, Hepatitis and Sexually Transmitted Infections Programmes, World Health Organization, Geneva, Switzerland
| | - Rebecca L. Anderson
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Quinton Hayre
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - R. Matthew Chico
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Olanrewaju Edun
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Jesse Knight
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Salome Kuchukhidze
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Evidence Majaya
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Domonique M. Reed
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Oliver Stevens
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Magdalene K Walters
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Remco PH Peters
- Department of Global HIV, Hepatitis and Sexually Transmitted Infections Programmes, World Health Organization, Geneva, Switzerland
| | - Anne Cori
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Marie-Claude Boily
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Jeffrey W. Imai-Eaton
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Chan M, Warner BM, Audet J, Barker D, Tailor N, Vendramelli R, Truong T, Tierney K, Boese AS, Qiu H, Holtsberg FW, Aman J, Kodihalli S, Kobasa D. Delayed treatment of cynomolgus macaques with a FVM04/CA45 monoclonal antibody cocktail provides complete protection against lethal Sudan virus infection. J Virol 2024; 98:e0124223. [PMID: 39012096 PMCID: PMC11334508 DOI: 10.1128/jvi.01242-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
Sudan ebolavirus (SUDV) is a member of the genus Ebolavirus (Family Filoviridae) and has caused sporadic outbreaks of Ebola disease (EBOD), or more specifically Sudan virus disease (SVD), with high mortality rates in Africa. Current vaccines and therapies that have been developed for filoviruses are almost all specific for Ebola virus (EBOV; of the species Zaire ebolavirus), and there is a current lack of therapeutics specific for SUDV. The recent SUDV outbreak in Uganda, which was distributed across multiple districts, including Kampala, a densely populated urban center, highlights the critical need for the development of novel SUDV-specific or pan-Ebola virus therapeutics. Previous work has characterized two monoclonal antibodies, FVM04 and CA45, which have neutralization capabilities against both EBOV and SUDV and have shown protective efficacy in animal challenge studies. Here, we expand upon this work, showing that treatment with a monoclonal antibody cocktail consisting of FVM04 and CA45 provides full protection against lethal SUDV infection in cynomolgus macaques. Studies that evaluate outcomes at late time points after infection, once clinical signs of illness are apparent, are vital for assessing the therapeutic efficacy of antibody therapeutics. We have shown that when treatment is initiated as late as 5 days after infection, with a second dose given on day 8, that treated groups showed few clinical signs or morbidity, with complete survival. This work provides further evidence that FVM04 and CA45 have strong therapeutic potential against SUDV and their development as a pan-Ebola virus therapeutic should be pursued. IMPORTANCE There are currently no approved vaccines or therapeutics for Sudan virus, a filovirus which is highly related to Ebola virus and causes similar disease and outbreaks. In this study, a cocktail of two potent monoclonal antibodies that effectively neutralize Sudan virus was tested in a nonhuman primate model of Sudan virus disease. Treatment was highly effective, even when initiated as late as 5 days after infection, when clinical signs of infection were already evident. All treated animals showed complete recovery from infection, with little evidence of disease, while all animals that received a control treatment succumbed to infection within 8 days. The study further demonstrated the strong therapeutic potential of the antibody treatment and supported further development for use in Sudan virus outbreaks.
Collapse
MESH Headings
- Animals
- Female
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Disease Models, Animal
- Ebolavirus/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/drug therapy
- Macaca fascicularis
- Treatment Delay
- Male
Collapse
Affiliation(s)
- Mable Chan
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Bryce M. Warner
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Jonathan Audet
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Douglas Barker
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | - Nikesh Tailor
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Robert Vendramelli
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Kevin Tierney
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Amrit S. Boese
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Honguy Qiu
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | | | - Javad Aman
- Integrated BioTherapeutics, Rockville, Maryland, USA
| | - Shantha Kodihalli
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
7
|
Ren H, Xu R. Prevention and control of Ebola virus transmission: mathematical modelling and data fitting. J Math Biol 2024; 89:25. [PMID: 38963509 DOI: 10.1007/s00285-024-02122-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/16/2023] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
The Ebola virus disease (EVD) has been endemic since 1976, and the case fatality rate is extremely high. EVD is spread by infected animals, symptomatic individuals, dead bodies, and contaminated environment. In this paper, we formulate an EVD model with four transmission modes and a time delay describing the incubation period. Through dynamical analysis, we verify the importance of blocking the infection source of infected animals. We get the basic reproduction number without considering the infection source of infected animals. And, it is proven that the model has a globally attractive disease-free equilibrium when the basic reproduction number is less than unity; the disease eventually becomes endemic when the basic reproduction number is greater than unity. Taking the EVD epidemic in Sierra Leone in 2014-2016 as an example, we complete the data fitting by combining the effect of the media to obtain the unknown parameters, the basic reproduction number and its time-varying reproduction number. It is shown by parameter sensitivity analysis that the contact rate and the removal rate of infected group have the greatest influence on the prevalence of the disease. And, the disease-controlling thresholds of these two parameters are obtained. In addition, according to the existing vaccination strategy, only the inoculation ratio in high-risk areas is greater than 0.4, the effective reproduction number can be less than unity. And, the earlier the vaccination time, the greater the inoculation ratio, and the faster the disease can be controlled.
Collapse
Affiliation(s)
- Huarong Ren
- Complex Systems Research Center, Shanxi University, Taiyuan, 030006, Shanxi, China
- School of Mathematical Sciences, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Rui Xu
- Complex Systems Research Center, Shanxi University, Taiyuan, 030006, Shanxi, China.
| |
Collapse
|
8
|
Anderson EM, Coller BAG. Translational success of fundamental virology: a VSV-vectored Ebola vaccine. J Virol 2024; 98:e0162723. [PMID: 38305150 PMCID: PMC10994820 DOI: 10.1128/jvi.01627-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Ebola virus disease (EVD) caused by Ebola virus (EBOV) is a severe, often fatal, hemorrhagic disease. A critical component of the public health response to curb EVD epidemics is the use of a replication-competent, recombinant vesicular stomatitis virus (rVSV)-vectored Ebola vaccine, rVSVΔG-ZEBOV-GP (ERVEBO). In this Gem, we will discuss the past and ongoing development of rVSVΔG-ZEBOV-GP, highlighting the importance of basic science and the strength of public-private partnerships to translate fundamental virology into a licensed VSV-vectored Ebola vaccine.
Collapse
|
9
|
Moso MA, Lim CK, Williams E, Marshall C, McCarthy J, Williamson DA. Prevention and post-exposure management of occupational exposure to Ebola virus. THE LANCET. INFECTIOUS DISEASES 2024; 24:e93-e105. [PMID: 37722397 DOI: 10.1016/s1473-3099(23)00376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 09/20/2023]
Abstract
There have been significant advances in the prevention and management of Ebola virus disease (EVD) caused by Zaire Ebola virus (ZEBOV), including the development of two effective vaccines, rVSV-ZEBOV and Ad26.ZEBOV/MVA-BN-Filo. In addition, ZEBOV monoclonal antibodies have become first-line therapy for EVD. However, the 2022-23 outbreak of Sudan Ebola virus (SUDV) in Uganda has highlighted the gap in current therapies and vaccines, whose efficacy is uncertain against non-ZEBOV species. Health-care and laboratory staff working in EVD treatment centres or Ebola virus diagnostic and research laboratories face unique risks relating to potential occupational exposure to Ebola viruses. Given the substantial morbidity and mortality associated with EVD, facilities should have strategies in place to manage occupational exposures, including consideration of post-exposure therapies. In this Review, we discuss currently available evidence for prevention and post-exposure prophylaxis of EVD, including therapies currently under evaluation for SUDV.
Collapse
Affiliation(s)
- Michael A Moso
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | - Chuan K Lim
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eloise Williams
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Caroline Marshall
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - James McCarthy
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
10
|
Bushmaker T, Feldmann F, Lovaglio J, Saturday G, Griffin AJ, O’Donnell KL, Strong JE, Sprecher A, Kobinger G, Geisbert TW, Marzi A, Feldmann H. Limited Benefit of Postexposure Prophylaxis With VSV-EBOV in Ebola Virus-Infected Rhesus Macaques. J Infect Dis 2023; 228:S721-S729. [PMID: 37474155 PMCID: PMC10651186 DOI: 10.1093/infdis/jiad280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Vesicular stomatitis virus-Ebola virus (VSV-EBOV) vaccine has been successfully used in ring vaccination approaches during EBOV disease outbreaks demonstrating its general benefit in short-term prophylactic vaccination, but actual proof of its benefit in true postexposure prophylaxis (PEP) for humans is missing. Animal studies have indicated PEP efficacy when VSV-EBOV was used within hours of lethal EBOV challenge. Here, we used a lower EBOV challenge dose and a combined intravenous and intramuscular VSV-EBOV administration to improve PEP efficacy in the rhesus macaque model. VSV-EBOV treatment 1 hour after EBOV challenge resulted in delayed disease progression but little benefit in outcome. Thus, we could not confirm previous results indicating questionable benefit of VSV-EBOV for EBOV PEP in a nonhuman primate model.
Collapse
Affiliation(s)
- Trenton Bushmaker
- Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Amanda J Griffin
- Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana
| | - Kyle L O’Donnell
- Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana
| | - James E Strong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | - Gary Kobinger
- Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Andrea Marzi
- Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana
| |
Collapse
|
11
|
Ouyang MJ, Ao Z, Olukitibi TA, Lawrynuik P, Shieh C, Kung SKP, Fowke KR, Kobasa D, Yao X. Oral Immunization with rVSV Bivalent Vaccine Elicits Protective Immune Responses, Including ADCC, against Both SARS-CoV-2 and Influenza A Viruses. Vaccines (Basel) 2023; 11:1404. [PMID: 37766083 PMCID: PMC10534613 DOI: 10.3390/vaccines11091404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 and influenza both cause enormous disease burdens, and vaccines are the primary measures for their control. Since these viral diseases are transmitted through the mucosal surface of the respiratory tract, developing an effective and convenient mucosal vaccine should be a high priority. We previously reported a recombinant vesicular stomatitis virus (rVSV)-based bivalent vaccine (v-EM2/SPΔC1Delta) that protects animals from both SARS-CoV-2 and influenza viruses via intramuscular and intranasal immunization. Here, we further investigated the immune response induced by oral immunization with this vaccine and its protective efficacy in mice. The results demonstrated that the oral delivery, like the intranasal route, elicited strong and protective systemic immune responses against SARS-CoV-2 and influenza A virus. This included high levels of neutralizing antibodies (NAbs) against SARS-CoV-2, as well as strong anti-SARS-CoV-2 spike protein (SP) antibody-dependent cellular cytotoxicity (ADCC) and anti-influenza M2 ADCC responses in mice sera. Furthermore, it provided efficient protection against challenge with influenza H1N1 virus in a mouse model, with a 100% survival rate and a significantly low lung viral load of influenza virus. All these findings provide substantial evidence for the effectiveness of oral immunization with the rVSV bivalent vaccine.
Collapse
Affiliation(s)
- Maggie Jing Ouyang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Titus A. Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Peter Lawrynuik
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Christopher Shieh
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Sam K. P. Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W3, Canada;
| | - Keith R. Fowke
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Darwyn Kobasa
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| |
Collapse
|
12
|
Parham KA, Kim GN, Richer CG, Ninkov M, Wu K, Saeedian N, Li Y, Rashu R, Barr SD, Arts EJ, Haeryfar SMM, Kang CY, Troyer RM. Monovalent and trivalent VSV-based COVID-19 vaccines elicit neutralizing antibodies and CD8 + T cells against SARS-CoV-2 variants. iScience 2023; 26:106292. [PMID: 36915805 PMCID: PMC9970654 DOI: 10.1016/j.isci.2023.106292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Recombinant vesicular stomatitis virus (rVSV) vaccines expressing spike proteins of Wuhan, Beta, and/or Delta variants of SARS-CoV-2 were generated and tested for induction of antibody and T cell immune responses following intramuscular delivery to mice. rVSV-Wuhan and rVSV-Delta vaccines and an rVSV-Trivalent (mixed rVSV-Wuhan, -Beta, -Delta) vaccine elicited potent neutralizing antibodies (nAbs) against live SARS-CoV-2 Wuhan (USAWA1), Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529) viruses. Prime-boost vaccination with rVSV-Beta was less effective in this capacity. Heterologous boosting of rVSV-Wuhan with rVSV-Delta induced strong nAb responses against Delta and Omicron viruses, with the rVSV-Trivalent vaccine consistently effective in inducing nAbs against all the SARS-CoV-2 variants tested. All vaccines, including rVSV-Beta, elicited a spike-specific immunodominant CD8+ T cell response. Collectively, rVSV vaccines targeting SARS-CoV-2 variants of concern may be considered in the global fight against COVID-19.
Collapse
Affiliation(s)
- Kate A Parham
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Gyoung Nyoun Kim
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Connor G Richer
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Marina Ninkov
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Kunyu Wu
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Nasrin Saeedian
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Yue Li
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Rasheduzzaman Rashu
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Stephen D Barr
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Eric J Arts
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - C Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| | - Ryan M Troyer
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 3K7, Canada
| |
Collapse
|
13
|
Vijver SV, Danklmaier S, Pipperger L, Gronauer R, Floriani G, Hackl H, Das K, Wollmann G. Prediction and validation of murine MHC class I epitopes of the recombinant virus VSV-GP. Front Immunol 2023; 13:1100730. [PMID: 36741416 PMCID: PMC9893851 DOI: 10.3389/fimmu.2022.1100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Oncolytic viruses are currently tested as a novel platform for cancer therapy. These viruses preferentially replicate in and kill malignant cells. Due to their microbial origin, treatment with oncolytic viruses naturally results in anti-viral responses and general immune activation. Consequently, the oncolytic virus treatment also induces anti-viral T cells. Since these can constitute the dominant activated T cell pool, monitoring of the anti-viral T cell response may aid in better understanding of the immune responses post oncolytic virotherapy. This study aimed to identify the anti-viral T cells raised by VSV-GP virotherapy in C57BL/6J mice, one of the most widely used models for preclinical studies. VSV-GP is a novel oncolytic agent that recently entered a clinical phase I study. To identify the VSV-GP epitopes to which mouse anti-viral T cells react, we used a multilevel adapted bioinformatics viral epitope prediction approach based on the tools netMHCpan, MHCflurry and netMHCstabPan, which are commonly used in neoepitope identification. Predicted viral epitopes were ranked based on consensus binding strength categories, predicted stability, and dissimilarity to the mouse proteome. The top ranked epitopes were selected and included in the peptide candidate matrix in order to use a matrix deconvolution approach. Using ELISpot, we showed which viral epitopes presented on C57BL/6J mouse MHC-I alleles H2-Db and H2-Kb trigger IFN-γ secretion due to T cell activation. Furthermore, we validated these findings using an intracellular cytokine staining. Collectively, identification of the VSV-GP T cell epitopes enables monitoring of the full range of anti-viral T cell responses upon VSV-GP virotherapy in future studies with preclinical mouse models to more comprehensively delineate anti-viral from anti-tumor T cell responses. These findings also support the development of novel VSV-GP variants expressing immunomodulatory transgenes and can improve the assessment of anti-viral immunity in preclinical models.
Collapse
Affiliation(s)
- Saskia V. Vijver
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Danklmaier
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Pipperger
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Raphael Gronauer
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriel Floriani
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Guido Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Sun Y, Shi X, Lu F, Fu H, Yin Y, Xu J, Jin C, Han ET, Huang X, Chen Y, Dong C, Cheng Y. Vesicular stomatitis virus-based vaccine targeting plasmodium blood-stage antigens elicits immune response and protects against malaria with protein booster strategy. Front Microbiol 2022; 13:1042414. [PMID: 36504817 PMCID: PMC9731671 DOI: 10.3389/fmicb.2022.1042414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Merozoite invasion of the erythrocytes in humans is a key step in the pathogenesis of malaria. The proteins involved in the merozoite invasion could be potential targets for the development of malaria vaccines. Novel viral-vector-based malaria vaccine regimens developed are currently under clinical trials. Vesicular stomatitis virus (VSV) is a single-stranded negative-strand RNA virus widely used as a vector for virus or cancer vaccines. Whether the VSV-based malarial vaccine is more effective than conventional vaccines based on proteins involved in parasitic invasion is still unclear. In this study, we have used the reverse genetics system to construct recombinant VSVs (rVSVs) expressing apical membrane protein 1 (AMA1), rhoptry neck protein 2 (RON2), and reticulocyte-binding protein homolog 5 (RH5), which are required for Plasmodium falciparum invasion. Our results showed that VSV-based viral vaccines significantly increased Plasmodium-specific IgG levels and lymphocyte proliferation. Also, VSV-PyAMA1 and VSV-PyRON2sp prime-boost regimens could significantly increase the levels of IL-2 and IFN-γ-producing by CD4+ and CD8+ T cells and suppress invasion in vitro. The rVSV prime-protein boost regimen significantly increase Plasmodium antigen-specific IgG levels in the serum of mice compared to the homologous rVSV prime-boost. Furthermore, the protective efficacy of rVSV prime protein boost immunization in the mice challenged with P. yoelii 17XL was better compared to traditional antigen immunization. Together, our results show that VSV vector is a novel strategy for malarial vaccine development and preventing the parasitic diseases.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China,Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaodan Shi
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Haitian Fu
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China,Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yi Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jiahui Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Cheng Jin
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Eun-taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | - Xuan Huang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China,*Correspondence: Chunsheng Dong,
| | - Yang Cheng
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China,Yang Cheng,
| |
Collapse
|
15
|
Alfson KJ, Goez-Gazi Y, Gazi M, Chou YL, Niemuth NA, Mattix ME, Staples H, Klaffke B, Rodriguez GF, Escareno P, Bartley C, Ticer A, Clemmons EA, Dutton III JW, Griffiths A, Meister GT, Sanford DC, Cirimotich CM, Carrion R. Development of a Well-Characterized Cynomolgus Macaque Model of Sudan Virus Disease for Support of Product Development. Vaccines (Basel) 2022; 10:1723. [PMID: 36298588 PMCID: PMC9611481 DOI: 10.3390/vaccines10101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
The primary objective of this study was to characterize the disease course in cynomolgus macaques exposed to Sudan virus (SUDV), to determine if infection in this species is an appropriate model for the evaluation of filovirus countermeasures under the FDA Animal Rule. Sudan virus causes Sudan virus disease (SVD), with an average case fatality rate of approximately 50%, and while research is ongoing, presently there are no approved SUDV vaccines or therapies. Well characterized animal models are crucial for further developing and evaluating countermeasures for SUDV. Twenty (20) cynomolgus macaques were exposed intramuscularly to either SUDV or sterile phosphate-buffered saline; 10 SUDV-exposed animals were euthanized on schedule to characterize pathology at defined durations post-exposure and 8 SUDV-exposed animals were not part of the scheduled euthanasia cohort. Survival was assessed, along with clinical observations, body weights, body temperatures, hematology, clinical chemistry, coagulation, viral load (serum and tissues), macroscopic observations, and histopathology. There were statistically significant differences between SUDV-exposed animals and mock-exposed animals for 26 parameters, including telemetry body temperature, clinical chemistry parameters, hematology parameters, activated partial thromboplastin time, serum viremia, and biomarkers that characterize the disease course of SUDV in cynomolgus macaques.
Collapse
Affiliation(s)
- Kendra J. Alfson
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Michal Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Ying-Liang Chou
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Nancy A. Niemuth
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina, OH 44256, USA
| | - Hilary Staples
- Current affiliation: National Emerging Infectious Diseases Laboratory, Department of Microbiology, Boston University School of Medicine, 620 Albany St, Boston, MA 02118, USA
| | - Benjamin Klaffke
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gloria F. Rodriguez
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Priscilla Escareno
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Carmen Bartley
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anysha Ticer
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Elizabeth A. Clemmons
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - John W. Dutton III
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anthony Griffiths
- Current affiliation: National Emerging Infectious Diseases Laboratory, Department of Microbiology, Boston University School of Medicine, 620 Albany St, Boston, MA 02118, USA
| | - Gabe T. Meister
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Daniel C. Sanford
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Chris M. Cirimotich
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| |
Collapse
|
16
|
Lewin B, Qian L, Huang R, Sy LS, Goddard K, Naleway AL, DeSilva M, Daley MF, McNeil MM, Jackson LA, Jacobsen SJ. Travelers and travel vaccines at six health care systems in the Vaccine Safety Datalink. Vaccine 2022; 40:5904-5911. [PMID: 36064668 PMCID: PMC10883331 DOI: 10.1016/j.vaccine.2022.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Studying the safety of travel vaccines poses challenges since recipients may be traveling during the risk window for adverse events and the identification of a suitable comparison group can also be difficult. The examination of traveler characteristics, travel vaccination patterns, and health care utilization using electronic health record (EHR) data can inform the feasibility of future travel vaccine safety studies. METHODS A retrospective cohort study of health plan members in the Vaccine Safety Datalink Project aged 9 months and older who had a travel-related encounter or received a travel vaccine from 2009 to 2018 was performed. Travel regions visited, travel duration, type of travel vaccine received (typhoid, yellow fever, Japanese encephalitis, rabies, and cholera), and timing of vaccination date before departure date were described. Sociodemographic information, clinical characteristics, and health care utilization were compared between travelers who received travel vaccines and travelers who did not. RESULTS A total of 1,026,822 unique travelers departing from the United States were identified; 612,795 travelers received 898,196 doses of travel vaccines. The most commonly administered travel vaccine was typhoid vaccine and 77% of all travel vaccines were given more than one week prior to departure. Compared with travelers without travel vaccines, travelers with travel vaccines were overall similar but as a group were slightly younger, healthier, and had lower Hispanic representation. Health care utilization dramatically decreased during travel. Outpatient visits decreased from 294.8 visits per 10,000 person-days before travel to 24.2 visits per 10,000 person-days during reported travel dates. CONCLUSIONS Through the EHR information from almost a million travelers, a departure date and duration of travel were successfully captured for the majority of travelers with corresponding health care utilization data. Time after vaccination and prior to departure can potentially be used in the future to compare travelers who receive travel vaccines with travelers who do not receive travel vaccines when looking at adverse events of interest after vaccination.
Collapse
Affiliation(s)
- Bruno Lewin
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S Los Robles, Pasadena, CA 91101, USA.
| | - Lei Qian
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S Los Robles, Pasadena, CA 91101, USA
| | - Runxin Huang
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S Los Robles, Pasadena, CA 91101, USA
| | - Lina S Sy
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S Los Robles, Pasadena, CA 91101, USA
| | - Kristin Goddard
- Kaiser Permanente Vaccine Study Center, Kaiser Permanente Northern California, 1 Kaiser Plaza 16th Floor, Oakland, CA 94612, USA
| | - Allison L Naleway
- Center for Health Research, Kaiser Permanente Northwest, 3800 N. Interstate Ave, Portland, OR 97227, USA
| | - Malini DeSilva
- HealthPartners Institute, 8170 33rd Avenue South PO Box 1524, Minneapolis, MN 55440, USA
| | - Matthew F Daley
- Institute for Health Research, Kaiser Permanente Colorado, 10065 E. Harvard Suite 300, Denver, CO 8023, USA
| | - Michael M McNeil
- Immunization Safety Office, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30333, USA
| | - Lisa A Jackson
- Kaiser Permanente Washington Health Research Institute, 1730 Minor Ave Suite 1600, Seattle, WA 98101, USA
| | - Steven J Jacobsen
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S Los Robles, Pasadena, CA 91101, USA
| |
Collapse
|
17
|
Lessons Learned from the Development and Roll-Out of the rVSVΔG-ZEBOV-GP Zaire ebolavirus Vaccine to Inform Marburg Virus and Sudan ebolavirus Vaccines. Vaccines (Basel) 2022; 10:vaccines10091446. [PMID: 36146524 PMCID: PMC9505064 DOI: 10.3390/vaccines10091446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
This review describes key aspects of the development of the rVSVΔG-ZEBOV-GP Ebola vaccine and key activities which are continuing to further expand our knowledge of the product. Extensive partnerships and innovative approaches were used to address the various challenges encountered during this process. The rVSVΔG-ZEBOV-GP Ebola vaccine was initially approved by the European Medicines Agency and prequalified by the World Health Organization in November 2019. It was approved by the United States Food and Drug Administration in December 2019 and approved in five African countries within 90 days of prequalification. The development resulted in the first stockpile of a registered Ebola vaccine that is available to support outbreak response. This also provides insights into how the example of rVSVΔG-ZEBOV-GP can inform the development of vaccines for Sudan ebolavirus, Marburg virus, and other emerging epidemic diseases in terms of the types of approaches and data needed to support product registration, availability, and the use of a filovirus vaccine.
Collapse
|
18
|
Alfson KJ, Goez-Gazi Y, Gazi M, Chou YL, Niemuth NA, Mattix ME, Staples HM, Klaffke B, Rodriguez GF, Bartley C, Ticer A, Clemmons EA, Dutton JW, Griffiths A, Meister GT, Sanford DC, Cirimotich CM, Carrion R. Development of a Well-Characterized Cynomolgus Macaque Model of Marburg Virus Disease for Support of Vaccine and Therapy Development. Vaccines (Basel) 2022; 10:1314. [PMID: 36016203 PMCID: PMC9414819 DOI: 10.3390/vaccines10081314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 12/03/2022] Open
Abstract
Marburg virus (MARV) is a filovirus that can infect humans and nonhuman primates (NHPs), causing severe disease and death. Of the filoviruses, Ebola virus (EBOV) has been the primary target for vaccine and therapeutic development. However, MARV has an average case fatality rate of approximately 50%, the infectious dose is low, and there are currently no approved vaccines or therapies targeted at infection with MARV. The purpose of this study was to characterize disease course in cynomolgus macaques intramuscularly exposed to MARV Angola variant. There were several biomarkers that reliably correlated with MARV-induced disease, including: viral load; elevated total clinical scores; temperature changes; elevated ALT, ALP, BA, TBIL, CRP and decreased ALB values; decreased lymphocytes and platelets; and prolonged PTT. A scheduled euthanasia component also provided the opportunity to study the earliest stages of the disease. This study provides evidence for the application of this model to evaluate potential vaccines and therapies against MARV and will be valuable in improving existing models.
Collapse
Affiliation(s)
- Kendra J. Alfson
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Michal Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Ying-Liang Chou
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Nancy A. Niemuth
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina, OH 44256, USA
| | - Hilary M. Staples
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Benjamin Klaffke
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gloria F. Rodriguez
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Carmen Bartley
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anysha Ticer
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Elizabeth A. Clemmons
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - John W. Dutton
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anthony Griffiths
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gabe T. Meister
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Daniel C. Sanford
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Chris M. Cirimotich
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| |
Collapse
|
19
|
Fathi A, Dahlke C, Krähling V, Kupke A, Okba NMA, Raadsen MP, Heidepriem J, Müller MA, Paris G, Lassen S, Klüver M, Volz A, Koch T, Ly ML, Friedrich M, Fux R, Tscherne A, Kalodimou G, Schmiedel S, Corman VM, Hesterkamp T, Drosten C, Loeffler FF, Haagmans BL, Sutter G, Becker S, Addo MM. Increased neutralization and IgG epitope identification after MVA-MERS-S booster vaccination against Middle East respiratory syndrome. Nat Commun 2022; 13:4182. [PMID: 35853863 PMCID: PMC9295877 DOI: 10.1038/s41467-022-31557-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
Vaccine development is essential for pandemic preparedness. We previously conducted a Phase 1 clinical trial of the vector vaccine candidate MVA-MERS-S against the Middle East respiratory syndrome coronavirus (MERS-CoV), expressing its full spike glycoprotein (MERS-CoV-S), as a homologous two-dose regimen (Days 0 and 28). Here, we evaluate the safety (primary objective) and immunogenicity (secondary and exploratory objectives: magnitude and characterization of vaccine-induced humoral responses) of a third vaccination with MVA-MERS-S in a subgroup of trial participants one year after primary immunization. MVA-MERS-S booster vaccination is safe and well-tolerated. Both binding and neutralizing anti-MERS-CoV antibody titers increase substantially in all participants and exceed maximum titers observed after primary immunization more than 10-fold. We identify four immunogenic IgG epitopes, located in the receptor-binding domain (RBD, n = 1) and the S2 subunit (n = 3) of MERS-CoV-S. The level of baseline anti-human coronavirus antibody titers does not impact the generation of anti-MERS-CoV antibody responses. Our data support the rationale of a booster vaccination with MVA-MERS-S and encourage further investigation in larger trials. Trial registration: Clinicaltrials.gov NCT03615911.
Collapse
Affiliation(s)
- Anahita Fathi
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - Christine Dahlke
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Verena Krähling
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Alexandra Kupke
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Nisreen M A Okba
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Matthijs P Raadsen
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Jasmin Heidepriem
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Marcel A Müller
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Grigori Paris
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Susan Lassen
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Michael Klüver
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Asisa Volz
- University of Veterinary Medicine Hanover, Institute of Virology, Hanover, Germany
- German Center for Infection Research, partner site Hanover-Brunswick, Hanover, Germany
| | - Till Koch
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - My L Ly
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Monika Friedrich
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Robert Fux
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Alina Tscherne
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Georgia Kalodimou
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Stefan Schmiedel
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - Victor M Corman
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Thomas Hesterkamp
- German Center for Infection Research, Translational Project Management Office, Brunswick, Germany
| | - Christian Drosten
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Felix F Loeffler
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Bart L Haagmans
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Gerd Sutter
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Stephan Becker
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Marylyn M Addo
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany.
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany.
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
20
|
Natural History of Sudan ebolavirus to Support Medical Countermeasure Development. Vaccines (Basel) 2022; 10:vaccines10060963. [PMID: 35746571 PMCID: PMC9228702 DOI: 10.3390/vaccines10060963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/23/2022] Open
Abstract
Sudan ebolavirus (SUDV) is one of four members of the Ebolavirus genus known to cause Ebola Virus Disease (EVD) in humans, which is characterized by hemorrhagic fever and a high case fatality rate. While licensed therapeutics and vaccines are available in limited number to treat infections of Zaire ebolavirus, there are currently no effective licensed vaccines or therapeutics for SUDV. A well-characterized animal model of this disease is needed for the further development and testing of vaccines and therapeutics. In this study, twelve cynomolgus macaques (Macaca fascicularis) were challenged intramuscularly with 1000 PFUs of SUDV and were followed under continuous telemetric surveillance. Clinical observations, body weights, temperature, viremia, hematology, clinical chemistry, and coagulation were analyzed at timepoints throughout the study. Death from SUDV disease occurred between five and ten days after challenge at the point that each animal met the criteria for euthanasia. All animals were observed to exhibit clinical signs and lesions similar to those observed in human cases which included: viremia, fever, dehydration, reduced physical activity, macular skin rash, systemic inflammation, coagulopathy, lymphoid depletion, renal tubular necrosis, hepatocellular degeneration and necrosis. The results from this study will facilitate the future preclinical development and evaluation of vaccines and therapeutics for SUDV.
Collapse
|
21
|
Alizadeh M, Amini-Khoei H, Tahmasebian S, Ghatrehsamani M, Ghatreh Samani K, Edalatpanah Y, Rostampur S, Salehi M, Ghasemi-Dehnoo M, Azadegan-Dehkordi F, Sanami S, Bagheri N. Designing a novel multi‑epitope vaccine against Ebola virus using reverse vaccinology approach. Sci Rep 2022; 12:7757. [PMID: 35545650 PMCID: PMC9094136 DOI: 10.1038/s41598-022-11851-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/19/2022] [Indexed: 12/18/2022] Open
Abstract
Ebola virus (EBOV) is a dangerous zoonotic infectious disease. To date, more than 25 EBOV outbreaks have been documented, the majority of which have occurred in Central Africa. The rVSVG-ZEBOV-GP vaccine (ERVEBO), a live attenuated vaccine, has been approved by the US Food and Drug Administration (FDA) to combat EBOV. Because of the several drawbacks of live attenuated vaccines, multi-epitope vaccines probably appear to be safer than live attenuated vaccines. In this work, we employed immunoinformatics tools to design a multi-epitope vaccine against EBOV. We collected sequences of VP35, VP24, VP30, VP40, GP, and NP proteins from the NCBI database. T-cell and linear B-cell epitopes from target proteins were identified and tested for antigenicity, toxicity, allergenicity, and conservancy. The selected epitopes were then linked together in the vaccine's primary structure using appropriate linkers, and the 50S ribosomal L7/L12 (Locus RL7 MYCTU) sequence was added as an adjuvant to the vaccine construct's N-terminal. The physicochemical, antigenicity, and allergenicity parameters of the vaccine were all found to be satisfactory. The 3D model of the vaccine was predicted, refined, and validated. The vaccine construct had a stable and strong interaction with toll-like receptor 4 (TLR4) based on molecular docking and molecular dynamic simulation (MD) analysis. The results of codon optimization and in silico cloning revealed that the proposed vaccine was highly expressed in Escherichia coli (E. coli). The findings of this study are promising; however, experimental validations should be carried out to confirm these findings.
Collapse
Affiliation(s)
- Morteza Alizadeh
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Tahmasebian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatrehsamani
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Keihan Ghatreh Samani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Yadolah Edalatpanah
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Susan Rostampur
- Department of Molecular Medicine, School of Advanced Medical Science and Technology, Shiraz University of Medical Science, Shiraz, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Maryam Ghasemi-Dehnoo
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Azadegan-Dehkordi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Sanami
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Nader Bagheri
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
22
|
Woolsey C, Cross RW, Agans KN, Borisevich V, Deer DJ, Geisbert JB, Gerardi C, Latham TE, Fenton KA, Egan MA, Eldridge JH, Geisbert TW, Matassov D. A highly attenuated Vesiculovax vaccine rapidly protects nonhuman primates against lethal Marburg virus challenge. PLoS Negl Trop Dis 2022; 16:e0010433. [PMID: 35622847 PMCID: PMC9182267 DOI: 10.1371/journal.pntd.0010433] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/09/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Marburg virus (MARV), an Ebola-like virus, remains an eminent threat to public health as demonstrated by its high associated mortality rate (23-90%) and recent emergence in West Africa for the first time. Although a recombinant vesicular stomatitis virus (rVSV)-based vaccine (Ervebo) is licensed for Ebola virus disease (EVD), no approved countermeasures exist against MARV. Results from clinical trials indicate Ervebo prevents EVD in 97.5-100% of vaccinees 10 days onwards post-immunization. METHODOLOGY/FINDINGS Given the rapid immunogenicity of the Ervebo platform against EVD, we tested whether a similar, but highly attenuated, rVSV-based Vesiculovax vector expressing the glycoprotein (GP) of MARV (rVSV-N4CT1-MARV-GP) could provide swift protection against Marburg virus disease (MVD). Here, groups of cynomolgus monkeys were vaccinated 7, 5, or 3 days before exposure to a lethal dose of MARV (Angola variant). All subjects (100%) immunized one week prior to challenge survived; 80% and 20% of subjects survived when vaccinated 5- and 3-days pre-exposure, respectively. Lethality was associated with higher viral load and sustained innate immunity transcriptional signatures, whereas survival correlated with development of MARV GP-specific antibodies and early expression of predicted NK cell-, B-cell-, and cytotoxic T-cell-type quantities. CONCLUSIONS/SIGNIFICANCE These results emphasize the utility of Vesiculovax vaccines for MVD outbreak management. The highly attenuated nature of rVSV-N4CT1 vaccines, which are clinically safe in humans, may be preferable to vaccines based on the same platform as Ervebo (rVSV "delta G" platform), which in some trial participants induced vaccine-related adverse events in association with viral replication including arthralgia/arthritis, dermatitis, and cutaneous vasculitis.
Collapse
Affiliation(s)
- Courtney Woolsey
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Robert W. Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Krystle N. Agans
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Viktoriya Borisevich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Daniel J. Deer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Joan B. Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Cheryl Gerardi
- Department of Viral Vaccine Development, Auro Vaccines, Pearl River, New York, United States of America
| | - Theresa E. Latham
- Department of Viral Vaccine Development, Auro Vaccines, Pearl River, New York, United States of America
| | - Karla A. Fenton
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael A. Egan
- Department of Immunology, Auro Vaccines, Pearl River, New York, United States of America
| | - John H. Eldridge
- Department of Immunology, Auro Vaccines, Pearl River, New York, United States of America
| | - Thomas W. Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Demetrius Matassov
- Department of Viral Vaccine Development, Auro Vaccines, Pearl River, New York, United States of America
| |
Collapse
|
23
|
Malenfant JH, Joyce A, Choi MJ, Cossaboom CM, Whitesell AN, Harcourt BH, Atmar RL, Villanueva JM, Bell BP, Hahn C, Loehr J, Davey RT, Sprecher A, Kraft CS, Shoemaker T, Montgomery JM, Helfand R, Damon IK, Frey SE, Chen WH. Use of Ebola Vaccine: Expansion of Recommendations of the Advisory Committee on Immunization Practices To Include Two Additional Populations - United States, 2021. MMWR. MORBIDITY AND MORTALITY WEEKLY REPORT 2022; 71:290-292. [PMID: 35202354 DOI: 10.15585/mmwr.mm7108a2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
On December 19, 2019, the Food and Drug Administration (FDA) approved rVSVΔG-ZEBOV-GP Ebola vaccine (ERVEBO, Merck) for the prevention of Ebola virus disease (EVD) caused by infection with Ebola virus, species Zaire ebolavirus, in adults aged ≥18 years. In February 2020, the Advisory Committee on Immunization Practices (ACIP) recommended preexposure vaccination with ERVEBO for adults aged ≥18 years in the United States who are at highest risk for potential occupational exposure to Ebola virus because they are responding to an outbreak of EVD, work as health care personnel at federally designated Ebola treatment centers in the United States, or work as laboratorians or other staff members at biosafety level 4 facilities in the United States (1).
Collapse
|
24
|
Maiese A, Baronti A, Manetti AC, Di Paolo M, Turillazzi E, Frati P, Fineschi V. Death after the Administration of COVID-19 Vaccines Approved by EMA: Has a Causal Relationship Been Demonstrated? Vaccines (Basel) 2022; 10:308. [PMID: 35214765 PMCID: PMC8875435 DOI: 10.3390/vaccines10020308] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
More than eight billion doses of COVID-19 vaccines have been administered globally so far and 44.29% of people are fully vaccinated. Pre-authorization clinical trials were carried out and the safety of vaccines is still continuously monitored through post-commercialization surveillance. However, some people are afraid of vaccine side effects, claiming they could lead to death, and hesitate to get vaccinated. Herein, a literature review of COVID-19-vaccine-related deaths has been carried out according to the PRISMA standards to understand if there is a causal relationship between vaccination and death and to highlight the real extent of such events. There have been 55 cases of death after COVID-19 vaccination reported and a causal relationship has been excluded in 17 cases. In the remaining cases, the causal link between the vaccine and the death was not specified (8) or considered possible (15), probable (1), or very probable/demonstrated (14). The causes of deaths among these cases were: vaccine-induced immune thrombotic thrombocytopenia (VITT) (32), myocarditis (3), ADEM (1), myocardial infarction (1), and rhabdomyolysis (1). In such cases, the demonstration of a causal relationship is not obvious, and more studies, especially with post-mortem investigations, are needed to deepen understanding of the possible pathophysiological mechanisms of fatal vaccine side effects. In any event, given the scarcity of fatal cases, the benefits of vaccination outweigh the risks and the scientific community needs to be cohesive in asserting that vaccination is fundamental to containing the spread of SARS-CoV-2.
Collapse
Affiliation(s)
- Aniello Maiese
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Institute of Legal Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (A.M.); (A.B.); (A.C.M.); (M.D.P.); (E.T.)
| | - Arianna Baronti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Institute of Legal Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (A.M.); (A.B.); (A.C.M.); (M.D.P.); (E.T.)
| | - Alice Chiara Manetti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Institute of Legal Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (A.M.); (A.B.); (A.C.M.); (M.D.P.); (E.T.)
| | - Marco Di Paolo
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Institute of Legal Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (A.M.); (A.B.); (A.C.M.); (M.D.P.); (E.T.)
| | - Emanuela Turillazzi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Institute of Legal Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (A.M.); (A.B.); (A.C.M.); (M.D.P.); (E.T.)
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Institute of Legal Medicine, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy;
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Institute of Legal Medicine, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy;
| |
Collapse
|
25
|
Tsakiri M, Naziris N, Demetzos C. Innovative vaccine platforms against infectious diseases: Under the scope of the COVID-19 pandemic. Int J Pharm 2021; 610:121212. [PMID: 34687816 PMCID: PMC8527590 DOI: 10.1016/j.ijpharm.2021.121212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022]
Abstract
While classic vaccines have proved greatly efficacious in eliminating serious infectious diseases, innovative vaccine platforms open a new pathway to overcome dangerous pandemics via the development of safe and effective formulations. Such platforms play a key role either as antigen delivery systems or as immune-stimulators that induce both innate and adaptive immune responses. Liposomes or lipid nanoparticles, virus-like particles, nanoemulsions, polymeric or inorganic nanoparticles, as well as viral vectors, all belong to the nanoscale and are the main categories of innovative vaccines that are currently on the market or in clinical and preclinical phases. In this paper, we review the above formulations used in vaccinology and we discuss their connection with the development of safe and effective prophylactic vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Nikolaos Naziris
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece.
| |
Collapse
|
26
|
Shenoy ES, Weber DJ. Occupational Health Update: Approach to Evaluation of Health Care Personnel and Preexposure Prophylaxis. Infect Dis Clin North Am 2021; 35:717-734. [PMID: 34362540 PMCID: PMC8331250 DOI: 10.1016/j.idc.2021.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Erica S Shenoy
- Infection Control Unit, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 334, Boston, MA 02114, USA; Division of Infectious Diseases, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 334, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - David J Weber
- Division of Infectious Disease, School of Medicine, University of North Carolina at Chapel Hill, Bioinformatics Building, Suite 2163, Campus Box 7030, 130 Mason Farm Road, Chapel Hill, NC 27599-7030, USA; Department of Hospital Epidemiology, UNC Medical Center, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Ghosh S, Saha A, Samanta S, Saha RP. Genome structure and genetic diversity in the Ebola virus. Curr Opin Pharmacol 2021; 60:83-90. [PMID: 34364102 DOI: 10.1016/j.coph.2021.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022]
Abstract
Ebola is a deadly pathogen responsible for Ebola virus disease, first came to prominence in the year 1976. This rapidly evolving virus imposed a serious threat to the human population in the last few decades and also continues to be a probable threat to our race. A better understanding of the virus in terms of its genomic structure is very much needed to develop an effective antiviral therapy against this deadly pathogen. Complete knowledge of its genomic structure and variations will help us and the entire scientific community to design effective therapy in terms of either vaccine development or the development of proper antiviral medicine.
Collapse
Affiliation(s)
- Sanmitra Ghosh
- Department of Microbiology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Saikat Samanta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India
| | - Rudra P Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, 700126, India.
| |
Collapse
|
28
|
Sharma AR, Lee YH, Nath S, Lee SS. Recent developments and strategies of Ebola virus vaccines. Curr Opin Pharmacol 2021; 60:46-53. [PMID: 34329960 DOI: 10.1016/j.coph.2021.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/11/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022]
Abstract
The Filovirus family member, Ebola virus (EBOV), is a highly infectious pathogen responsible for viral hemorrhagic fever. EBOV has a fatality rate in the range 50%-90% in primates. The lethal viral hemorrhagic attack in 2014 by EBOV has forced the human race to look for rapid countermeasures. Fortunately, owing to continuous efforts and several vaccine platforms, few potential vaccine candidates are emerging, such as replicative and non-replicative vectored vaccines, polyepitopic or monovalent vaccines, and DNA vaccines. This article reviewed various kinds of EBOV vaccines in different clinical trial phases and their approval status. Updated knowledge of vaccine development progress might stimulate the researchers to look for more potent and effective vaccine candidates against EBOV.
Collapse
Affiliation(s)
- Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| | - Yeon-Hee Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Sudarshini Nath
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| |
Collapse
|
29
|
Tomori O, Kolawole MO. Ebola virus disease: current vaccine solutions. Curr Opin Immunol 2021; 71:27-33. [PMID: 33873076 DOI: 10.1016/j.coi.2021.03.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
Ebola Virus Disease (EVD) is an emerging zoonotic disease with intermittent outbreaks in Central and West African countries. The unpredictable high case fatality rate has made it a disease of public health concern. Different vaccine platforms have shown prophylactic protection in human and non-human primates, with the progress towards a licensed vaccine greatly accelerated in response to the devastating outbreak of EVD in West Africa from 2013-2016. Currently, two vaccines: Ervebo (rVSV-ZEBOV) and a two-dose combination of Zabdeno (Ad26.ZEBOV) and Mvabea (MVA-BN-Filo) have been licensed and in use. The licensing of an Ebola vaccine for use is challenging for several reasons, including the sporadic and limited nature of EVD outbreaks and the enormous resources needed to bring a vaccine to licensure. While vaccine solutions remain important in reducing the fatality of EVD, other strategic interventions are necessary for the prevention and control of EVD.
Collapse
Affiliation(s)
| | - Matthew Olatunji Kolawole
- Infectious Diseases and Environmental Health Research Group (IDEHRG), Department of Microbiology, University of Ilorin, Ilorin, Kwara State, Nigeria
| |
Collapse
|
30
|
Chung EH. Coronavirus disease 2019 vaccines and relevant adverse reactions. ALLERGY ASTHMA & RESPIRATORY DISEASE 2021. [DOI: 10.4168/aard.2021.9.3.124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Eun Hee Chung
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon Atopy·Asthma Education Information Center, Daejeon, Korea
| |
Collapse
|