1
|
Valayer S, Alexandre M, Prague M, Beavogui AH, Doumbia S, Kieh M, Greenwood B, Leigh B, Poupelin M, Schwimmer C, Sow SO, Berry IM, Kuhn JH, Fusco D, Cauwelaert ND, Watson-Jones D, Thiébaut R, Lévy Y, Yazdanpanah Y, Richert L, Lhomme E. Evaluation of waning of IgG antibody responses after rVSVΔG-ZEBOV-GP and Ad26.ZEBOV, MVA-BN-Filo Ebola virus disease vaccines: a modelling study from the PREVAC randomized trial. Emerg Microbes Infect 2025; 14:0. [PMID: 39559990 PMCID: PMC11632942 DOI: 10.1080/22221751.2024.2432353] [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: 07/16/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
rVSVΔG-ZEBOV-GP and Ad26.ZEBOV, MVA-BN-Filo are WHO-prequalified vaccination regimens against Ebola virus disease (EVD). Challenges associated with measuring long-term clinical protection warrant the evaluation of immune response kinetics after vaccination. Data from a large phase 2 randomized double-blind clinical trial (PREVAC) were used to evaluate waning of anti-Ebola virus (EBOV) glycoprotein (GP1,2) antibody concentrations after rVSVΔG-ZEBOV-GP or Ad26.ZEBOV, MVA-BN-Filo vaccination with linear mixed-effect regression models. After a post-vaccination peak, each vaccination strategy was associated with a decrease of anti-EBOV GP1,2 antibody concentrations with distinct kinetics, highlighting a less-rapid decline in antibody levels after vaccination by rVSVΔG-ZEBOV-GP. One year after administration of the vaccine, antibody concentrations were higher in children compared to adults for both vaccines, although with different effect sizes: 1.74-fold higher concentrations (95% confidence interval [CI] [1.48; 2.02]) for children 12-17 years old to 3.10-fold higher concentrations (95% CI [2.58; 3.69]) for those 1-4 years old compared to adults for Ad26.ZEBOV, MVA-BN-Filo versus 1.36-fold (95% CI [1.12; 1.61]) to 1.41-fold (95% CI [1.21; 1.62]) higher than these values for adults, with relatively small changes from one age category of children to another, for rVSVΔG-ZEBOV-GP. Antibody concentrations also differed according to geographical location, pre-vaccination antibody concentration, and sex. In combination with knowledge on memory response, characterization of the major determinants of immune response durability of both vaccinations may guide future EVD control protocols.Trial registration: ClinicalTrials.gov identifier: NCT02876328.
Collapse
Affiliation(s)
- Simon Valayer
- Infection, Antimicrobials, Modelling, Evolution (IAME), Université Sorbonne Paris Nord, Université Sorbonne Paris Cité, and Institut national de la santé et de la recherche médicale (Inserm), Paris, France
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
| | - Marie Alexandre
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
| | - Mélanie Prague
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
| | - Abdoul Habib Beavogui
- Centre National de Formation et de Recherche en Santé Rurale (CNFRSR) de Mafèrinyah, Ministère de la Santé et de l’Hygiène Publique, Mafèrinyah, Guinea
| | - Seydou Doumbia
- University Clinical Research Center, University of Sciences, Technique and Technology of Bamako, Bamako, Mali
| | - Mark Kieh
- Partnership for Research on Ebola Virus in Liberia (PREVAIL), Monrovia, Liberia
| | - Brian Greenwood
- London School of Hygiene & Tropical Medicine (LSHTM), London, UK
| | - Bailah Leigh
- College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Marie Poupelin
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
| | - Christine Schwimmer
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
| | - Samba O. Sow
- Centre pour le Développement des Vaccins, Ministère de la Santé et du Développement Social du Mali, Bamako, Mali
| | - Irina Maljkovic Berry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, MD, USA
| | - Daniela Fusco
- French Agency for Research on AIDS and Viral Hepatitis (ANRS), Emerging Infectious Diseases, PariSanté Campus, Paris, France
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Rodolphe Thiébaut
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- Inserm, Paris Cedex 13, France
- Service d’Immunologie Clinique et Maladies Infectieuses, Hôpital Henri-Mondor, Créteil, France
| | - Yazdan Yazdanpanah
- Infection, Antimicrobials, Modelling, Evolution (IAME), Université Sorbonne Paris Nord, Université Sorbonne Paris Cité, and Institut national de la santé et de la recherche médicale (Inserm), Paris, France
- Inserm, Paris Cedex 13, France
- Service de Maladies Infectieuses et Tropicales, Hôpital Bichat, Paris, France
| | - Laura Richert
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - Edouard Lhomme
- Bordeaux Population Health Research Centre, Université de Bordeaux, Inserm, and Inria, Bordeaux, France
- Vaccine Research Institute, Faculté de Médicine, Université Paris-Est Créteil, Créteil, France
- EUropean CLInical Trials Platform & Development (EUCLID), Université de Bordeaux, Centre Hospitalier Universitaire Bordeaux, and Inserm, ISPED, Bordeaux, France
- Pôle de Santé Publique, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
| | - the PREVAC Study Team
- Edouard Lhomme SISTM Team – Inserm U1219 Bordeaux Population Health, 146 Rue Léo Saignat, 33000Bordeaux, France
| |
Collapse
|
2
|
Merritt S, Halbrook M, Kompany JP, Chandrasekaran P, Smith OA, Hoff NA, Tambu M, Martin SA, Wong TA, Jarra A, Barrall AL, Musene K, Beya M, Orr R, Myers T, MacGill T, Hensley LE, Muyembe-Tamfum JJ, Kaba D, Berry IM, Mbala-Kingebeni P, Lehrer AT, Rimoin AW. Comparison of EBOV GP IgG antibody reactivity: Results from two immunoassays in the Democratic Republic of the Congo. J Virol Methods 2025; 336:115154. [PMID: 40194662 DOI: 10.1016/j.jviromet.2025.115154] [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] [Received: 08/30/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Ebola virus (EBOV) is a highly infectious pathogen, and its long-term consequences continue to be investigated. With its high fatality rate and potential for reinfection or latent infection, continued development of research tools is of utmost importance. Using a cohort (n = 503) of existing bio-banked specimens from the Democratic Republic of the Congo (DRC) two EBOV glycoprotein (GP) immunoglobulin G (IgG) antibody-detection assays were compared: the gold-standard Filovirus Animal Non-Clinical Group (FANG) and a Multiplex bead-based Immunoassay (MIA) with seven pan-filoviral targets. As not all immunoassays have been shown to detect a vaccine-induced immune response, and previous EBOV serosurveillance has been primarily conducted with singleplex technology, this MIA was assessed as an additional resource. Among the cohort, as sample seroreactivity increased, assay correlation increased (r2=0.80). Correlation was sustained among sub-populations of the cohort-in detecting natural immunity among survivors and vaccine-derived responses. Additionally, when results were binarized by seroreactivity, there was high correlation between the two assays (kappa=0.70) with 71 serodiscordant samples. These data indicate that the MIA is an apt alternative to the singleplex FANG assay in detecting relative seroreactivity and can be used as a potential tool for widespread pan-filovirus serosurveillance in the DRC and similar contexts.
Collapse
Affiliation(s)
- Sydney Merritt
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Megan Halbrook
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Jean Paul Kompany
- Institut National de Recherche Biomédicale (INRB), Department of Epidemiology and Global Health, Kinshasa, Democratic Republic of the Congo
| | - Prabha Chandrasekaran
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Olivia A Smith
- University of Hawaii at Manoa, Department of Tropical Medicine, Medical Microbiology and Pharmacology, Honolulu, HI, USA
| | - Nicole A Hoff
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Merly Tambu
- Institut National de Recherche Biomédicale (INRB), Department of Epidemiology and Global Health, Kinshasa, Democratic Republic of the Congo
| | - Skylar A Martin
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Teri Ann Wong
- University of Hawaii at Manoa, Department of Tropical Medicine, Medical Microbiology and Pharmacology, Honolulu, HI, USA
| | - Amie Jarra
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Angelica L Barrall
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Kamy Musene
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA
| | - Michael Beya
- Kinshasa School of Public Health, Univeristy of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Robert Orr
- Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Todd Myers
- Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Tracy MacGill
- Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Lisa E Hensley
- Zoonotic and Emerging Diseases Research Unit (ZEDRU), United States Department of Agriculture Agricultural Research Service (USDA-ARS), United States Department of Agriculture, Manhattan, KS, USA
| | - Jean-Jacques Muyembe-Tamfum
- Institut National de Recherche Biomédicale (INRB), Department of Epidemiology and Global Health, Kinshasa, Democratic Republic of the Congo
| | - Didine Kaba
- Kinshasa School of Public Health, Univeristy of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Irina Maljkovic Berry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Placide Mbala-Kingebeni
- Institut National de Recherche Biomédicale (INRB), Department of Epidemiology and Global Health, Kinshasa, Democratic Republic of the Congo
| | - Axel T Lehrer
- University of Hawaii at Manoa, Department of Tropical Medicine, Medical Microbiology and Pharmacology, Honolulu, HI, USA
| | - Anne W Rimoin
- University of California Los Angeles, Department of Epidemiology, Fielding School of Public Health, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Cooper CL, Morrow G, Yuan M, Postler TS, Neal ML, Cross RW, Woolsey C, Agans KN, Borisevich V, McNamara RP, Atyeo C, Roy V, Germosen D, Hou F, Li SL, Reiserova L, Choi Y, Wilson A, Wagner D, Wallace-Selman O, Carpov A, Geng F, Frederick DJ, DeStefano J, Ercolini AM, Enriquez AS, Hastie KM, Ramos da Silva S, Sayeed E, Coleman JW, Kilianski A, Alter G, Saphire EO, Aitchison JD, Geisbert TW, Gupta SB, Feinberg MB, Parks CL. Preclinical development of a replication-competent vesicular stomatitis virus-based Lassa virus vaccine candidate advanced into human clinical trials. EBioMedicine 2025; 114:105647. [PMID: 40157130 PMCID: PMC11994357 DOI: 10.1016/j.ebiom.2025.105647] [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] [Received: 05/24/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Lassa fever (LF) is a zoonotic haemorrhagic disease caused by Lassa virus (LASV), which is endemic in West African countries. The multimammate rat is the main animal reservoir and its geographic range is expected to expand due to influences like climate change and land usage, and this will place larger parts of Africa at risk. We conducted preclinical development on a promising experimental vaccine that allowed its advancement into human trials. METHODS The LF vaccine is based on a vesicular stomatitis virus (VSV) vector in which the VSV glycoprotein (G) was replaced with the LASV glycoprotein complex (GPC). Earlier studies showed that this vaccine (VSVΔG-LASV-GPC) was efficacious in macaques, thus we regenerated VSVΔG-LASV-GPC using laboratory and documentation practices required to support vaccine manufacturing and human trials. The efficacy of the clinical vaccine candidate was assessed in cynomolgus macaques and more extensive immunologic analysis was performed than previously to investigate immune responses associated with protection. FINDINGS A single VSVΔG-LASV-GPC vaccination elicited innate, humoural and cellular immune responses, prevented development of substantial LASV viraemia, and protected animals from disease. Vaccinated macaques developed polyfunctional antibodies and serum was shown to neutralize virus expressing GPCs representative of geographically diverse LASV lineages. INTERPRETATION The VSVΔG-LASV-GPC clinical candidate elicited immunity that protected 10 of 10 vaccinated macaques from disease supporting its use in a clinical development program, which recently progressed to phase 2 clinical trials. Moreover, immunologic analysis showed that virus-neutralizing serum antibodies likely played a role in preventing LASV disease in vaccinated macaques. FUNDING This work was supported by the Coalition for Epidemic Preparedness Innovations (CEPI), The National Institute of Allergy and Infectious Diseases (NIAID)/National Institutes of Health (NIH), The Bill and Melinda Gates Global Vaccine Accelerator Program, the Burroughs Wellcome Fund, and financial gifts and support by Nancy Zimmerman, Mark and Lisa Schwartz, and Terry and Susan Ragon.
Collapse
Affiliation(s)
| | - Gavin Morrow
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Maoli Yuan
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Thomas S Postler
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Maxwell L Neal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ryan P McNamara
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Caroline Atyeo
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Daritza Germosen
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Fuxiang Hou
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Shui L Li
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Lucia Reiserova
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Yesle Choi
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Aaron Wilson
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Denise Wagner
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Alexei Carpov
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Fuqiang Geng
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Joanne DeStefano
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Anne M Ercolini
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | - Adrian S Enriquez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | - John W Coleman
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA 02139, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Departments of Pediatrics and Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | - Christopher L Parks
- IAVI, Vaccine Design and Development Laboratory, Jersey City, NJ 07302, USA.
| |
Collapse
|
4
|
Fatima M, An T, Park PG, Hong KJ. Advancements and Challenges in Addressing Zoonotic Viral Infections with Epidemic and Pandemic Threats. Viruses 2025; 17:352. [PMID: 40143281 PMCID: PMC11946417 DOI: 10.3390/v17030352] [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: 01/13/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Zoonotic viruses have significant pandemic potential, as evidenced by the coronavirus pandemic, which underscores that zoonotic infections have historically caused numerous outbreaks and millions of deaths over centuries. Zoonotic viruses induce numerous types of illnesses in their natural hosts. These viruses are transmitted to humans via biological vectors, direct contact with infected animals or their bites, and aerosols. Zoonotic viruses continuously evolve and adapt to human hosts, resulting in devastating consequences. It is very important to understand pathogenesis pathways associated with zoonotic viral infections across various hosts and develop countermeasure strategies accordingly. In this review, we briefly discuss advancements in diagnostics and therapeutics for zoonotic viral infections. It provides insight into recent outbreaks, viral dynamics, licensed vaccines, as well as vaccine candidates progressing to clinical investigations. Despite advancements, challenges persist in combating zoonotic viruses due to immune evasion, unpredicted outbreaks, and the complexity of the immune responses. Most of these viruses lack effective treatments and vaccines, relying entirely on supportive care and preventive measures. Exposure to animal reservoirs, limited vaccine access, and insufficient coverage further pose challenges to preventive efforts. This review highlights the critical need for ongoing interdisciplinary research and collaboration to strengthen preparedness and response strategies against emerging infectious threats.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea; (M.F.)
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Pil-Gu Park
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea; (M.F.)
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea; (M.F.)
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
5
|
Bixler SL, Shurtleff AC, Sunay MME, Liu K, Chen Z, Eichberg M, Simon JK, Coller BAG, Dubey S. Durability of Immunogenicity and Protection of rVSV∆G-ZEBOV-GP Vaccine in a Nonhuman Primate EBOV Challenge Model. Viruses 2025; 17:342. [PMID: 40143270 PMCID: PMC11945958 DOI: 10.3390/v17030342] [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: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
The rVSVΔG-ZEBOV-GP vaccine demonstrated efficacy in preventing Ebola virus (EBOV) disease in a ring vaccination clinical trial conducted during the 2014-2016 West Africa outbreak and is licensed by regulatory agencies, including the US FDA and the EMA. Here, we present two studies that evaluated the durability of immunogenicity and protection from an EBOV challenge up to ~12 months following vaccination with rVSVΔG-ZEBOV-GP in nonhuman primates (NHPs). Cynomolgus macaques were vaccinated with either one or two doses of rVSVΔG-ZEBOV-GP or a saline control and were challenged intramuscularly with EBOV at a target dose of 1000 pfu at ~4 months (Study 1) or ~8 or ~12 months (Study 2) after the last vaccination. All vaccinated animals developed robust ZEBOV-GP-specific IgG and neutralizing antibody titers, which were sustained until the last time point tested prior to the challenge. The majority of animals (88-93%) challenged with EBOV at ~4 or ~8 months post-vaccination survived, whereas the survival rate was lower (53%) in animals challenged ~12 months post-vaccination. These results demonstrate that both one-dose and two-dose regimens of the rVSVΔG-ZEBOV-GP vaccine induced durable ZEBOV-GP-specific antibody titers in NHPs and provided high levels of protection against a lethal EBOV challenge up to ~8 months post-vaccination. In this stringent challenge model, decreased protection was observed at ~12 months post-vaccination despite sustained antibody levels.
Collapse
Affiliation(s)
- Sandra L. Bixler
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Amy C. Shurtleff
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Melek M. E. Sunay
- US Army Medical Research Institute of Infectious Diseases, Porter Street, Fort Detrick, MD 21702, USA; (S.L.B.); (A.C.S.); (M.M.E.S.)
| | - Kenneth Liu
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Ziqiang Chen
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Michael Eichberg
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Jakub K. Simon
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Beth-Ann G. Coller
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| | - Sheri Dubey
- Merck & Co., Inc., Lincoln Ave., Rahway, NJ 07065, USA; (K.L.); (Z.C.); (M.E.); (J.K.S.); (B.-A.G.C.)
| |
Collapse
|
6
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
7
|
Lee AW, Liu K, Lhomme E, Blie J, McCullough J, Onorato MT, Connor L, Simon JK, Dubey S, VanRheenen S, Deutsch J, Owens A, Morgan A, Welebob C, Hyatt D, Nair S, Hamzé B, Guindo O, Sow SO, Beavogui AH, Leigh B, Samai M, Akoo P, Serry-Bangura A, Fleck S, Secka F, Lowe B, Watson-Jones D, Roy C, Hensley LE, Kieh M, Coller BAG. Immunogenicity and Vaccine Shedding After 1 or 2 Doses of rVSVΔG-ZEBOV-GP Ebola Vaccine (ERVEBO®): Results From a Phase 2, Randomized, Placebo-controlled Trial in Children and Adults. Clin Infect Dis 2024; 78:870-879. [PMID: 37967326 PMCID: PMC11006114 DOI: 10.1093/cid/ciad693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND The rVSVΔG-ZEBOV-GP vaccine (ERVEBO®) is a single-dose, live-attenuated, recombinant vesicular stomatitis virus vaccine indicated for the prevention of Ebola virus disease (EVD) caused by Zaire ebolavirus in individuals 12 months of age and older. METHODS The Partnership for Research on Ebola VACcination (PREVAC) is a multicenter, phase 2, randomized, double-blind, placebo-controlled trial of 3 vaccine strategies in healthy children (ages 1-17) and adults, with projected 5 years of follow-up (NCT02876328). Using validated assays (GP-ELISA and PRNT), we measured antibody responses after 1-dose rVSVΔG-ZEBOV-GP, 2-dose rVSVΔG-ZEBOV-GP (given on Day 0 and Day 56), or placebo. Furthermore, we quantified vaccine virus shedding in a subset of children's saliva using RT-PCR. RESULTS In total, 819 children and 783 adults were randomized to receive rVSVΔG-ZEBOV-GP (1 or 2 doses) or placebo. A single dose of rVSVΔG-ZEBOV-GP increased antibody responses by Day 28 that were sustained through Month 12. A second dose of rVSVΔG-ZEBOV-GP given on Day 56 transiently boosted antibody concentrations. In vaccinated children, GP-ELISA titers were superior to placebo and non-inferior to vaccinated adults. Vaccine virus shedding was observed in 31.7% of children, peaking by Day 7, with no shedding observed after Day 28 post-dose 1 or any time post-dose 2. CONCLUSIONS A single dose of rVSVΔG-ZEBOV-GP induced robust antibody responses in children that was non-inferior to the responses induced in vaccinated adults. Vaccine virus shedding in children was time-limited and only observed after the first dose. Overall, these data support the use of rVSVΔG-ZEBOV-GP for the prevention of EVD in at-risk children. Clinical Trials Registration. The study is registered at ClinicalTrials.gov (NCT02876328), the Pan African Clinical Trials Registry (PACTR201712002760250), and the European Clinical Trials Register (EudraCT number: 2017-001798-18).
Collapse
Affiliation(s)
| | - Ken Liu
- Merck & Co., Inc., Rahway, New Jersey, USA
| | - Edouard Lhomme
- Inserm, CHU Bordeaux, CIC 1401, EUCLID/F-CRIN Clinical Trials Platform, University of Bordeaux, Bordeaux, France
| | - Julie Blie
- Partnership for Research on Ebola Vaccines in Liberia (PREVAIL), Monrovia, Liberia
| | - John McCullough
- Advanced BioMedical Laboratories (ABML), Cinnaminson, New Jersey, USA
| | | | | | | | | | | | | | | | - Amy Morgan
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | | | | | - Benjamin Hamzé
- Pôle Recherche Clinique, Institut National de la Santé et de la Recherche Médicale (Inserm), Paris, France
| | - Oumar Guindo
- University Clinical Research Center (UCRC), Bamako, Mali
| | | | - Abdoul H Beavogui
- Centre National de Formation et de Recherche en Santé Rurale (CNFRSR), Maferinyah, Guinea
| | - Bailah Leigh
- College of Medicine & Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Mohamed Samai
- College of Medicine & Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Pauline Akoo
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine (LSHTM), London, United Kingdom
| | - Alimamy Serry-Bangura
- College of Medicine & Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Suzanne Fleck
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine (LSHTM), London, United Kingdom
| | - Fatou Secka
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine (LSHTM), London, United Kingdom
| | - Brett Lowe
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine (LSHTM), London, United Kingdom
| | - Deborah Watson-Jones
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine (LSHTM), London, United Kingdom
- Mwanza Intervention Trials Unit, National Institute for Medical Research, Mwanza, Tanzania
| | - Céline Roy
- Inserm, CHU Bordeaux, CIC 1401, EUCLID/F-CRIN Clinical Trials Platform, University of Bordeaux, Bordeaux, France
- University of Bordeaux, INSERM, MART, UMS 54, F-33000 Bordeaux, France
| | - Lisa E Hensley
- National Bio and Agro-Defense Facility (NBAF), United States Department of Agriculture (USDA), Manhattan, Kansas, USA
| | - Mark Kieh
- Partnership for Research on Ebola Vaccines in Liberia (PREVAIL), Monrovia, Liberia
| | | |
Collapse
|
8
|
Yin J, Zhang L, Wang C, Qin C, Miao M. Immunogenicity and safety of ebolavirus vaccines in healthy adults: a systematic review and meta-analysis of randomized controlled trials. Expert Rev Vaccines 2024; 23:148-159. [PMID: 38112249 DOI: 10.1080/14760584.2023.2296937] [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] [Received: 09/19/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND This review aimed to systematically evaluate the immunogenicity and safety of the candidate Ebola virus vaccine (EVV). METHODS We searched five databases for randomized controlled trials (RCTs) evaluating the effects of EVV on healthy adults. The primary outcomes were relative risk (RR) of sero-conversion or sero-response of EVV in healthy adults between the groups that received EVV and the controls. RESULTS Twenty-nine RCTs (n = 23573) were included. There was a significant difference in RR of sero-conversion of EVV (RR 13.18; 95% CI 11.28-15.41; I2 = 33%; P < 0.01) between the two groups. There was a significant difference in RR of adverse events (AEs) of EVV (RR 1.49; 95% CI 1.27-1.74; I2 = 88%; P < 0.01), although no difference in RR of serious AE (SAE) between the two groups. Subgroup analysis showed that there was no significant difference in RR of AEs for DNAEBO, EBOV-GP, MVA, and rVSVN4CT1 vaccines, compared with controls. CONCLUSIONS The DNAEBO, EBOV-GP, MVA, and rVSVN4CT1 vaccines are likely to be safe and immunogenic, tending to support the vaccination against Ebola disease. These findings should provide much-needed evidence for public health policy makers to develop preventive measures based on disease prevalence features and socio-economic conditions.
Collapse
Affiliation(s)
- Juntao Yin
- Department of Pharmacy, Huaihe Hospital, Henan University, Kaifeng, Henan, China
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, zhengzhou, Henan, China
| | - Liang Zhang
- School of Medicine, Henan Technical Institute, Zhengzhou, China
| | - Chaoyang Wang
- Department of General Surgery, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Changjiang Qin
- Department of General Surgery, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Mingsan Miao
- National International Cooperation Base of Chinese Medicine, Henan University of Chinese Medicine, zhengzhou, Henan, China
| |
Collapse
|
9
|
Guttieres D, Diepvens C, Decouttere C, Vandaele N. Modeling Supply and Demand Dynamics of Vaccines against Epidemic-Prone Pathogens: Case Study of Ebola Virus Disease. Vaccines (Basel) 2023; 12:24. [PMID: 38250837 PMCID: PMC10819028 DOI: 10.3390/vaccines12010024] [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: 11/10/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Health emergencies caused by epidemic-prone pathogens (EPPs) have increased exponentially in recent decades. Although vaccines have proven beneficial, they are unavailable for many pathogens. Furthermore, achieving timely and equitable access to vaccines against EPPs is not trivial. It requires decision-makers to capture numerous interrelated factors across temporal and spatial scales, with significant uncertainties, variability, delays, and feedback loops that give rise to dynamic and unexpected behavior. Therefore, despite progress in filling R&D gaps, the path to licensure and the long-term viability of vaccines against EPPs continues to be unclear. This paper presents a quantitative system dynamics modeling framework to evaluate the long-term sustainability of vaccine supply under different vaccination strategies. Data from both literature and 50 expert interviews are used to model the supply and demand of a prototypical Ebolavirus Zaire (EBOV) vaccine. Specifically, the case study evaluates dynamics associated with proactive vaccination ahead of an outbreak of similar magnitude as the 2018-2020 epidemic in North Kivu, Democratic Republic of the Congo. The scenarios presented demonstrate how uncertainties (e.g., duration of vaccine-induced protection) and design criteria (e.g., priority geographies and groups, target coverage, frequency of boosters) lead to important tradeoffs across policy aims, public health outcomes, and feasibility (e.g., technical, operational, financial). With sufficient context and data, the framework provides a foundation to apply the model to a broad range of additional geographies and priority pathogens. Furthermore, the ability to identify leverage points for long-term preparedness offers directions for further research.
Collapse
Affiliation(s)
- Donovan Guttieres
- Access-to-Medicines Research Centre, Faculty of Economics & Business, KU Leuven, 3000 Leuven, Belgium; (C.D.); (C.D.); (N.V.)
| | | | | | | |
Collapse
|
10
|
Alexandre M, Prague M, McLean C, Bockstal V, Douoguih M, Thiébaut R. Prediction of long-term humoral response induced by the two-dose heterologous Ad26.ZEBOV, MVA-BN-Filo vaccine against Ebola. NPJ Vaccines 2023; 8:174. [PMID: 37940656 PMCID: PMC10632397 DOI: 10.1038/s41541-023-00767-y] [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: 03/23/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
The persistence of the long-term immune response induced by the heterologous Ad26.ZEBOV, MVA-BN-Filo two-dose vaccination regimen against Ebola has been investigated in several clinical trials. Longitudinal data on IgG-binding antibody concentrations were analyzed from 487 participants enrolled in six Phase I and Phase II clinical trials conducted by the EBOVAC1 and EBOVAC2 consortia. A model based on ordinary differential equations describing the dynamics of antibodies and short- and long-lived antibody-secreting cells (ASCs) was used to model the humoral response from 7 days after the second vaccination to a follow-up period of 2 years. Using a population-based approach, we first assessed the robustness of the model, which was originally estimated based on Phase I data, against all data. Then we assessed the longevity of the humoral response and identified factors that influence these dynamics. We estimated a half-life of the long-lived ASC of at least 15 years and found an influence of geographic region, sex, and age on the humoral response dynamics, with longer antibody persistence in Europeans and women and higher production of antibodies in younger participants.
Collapse
Affiliation(s)
- Marie Alexandre
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mélanie Prague
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Chelsea McLean
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Viki Bockstal
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- ExeVir, Ghent, Belgium
| | | | - Rodolphe Thiébaut
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France.
- Vaccine Research Institute, Créteil, France.
| |
Collapse
|
11
|
Kovyrshina AV, Sizikova TE, Lebedev VN, Borisevich SV, Dolzhikova IV, Logunov DY, Gintsburg AL. [Vaccines to prevent Ebola virus disease: current challenges and perspectives]. Vopr Virusol 2023; 68:372-384. [PMID: 38156572 DOI: 10.36233/0507-4088-193] [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] [Received: 09/25/2023] [Indexed: 12/30/2023]
Abstract
RELEVANCE Ebola virus disease (EVD) is an acute infectious disease with an extremely high case fatality rate reaching up to 90%. EVD has become widely known since 2014-2016, when outbreak in West Africa occurred and led to epidemic, which caused travel-related cases on the territory of other continents. There are two vaccines against EVD, prequalified by WHO for emergency use, as well as a number of vaccines, approved by local regulators in certain countries. However, even with the availability of effective vaccines, the lack of data on immune correlates of protection and duration of protective immune response in humans and primates is limiting factor for effectively preventing the spread of EVD outbreaks. AIMS This review highlights experience of use of EVD vaccines during outbreaks in endemic areas, summarizes data on vaccine immunogenicity in clinical trials, and discusses perspectives for further development and use of effective EVD vaccines.
Collapse
Affiliation(s)
- A V Kovyrshina
- National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| | - T E Sizikova
- 48 Central Scientific Research Institute of the Ministry of Defence of the Russian Federation
| | - V N Lebedev
- 48 Central Scientific Research Institute of the Ministry of Defence of the Russian Federation
| | - S V Borisevich
- 48 Central Scientific Research Institute of the Ministry of Defence of the Russian Federation
| | - I V Dolzhikova
- National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| | - D Y Logunov
- National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| | - A L Gintsburg
- National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| |
Collapse
|
12
|
Hu Q, Zhang Y, Jiang J, Zheng A. Two Point Mutations in the Glycoprotein of SFTSV Enhance the Propagation Recombinant Vesicular Stomatitis Virus Vectors at Assembly Step. Viruses 2023; 15:800. [PMID: 36992507 PMCID: PMC10052781 DOI: 10.3390/v15030800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen for which approved therapeutic drugs or vaccines are not available. We previously developed a recombinant vesicular stomatitis virus-based vaccine candidate (rVSV-SFTSV) by replacing the original glycoprotein with Gn/Gc from SFTSV, which conferred complete protection in a mouse model. Here, we found that two spontaneous mutations, M749T/C617R, emerged in the Gc glycoprotein during passaging that could significantly increase the titer of rVSV-SFTSV. M749T/C617R enhanced the genetic stability of rVSV-SFTSV, and no further mutations appeared after 10 passages. Using immunofluorescence analysis, we found that M749T/C617R could increase glycoprotein traffic to the plasma membrane, thus facilitating virus assembly. Remarkably, the broad-spectrum immunogenicity of rVSV-SFTSV was not affected by the M749T/C617R mutations. Overall, M749T/C617R could enhance the further development of rVSV-SFTSV into an effective vaccine in the future.
Collapse
Affiliation(s)
- Qiang Hu
- College of Life Science, Hebei University, Baoding 071002, China
| | - Yuhang Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiafu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
13
|
Raabe V, Lai L, Morales J, Xu Y, Rouphael N, Davey RT, Mulligan MJ. Cellular and humoral immunity to Ebola Zaire glycoprotein and viral vector proteins following immunization with recombinant vesicular stomatitis virus-based Ebola vaccine (rVSVΔG-ZEBOV-GP). Vaccine 2023; 41:1513-1523. [PMID: 36725433 PMCID: PMC10021073 DOI: 10.1016/j.vaccine.2023.01.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
While effective at preventing Zaire ebolavirus (ZEBOV) disease, cellular immunity to ZEBOV and vector-directed immunity elicited by the recombinant vesicular stomatitis virus expressing ZEBOV glycoprotein (rVSVΔG-ZEBOV-GP) vaccine remain poorly understood. Sera and peripheral blood mononuclear cells were collected from 32 participants enrolled in a prospective multicenter study [ClinicalTrials.gov NCT02788227] before vaccination and up to six months post-vaccination. IgM and IgG antibodies, IgG-producing memory B cells (MBCs), and T cell reactivity to ZEBOV glycoprotein (ZEBOV-GP), vesicular stomatitis virus-Indiana strain (VSV-I) matrix (M) protein, and VSV-I nucleoprotein (NP) were measured using ELISA, ELISpot, and flow cytometry, respectively. 11/32 (34.4%) participants previously received a different investigational ZEBOV vaccine prior to enrollment and 21/32 (65.6%) participants were ZEBOV vaccine naïve. Both ZEBOV vaccine naïve and experienced participants had increased ZEBOV-GP IgG optical densities (ODs) post-rVSVΔG-ZEBOV-GP vaccination while only ZEBOV vaccine naïve participants had increased ZEBOV-GP IgM ODs. Transient IgM and IgG antibody responses to VSV-I M protein and NP were observed in a minority of participants. All participants had detectable ZEBOV-GP specific IgG-producing MBCs by 6 months post-vaccination while no changes were observed in the median IgG-producing MBCs to VSV-I proteins. T cell responses to ZEBOV-GP differed between ZEBOV vaccine experienced and ZEBOV vaccine naïve participants. T cell responses to both VSV-I M protein and VSV-I NP were observed, but were of a low magnitude. The rVSVΔG-ZEBOV-GP vaccine elicits robust humoral and memory B cell responses to ZEBOV glycoprotein in both ZEBOV vaccine naïve and experienced individuals and can generate vector-directed T cell immunity. Further research is needed to understand the significance of pre-existing vector and target antigen immunity on responses to booster doses of rVSVΔG-ZEBOV-GP and other rVSV-vectored vaccines.
Collapse
Affiliation(s)
- Vanessa Raabe
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| | - Lilin Lai
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| | - Juliet Morales
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| | - Richard T Davey
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center Drive, Rm. 4-1479, MSC 1460, Bethesda, MD 20892, USA.
| | - Mark J Mulligan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, GA 30030, USA.
| |
Collapse
|