1
|
Mdluli T, Wollen-Roberts S, Merbah M, Beckman B, Li Y, Alrubayyi A, Curtis DJ, Shubin Z, Barrera MD, Boeckelman J, Duncan S, Thapa P, Kim D, Costanzo MC, Bai H, Dearlove BL, Hooper JW, Kwilas SA, Paquin-Proulx D, Eller MA, Eller LA, Kibuuka H, Mwesigwa B, Kosgei J, Sawe F, Oyieko J, Ntinginya N, Mwakisisile J, Jani I, Viegas E, Iroezindu M, Akintunde A, Paolino K, Robb ML, Ward L, McLean C, Luhn K, Robinson C, Ake JA, Rolland M. Ebola virus vaccination elicits Ebola virus-specific immune responses without substantial cross-reactivity to other filoviruses. Sci Transl Med 2025; 17:eadq2496. [PMID: 40173257 DOI: 10.1126/scitranslmed.adq2496] [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: 05/04/2024] [Revised: 08/30/2024] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
The Janssen Ebola virus (EBOV) vaccine consists of the adenovirus type 26 vector encoding the EBOV glycoprotein (GP) (Ad26.ZEBOV) and the modified vaccinia Ankara (MVA) vector encoding GP from EBOV, Sudan virus, and Marburg virus and nucleoprotein from Tai Forest virus (MVA-BN-Filo) administered 8 weeks later. We conducted a systems immunology analysis of antibody-mediated and cellular immune responses induced after two immunizations with either vaccine used first. The response to vaccination was EBOV GP specific and defined by high antibody binding, Fc effector, and neutralizing responses with CD4 T cell responses also contributing. The vaccine-induced antibody profile did not distinguish people living with or without HIV-1. Samples from 48 survivors and 121 contacts from the 2007 Ugandan Bundibugyo virus epidemic also showed minimal cross-reactivity to other filovirus proteins after infection and exposure. The lack of cross-reactivity suggests that different multivalent vaccine candidates are required to provide broad protection across filoviruses.
Collapse
Affiliation(s)
- Thembi Mdluli
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Suzanne Wollen-Roberts
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Mélanie Merbah
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Bradley Beckman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Yifan Li
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Aljawharah Alrubayyi
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Daniel J Curtis
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Zhanna Shubin
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Michael D Barrera
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Jacob Boeckelman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Shayla Duncan
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Pallavi Thapa
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Dohoon Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Margaret C Costanzo
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Hongjun Bai
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Bethany L Dearlove
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Jay W Hooper
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA
| | - Steven A Kwilas
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD 21702, USA
| | - Dominic Paquin-Proulx
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Michael A Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Leigh Anne Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Betty Mwesigwa
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Josphat Kosgei
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Fredrick Sawe
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Janet Oyieko
- Henry M. Jackson Foundation Medical Research International, Nairobi, Kenya
- Kenya Medical Research Institute, US Army Medical Research Directorate-Africa, Kericho, Kenya
| | - Nyanda Ntinginya
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Joel Mwakisisile
- National Institute for Medical Research-Mbeya Medical Research Center, Mbeya, Tanzania
| | - Ilesh Jani
- Polana Caniço Health Research and Training Center, Maputo, Mozambique
| | - Edna Viegas
- Polana Caniço Health Research and Training Center, Maputo, Mozambique
| | - Michael Iroezindu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation Medical Research International, Abuja, Nigeria
| | - Akindiran Akintunde
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
- Henry M. Jackson Foundation Medical Research International, Abuja, Nigeria
- US Army Medical Research Directorate-Africa, Abuja, Nigeria
| | - Kristopher Paolino
- Clinical Trials Center, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Institute of Global Health and Translational Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Merlin L Robb
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Lucy Ward
- US Department of Defense (DOD) Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO-CBRND), Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA
| | | | - Kerstin Luhn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | - Julie A Ake
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Morgane Rolland
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| |
Collapse
|
2
|
Fan P, Sun B, Liu Z, Fang T, Ren Y, Zhao X, Song Z, Yang Y, Li J, Yu C, Chen W. A pan-orthoebolavirus neutralizing antibody encoded by mRNA effectively prevents virus infection. Emerg Microbes Infect 2024; 13:2432366. [PMID: 39560055 PMCID: PMC11590195 DOI: 10.1080/22221751.2024.2432366] [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: 09/09/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
Orthoebolavirus is a genus of hazardous pathogens that has caused over 30 outbreaks. However, currently approved therapies are limited in scope, as they are only effective against the Ebola virus and lack cross-protection against other orthoebolaviruses. Here, we demonstrate that a previously isolated human-derived antibody, 2G1, can recognize the glycoprotein (GP) of every orthoebolavirus species. The cryo-electron microscopy structure of 2G1 Fab in complex with the GPΔMucin trimer reveals that 2G1 binds a quaternary pocket formed by three subunits from two GP protomers. 2G1 recognizes highly conserved epitopes among filoviruses and achieves neutralization by blocking GP proteolysis. We designed an efficient mRNA module capable of producing test antibodies at expression levels exceeding 1500 ng/mL in vitro. The lipid nanoparticle (LNP)-encapsulated mRNA-2G1 exhibited potent neutralizing activities against the HIV-pseudotyped Ebola and Sudan viruses that were 19.8 and 12.5 times that of IgG format, respectively. In mice, the antibodies encoded by the mRNA-2G1-LNP peaked within 24 h, effectively blocking the invasion of pseudoviruses with no apparent liver toxicity. This study suggests that the 2G1 antibody and its mRNA formulation represent promising candidate interventions for orthoebolavirus disease, and it provides an efficient mRNA framework applicable to antibody-based therapies.
Collapse
Affiliation(s)
- Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yi Ren
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zhenwei Song
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Wei Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| |
Collapse
|
3
|
Davey RT, Collins GL, Rouphael N, Poliquin G, McConnell R, Grubbs G, Moir SL, Langley JM, Teitelbaum M, Hewlett AL, McLellan SLF, Bhadelia N, Raabe VN, Mulligan MJ, Maljkovic Berry I, Dighero-Kemp B, Kurtz JR, Hensley LE, Dozier NCE, Marron LCB, DuChene A, Kuhn JH, Brown SK, Khurana S, Lane HC, Neaton JD. Safety and immunogenicity of a delayed booster dose of the rVSVΔG-ZEBOV-GP vaccine for prevention of Ebola virus disease: a multicentre, open-label, phase 2 randomised controlled trial. THE LANCET. MICROBE 2024; 5:100923. [PMID: 39374605 PMCID: PMC11560587 DOI: 10.1016/s2666-5247(24)00163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND rVSVΔG-ZEBOV-GP is the first approved vaccine with clinical efficacy against Ebola virus disease. Although a seroprotective threshold has not been defined for those at occupational risk of exposure, the current vaccine strategy is to attain a sustained high level of antibody titres. The aim of this trial was to explore the effects of delayed boosting upon both the height and duration of antibody titres following primary immunisation. METHODS In this open-label phase 2 randomised controlled trial, we compared antibody titres at month 36 in participants who had received a homologous booster dose at month 18 following primary immunisation with those who had received no booster. From Oct 25, 2016, to Jan 29, 2020, healthy adults aged 18 years or older deemed at occupational risk of exposure to Ebola virus due to laboratory work, clinical duties, or travel to an active endemic region were recruited from four hospital clinics in the USA and one hospital clinic in Canada and received primary vaccination with 2×107 plaque-forming unit per mL of VSVΔG-ZEBOV-GP. 18 months later, individuals who consented and were still eligible were randomly assigned 1:1 to receive either a homologous booster dose or no booster. Study visits for safety and serial blood collections for antibody titres were done on enrolled participants at months 0, 1, 3, 6, 12, 18, 19, 24, 30, and 36. Through July, 2021, a web-based application was used for randomisation, including assignments with schedules for each of the five sites using mixed permuted blocks. The trial was not masked to participants or site staff. The primary endpoint was a comparison of geometric mean titres (GMTs) of anti-Ebola virus glycoprotein IgG antibody at month 36 (ie, 18 months after randomisation) for all randomly assigned participants who completed the 36 months of follow-up (primary analysis cohort). Investigators were aware of antibody titres from baseline (enrolment) through month 18 but were masked to summary data by randomisation group after month 18. This study is registered with ClinicalTrials.gov (NCT02788227). FINDINGS Of the 248 participants who enrolled and received their primary immunisation, 114 proceeded to the randomisation step at month 18. The two randomisation groups were balanced: 57 participants (24 [42%] of whom were female; median age was 42 years [IQR 35-50]) were randomly assigned to the booster group and 57 (24 [42%] of whom were female; median age was 42 years [IQR 36-51]) to the no-booster group. Of those randomly assigned, 92 participants (45 in the booster group and 47 in the no-booster group) completed 36 months of follow-up. At 18 months after primary immunisation, GMTs in the no-booster group increased from a baseline of 10 ELISA units (EU)/mL (95% CI 7-14) to 1451 EU/mL (1118-1882); GMTs in the booster group increased from 9 EU/mL (6-16) to 1769 EU/mL (1348-2321). At month 19, GMTs were 31 408 EU/mL (23 181-42 554) for the booster group and 1406 EU/mL (1078-1833) for the no-booster group; at month 36, GMTs were 10 146 EU/mL (7960-12 933) for the booster group and 1240 EU/mL (984-1563) for the no-booster group. Accordingly, the geometric mean ratio (GMR) of antibody titres had increased almost 21-fold more in the booster versus no-booster group at 1 month after booster administration (GMR 20·6; 95% CI 18·2-23·0; p<0·0001) and was still over 7-fold higher at month 36 (GMR 7·8; 95% CI 5·5-10·2; p<0·0001). Consistent with previous reports of this vaccine's side-effects, transient mono-articular or oligo-articular arthritis was diagnosed in 18 (9%) of 207 primary vaccination recipients; after randomisation, arthritis was diagnosed in one (2%) of 57 participants in the no-booster group. No new cases of arthritis developed after booster administration. Four serious adverse events occurred following randomisation: one (epistaxis) in the booster group and three (gastrointestinal haemorrhage, prostate cancer, and tachyarrhythmia) in the no-booster group. None of the serious adverse events was judged attributable to the booster vaccination assignment. INTERPRETATION In addition to no new safety concerns and in marked contrast to earlier trials evaluating short-term boosting, delaying a rVSVΔG-ZEBOV-GP booster until month 18 resulted in an increase in GMT that remained several-fold above the no-booster group GMT for at least 18 months. These findings could have implications for defining the optimal timing of booster doses as pre-exposure prophylaxis in populations at ongoing risk for Ebola virus exposure. FUNDING The Division of Intramural Research and the Division of Clinical Research of the National Institute of Allergy and Infectious Diseases at the US National Institutes of Health, Canadian Immunization Research Network through the Public Health Agency of Canada, Canadian Institutes of Health Research, and the US Defense Threat Reduction Agency.
Collapse
Affiliation(s)
- Richard T Davey
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Gary L Collins
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Nadine Rouphael
- Hope Clinic of Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Guillaume Poliquin
- Public Health Agency of Canada, Children's Hospital Research Institute, Winnipeg, MB, Canada
| | - Rosemary McConnell
- Critical Care Medicine Department, Clinical Research Center, National Institutes of Health, Bethesda, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, Silver Spring, MD, USA
| | - Susan L Moir
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joanne M Langley
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS, Canada
| | - Marc Teitelbaum
- Clinical Monitoring Research Program, Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Angela L Hewlett
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susan L F McLellan
- Division of Infectious Diseases, Dept of Medicine, School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Nahid Bhadelia
- Boston University Center for Emerging Infectious Diseases Policy and Research, Boston University, Boston, MA, USA
| | - Vanessa N Raabe
- New York University Grossman School of Medicine, New York, NY, USA
| | - Mark J Mulligan
- New York University Grossman School of Medicine, New York, NY, USA
| | - Irina Maljkovic Berry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Bonnie Dighero-Kemp
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jonathan R Kurtz
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Lisa E Hensley
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Nelson C E Dozier
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Lindsay C B Marron
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Alain DuChene
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Shawn K Brown
- Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food & Drug Administration, Silver Spring, MD, USA
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James D Neaton
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
4
|
Kainulainen MH, Harmon JR, Karaaslan E, Kyondo J, Whitesell A, Twongyeirwe S, Malenfant JH, Baluku J, Kofman A, Bergeron É, Waltenburg MA, Nyakarahuka L, Balinandi S, Cossaboom CM, Choi MJ, Shoemaker TR, Montgomery JM, Spiropoulou CF. A public, cross-reactive glycoprotein epitope confounds Ebola virus serology. J Med Virol 2024; 96:e29946. [PMID: 39370872 PMCID: PMC11874798 DOI: 10.1002/jmv.29946] [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: 06/25/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Ebola disease (EBOD) in humans is a severe disease caused by at least four related viruses in the genus Orthoebolavirus, most often by the eponymous Ebola virus. Due to human-to-human transmission and incomplete success in treating cases despite promising therapeutic development, EBOD is a high priority in public health research. Yet despite almost 50 years since EBOD was first described, the sources of these viruses remain undefined and much remains to be understood about the disease epidemiology and virus emergence and spread. One important approach to improve our understanding is detection of antibodies that can reveal past human infections. However, serosurveys routinely describe seroprevalences that imply infection rates much higher than those clinically observed. Proposed hypotheses to explain this difference include existence of common but less pathogenic strains or relatives of these viruses, misidentification of EBOD as something else, and a higher proportion of subclinical infections than currently appreciated. The work presented here maps B-cell epitopes in the spike protein of Ebola virus and describes a single epitope that is cross-reactive with an antigen seemingly unrelated to orthoebolaviruses. Antibodies against this epitope appear to explain most of the unexpected reactivity towards the spike, arguing against common but unidentified infections in the population. Importantly, antibodies of cross-reactive donors from within and outside the known EBOD geographic range bound the same epitope. In light of this finding, it is plausible that epitope mapping enables broadly applicable specificity improvements in the field of serology.
Collapse
Affiliation(s)
- Markus H. Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica R. Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jackson Kyondo
- VHF Diagnostics Laboratory, Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Amy Whitesell
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sam Twongyeirwe
- VHF Diagnostics Laboratory, Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jason H. Malenfant
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jimmy Baluku
- VHF Diagnostics Laboratory, Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Aaron Kofman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michelle A. Waltenburg
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Luke Nyakarahuka
- VHF Diagnostics Laboratory, Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
- Department of Biosecurity, Ecosystems, and Veterinary Public Health, College of Veterinary Medicine, Animal Resources, and Biosecurity, Makerere University, Kampala, Uganda
| | - Stephen Balinandi
- VHF Diagnostics Laboratory, Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Caitlin M. Cossaboom
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mary J. Choi
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Trevor R. Shoemaker
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Joel M. Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
5
|
Khurana S, Grubbs G, Ravichandran S, Cluff E, Kim J, Kuehne AI, Zak S, Dye JM, Lutwama JJ, Herbert AS. Longitudinal proteome-wide antibody profiling in Marburg virus survivors identifies wing domain immunogen for vaccine design. Nat Commun 2024; 15:8133. [PMID: 39285186 PMCID: PMC11405854 DOI: 10.1038/s41467-024-51021-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/25/2024] [Indexed: 09/19/2024] Open
Abstract
Limited knowledge exists on the quality of polyclonal antibody responses generated following Marburg virus (MARV) infection and its evolution in survivors. In this study, we evaluate MARV proteome-wide antibody repertoire longitudinally in convalescent phase approximately every six months for five years following MARV infection in ten human survivors. Differential kinetics were observed for IgM vs IgG vs IgA epitope diversity, antibody binding, antibody affinity maturation and Fc-receptor interaction to MARV proteins. Durability of MARV-neutralizing antibodies is low in survivors. MARV infection induces a diverse epitope repertoire with predominance against GP, VP40, VP30 and VP24 that persisted up to 5 years post-exposure. However, the IgM and IgA repertoire declines over time. Within MARV-GP, IgG recognize antigenic sites predominantly in the amino-terminus, wing domain and GP2-heptad repeat. Interestingly, MARV infection generates robust durable FcɣRI, FcɣRIIA and FcɣRIIIA IgG-Fc receptor interactions. Immunization with immunodominant MARV epitopes reveals conserved wing region between GP1 and GP2, induces neutralizing antibodies against MARV. These findings demonstrate that MARV infection generates a diverse, long-lasting, non-neutralizing, IgG antibody repertoire that perturbs disease by FcɣR activity. This information, along with discovery of neutralizing immunogen in wing domain, could aid in development of effective therapeutics and vaccines against Marburg virus.
Collapse
Affiliation(s)
- Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA.
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Emily Cluff
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - JungHyun Kim
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Ana I Kuehne
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Samantha Zak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - John M Dye
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Julius J Lutwama
- Department of Arbovirology, Emerging, and Re-emerging Infection, Uganda Virus Research Institute, Entebbe, Uganda
| | - Andrew S Herbert
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| |
Collapse
|
6
|
Filipić B, Pantelić I, Nikolić I, Majhen D, Stojić-Vukanić Z, Savić S, Krajišnik D. Nanoparticle-Based Adjuvants and Delivery Systems for Modern Vaccines. Vaccines (Basel) 2023; 11:1172. [PMID: 37514991 PMCID: PMC10385383 DOI: 10.3390/vaccines11071172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Ever since the development of the first vaccine, vaccination has had the great impact on global health, leading to the decrease in the burden of numerous infectious diseases. However, there is a constant need to improve existing vaccines and develop new vaccination strategies and vaccine platforms that induce a broader immune response compared to traditional vaccines. Modern vaccines tend to rely on certain nanotechnology platforms but are still expected to be readily available and easy for large-scale manufacturing and to induce a durable immune response. In this review, we present an overview of the most promising nanoadjuvants and nanoparticulate delivery systems and discuss their benefits from tehchnological and immunological standpoints as well as their objective drawbacks and possible side effects. The presented nano alums, silica and clay nanoparticles, nanoemulsions, adenoviral-vectored systems, adeno-associated viral vectors, vesicular stomatitis viral vectors, lentiviral vectors, virus-like particles (including bacteriophage-based ones) and virosomes indicate that vaccine developers can now choose different adjuvants and/or delivery systems as per the requirement, specific to combatting different infectious diseases.
Collapse
Affiliation(s)
- Brankica Filipić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ivana Pantelić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Ines Nikolić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
- Section of Pharmaceutical Sciences, University of Geneva, 1206 Geneva, Switzerland
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Snežana Savić
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| | - Danina Krajišnik
- Department of Pharmaceutical Technology and Cosmetology, University of Belgrade-Faculty of Pharmacy, 11000 Belgrade, Serbia
| |
Collapse
|
7
|
Rijal P, Donnellan FR. A review of broadly protective monoclonal antibodies to treat Ebola virus disease. Curr Opin Virol 2023; 61:101339. [PMID: 37392670 DOI: 10.1016/j.coviro.2023.101339] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/26/2023] [Accepted: 05/28/2023] [Indexed: 07/03/2023]
Abstract
The filovirus vaccine and the therapeutic monoclonal antibody (mAb) research have made substantial progress. However, existing vaccines and mAbs approved for use in humans are specific to Zaire ebolavirus (EBOV). Since other Ebolavirus species are a continuing threat to public health, the search for broadly protective mAbs has drawn attention. Here, we review viral glycoprotein-targeting mAbs that have proved their broader protective efficacy in animal models. MBP134AF, the most advanced of these new-generation mAb therapies, has recently been deployed in Uganda during the Sudan ebolavirus outbreak. Furthermore, we discuss the measures associated with enhancing antibody therapies and the risks associated with them, including the rise of escape mutations following the mAb treatment and naturally occurring EBOV variants.
Collapse
Affiliation(s)
- Pramila Rijal
- Center for Translational Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine, University of Oxford, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, United Kingdom.
| | - Francesca R Donnellan
- Department of Biochemistry, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, UK.
| |
Collapse
|
8
|
Gunn BM, McNamara RP, Wood L, Taylor S, Devadhasan A, Guo W, Das J, Nilsson A, Shurtleff A, Dubey S, Eichberg M, Suscovich TJ, Saphire EO, Lauffenburger D, Coller BA, Simon JK, Alter G. Antibodies against the Ebola virus soluble glycoprotein are associated with long-term vaccine-mediated protection of non-human primates. Cell Rep 2023; 42:112402. [PMID: 37061918 PMCID: PMC10576837 DOI: 10.1016/j.celrep.2023.112402] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/30/2023] [Accepted: 03/31/2023] [Indexed: 04/17/2023] Open
Abstract
The 2013 Ebola epidemic in Central and West Africa heralded the emergence of wide-spread, highly pathogenic viruses. The successful recombinant vector vaccine against Ebola (rVSVΔG-ZEBOV-GP) will limit future outbreaks, but identifying mechanisms of protection is essential to protect the most vulnerable. Vaccine-induced antibodies are key determinants of vaccine efficacy, yet the mechanism by which vaccine-induced antibodies prevent Ebola infection remains elusive. Here, we exploit a break in long-term vaccine efficacy in non-human primates to identify predictors of protection. Using unbiased humoral profiling that captures neutralization and Fc-mediated functions, we find that antibodies specific for soluble glycoprotein (sGP) drive neutrophil-mediated phagocytosis and predict vaccine-mediated protection. Similarly, we show that protective sGP-specific monoclonal antibodies have elevated neutrophil-mediated phagocytic activity compared with non-protective antibodies, highlighting the importance of sGP in vaccine protection and monoclonal antibody therapeutics against Ebola virus.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| | - Lianna Wood
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Sabian Taylor
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Wenyu Guo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Jishnu Das
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Avlant Nilsson
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Amy Shurtleff
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | | | | | | | | | - Douglas Lauffenburger
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
9
|
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
|
10
|
Sparks R, Lau WW, Liu C, Han KL, Vrindten KL, Sun G, Cox M, Andrews SF, Bansal N, Failla LE, Manischewitz J, Grubbs G, King LR, Koroleva G, Leimenstoll S, Snow L, Chen J, Tang J, Mukherjee A, Sellers BA, Apps R, McDermott AB, Martins AJ, Bloch EM, Golding H, Khurana S, Tsang JS. Influenza vaccination reveals sex dimorphic imprints of prior mild COVID-19. Nature 2023; 614:752-761. [PMID: 36599369 PMCID: PMC10481789 DOI: 10.1038/s41586-022-05670-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023]
Abstract
Acute viral infections can have durable functional impacts on the immune system long after recovery, but how they affect homeostatic immune states and responses to future perturbations remain poorly understood1-4. Here we use systems immunology approaches, including longitudinal multimodal single-cell analysis (surface proteins, transcriptome and V(D)J sequences) to comparatively assess baseline immune statuses and responses to influenza vaccination in 33 healthy individuals after recovery from mild, non-hospitalized COVID-19 (mean, 151 days after diagnosis) and 40 age- and sex-matched control individuals who had never had COVID-19. At the baseline and independent of time after COVID-19, recoverees had elevated T cell activation signatures and lower expression of innate immune genes including Toll-like receptors in monocytes. Male individuals who had recovered from COVID-19 had coordinately higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared with healthy male individuals and female individuals who had recovered from COVID-19, in part because male recoverees had monocytes with higher IL-15 responses early after vaccination coupled with elevated prevaccination frequencies of 'virtual memory'-like CD8+ T cells poised to produce more IFNγ after IL-15 stimulation. Moreover, the expression of the repressed innate immune genes in monocytes increased by day 1 to day 28 after vaccination in recoverees, therefore moving towards the prevaccination baseline of the healthy control individuals. By contrast, these genes decreased on day 1 and returned to the baseline by day 28 in the control individuals. Our study reveals sex-dimorphic effects of previous mild COVID-19 and suggests that viral infections in humans can establish new immunological set-points that affect future immune responses in an antigen-agnostic manner.
Collapse
Affiliation(s)
- Rachel Sparks
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - William W Lau
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Can Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Graduate Program in Biological Sciences, University of Maryland, College Park, MD, USA
| | - Kyu Lee Han
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Kiera L Vrindten
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Guangping Sun
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Milann Cox
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Laura E Failla
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Lisa R King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Galina Koroleva
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - LaQuita Snow
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
- Johns Hopkins University, Baltimore, MD, USA
| | - Jinguo Chen
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | | | - Brian A Sellers
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Richard Apps
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - Andrew J Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA.
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA.
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
11
|
Simon JK, Kennedy SB, Mahon BE, Dubey SA, Grant-Klein RJ, Liu K, Hartzel J, Coller BAG, Welebob C, Hanson ME, Grais RF. Immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine (ERVEBO®) in African clinical trial participants by age, sex, and baseline GP-ELISA titer: A post hoc analysis of three Phase 2/3 trials. Vaccine 2022; 40:6599-6606. [PMID: 36208978 DOI: 10.1016/j.vaccine.2022.09.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND ERVEBO®, a live recombinant vesicular stomatitis virus (VSV) vaccine containing the Zaire ebolavirus glycoprotein (GP) in place of the VSV GP (rVSVΔG-ZEBOV-GP), was advanced through clinical development by Merck & Co., Inc., Rahway, NJ, USA in collaboration with multiple partners to prevent Ebola virus disease (EVD) and has been approved for human use in several countries. METHODS We evaluated data from three Phase 2/3 clinical trials conducted in Liberia (PREVAIL), Guinea (FLW), and Sierra Leone (STRIVE) during the 2013-2016 West African EVD outbreak to assess immune responses using validated assays. We performed a post hoc analysis of the association of vaccine response with sex, age (18-50 yrs & >50 yrs), and baseline (BL) GP-enzyme-linked immunosorbent assay (ELISA) titer (<200 & ≥200 EU/mL), including individual study (PREVAIL, FLW, or STRIVE) data and pooled data from all 3 studies. The endpoints were total IgG antibody response (EU/mL) measured by the GP-ELISA and neutralizing antibody response measured by the plaque reduction neutralization test (PRNT) to rVSVΔG-ZEBOV-GP at Days 28, 180, and 365 postvaccination. RESULTS In the overall pooled population, in all subgroups, and in each trial independently, GP-ELISA and PRNT geometric mean titers increased from BL, generally peaking at Day 28 and persisting through Day 365. Immune responses were greater in women and participants with BL GP-ELISA ≥ 200 EU/mL, but did not differ across age groups. CONCLUSION These data demonstrate that rVSVΔG-ZEBOV-GP elicits a robust and durable immune response through 12 months postvaccination in participants regardless of age, sex, or BL GP-ELISA titer. The higher immune responses observed in women and participants with pre-existing immunity are consistent with those described previously and for other vaccines. Trials were registered as follows: PREVAIL: ClinicalTrials.gov NCT02344407; FLW: Pan African Clinical Trials Registry PACTR201503001057193; STRIVE: ClinicalTrials.gov NCT02378753. Protocols V920-009, 011, and 018.
Collapse
Affiliation(s)
| | - Stephen B Kennedy
- Partnership for Research on Ebola Vaccines in Liberia (PREVAIL), Monrovia, Liberia
| | - Barbara E Mahon
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Ken Liu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | | | | |
Collapse
|
12
|
Pogostin BH, Yu MH, Azares AR, Euliano EM, Lai CSE, Saenz G, Wu SX, Farsheed AC, Melhorn SM, Graf TP, Woodside DG, Hartgerink JD, McHugh KJ. Multidomain peptide hydrogel adjuvants elicit strong bias towards humoral immunity. Biomater Sci 2022; 10:6217-6229. [PMID: 36102692 PMCID: PMC9717470 DOI: 10.1039/d2bm01242a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Adjuvants play a critical role in enhancing vaccine efficacy; however, there is a need to develop new immunomodulatory compounds to address emerging pathogens and to expand the use of immunotherapies. Multidomain peptides (MDPs) are materials composed of canonical amino acids that form injectable supramolecular hydrogels under physiological salt and pH conditions. MDP hydrogels are rapidly infiltrated by immune cells in vivo and have previously been shown to influence cytokine production. Therefore, we hypothesized that these immunostimulatory characteristics would allow MDPs to function as vaccine adjuvants. Herein, we demonstrate that loading antigen into MDP hydrogels does not interfere with their rheological properties and that positively charged MDPs can act as antigen depots, as demonstrated by their ability to release ovalbumin (OVA) over a period of 7-9 days in vivo. Mice vaccinated with MDP-adjuvanted antigen generated significantly higher IgG titers than mice treated with the unadjuvanted control, suggesting that these hydrogels potentiate humoral immunity. Interestingly, MDP hydrogels did not elicit a robust cellular immune response, as indicated by the lower production of IgG2c and smaller populations of tetramer-positive CD8+ T splenocytes compared to mice vaccinated alum-adjuvanted OVA. Together, the data suggest that MDP hydrogel adjuvants strongly bias the immune response towards humoral immunity while evoking a very limited cellular immune response. As a result, MDPs may have the potential to serve as adjuvants for applications that benefit exclusively from humoral immunity.
Collapse
Affiliation(s)
- Brett H Pogostin
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Marina H Yu
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Alon R Azares
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, TX, 77030, USA
| | - Erin M Euliano
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | | | - Gabriel Saenz
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Samuel X Wu
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Adam C Farsheed
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Sarah M Melhorn
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Tyler P Graf
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Darren G Woodside
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, TX, 77030, USA
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
13
|
Xu HM, Xu WM, Zhang L. Current Status of Phage Therapy against Infectious Diseases and Potential Application beyond Infectious Diseases. Int J Clin Pract 2022; 2022:4913146. [PMID: 36263241 PMCID: PMC9550513 DOI: 10.1155/2022/4913146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/19/2022] [Indexed: 12/02/2022] Open
Abstract
Intestinal microbiota plays a key role in regulating the pathogenesis of human disease and maintaining health. Many diseases, mainly induced by bacteria, are on the rise due to the emergence of antibiotic-resistant strains. Intestinal microorganisms include organisms such as bacteria, viruses, and fungi. They play an important role in maintaining human health. Among these microorganisms, phages are the main members of intestinal viromes. In particular, the viral fraction, composed essentially of phages, affects homeostasis by exerting selective pressure on bacterial communities living in the intestinal tract. In recent years, with the widespread use and even abuse of antibacterial drugs, more and more drug-resistant bacteria have been found, and they show a trend of high drug resistance and multidrug resistance. Therefore, it has also become increasingly difficult to treat serious bacterial infections. Phages, a natural antibacterial agent with strong specificity and rapid proliferation, have come back to the field of vision of clinicians and scholars. In this study, the current state of research on intestinal phages was discussed, with an exploration of the impact of phage therapy against infectious diseases, as well as potential application beyond infectious diseases.
Collapse
Affiliation(s)
- Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Wen-Min Xu
- Department of Endoscopy, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510091, China
| | - Long Zhang
- Department of Endoscopy, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510091, China
| |
Collapse
|
14
|
Sparks R, Lau WW, Liu C, Han KL, Vrindten KL, Sun G, Cox M, Andrews SF, Bansal N, Failla LE, Manischewitz J, Grubbs G, King LR, Koroleva G, Leimenstoll S, Snow L, Chen J, Tang J, Mukherjee A, Sellers BA, Apps R, McDermott AB, Martins AJ, Bloch EM, Golding H, Khurana S, Tsang JS. Influenza vaccination and single cell multiomics reveal sex dimorphic immune imprints of prior mild COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.02.17.22271138. [PMID: 35233581 PMCID: PMC8887138 DOI: 10.1101/2022.02.17.22271138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Viral infections can have profound and durable functional impacts on the immune system. There is an urgent need to characterize the long-term immune effects of SARS-CoV-2 infection given the persistence of symptoms in some individuals and the continued threat of novel variants. Here we use systems immunology, including longitudinal multimodal single cell analysis (surface proteins, transcriptome, and V(D)J sequences) from 33 previously healthy individuals after recovery from mild, non-hospitalized COVID-19 and 40 age- and sex-matched healthy controls with no history of COVID-19 to comparatively assess the post-infection immune status (mean: 151 days after diagnosis) and subsequent innate and adaptive responses to seasonal influenza vaccination. Identification of both sex-specific and -independent temporally stable changes, including signatures of T-cell activation and repression of innate defense/immune receptor genes (e.g., Toll-like receptors) in monocytes, suggest that mild COVID-19 can establish new post-recovery immunological set-points. COVID-19-recovered males had higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared to healthy males and COVID-19-recovered females, partly attributable to elevated pre-vaccination frequencies of a GPR56 expressing CD8+ T-cell subset in male recoverees that are "poised" to produce higher levels of IFNγ upon inflammatory stimulation. Intriguingly, by day 1 post-vaccination in COVID-19-recovered subjects, the expression of the repressed genes in monocytes increased and moved towards the pre-vaccination baseline of healthy controls, suggesting that the acute inflammation induced by vaccination could partly reset the immune states established by mild COVID-19. Our study reveals sex-dimorphic immune imprints and in vivo functional impacts of mild COVID-19 in humans, suggesting that prior COVID-19, and possibly respiratory viral infections in general, could change future responses to vaccination and in turn, vaccines could help reset the immune system after COVID-19, both in an antigen-agnostic manner.
Collapse
Affiliation(s)
- Rachel Sparks
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - William W. Lau
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,These authors contributed equally
| | - Can Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Graduate Program in Biological Sciences, University of Maryland, College Park, MD, USA,These authors contributed equally
| | - Kyu Lee Han
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Kiera L. Vrindten
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Guangping Sun
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Milann Cox
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - Neha Bansal
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Laura E. Failla
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Lisa R. King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Galina Koroleva
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - LaQuita Snow
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | | | - Jinguo Chen
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | | | | | - Richard Apps
- NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA
| | | | - Andrew J. Martins
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - John S. Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA,NIH Center for Human Immunology, NIAID, NIH, Bethesda, MD, USA,Correspondence:
| |
Collapse
|
15
|
Bellusci L, Grubbs G, Zahra FT, Forgacs D, Golding H, Ross TM, Khurana S. Antibody affinity and cross-variant neutralization of SARS-CoV-2 Omicron BA.1, BA.2 and BA.3 following third mRNA vaccination. Nat Commun 2022; 13:4617. [PMID: 35941152 PMCID: PMC9358642 DOI: 10.1038/s41467-022-32298-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/25/2022] [Indexed: 11/11/2022] Open
Abstract
There is limited knowledge on durability of neutralization capacity and antibody affinity maturation generated following two versus three doses of SARS-CoV-2 mRNA vaccines in naïve versus convalescent individuals (hybrid immunity) against the highly transmissible Omicron BA.1, BA.2 and BA.3 subvariants. Virus neutralization titers against the vaccine-homologous strain (WA1) and Omicron sublineages are measured in a pseudovirus neutralization assay (PsVNA). In addition, antibody binding and antibody affinity against spike proteins from WA1, BA.1, and BA.2 is determined using surface plasmon resonance (SPR). The convalescent individuals who after SARS-CoV-2 infection got vaccinated develop hybrid immunity that shows broader neutralization activity and cross-reactive antibody affinity maturation against the Omicron BA.1 and BA.2 after either second or third vaccination compared with naïve individuals. Neutralization activity correlates with antibody affinity against Omicron subvariants BA.1 and BA.2 spikes. Importantly, at four months post-third vaccination the neutralization activity and antibody affinity against the Omicron subvariants is maintained and trended higher for the individuals with hybrid immunity compared with naïve adults. These findings about hybrid immunity resulting in superior immune kinetics, breadth, and durable high affinity antibodies support the need for booster vaccinations to provide effective protection from emerging SARS-CoV-2 variants like the rapidly spreading Omicron subvariants. Here the authors show that a third SARS-CoV-2 vaccination significantly boosts neutralizing antibodies against Omicron subvariants and that hybrid immunity (infection and vaccination) results in broader neutralization activity and cross-reactive antibody affinity maturation.
Collapse
Affiliation(s)
- Lorenza Bellusci
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20871, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20871, USA
| | - Fatema Tuz Zahra
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20871, USA
| | - David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, 30602, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20871, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, 30602, USA.,Department of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20871, USA.
| |
Collapse
|
16
|
Ebola-Detect: A differential serodiagnostic assay for Ebola virus infections and surveillance in the presence of vaccine-induced antibodies. EBioMedicine 2022; 82:104186. [PMID: 35901660 PMCID: PMC9326332 DOI: 10.1016/j.ebiom.2022.104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/05/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Background Ebola virus (EBOV) vaccines containing glycoprotein (GP) provide protection against severe Ebola virus disease (EVD). EBO vaccinations elicit antibodies that are detectable in Ebola serodiagnostic tests, as EBOV GP is a major target antigen. This vaccine-induced seropositivity presents issues with early detection of natural EBOV infections, following vaccination and during surveillance, leading to ‘uninfected’ vaccine trial participants being falsely diagnosed as ‘EBOV infected’ potentially resulting in long-term social and economic distress. Since mass vaccinations are being employed to curtail the recurrent EBOV epidemics in multiple African countries, it is, therefore, essential to differentiate vaccine-induced from natural infection–induced antibodies by a differential serodiagnosis assay for accurate detection of Ebola virus infections. Methods To develop a serodiagnostic test that can differentiate between individuals with EBOV infection-induced antibodies and individuals with EBOV vaccine-induced antibodies, we analysed peptides of EBOV viral protein 40 (VP40), viral protein 35 (VP35) and nucleocapsid protein (NP) using an ELISA with a panel of 181 human sera collected from healthy controls, EBO vaccinees, and EBOV-infected survivors. Receiver Operating Characteristic (ROC) curve analysis was used to calculate sensitivity and specificity of the assay. A simple peptide-based serodiagnostic assay was used to evaluate detection of breakthrough EBOV infections in vaccinated non-human primates (NHP) in EBOV challenge studies. Findings We identified conserved peptide sequences in EBOV VP40, VP35 and NP, produced soon after EBOV infection that are not part of the current EBO vaccine target antigens. The new ELISA-based differential serodetection assay termed ‘EBOV-Detect’ demonstrated >94% specificity and 96% sensitivity for diagnosis of EBOV infection. Importantly, the uninfected vaccine-trial participants scored negative in ‘EBOV-Detect’ assay. The results from the NHPs EBOV challenge study established that post-EBO vaccination serum scored negative in ‘EBOV-Detect’ and all NHPs with Ebola breakthrough infections, following EBOV challenge, were serodiagnosed positively with EBOV-Detect. Interpretation The new ‘EBOV-Detect’ is a simple and sensitive serodiagnostic assay that can specifically differentiate between natural Ebola virus infected and those with vaccine-induced immunity. This could potentially be implemented as a robust diagnostic tool for epidemiology and surveillance of EBOV infections during and after outbreaks, especially in countries with mass Ebola vaccinations. Funding The antibody characterization work described in this manuscript was supported by FDA Office of Counterterrorism and Emerging Threats (OCET) - Medical Countermeasures initiative (MCMi) grant- OCET 2019-1018 and Defense Threat Reduction Agency (HDTRA1930447) funds to S.K.
Collapse
|
17
|
Rudge TL, Machesky NJ, Sankovich KA, Lemmon EE, Badorrek CS, Overman R, Niemuth NA, Anderson MS. Assays for the Evaluation of the Immune Response to Marburg and Ebola Sudan Vaccination-Filovirus Animal Nonclinical Group Anti-Marburg Virus Glycoprotein Immunoglobulin G Enzyme-Linked Immunosorbent Assay and a Pseudovirion Neutralization Assay. Vaccines (Basel) 2022; 10:1211. [PMID: 36016099 PMCID: PMC9413256 DOI: 10.3390/vaccines10081211] [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: 06/06/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Since the discovery of the Marburg virus (MARV) in 1967 and Ebola virus (EBOV) in 1976, there have been over 40 reported outbreaks of filovirus disease with case fatality rates greater than 50%. This underscores the need for efficacious vaccines against these highly pathogenic filoviruses. Due to the sporadic and unpredictable nature of filovirus outbreaks, such a vaccine would likely need to be vetted through the U.S. Food and Drug Administration (FDA), following the Animal Rule or similar European Medicines Agency (EMA) regulatory pathway. Under the FDA Animal Rule, vaccine-induced immune responses correlating with survival of non-human primates (NHPs), or another well-characterized animal model, following lethal challenge, will need to be bridged for human immune response distributions in clinical trials. A correlate of protection has not yet been identified for the filovirus disease, but antibodies, specifically anti-glycoprotein (GP) antibodies, are believed to be critical in providing protection against the filovirus disease following vaccination and are thus a strong candidate for a correlate of protection. Thus, species-neutral methods capable of the detection and bridging of these antibody immune responses, such as methods to quantify anti-GP immunoglobulin G (IgG)-binding antibodies and neutralizing antibodies, are needed. Reported here is the development and qualification of two Filovirus Animal Nonclinical Group (FANG) anti-GP IgG Enzyme-Linked Immunosorbent Assays (ELISAs) to quantify anti-MARV and anti-Sudan virus (SUDV) IgG antibodies in human and NHP serum samples, as well as the development of pseudovirion neutralization assays (PsVNAs) to quantify MARV- and SUDV-neutralizing antibodies in human and NHP serum samples.
Collapse
Affiliation(s)
- Thomas L. Rudge
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| | - Nicholas J. Machesky
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| | - Karen A. Sankovich
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| | - Erin E. Lemmon
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| | - Christopher S. Badorrek
- Contract Support for the U.S. Department of Defense (DOD) Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense (JPEO-CBRND) Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA;
| | - Rachel Overman
- U.S. Department of Defense (DOD) Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense (JPEO-CBRND) Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, MD 21702, USA;
| | - Nancy A. Niemuth
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| | - Michael S. Anderson
- Battelle, West Jefferson, OH 43162, USA; (N.J.M.); (K.A.S.); (E.E.L.); (N.A.N.); (M.S.A.)
| |
Collapse
|
18
|
Travieso T, Li J, Mahesh S, Mello JDFRE, Blasi M. The use of viral vectors in vaccine development. NPJ Vaccines 2022; 7:75. [PMID: 35787629 PMCID: PMC9253346 DOI: 10.1038/s41541-022-00503-y] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/15/2022] [Indexed: 12/22/2022] Open
Abstract
Vaccines represent the single most cost-efficient and equitable way to combat and eradicate infectious diseases. While traditional licensed vaccines consist of either inactivated/attenuated versions of the entire pathogen or subunits of it, most novel experimental vaccines against emerging infectious diseases employ nucleic acids to produce the antigen of interest directly in vivo. These include DNA plasmid vaccines, mRNA vaccines, and recombinant viral vectors. The advantages of using nucleic acid vaccines include their ability to induce durable immune responses, high vaccine stability, and ease of large-scale manufacturing. In this review, we present an overview of pre-clinical and clinical data on recombinant viral vector vaccines and discuss the advantages and limitations of the different viral vector platforms.
Collapse
Affiliation(s)
- Tatianna Travieso
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jenny Li
- Duke University, Durham, NC, USA
| | - Sneha Mahesh
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Juliana Da Fonzeca Redenze E Mello
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maria Blasi
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA. .,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
19
|
Kurtović T, Ravlić S, Štimac A, Mateljak Lukačević S, Hećimović A, Kazazić S, Halassy B. Efficient and Sustainable Platform for Preparation of a High-Quality Immunoglobulin G as an Urgent Treatment Option During Emerging Virus Outbreaks. Front Immunol 2022; 13:889736. [PMID: 35655779 PMCID: PMC9152316 DOI: 10.3389/fimmu.2022.889736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
During the pre-vaccine era of the COVID-19 pandemic convalescent plasma has once again emerged as a major potential therapeutic form of passive immunization that in specific cases still represents irreplaceable treatment option. There is a growing concern that variable concentration of neutralizing antibodies, present in convalescent plasma which originates from different donors, apparently affects its effectiveness. The drawback can be overcome through the downstream process of immunoglobulin fraction purification into a standardized product of improved safety and efficacy. All modern procedures are quite lengthy processes. They are also based on fractionation of large plasma quantities whose collection is not attainable during an epidemic. When outbreaks of infectious diseases are occurring more frequently, there is a great need for a more sustainable production approach that would be goal-oriented towards assuring easily and readily available immunoglobulin of therapeutic relevance. We propose a refinement strategy for the IgG preparation achieved through simplification and reduction of the processing steps. It was designed as a small but scalable process to offer an immediately available treatment option that would simultaneously be harmonized with an increased availability of convalescent plasma over the viral outbreak time-course. Concerning the ongoing pandemic status of the COVID-19, the proof of concept was demonstrated on anti-SARS-CoV-2 convalescent plasma but is likely applicable to any other type depending on the current needs. It was guided by the idea of persistent keeping of IgG molecules in the solution, so that protection of their native structure could be assured. Our manufacturing procedure provided a high-quality IgG product of above the average recovery whose composition profile was analyzed by mass spectrometry as quality control check. It was proved free from IgA and IgM as mediators of adverse transfusion reactions, as well as of any other residual impurities, since only IgG fragments were identified. The proportion of S protein-specific IgGs remained unchanged relative to the convalescent plasma. Undisturbed IgG subclass composition was accomplished as well. However, the fractionation principle affected the final product's capacity to neutralize wild-type SARS-CoV-2 infectivity, reducing it by half. Decrease in neutralization potency significantly correlated with the amount of IgM in the starting material.
Collapse
Affiliation(s)
- Tihana Kurtović
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, Zagreb, Croatia
| | - Sanda Ravlić
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, Zagreb, Croatia
| | - Adela Štimac
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, Zagreb, Croatia
| | - Sanja Mateljak Lukačević
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, Zagreb, Croatia
| | - Ana Hećimović
- Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Saša Kazazić
- Division of Physical Chemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Virus Immunology and Vaccines, Zagreb, Croatia
| |
Collapse
|
20
|
Vaccine-Associated Enhanced Respiratory Disease following Influenza Virus Infection in Ferrets Recapitulates the Model in Pigs. J Virol 2022; 96:e0172521. [PMID: 34985999 DOI: 10.1128/jvi.01725-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Influenza A virus (IAV) causes respiratory disease in swine and humans. Vaccines are used to prevent influenza illness in both populations but must be frequently updated due to rapidly evolving strains. Mismatch between the circulating strains and the strains contained in vaccines may cause loss of efficacy. Whole inactivated virus (WIV) vaccines with adjuvant, utilized by the swine industry, are effective against antigenically similar viruses; however, vaccine-associated enhanced respiratory disease (VAERD) may happen when the WIV is antigenically mismatched with the infecting virus. VAERD is a repeatable model in pigs, but had yet to be experimentally demonstrated in other mammalian species. We recapitulated VAERD in ferrets, a standard benchmark animal model for studying human influenza infection, in a direct comparison to VAERD in pigs. Both species were vaccinated with WIV with oil-in-water adjuvant containing a δ-1 H1N2 (1B.2.2) derived from the pre-2009 human seasonal lineage, then challenged with a 2009 pandemic H1N1 (H1N1pdm09, 1A.3.3.2) 5 weeks after vaccination. Nonvaccinated and challenged groups showed typical signs of influenza disease, but the mismatched vaccinated and challenged pigs and ferrets showed elevated clinical signs, despite similar viral loads. VAERD-affected pigs exhibited a 2-fold increase in lung lesions, while VAERD-affected ferrets showed a 4-fold increase. Similar to pigs, antibodies from VAERD-affected ferrets preferentially bound to the HA2 domain of the H1N1pdm09 challenge strain. These results indicate that VAERD is not limited to pigs, as demonstrated here in ferrets, and the need to consider VAERD when evaluating new vaccine platforms and strategies. IMPORTANCE We demonstrated the susceptibility of ferrets, a laboratory model species for human influenza A virus research, to vaccine-associated enhanced respiratory disease (VAERD) using an experimental model previously demonstrated in pigs. Ferrets developed clinical characteristics of VAERD very similar to that in pigs. The hemagglutinin (HA) stalk is a potential vaccine target to develop more efficacious, broadly reactive influenza vaccine platforms and strategies. However, non-neutralizing antibodies directed toward a conserved epitope on the HA stalk induced by an oil-in-water, adjuvanted, whole influenza virus vaccine were previously shown in VAERD-affected pigs and were also identified here in VAERD-affected ferrets. The induction of VAERD in ferrets highlights the potential risk of mismatched influenza vaccines for humans and the need to consider VAERD when designing and evaluating vaccine strategies.
Collapse
|
21
|
Characterization of human IgM and IgG repertoires in individuals with chronic HIV-1 infection. Virol Sin 2022; 37:370-379. [PMID: 35247647 PMCID: PMC9243603 DOI: 10.1016/j.virs.2022.02.010] [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: 09/25/2021] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
Advancements in high-throughput sequencing (HTS) of antibody repertoires (Ig-Seq) have unprecedentedly improved our ability to characterize the antibody repertoires on a large scale. However, currently, only a few studies explored the influence of chronic HIV-1 infection on human antibody repertoires and many of them reached contradictory conclusions, possibly limited by inadequate sequencing depth and throughput. To better understand how HIV-1 infection would impact humoral immune system, in this study, we systematically analyzed the differences between the IgM (HIV-IgM) and IgG (HIV-IgG) heavy chain repertoires of HIV-1 infected patients, as well as between antibody repertoires of HIV-1 patients and healthy donors (HH). Notably, the public unique clones accounted for only a negligible proportion between the HIV-IgM and HIV-IgG repertoires libraries, and the diversity of unique clones in HIV-IgG remarkably reduced. In aspect of somatic mutation rates of CDR1 and CDR2, the HIV-IgG repertoire was higher than HIV-IgM. Besides, the average length of CDR3 region in HIV-IgM was significant longer than that in the HH repertoire, presumably caused by the great number of novel VDJ rearrangement patterns, especially a massive use of IGHJ6. Moreover, some of the B cell clonotypes had numerous clones, and somatic variants were detected within the clonotype lineage in HIV-IgG, indicating HIV-1 neutralizing activities. The in-depth characterization of HIV-IgG and HIV-IgM repertoires enriches our knowledge in the profound effect of HIV-1 infection on human antibody repertoires and may have practical value for the discovery of therapeutic antibodies. Ultra-deep sequencing of both IgM and IgG repertoires in chronic HIV-1 infection. VDJ gene rearrangement patterns can be dramatically changed by HIV-1 infection. Multiple mechanisms cause the high complexity of HIV-1-experienced antibodies. Discovery of promising neutralizing HIV-1 antibodies from antibody repertoires.
Collapse
|
22
|
Carnino L, Vetter P, Peyraud N, Aebischer-Perone S, Chappuis F, Huttner A, Kaiser L, Eperon G. Feasibility and safety of rVSV-ZEBOV vaccination of humanitarian health workers against Ebola virus disease: an observational study. J Travel Med 2021; 28:6299197. [PMID: 34128975 DOI: 10.1093/jtm/taab086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 11/14/2022]
Abstract
BACKGROUND AND RATIONALE Geneva University Hospitals were granted a temporary authorization to administer the recombinant live vesicular stomatitis virus rVSV-ZEBOV (Ervebo®) vaccine to expatriate humanitarian frontline workers (FLWs) prior to mission deployment. OBJECTIVES Our aims were to assess the feasibility of FLW vaccination before deployment and to report adverse events (AEs). METHODS FLWs received a single injection of rVSV-ZEBOV (>7.2E7 plaque forming unit) during their pre-deployment medical check-up at the Travel Medicine Clinic of the Geneva University Hospitals (Day 0). A safety questionnaire regarding potential AEs was emailed to FLWs on Days 3 and 21. Early and delayed AEs were those starting within 3 or 21 days of vaccination, respectively. RESULTS Between 1 August 2019 and 30 June 2020, 124 FLWs received the rVSV-ZEBOV vaccine. Eighty-six volunteers (86/124; 69%) received a concomitant vaccine. The response rate to the follow-up questionnaire was 88 and 55% at Days 3 and 21, respectively. Most respondents (105/109; 96.3%), experienced at least one AE, with a mean of three (±SD 1.75) AEs per person. The most common AE was injection site pain, followed by fever (53/109; 48.6%), fatigue (51/109; 46.7%) and myalgia (49/109; 44.9%). Most early AEs (360/377; 95.4%) resolved within 3 days, reflecting vaccine reactogenicity. Delayed AEs were reported by 6/69 (7.2%) subjects, the median time to symptom onset was 11 days (range: 5-14); half of them were joint-related AEs (3/6). Four serious adverse events (SAE) were observed: two cases of high grade fever, one rash and one case of arthritis. Two suspected unexpected serious adverse reactions were observed: one case of continuing recurrent transient dizziness and fatigue considered related to the vaccine; and one case of presbyopia that was deemed unrelated. CONCLUSION AEs to rVSV-ZEBOV were common but in general transient and were well tolerated, pre-deployment rVSV-ZEBOV vaccination in FLW is feasible and can be included with pre-mission check-up.
Collapse
Affiliation(s)
- Luisa Carnino
- Division of Tropical and Humanitarian Medicine, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland.,University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland
| | - Pauline Vetter
- University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland.,Geneva Center for Emerging Viral Diseases, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva 1205, Switzerland.,Division of Infectious Diseases, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland
| | | | - Sigiriya Aebischer-Perone
- Division of Tropical and Humanitarian Medicine, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland.,Health Unit, International Committee of the Red Cross (ICRC), Geneva 1205, Switzerland
| | - François Chappuis
- Division of Tropical and Humanitarian Medicine, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland.,University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland
| | - Angela Huttner
- University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland.,Division of Infectious Diseases, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland.,Center for Vaccinology, University and University Hospitals of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland
| | - Laurent Kaiser
- University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland.,Geneva Center for Emerging Viral Diseases, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 4, Geneva 1205, Switzerland.,Division of Infectious Diseases, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland
| | - Gilles Eperon
- Division of Tropical and Humanitarian Medicine, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, Geneva 1205, Switzerland.,University of Geneva, Rue Michel-Servet 1, Geneva 1205, Switzerland
| |
Collapse
|
23
|
Pinski AN, Messaoudi I. Therapeutic vaccination strategies against EBOV by rVSV-EBOV-GP: the role of innate immunity. Curr Opin Virol 2021; 51:179-189. [PMID: 34749265 DOI: 10.1016/j.coviro.2021.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022]
Abstract
Zaire Ebola virus (EBOV) is a member of the Filoviridae family. Infection with EBOV causes Ebola virus disease (EVD) characterized by excessive inflammation, lymphocyte death, coagulopathy, and multi-organ failure. In 2019, the FDA-approved the first anti-EBOV vaccine, rVSV-EBOV-GP (Ervebo® by Merck). This live-recombinant vaccine confers both prophylactic and therapeutic protection to nonhuman primates and humans. While mechanisms conferring prophylactic protection are well-investigated, those underlying protection conferred shortly before and after exposure to EBOV remain poorly understood. In this review, we review data from in vitro and in vivo studies analyzing early immune responses to rVSV-EBOV-GP and discuss the role of innate immune activation in therapeutic protection.
Collapse
Affiliation(s)
- Amanda N Pinski
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
24
|
Gunn BM, Bai S. Building a better antibody through the Fc: advances and challenges in harnessing antibody Fc effector functions for antiviral protection. Hum Vaccin Immunother 2021; 17:4328-4344. [PMID: 34613865 PMCID: PMC8827636 DOI: 10.1080/21645515.2021.1976580] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Antibodies can provide antiviral protection through neutralization and recruitment of innate effector functions through the Fc domain. While neutralization has long been appreciated for its role in antibody-mediated protection, a growing body of work indicates that the antibody Fc domain also significantly contributes to antiviral protection. Recruitment of innate immune cells such as natural killer cells, neutrophils, monocytes, macrophages, dendritic cells and the complement system by antibodies can lead to direct restriction of viral infection as well as promoting long-term antiviral immunity. Monoclonal antibody therapeutics against viruses are increasingly incorporating Fc-enhancing features to take advantage of the Fc domain, uncovering a surprising breadth of mechanisms through which antibodies can control viral infection. Here, we review the recent advances in our understanding of antibody-mediated innate immune effector functions in protection from viral infection and review the current approaches and challenges to effectively leverage innate immune cells via antibodies.
Collapse
Affiliation(s)
- Bronwyn M. Gunn
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Shuangyi Bai
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
25
|
Marzi A, Jankeel A, Menicucci AR, Callison J, O’Donnell KL, Feldmann F, Pinski AN, Hanley PW, Messaoudi I. Single Dose of a VSV-Based Vaccine Rapidly Protects Macaques From Marburg Virus Disease. Front Immunol 2021; 12:774026. [PMID: 34777392 PMCID: PMC8578864 DOI: 10.3389/fimmu.2021.774026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Marburg virus (MARV) is a member of the filovirus family that causes hemorrhagic disease with high case fatality rates. MARV is on the priority list of the World Health Organization for countermeasure development highlighting its potential impact on global public health. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the MARV glycoprotein (VSV-MARV) and previously demonstrated uniform protection of nonhuman primates (NHPs) with a single dose. Here, we investigated the fast-acting potential of this vaccine by challenging NHPs with MARV 14, 7 or 3 days after a single dose vaccination with VSV-MARV. We found that 100% of the animals survived when vaccinated 7 or 14 days and 75% of the animal survived when vaccinated 3 days prior to lethal MARV challenge. Transcriptional analysis of whole blood samples indicated activation of B cells and antiviral defense after VSV-MARV vaccination. In the day -14 and -7 groups, limited transcriptional changes after challenge were observed with the exception of day 9 post-challenge in the day -7 group where we detected gene expression profiles indicative of a recall response. In the day -3 group, transcriptional analysis of samples from surviving NHPs revealed strong innate immune activation. In contrast, the animal that succumbed to disease in this group lacked signatures of antiviral immunity. In summary, our data demonstrate that the VSV-MARV is a fast-acting vaccine suitable for the use in emergency situations like disease outbreaks in Africa.
Collapse
Affiliation(s)
- Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Andrea R. Menicucci
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Kyle L. O’Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Amanda N. Pinski
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
26
|
SARS-CoV-2 immune repertoire in MIS-C and pediatric COVID-19. Nat Immunol 2021; 22:1452-1464. [PMID: 34611361 DOI: 10.1038/s41590-021-01051-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023]
Abstract
There is limited understanding of the viral antibody fingerprint following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children. Herein, SARS-CoV-2 proteome-wide immunoprofiling of children with mild/moderate or severe coronavirus disease 2019 (COVID-19) versus multisystem inflammatory syndrome in children versus hospitalized control patients revealed differential cytokine responses, IgM/IgG/IgA epitope diversity, antibody binding and avidity. Apart from spike and nucleocapsid, IgG/IgA recognized epitopes in nonstructural protein (NSP) 2, NSP3, NSP12-NSP14 and open reading frame (ORF) 3a-ORF9. Peptides representing epitopes in NSP12, ORF3a and ORF8 demonstrated SARS-CoV-2 serodiagnosis. Antibody-binding kinetics with 24 SARS-CoV-2 proteins revealed antibody parameters that distinguish children with mild/moderate versus severe COVID-19 or multisystem inflammatory syndrome in children. Antibody avidity to prefusion spike correlated with decreased illness severity and served as a clinical disease indicator. The fusion peptide and heptad repeat 2 region induced SARS-CoV-2-neutralizing antibodies in rabbits. Thus, we identified SARS-CoV-2 antibody signatures in children associated with disease severity and delineate promising serodiagnostic and virus neutralization targets. These findings might guide the design of serodiagnostic assays, prognostic algorithms, therapeutics and vaccines in this important but understudied population.
Collapse
|
27
|
Hargreaves A, Brady C, Mellors J, Tipton T, Carroll MW, Longet S. Filovirus Neutralising Antibodies: Mechanisms of Action and Therapeutic Application. Pathogens 2021; 10:pathogens10091201. [PMID: 34578233 PMCID: PMC8468515 DOI: 10.3390/pathogens10091201] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/02/2022] Open
Abstract
Filoviruses, especially Ebola virus, cause sporadic outbreaks of viral haemorrhagic fever with very high case fatality rates in Africa. The 2013–2016 Ebola epidemic in West Africa provided large survivor cohorts spurring a large number of human studies which showed that specific neutralising antibodies played a key role in protection following a natural Ebola virus infection, as part of the overall humoral response and in conjunction with the cellular adaptive response. This review will discuss the studies in survivors and animal models which described protective neutralising antibody response. Their mechanisms of action will be detailed. Furthermore, the importance of neutralising antibodies in antibody-based therapeutics and in vaccine-induced responses will be explained, as well as the strategies to avoid immune escape from neutralising antibodies. Understanding the neutralising antibody response in the context of filoviruses is crucial to furthering our understanding of virus structure and function, in addition to improving current vaccines & antibody-based therapeutics.
Collapse
Affiliation(s)
- Alexander Hargreaves
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Caolann Brady
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Jack Mellors
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7ZX, UK
| | - Tom Tipton
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Miles W. Carroll
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Stephanie Longet
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Correspondence: ; Tel.: +44-18-6561-7892
| |
Collapse
|
28
|
Liu G, Cao W, Salawudeen A, Zhu W, Emeterio K, Safronetz D, Banadyga L. Vesicular Stomatitis Virus: From Agricultural Pathogen to Vaccine Vector. Pathogens 2021; 10:1092. [PMID: 34578125 PMCID: PMC8470541 DOI: 10.3390/pathogens10091092] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Vesicular stomatitis virus (VSV), which belongs to the Vesiculovirus genus of the family Rhabdoviridae, is a well studied livestock pathogen and prototypic non-segmented, negative-sense RNA virus. Although VSV is responsible for causing economically significant outbreaks of vesicular stomatitis in cattle, horses, and swine, the virus also represents a valuable research tool for molecular biologists and virologists. Indeed, the establishment of a reverse genetics system for the recovery of infectious VSV from cDNA transformed the utility of this virus and paved the way for its use as a vaccine vector. A highly effective VSV-based vaccine against Ebola virus recently received clinical approval, and many other VSV-based vaccines have been developed, particularly for high-consequence viruses. This review seeks to provide a holistic but concise overview of VSV, covering the virus's ascension from perennial agricultural scourge to promising medical countermeasure, with a particular focus on vaccines.
Collapse
Affiliation(s)
- Guodong Liu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Wenguang Cao
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Abdjeleel Salawudeen
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Wenjun Zhu
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada
| | - Karla Emeterio
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - David Safronetz
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| |
Collapse
|
29
|
Grenache DG, Ye C, Bradfute SB. Correlation of SARS-CoV-2 Neutralizing Antibodies to an Automated Chemiluminescent Serological Immunoassay. J Appl Lab Med 2021; 6:491-495. [PMID: 33098417 PMCID: PMC7665545 DOI: 10.1093/jalm/jfaa195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/06/2020] [Indexed: 01/05/2023]
Abstract
Introduction Neutralizing antibodies (NAbs) are capable of binding to a virus to render incapable of infection. The ability of commercially available SARS-CoV-2 serological tests to detect NAbs has not been widely reported. We sought to correlate the antibodies detected by an automated chemiluminescent immunoassay with NAbs. Methods Residual serum samples from 35 patients that had a positive antibody test using the LIAISON® SARS-CoV-2 S1/S2 IgG chemiluminescent immunoassay and two antibody-negative control sera were tested for NAbs using a plaque reduction neutralization test (PRNT). Results NAbs were detected in 66% (23/35) of the antibody-positive samples. The immunoassay signal value ranged from 21.7 to 131.3 AU/mL (median, 90.5) with significant correlation between it and the PRNT (r = 0.61, p = 0.002). In the samples without NAbs, the immunoassay signal ranged from 16.3 to 66.2 AU/mL (median, 27.2). An immunoassay signal cutoff of > 41 AU/mL was 91% sensitive and 92% specific for the detection of NAbs. Discussion It is important that correlates of immunity to SARS-CoV-2 be identified and NAbs are considered to be central indicators of such. PRNT is the gold-standard test for identifying NAbs but it cannot be used for large-scale testing of populations. It is necessary to establish relationships between it and widely used commercial serological assays for SARS-CoV-2.
Collapse
Affiliation(s)
- David G Grenache
- TriCore Reference Laboratories, Albuquerque, NM.,Department of Pathology, University of New Mexico, Albuquerque, NM
| | - Chunyan Ye
- Department of Internal Medicine, University of New Mexico, Center for Global Health, Albuquerque, NM
| | - Steven B Bradfute
- Department of Internal Medicine, University of New Mexico, Center for Global Health, Albuquerque, NM
| |
Collapse
|
30
|
Ravichandran S, Lee Y, Grubbs G, Coyle EM, Klenow L, Akasaka O, Koga M, Adachi E, Saito M, Nakachi I, Ogura T, Baba R, Ito M, Kiso M, Yasuhara A, Yamada S, Sakai-Tagawa Y, Iwatsuki-Horimoto K, Imai M, Yamayoshi S, Yotsuyanagi H, Kawaoka Y, Khurana S. Longitudinal antibody repertoire in "mild" versus "severe" COVID-19 patients reveals immune markers associated with disease severity and resolution. SCIENCE ADVANCES 2021; 7:7/10/eabf2467. [PMID: 33674317 PMCID: PMC7935365 DOI: 10.1126/sciadv.abf2467] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/21/2021] [Indexed: 05/05/2023]
Abstract
Limited knowledge exists on immune markers associated with disease severity or recovery in patients with coronavirus disease 2019 (COVID-19). Here, we elucidated longitudinal evolution of SARS-CoV-2 antibody repertoire in patients with acute COVID-19. Differential kinetics was observed for immunoglobulin M (IgM)/IgG/IgA epitope diversity, antibody binding, and affinity maturation in "severe" versus "mild" COVID-19 patients. IgG profile demonstrated immunodominant antigenic sequences encompassing fusion peptide and receptor binding domain (RBD) in patients with mild COVID-19 who recovered early compared with "fatal" COVID-19 patients. In patients with severe COVID-19, high-titer IgA were observed, primarily against RBD, especially in patients who succumbed to SARS-CoV-2 infection. The patients with mild COVID-19 showed marked increase in antibody affinity maturation to prefusion SARS-CoV-2 spike that associated with faster recovery from COVID-19. This study revealed antibody markers associated with disease severity and resolution of clinical disease that could inform development and evaluation of effective immune-based countermeasures against COVID-19.
Collapse
Affiliation(s)
- Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Youri Lee
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Osamu Akasaka
- Emergency Medical Center, Fujisawa City Hospital 2-6-1 Fujisawa, Fujisawa City, Kanagawa 251-8550, Japan
| | - Michiko Koga
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Eisuke Adachi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Makoto Saito
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ichiro Nakachi
- Pulmonary Division, Department of Internal Medicine, Saiseikai Utsunomiya Hospital 911-1 Takebayashimachi, Utsunomiya 321-0974, Japan
| | - Takayuki Ogura
- Department of Emergency & Intensive care, Saiseikai Utsunomiya Hospital 911-1 Takebayashimachi, Utsunomiya 321-0974, Japan
| | - Rie Baba
- Pulmonary Division, Department of Internal Medicine, Saiseikai Utsunomiya Hospital 911-1 Takebayashimachi, Utsunomiya 321-0974, Japan
| | - Mutsumi Ito
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Maki Kiso
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Atsuhiro Yasuhara
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Shinya Yamada
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yuko Sakai-Tagawa
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kiyoko Iwatsuki-Horimoto
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Masaki Imai
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seiya Yamayoshi
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hiroshi Yotsuyanagi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA.
| |
Collapse
|
31
|
Tang J, Ravichandran S, Lee Y, Grubbs G, Coyle EM, Klenow L, Genser H, Golding H, Khurana S. Antibody affinity maturation and plasma IgA associate with clinical outcome in hospitalized COVID-19 patients. Nat Commun 2021; 12:1221. [PMID: 33619281 PMCID: PMC7900119 DOI: 10.1038/s41467-021-21463-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/27/2021] [Indexed: 01/10/2023] Open
Abstract
Hospitalized COVID-19 patients often present with a large spectrum of clinical symptoms. There is a critical need to better understand the immune responses to SARS-CoV-2 that lead to either resolution or exacerbation of the clinical disease. Here, we examine longitudinal plasma samples from hospitalized COVID-19 patients with differential clinical outcome. We perform immune-repertoire analysis including cytokine, hACE2-receptor inhibition, neutralization titers, antibody epitope repertoire, antibody kinetics, antibody isotype and antibody affinity maturation against the SARS-CoV-2 prefusion spike protein. Fatal cases demonstrate high plasma levels of IL-6, IL-8, TNFα, and MCP-1, and sustained high percentage of IgA-binding antibodies to prefusion spike compared with non-ICU survivors. Disease resolution in non-ICU and ICU patients associates with antibody binding to the receptor binding motif and fusion peptide, and antibody affinity maturation to SARS-CoV-2 prefusion spike protein. Here, we provide insight into the immune parameters associated with clinical disease severity and disease-resolution outcome in hospitalized patients that could inform development of vaccine/therapeutics against COVID-19. SARS-CoV2 infection has been linked to a wide range of clinical severities and the immunopathology is still under intense scrutiny. Here, the authors uncover an association of antibody affinity maturation and plasma IgA levels with clinical outcome in patients with COVID-19 disease.
Collapse
Affiliation(s)
- Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Youri Lee
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Hollie Genser
- Quest Diagnostics at Adventist Healthcare, Rockville, MD, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, USA.
| |
Collapse
|
32
|
Heiss K, Heidepriem J, Fischer N, Weber LK, Dahlke C, Jaenisch T, Loeffler FF. Rapid Response to Pandemic Threats: Immunogenic Epitope Detection of Pandemic Pathogens for Diagnostics and Vaccine Development Using Peptide Microarrays. J Proteome Res 2020; 19:4339-4354. [PMID: 32892628 PMCID: PMC7640972 DOI: 10.1021/acs.jproteome.0c00484] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Emergence and re-emergence of pathogens bearing the risk of becoming a pandemic threat are on the rise. Increased travel and trade, growing population density, changes in urbanization, and climate have a critical impact on infectious disease spread. Currently, the world is confronted with the emergence of a novel coronavirus SARS-CoV-2, responsible for yet more than 800 000 deaths globally. Outbreaks caused by viruses, such as SARS-CoV-2, HIV, Ebola, influenza, and Zika, have increased over the past decade, underlining the need for a rapid development of diagnostics and vaccines. Hence, the rational identification of biomarkers for diagnostic measures on the one hand, and antigenic targets for vaccine development on the other, are of utmost importance. Peptide microarrays can display large numbers of putative target proteins translated into overlapping linear (and cyclic) peptides for a multiplexed, high-throughput antibody analysis. This enabled for example the identification of discriminant/diagnostic epitopes in Zika or influenza and mapping epitope evolution in natural infections versus vaccinations. In this review, we highlight synthesis platforms that facilitate fast and flexible generation of high-density peptide microarrays. We further outline the multifaceted applications of these peptide array platforms for the development of serological tests and vaccines to quickly encounter pandemic threats.
Collapse
Affiliation(s)
- Kirsten Heiss
- PEPperPRINT
GmbH, Rischerstrasse
12, 69123 Heidelberg, Germany
| | - Jasmin Heidepriem
- Max
Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Muehlenberg 1, 14476 Potsdam, Germany
| | - Nico Fischer
- Section
Clinical Tropical Medicine, Department of Infectious Diseases, Heidelberg University Hospital, INF 324, 69120 Heidelberg, Germany
| | - Laura K. Weber
- PEPperPRINT
GmbH, Rischerstrasse
12, 69123 Heidelberg, Germany
- Institute
of Microstructure Technology, Karlsruhe
Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Christine Dahlke
- Division
of Infectious Diseases, First Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department
of Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German
Center for Infection Research, Partner Site
Hamburg-Lübeck-Borstel-Riems, 38124 Braunschweig, Germany
| | - Thomas Jaenisch
- Heidelberg
Institute of Global Health (HIGH), Heidelberg
University Hospital, Im Neuenheimer Feld 130, 69120 Heidelberg, Germany
- Center
for Global Health, Colorado School of Public Health, University of Colorado, Aurora, Colorado 80045, United States
- Department
of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado 80045, United States
| | - Felix F. Loeffler
- Max
Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Muehlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
33
|
Niemuth NA, Rudge TL, Sankovich KA, Anderson MS, Skomrock ND, Badorrek CS, Sabourin CL. Method feasibility for cross-species testing, qualification, and validation of the Filovirus Animal Nonclinical Group anti-Ebola virus glycoprotein immunoglobulin G enzyme-linked immunosorbent assay for non-human primate serum samples. PLoS One 2020; 15:e0241016. [PMID: 33119638 PMCID: PMC7595334 DOI: 10.1371/journal.pone.0241016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
An anti-Zaire Ebola virus (EBOV) glycoprotein (GP) immunoglobulin G (IgG) enzyme linked immunosorbent assay (ELISA) was developed to quantify the serum levels of anti-EBOV IgG in human and non-human primate (NHP) serum following vaccination and/or exposure to EBOV. This method was validated for testing human serum samples as previously reported. However, for direct immunobridging comparability between humans and NHPs, additional testing was warranted. First, method feasibility experiments were performed to assess cross-species reactivity and parallelism between human and NHP serum samples. During these preliminary assessments, the goat anti-human IgG secondary antibody conjugate used in the previous human validation was found to be favorably cross-reactive with NHP samples when tested at the same concentrations previously used in the validated assay for human sample testing. Further, NHP serum samples diluted in parallel with human serum when tested side-by-side in the ELISA. A subsequent NHP matrix qualification and partial validation in the anti-GP IgG ELISA were performed based on ICH and FDA guidance, to characterize assay performance for NHP test samples and supplement the previous validation for human sample testing. Based on our assessments, the anti-EBOV GP IgG ELISA method is considered suitable for the intended use of testing with both human and NHP serum samples in the same assay for immunobridging purposes.
Collapse
Affiliation(s)
- Nancy A. Niemuth
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
- * E-mail:
| | - Thomas L. Rudge
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| | - Karen A. Sankovich
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| | - Michael S. Anderson
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| | - Nicholas D. Skomrock
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| | - Christopher S. Badorrek
- Contract Support for the U.S. Department of Defense (DOD) Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense (JPEO-CBRND) Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, Maryland, United States of America
| | - Carol L. Sabourin
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| |
Collapse
|
34
|
Scher G, Schnell MJ. Rhabdoviruses as vectors for vaccines and therapeutics. Curr Opin Virol 2020; 44:169-182. [PMID: 33130500 PMCID: PMC8331071 DOI: 10.1016/j.coviro.2020.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/12/2020] [Accepted: 09/13/2020] [Indexed: 12/24/2022]
Abstract
Appropriate choice of vaccine vector is crucial for effective vaccine development. Rhabdoviral vectors, such as rabies virus and vesicular stomatitis virus, have been used in a variety of vaccine strategies. These viruses have small, easily manipulated genomes that can stably express foreign glycoproteins due to a well-established reverse genetics system for virus recovery. Both viruses have well-described safety profiles and have been demonstrated to be effective vaccine vectors. This review will describe how these Rhabdoviruses can be manipulated for use as vectors, their various applications as vaccines or therapeutics, and the advantages and disadvantages of their use.
Collapse
Affiliation(s)
- Gabrielle Scher
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA; Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
35
|
To B or Not to B: Mechanisms of Protection Conferred by rVSV-EBOV-GP and the Roles of Innate and Adaptive Immunity. Microorganisms 2020; 8:microorganisms8101473. [PMID: 32992829 PMCID: PMC7600878 DOI: 10.3390/microorganisms8101473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/28/2022] Open
Abstract
Zaire Ebola virus (EBOV) is a member of the Filoviridae family of negative sense, single-stranded RNA viruses. EBOV infection causes Ebola virus disease (EVD), characterized by coagulopathy, lymphopenia, and multi-organ failure, which can culminate in death. In 2019, the FDA approved the first vaccine against EBOV, a recombinant live-attenuated viral vector wherein the G protein of vesicular stomatitis virus is replaced with the glycoprotein (GP) of EBOV (rVSV-EBOV-GP, Ervebo® by Merck). This vaccine demonstrates high efficacy in nonhuman primates by providing prophylactic, rapid, and post-exposure protection. In humans, rVSV-EBOV-GP demonstrated 100% protection in several phase III clinical trials in over 10,000 individuals during the 2013–2016 West Africa epidemic. As of 2020, over 218,000 doses of rVSV-EBOV-GP have been administered to individuals with high risk of EBOV exposure. Despite licensure and robust preclinical studies, the mechanisms of rVSV-EBOV-GP-mediated protection are not fully understood. Such knowledge is crucial for understanding vaccine-mediated correlates of protection from EVD and to aid the further design and development of therapeutics against filoviruses. Here, we summarize the current literature regarding the host response to vaccination and EBOV exposure, and evidence regarding innate and adaptive immune mechanisms involved in rVSV-EBOV-GP-mediated protection, with a focus on the host transcriptional response. Current data strongly suggest a protective synergy between rapid innate and humoral immunity.
Collapse
|
36
|
Ebola Virus Disease Survivors Show More Efficient Antibody Immunity than Vaccinees Despite Similar Levels of Circulating Immunoglobulins. Viruses 2020; 12:v12090915. [PMID: 32825479 PMCID: PMC7552031 DOI: 10.3390/v12090915] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
The last seven years have seen the greatest surge of Ebola virus disease (EVD) cases in equatorial Africa, including the 2013-2016 epidemic in West Africa and the recent epidemics in the Democratic Republic of Congo (DRC). The vaccine clinical trials that took place in West Africa and the DRC, as well as follow-up studies in collaboration with EVD survivor communities, have for the first time allowed researchers to compare immune memory induced by natural infection and vaccination. These comparisons may be relevant to evaluate the putative effectiveness of vaccines and candidate medical countermeasures such as convalescent plasma transfer. In this study, we compared the long-term functionality of anti-EBOV glycoprotein (GP) antibodies from EVD survivors with that from volunteers who received the recombinant vesicular stomatitis virus vectored vaccine (rVSV-ZEBOV) during the Phase I clinical trial in Hamburg. Our study highlights important differences between EBOV vaccination and natural infection and provides a framework for comparison with other vaccine candidates.
Collapse
|
37
|
Gunn BM, Roy V, Karim MM, Hartnett JN, Suscovich TJ, Goba A, Momoh M, Sandi JD, Kanneh L, Andersen KG, Shaffer JG, Schieffelin JS, Garry RF, Grant DS, Alter G. Survivors of Ebola Virus Disease Develop Polyfunctional Antibody Responses. J Infect Dis 2020; 221:156-161. [PMID: 31301137 PMCID: PMC7184900 DOI: 10.1093/infdis/jiz364] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/11/2019] [Indexed: 11/21/2022] Open
Abstract
Monoclonal antibodies can mediate protection against Ebola virus (EBOV) infection through direct neutralization as well as through the recruitment of innate immune effector functions. However, the antibody functional response following survival of acute EBOV disease has not been well characterized. In this study, serum antibodies from Ebola virus disease (EVD) survivors from Sierra Leone were profiled to capture variation in overall subclass/isotype abundance, neutralizing activity, and innate immune effector functions. Antibodies from EVD survivors exhibited robust innate immune effector functions, mediated primarily by IgG1 and IgA1. In conclusion, development of functional antibodies follows survival of acute EVD.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
| | - Marcus M Karim
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
| | - Jessica N Hartnett
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Todd J Suscovich
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital.,Eastern Polytechnic University, Kenema, Sierra Leone
| | | | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital
| | - Kristian G Andersen
- Department of Immunology and Microbiology, Scripps Research Institute.,Scripps Research Translational Institute, La Jolla, California
| | - Jeffrey G Shaffer
- Department of Biostatistics and Bioinformatics, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - John S Schieffelin
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Donald S Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital.,Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts
| |
Collapse
|
38
|
Choi Y, Jeong S, Choi JM, Ndong C, Griswold KE, Bailey-Kellogg C, Kim HS. Computer-guided binding mode identification and affinity improvement of an LRR protein binder without structure determination. PLoS Comput Biol 2020; 16:e1008150. [PMID: 32866140 PMCID: PMC7485979 DOI: 10.1371/journal.pcbi.1008150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/11/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
Precise binding mode identification and subsequent affinity improvement without structure determination remain a challenge in the development of therapeutic proteins. However, relevant experimental techniques are generally quite costly, and purely computational methods have been unreliable. Here, we show that integrated computational and experimental epitope localization followed by full-atom energy minimization can yield an accurate complex model structure which ultimately enables effective affinity improvement and redesign of binding specificity. As proof-of-concept, we used a leucine-rich repeat (LRR) protein binder, called a repebody (Rb), that specifically recognizes human IgG1 (hIgG1). We performed computationally-guided identification of the Rb:hIgG1 binding mode and leveraged the resulting model to reengineer the Rb so as to significantly increase its binding affinity for hIgG1 as well as redesign its specificity toward multiple IgGs from other species. Experimental structure determination verified that our Rb:hIgG1 model closely matched the co-crystal structure. Using a benchmark of other LRR protein complexes, we further demonstrated that the present approach may be broadly applicable to proteins undergoing relatively small conformational changes upon target binding.
Collapse
Affiliation(s)
- Yoonjoo Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Sukyo Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Jung-Min Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Christian Ndong
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Karl E. Griswold
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center at Dartmouth, Lebanon, New Hampshire, United States of America
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Chris Bailey-Kellogg
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| |
Collapse
|
39
|
Matz KM, Marzi A, Feldmann H. Ebola vaccine trials: progress in vaccine safety and immunogenicity. Expert Rev Vaccines 2020; 18:1229-1242. [PMID: 31779496 DOI: 10.1080/14760584.2019.1698952] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: Ebolaviruses are non-segmented negative-strand RNA viruses in the Filoviridae family that cause a neglected infectious disease designated as Ebola virus disease (EVD). The most prominent member is the Ebola virus (EBOV), representing the Zaire ebolavirus species that has been responsible for the largest reported EVD outbreaks including the West African epidemic and the current outbreak in the Democratic Republic of the Congo. Today, the most advanced EVD vaccine approaches target EBOV and multiple phase 1-4 human trials have been performed over the past few years. The most advanced platforms include vectored vaccines based on vesicular stomatitis virus (VSV-EBOV), distinct human (Ad5 and Ad26) and chimpanzee (ChAd3) adenoviruses and modified vaccinia Ankara (MVA) as well as DNA-based vaccines administered as a prime-only or homologous or combined prime-boost immunization.Areas covered: Here, we review and discuss human trials with a focus on vaccine safety and immunogenicity.Expert opinion: Despite obvious progress and promising success in EBOV vaccine development, many shortcomings and challenges remain to be tackled in the future.
Collapse
Affiliation(s)
- Keesha M Matz
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| |
Collapse
|
40
|
Ravichandran S, Coyle EM, Klenow L, Tang J, Grubbs G, Liu S, Wang T, Golding H, Khurana S. Antibody signature induced by SARS-CoV-2 spike protein immunogens in rabbits. Sci Transl Med 2020; 12:scitranslmed.abc3539. [PMID: 32513867 PMCID: PMC7286538 DOI: 10.1126/scitranslmed.abc3539] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Multiple vaccine candidates against SARS-CoV-2 based on viral spike protein are under development. However, there is limited information on the quality of antibody responses generated with these vaccine modalities. To better understand antibody responses induced by spike protein-based vaccines, we performed a qualitative study by immunizing rabbits with various SARS-CoV-2 spike protein antigens: S ectodomain (S1+S2; amino acids 16 to 1213), which lacks the cytoplasmic and transmembrane domains (CT-TM), the S1 domain (amino acids 16 to 685), the receptor binding domain (RBD) (amino acids 319 to 541), and the S2 domain (amino acids 686 to 1213, lacking the RBD, as control). Resulting antibody quality and function were analyzed by enzyme-linked immunosorbent assay (ELISA), RBD competition assay, surface plasmon resonance (SPR) against different spike proteins in native conformation, and neutralization assays. All three antigens (S1+S2 ectodomain, S1 domain, and RBD), but not S2, generated strong neutralizing antibodies against SARS-CoV-2. Vaccination-induced antibody repertoire was analyzed by SARS-CoV-2 spike genome fragment phage display libraries (SARS-CoV-2 GFPDL), which identified immunodominant epitopes in the S1, S1-RBD, and S2 domains. Furthermore, these analyses demonstrated that the RBD immunogen elicited a higher antibody titer with five-fold higher affinity antibodies to native spike antigens compared with other spike antigens, and antibody affinity correlated strongly with neutralization titers. These findings may help guide rational vaccine design and facilitate development and evaluation of effective therapeutics and vaccines against COVID-19 disease.
Collapse
Affiliation(s)
- Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Shufeng Liu
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Tony Wang
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA.
| |
Collapse
|
41
|
A Bivalent, Spherical Virus-Like Particle Vaccine Enhances Breadth of Immune Responses against Pathogenic Ebola Viruses in Rhesus Macaques. J Virol 2020; 94:JVI.01884-19. [PMID: 32075939 DOI: 10.1128/jvi.01884-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/13/2020] [Indexed: 12/16/2022] Open
Abstract
The 2013-2016 Ebola outbreak in West Africa led to accelerated efforts to develop vaccines against these highly virulent viruses. A live, recombinant vesicular stomatitis virus-based vaccine has been deployed in outbreak settings and appears highly effective. Vaccines based on replication-deficient adenovirus vectors either alone or in combination with a multivalent modified vaccinia Ankara (MVA) Ebola vaccine also appear promising and are progressing in clinical evaluation. However, the ability of current live vector-based approaches to protect against multiple pathogenic species of Ebola is not yet established, and eliciting durable responses may require additional booster vaccinations. Here, we report the development of a bivalent, spherical Ebola virus-like particle (VLP) vaccine that incorporates glycoproteins (GPs) from Zaire Ebola virus (EBOV) and Sudan Ebola virus (SUDV) and is designed to extend the breadth of immunity beyond EBOV. Immunization of rabbits with bivalent Ebola VLPs produced antibodies that neutralized all four pathogenic species of Ebola viruses and elicited antibody-dependent cell-mediated cytotoxicity (ADCC) responses against EBOV and SUDV. Vaccination of rhesus macaques with bivalent VLPs generated strong humoral immune responses, including high titers of binding, as well as neutralizing antibodies and ADCC responses. VLP vaccination led to a significant increase in the frequency of Ebola GP-specific CD4 and CD8 T cell responses. These results demonstrate that a novel bivalent Ebola VLP vaccine elicits strong humoral and cellular immune responses against pathogenic Ebola viruses and support further evaluation of this approach as a potential addition to Ebola vaccine development efforts.IMPORTANCE Ebola outbreaks result in significant morbidity and mortality in affected countries. Although several leading candidate Ebola vaccines have been developed and advanced in clinical testing, additional vaccine candidates may be needed to provide protection against different Ebola species and to extend the durability of protection. A novel approach demonstrated here is to express two genetically diverse glycoproteins on a spherical core, generating a vaccine that can broaden immune responses against known pathogenic Ebola viruses. This approach provides a new method to broaden and potentially extend protective immune responses against Ebola viruses.
Collapse
|
42
|
O'Donnell K, Marzi A. The Ebola virus glycoprotein and its immune responses across multiple vaccine platforms. Expert Rev Vaccines 2020; 19:267-277. [PMID: 32129120 DOI: 10.1080/14760584.2020.1738225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: For over 40 years, ebolaviruses have been responsible for sporadic outbreaks of severe and often fatal hemorrhagic fever in humans and nonhuman primates across western and central Africa. In December 2013, an unprecedented Ebola virus (EBOV) epidemic began in West Africa and resulted in the largest outbreak to date. The past and current epidemics in West Africa and the Democratic Republic of the Congo has focused attention on the potential vaccine platforms developed over the past 20 years.Areas covered: This review summarizes the extraordinary progress using a variety of vaccination platforms including DNA, subunit, and several viral vector approaches, replicating and non-replicating, incorporating the primary antigen of EBOV, the glycoprotein. These vaccine constructs have shown varying degrees of protective efficacy in the 'gold-standard' nonhuman primate model for EBOV infections and were immunogenic in human clinical trials.Expert commentary: A number of these vaccine platforms have moved into phase III clinical trials over the past years and with the recent approval of the first EBOV vaccine in the European Union and the USA there is a strong potential to prevent future outbreaks/epidemics of EBOV infections on the scale of the West African epidemic.
Collapse
Affiliation(s)
- Kyle O'Donnell
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
43
|
Fuentes S, Ravichandran S, Coyle EM, Klenow L, Khurana S. Human Antibody Repertoire following Ebola Virus Infection and Vaccination. iScience 2020; 23:100920. [PMID: 32145646 PMCID: PMC7058406 DOI: 10.1016/j.isci.2020.100920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 01/14/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023] Open
Abstract
Limited knowledge exists on the quality of polyclonal antibody response generated following Ebola virus (EBOV) infection compared with vaccination. Polyclonal antibody repertoire in plasma following EBOV infection in survivors was compared with ChAd3-MVA prime-boost human vaccination. Higher antibody binding and affinity to GP was observed in survivors compared with vaccinated plasma that correlated with EBOV neutralization. Surprisingly, a predominant IgM response was generated after prime-boost vaccination, whereas survivors demonstrated IgG-dominant antibody response. EBOV infection induced more diverse antibody epitope repertoire compared with vaccination. A strong binding to antigenic sites in the fusion peptide and another in the highly conserved GP2-HR2 domain was preferentially recognized by EBOV survivors than vaccinated individuals that correlated strongly with EBOV neutralization titers. These findings will help development and evaluation of effective Ebola countermeasures including therapeutics and vaccines. EBOV infection induced a more diverse antibody repertoire than vaccination Ebola survivors demonstrated long-lasting, high-affinity, IgG antibody response Several novel antigenic sites recognized by post-EBOV infection sera
Collapse
Affiliation(s)
- Sandra Fuentes
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| |
Collapse
|
44
|
Khurana S, Ravichandran S, Hahn M, Coyle EM, Stonier SW, Zak SE, Kindrachuk J, Davey RT, Dye JM, Chertow DS. Longitudinal Human Antibody Repertoire against Complete Viral Proteome from Ebola Virus Survivor Reveals Protective Sites for Vaccine Design. Cell Host Microbe 2020; 27:262-276.e4. [PMID: 32053790 PMCID: PMC7071344 DOI: 10.1016/j.chom.2020.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/14/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022]
Abstract
Evolution of antibody repertoire against the Ebola virus (EBOV) proteome was characterized in an acutely infected patient receiving supportive care alone to elucidate virus-host interactions over time. Differential kinetics are observed for IgM-IgG-IgA epitope diversity, antibody binding, and affinity maturation to EBOV proteins. During acute illness, antibodies predominate to VP40 and glycoprotein (GP). At day 13 of clinical illness, a marked increase in antibody titers to most EBOV proteins and affinity maturation to GP is associated with rapid decline in viral replication and illness severity. At one year, despite undetectable virus, a diverse IgM repertoire against VP40 and GP epitopes is observed suggesting occult viral persistence. Rabbit immunization experiments identify key immunodominant sites of GP, while challenge studies in mice found these epitopes induce EBOV-neutralizing antibodies and protect against lethal EBOV challenge. This study reveals markers of viral persistence and provides promising approaches for development and evaluation of vaccines and therapeutics.
Collapse
Affiliation(s)
- Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA.
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Megan Hahn
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD 20871, USA
| | - Spencer W Stonier
- United States Army, Medical Research Institute of Infectious Diseases, Viral Immunology Branch, Virology Division, Fort Detrick, Frederick, MD 21702, USA
| | - Samantha E Zak
- United States Army, Medical Research Institute of Infectious Diseases, Viral Immunology Branch, Virology Division, Fort Detrick, Frederick, MD 21702, USA
| | - Jason Kindrachuk
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Richard T Davey
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John M Dye
- United States Army, Medical Research Institute of Infectious Diseases, Viral Immunology Branch, Virology Division, Fort Detrick, Frederick, MD 21702, USA
| | - Daniel S Chertow
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
45
|
Sanchez-Lockhart M, Reyes DS, Gonzalez JC, Garcia KY, Villa EC, Pfeffer BP, Trefry JC, Kugelman JR, Pitt ML, Palacios GF. Qualitative Profiling of the Humoral Immune Response Elicited by rVSV-ΔG-EBOV-GP Using a Systems Serology Assay, Domain Programmable Arrays. Cell Rep 2020; 24:1050-1059.e5. [PMID: 30044972 DOI: 10.1016/j.celrep.2018.06.077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 06/18/2018] [Indexed: 01/02/2023] Open
Abstract
Development of an effective vaccine became a worldwide priority after the devastating 2013-2016 Ebola disease outbreak. To qualitatively profile the humoral response against advanced filovirus vaccine candidates, we developed Domain Programmable Arrays (DPA), a systems serology platform to identify epitopes targeted after vaccination or filovirus infection. We optimized the assay using a panel of well-characterized monoclonal antibodies. After optimization, we utilized the system to longitudinally characterize the immunoglobulin (Ig) isotype-specific responses in non-human primates vaccinated with rVSV-ΔG-EBOV-glycoprotein (GP). Strikingly, we observed that, although the IgM response was directed against epitopes over the whole GP, the IgG and IgA responses were almost exclusively directed against the mucin-like domain (MLD) of the glycan cap. Further research will be needed to characterize this possible biased IgG and IgA response toward the MLD, but the results corroborate that DPA is a valuable tool to qualitatively measure the humoral response after vaccination.
Collapse
Affiliation(s)
- Mariano Sanchez-Lockhart
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; Departments of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daniel S Reyes
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; Departments of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jeanette C Gonzalez
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA
| | - Karla Y Garcia
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; Departments of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Erika C Villa
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bradley P Pfeffer
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA
| | - John C Trefry
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA
| | - Jeffrey R Kugelman
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA
| | - Margaret L Pitt
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA
| | - Gustavo F Palacios
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
46
|
Poetsch JH, Dahlke C, Zinser ME, Kasonta R, Lunemann S, Rechtien A, Ly ML, Stubbe HC, Krähling V, Biedenkopf N, Eickmann M, Fehling SK, Olearo F, Strecker T, Sharma P, Lang KS, Lohse AW, Schmiedel S, Becker S, Addo MM. Detectable Vesicular Stomatitis Virus (VSV)-Specific Humoral and Cellular Immune Responses Following VSV-Ebola Virus Vaccination in Humans. J Infect Dis 2019; 219:556-561. [PMID: 30452666 PMCID: PMC6350948 DOI: 10.1093/infdis/jiy565] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/13/2018] [Indexed: 01/16/2023] Open
Abstract
In response to the Ebola virus (EBOV) crisis of 2013–2016, a recombinant vesicular stomatitis virus (VSV)–based EBOV vaccine was clinically tested (NCT02283099). A single-dose regimen of VSV-EBOV revealed a safe and immunogenic profile and demonstrated clinical efficacy. While EBOV-specific immune responses to this candidate vaccine have previously been investigated, limited human data on immunity to the VSV vector are available. Within the scope of a phase 1 study, we performed a comprehensive longitudinal analysis of adaptive immune responses to internal VSV proteins following VSV-EBOV immunization. While no preexisting immunity to the vector was observed, more than one-third of subjects developed VSV-specific cytotoxic T-lymphocyte responses and antibodies.
Collapse
Affiliation(s)
- Joseph H Poetsch
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Christine Dahlke
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Madeleine E Zinser
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Rahel Kasonta
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems
| | - Sebastian Lunemann
- Leibniz Institute for Experimental Virology, Heinrich Pette Institute, Hamburg
| | - Anne Rechtien
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf.,Leibniz Institute for Experimental Virology, Heinrich Pette Institute, Hamburg
| | - My L Ly
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Hans C Stubbe
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, Department of Medicine II, Ludwig Maximilian University, Munich
| | - Verena Krähling
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Nadine Biedenkopf
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Markus Eickmann
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Sarah K Fehling
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Flaminia Olearo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Thomas Strecker
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Piyush Sharma
- Institute of Immunology, University Duisburg-Essen, Essen, Germany
| | - Karl S Lang
- Institute of Immunology, University Duisburg-Essen, Essen, Germany
| | - Ansgar W Lohse
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems
| | - Stefan Schmiedel
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Stephan Becker
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | | | - Marylyn M Addo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| |
Collapse
|
47
|
Suschak JJ, Schmaljohn CS. Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Hum Vaccin Immunother 2019; 15:2359-2377. [PMID: 31589088 DOI: 10.1080/21645515.2019.1651140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The filoviruses Ebola virus and Marburg virus are among the most dangerous pathogens in the world. Both viruses cause viral hemorrhagic fever, with case fatality rates of up to 90%. Historically, filovirus outbreaks had been relatively small, with only a few hundred cases reported. However, the recent West African Ebola virus outbreak underscored the threat that filoviruses pose. The three year-long outbreak resulted in 28,646 Ebola virus infections and 11,323 deaths. The lack of Food and Drug Administration (FDA) licensed vaccines and antiviral drugs hindered early efforts to contain the outbreak. In response, the global scientific community has spurred the advanced development of many filovirus vaccine candidates. Novel vaccine platforms, such as viral vectors and DNA vaccines, have emerged, leading to the investigation of candidate vaccines that have demonstrated protective efficacy in small animal and nonhuman primate studies. Here, we will discuss several of these vaccine platforms with a particular focus on approaches that have advanced into clinical development.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- Headquarters Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
48
|
Polyclonal and convergent antibody response to Ebola virus vaccine rVSV-ZEBOV. Nat Med 2019; 25:1589-1600. [PMID: 31591605 DOI: 10.1038/s41591-019-0602-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/04/2019] [Indexed: 11/08/2022]
Abstract
Recombinant vesicular stomatitis virus-Zaire Ebola virus (rVSV-ZEBOV) is the most advanced Ebola virus vaccine candidate and is currently being used to combat the outbreak of Ebola virus disease (EVD) in the Democratic Republic of the Congo (DRC). Here we examine the humoral immune response in a subset of human volunteers enrolled in a phase 1 rVSV-ZEBOV vaccination trial by performing comprehensive single B cell and electron microscopy structure analyses. Four studied vaccinees show polyclonal, yet reproducible and convergent B cell responses with shared sequence characteristics. EBOV-targeting antibodies cross-react with other Ebolavirus species, and detailed epitope mapping revealed overlapping target epitopes with antibodies isolated from EVD survivors. Moreover, in all vaccinees, we detected highly potent EBOV-neutralizing antibodies with activities comparable or superior to the monoclonal antibodies currently used in clinical trials. These include antibodies combining the IGHV3-15/IGLV1-40 immunoglobulin gene segments that were identified in all investigated individuals. Our findings will help to evaluate and direct current and future vaccination strategies and offer opportunities for novel EVD therapies.
Collapse
|
49
|
Fuentes S, Ravichandran S, Khurana S. Antibody Repertoire of Human Polyclonal Antibodies Against Ebola Virus Glycoprotein Generated After Deoxyribonucleic Acid and Protein Vaccination of Transchromosomal Bovines. J Infect Dis 2019; 218:S597-S602. [PMID: 29939294 PMCID: PMC7107430 DOI: 10.1093/infdis/jiy325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Several Ebola vaccines and therapeutics are under clinical development. However, limited knowledge exists on the quality of antibody response generated by different Ebola vaccines. In this study, antibody repertoire induced by vaccination of transchromosomal bovine (TcB) with Ebola virus (EBOV) glycoprotein ([GP]; recombinant GP [rGP]) encoded by either deoxyribonucleic acid (DNA) or nanoparticle-based vaccine platform was analyzed using EBOV genome fragment phage display library and surface plasmon resonance (SPR)-based real-time kinetics assay to measure antibody affinity maturation to both native and partially denatured Ebola GP as well as GP containing the receptor binding domain but lacking the mucin-like domain. Immunoglobulin (IgG) obtained from rGP nanoparticle-vaccinated TcB demonstrated ~4-fold higher binding affinity compared with DNA-vaccinated TcB-induced IgG against the native rGP’s. The rGP nanoparticle vaccine generated a more robust and diverse antibody immune response to the native EBOV-GP compared with the DNA vaccine, which may explain the protective efficacy observed for these antibody preparations.
Collapse
Affiliation(s)
- Sandra Fuentes
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Supriya Ravichandran
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
50
|
Davis CW, Jackson KJL, McElroy AK, Halfmann P, Huang J, Chennareddy C, Piper AE, Leung Y, Albariño CG, Crozier I, Ellebedy AH, Sidney J, Sette A, Yu T, Nielsen SCA, Goff AJ, Spiropoulou CF, Saphire EO, Cavet G, Kawaoka Y, Mehta AK, Glass PJ, Boyd SD, Ahmed R. Longitudinal Analysis of the Human B Cell Response to Ebola Virus Infection. Cell 2019; 177:1566-1582.e17. [PMID: 31104840 PMCID: PMC6908968 DOI: 10.1016/j.cell.2019.04.036] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/11/2019] [Accepted: 04/16/2019] [Indexed: 01/12/2023]
Abstract
Ebola virus (EBOV) remains a public health threat. We performed a longitudinal study of B cell responses to EBOV in four survivors of the 2014 West African outbreak. Infection induced lasting EBOV-specific immunoglobulin G (IgG) antibodies, but their subclass composition changed over time, with IgG1 persisting, IgG3 rapidly declining, and IgG4 appearing late. Striking changes occurred in the immunoglobulin repertoire, with massive recruitment of naive B cells that subsequently underwent hypermutation. We characterized a large panel of EBOV glycoprotein-specific monoclonal antibodies (mAbs). Only a small subset of mAbs that bound glycoprotein by ELISA recognized cell-surface glycoprotein. However, this subset contained all neutralizing mAbs. Several mAbs protected against EBOV disease in animals, including one mAb that targeted an epitope under evolutionary selection during the 2014 outbreak. Convergent antibody evolution was seen across multiple donors, particularly among VH3-13 neutralizing antibodies specific for the GP1 core. Our study provides a benchmark for assessing EBOV vaccine-induced immunity. Ebola virus infection causes massive recruitment of naive B cells Virus-specific antibodies continue to class-switch and mutate for months after acute infection Protective antibodies can be neutralizing or non-neutralizing and can appear early Convergent, protective antibody rearrangements are seen in multiple donors
Collapse
Affiliation(s)
- Carl W Davis
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Katherine J L Jackson
- Department of Pathology, Stanford University, Stanford, CA, USA; Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Anita K McElroy
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA, USA; Division of Pediatric Infectious Disease, Emory University, Atlanta, GA, USA; Division of Pediatric Infectious Disease, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
| | - Jessica Huang
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Chakravarthy Chennareddy
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Ashley E Piper
- Virology Division, United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD, USA
| | | | - César G Albariño
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ian Crozier
- Integrated Research Facility at Fort Detrick, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institutes, Frederick, MD, USA
| | - Ali H Ellebedy
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA; Division of Immunobiology, Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tianwei Yu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | | | - Arthur J Goff
- Virology Division, United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Erica Ollman Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA; La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA; Division of Virology, Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Aneesh K Mehta
- Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Pamela J Glass
- Virology Division, United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA.
| |
Collapse
|