1
|
Stephenson KE. Next-Generation Coronavirus Disease 2019 Vaccines: Clinical Data and Future Directions. Infect Dis Clin North Am 2025; 39:253-274. [PMID: 40090832 DOI: 10.1016/j.idc.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Current coronavirus disease 2019 (COVID-19) vaccines have prevented millions of deaths since the beginning of the pandemic. Yet, current vaccines have critical limitations: (1) they elicit poor mucosal immune responses with reduced protection against infection and viral transmission, (2) immune responses wane quickly, and (3) new immune evasive severe acute respiratory syndrome coronavirus 2 variants continue to emerge. Next-generation vaccines aim to induce improved mucosal immunity via mucosal administration, and to elicit a greater duration and breadth of immune responses through new platforms like self-amplifying RNA. This review focuses on next-generation COVID-19 vaccines that have publicly available clinical data and are most likely to proceed in further development.
Collapse
Affiliation(s)
- Kathryn E Stephenson
- Center for Virology and Vaccine Research and Division of Infectious Diseases, Beth Israel Deaconess Medical Center, 330 Brookline E/CLS-1037, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Klimova EM, Lavinska OV, Drozdova LA. The role of DAMP cytotoxic fractions in the immune markers' disruption in patients with urgent surgical pathology and against the background of post-COVID-19 syndrome. Immunol Lett 2025:107033. [PMID: 40409597 DOI: 10.1016/j.imlet.2025.107033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND As a result of the SARS-CoV-2 pandemic, various population groups were formed that had acute and asymptomatic COVID-19. A survey in these groups revealed with equal frequency an asthenic symptom complex, the so-called post-COVID-19 syndrome (PCS). The frequency of urgent surgical pathology against the background of PCS and structural and functional disorders of various organs was increased. The aim - to study the dynamics of immunoresistance factors changes in patients with urgent surgical pathology that developed against the background of PCS and to identify pathogenic markers of the severe course and the risk of mortality. MATERIALS AND METHODS To examine patients with PCS and urgent cardiovascular (n = 103) and abdominal (n = 106) pathology we used the following methods: fluorescence microscopy, confocal microscopy, flow cytometry, spectrophotometry, ELISA. RESULTS We revealed a temporal dependence of immune dysfunction in patients with a comorbid course of urgent surgical pathology and PCS. The nature of the DAMP (damage-associated molecular patterns) cytotoxic fractions ratio was associated with certain changes in innate and adaptive immunity factors, severity of the condition and risk of mortality. At the first stage (2020-2021), patients with PCS has disorders of the humoral and cellular components of innate immunity against the background of an increase in the oligopeptide and peptide DAMP fractions. At the second stage (2022-2024) of PCS development, changes in innate as well as adaptive immunity were observed against the background of an increase in the cytotoxic oligonucleotide DAMP fraction (mortality was 17.3%). CONCLUSIONS The identified markers of impaired immunoresistance in cardiovascular and abdominal urgent pathology can be used to select targeted therapy tactics.
Collapse
Affiliation(s)
- Elena M Klimova
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine.
| | - Olena V Lavinska
- Department of Molecular Biology and Biotechnology, V.N. Karazin Kharkiv National University, 4 Pl. Svobody, 61022 Kharkiv, Ukraine; Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| | - Larisa A Drozdova
- Diagnostic Laboratory with Enzyme Immunoassay and Immunofluorescence Analysis, State Institution "Zaycev V.T. Institute of General and Urgent Surgery of National Academy of Medical Sciences of Ukraine", vyizd Likarskyi, 1, 61103, Kharkiv, Ukraine
| |
Collapse
|
3
|
Pinto PBA, Timis J, Chuensirikulchai K, Li QH, Lu HH, Maule E, Nguyen M, Alves RPDS, Verma SK, Ana-Sosa-Batiz F, Valentine K, Landeras-Bueno S, Kim K, Hastie K, Saphire EO, Alves A, Elong Ngono A, Shresta S. Co-immunization with spike and nucleocapsid based DNA vaccines for long-term protective immunity against SARS-CoV-2 Omicron. NPJ Vaccines 2024; 9:252. [PMID: 39702529 PMCID: PMC11659323 DOI: 10.1038/s41541-024-01043-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
The continuing evolution of SARS-CoV-2 variants challenges the durability of existing spike (S)-based COVID-19 vaccines. We hypothesized that vaccines composed of both S and nucleocapsid (N) antigens would increase the durability of protection by strengthening and broadening cellular immunity compared with S-based vaccines. To test this, we examined the immunogenicity and efficacy of wild-type SARS-CoV-2 S- and N-based DNA vaccines administered individually or together to K18-hACE2 mice. S, N, and S + N vaccines all elicited polyfunctional CD4+ and CD8+ T cell responses and provided short-term cross-protection against Beta and Omicron BA.2 variants, but only co-immunization with S + N vaccines provided long-term protection against Omicron BA.2. Depletion of CD4+ and CD8+ T cells reduced the long-term efficacy, demonstrating a crucial role for T cells in the durability of protection. These findings underscore the potential to enhance long-lived protection against SARS-CoV-2 variants by combining S and N antigens in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Paolla Beatriz Almeida Pinto
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kantinan Chuensirikulchai
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Hsueh Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Michael Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | | | | | | | - Kristen Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- University Cardenal Herrera-CEU, CEU Universities, Valencia, 46113, Spain
| | - Kenneth Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, 92093, USA
| | - Ada Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, 92093, USA.
| |
Collapse
|
4
|
Reguzova A, Müller M, Pagallies F, Burri D, Salomon F, Rziha HJ, Bittner-Schrader Z, Verstrepen BE, Böszörményi KP, Verschoor EJ, Gerhauser I, Elbers K, Esen M, Manenti A, Monti M, Rammensee HG, Derouazi M, Löffler MW, Amann R. A multiantigenic Orf virus-based vaccine efficiently protects hamsters and nonhuman primates against SARS-CoV-2. NPJ Vaccines 2024; 9:191. [PMID: 39414789 PMCID: PMC11484955 DOI: 10.1038/s41541-024-00981-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Among the common strategies to design next-generation COVID-19 vaccines is broadening the antigenic repertoire thereby aiming to increase efficacy against emerging variants of concern (VoC). This study describes a new Orf virus-based vector (ORFV) platform to design a multiantigenic vaccine targeting SARS-CoV-2 spike and nucleocapsid antigens. Vaccine candidates were engineered, either expressing spike protein (ORFV-S) alone or co-expressing nucleocapsid protein (ORFV-S/N). Mono- and multiantigenic vaccines elicited comparable levels of spike-specific antibodies and virus neutralization in mice. Results from a SARS-CoV-2 challenge model in hamsters suggest cross-protective properties of the multiantigenic vaccine against VoC, indicating improved viral clearance with ORFV-S/N, as compared to equal doses of ORFV-S. In a nonhuman primate challenge model, vaccination with the ORFV-S/N vaccine resulted in long-term protection against SARS-CoV-2 infection. These results demonstrate the potential of the ORFV platform for prophylactic vaccination and represent a preclinical development program supporting first-in-man studies with the multiantigenic ORFV vaccine.
Collapse
Affiliation(s)
- Alena Reguzova
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Melanie Müller
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Felix Pagallies
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Dominique Burri
- Speransa Therapeutics, Bethmannstrasse 8, 60311, Frankfurt am Main, Germany
| | - Ferdinand Salomon
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Hanns-Joachim Rziha
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Zsofia Bittner-Schrader
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
- Department of Viroscience, Erasmus University Medical Center, Dr. Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Knut Elbers
- Boehringer Ingelheim International GmbH, Binger Strasse 173, 55216, Ingelheim am Rhein, Germany
- ViraTherapeutics GmbH, Bundesstraße 27, 6063, Rum, Austria
| | - Meral Esen
- Institute of Tropical Medicine, University of Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen; Cluster of Excellence (EXC2124) "Controlling Microbes to Fight Infection", Tübingen, Germany
| | - Alessandro Manenti
- VisMederi Srl., Strada del Petriccio e Belriguardo 35, 53100, Siena, Italy
| | - Martina Monti
- VisMederi Srl., Strada del Petriccio e Belriguardo 35, 53100, Siena, Italy
| | - Hans-Georg Rammensee
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Madiha Derouazi
- Speransa Therapeutics, Bethmannstrasse 8, 60311, Frankfurt am Main, Germany
| | - Markus W Löffler
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital Tübingen, Otfried-Müller-Str. 4/1, 72076, Tübingen, Germany.
- Centre for Clinical Transfusion Medicine, Otfried-Müller-Str. 4/1, 72076, Tübingen, Germany.
| | - Ralf Amann
- Institute of Immunology, University Hospital Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.
| |
Collapse
|
5
|
Huang Y, Chen J, Chen S, Huang C, Li B, Li J, Jin Z, Zhang Q, Pan P, Du W, Liu L, Liu Z. Molecular characterization of SARS-CoV-2 nucleocapsid protein. Front Cell Infect Microbiol 2024; 14:1415885. [PMID: 38846351 PMCID: PMC11153676 DOI: 10.3389/fcimb.2024.1415885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.
Collapse
Affiliation(s)
- Yanping Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Junkai Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Siwei Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixiong Jin
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Zhang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pan Pan
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Weixing Du
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Long Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixin Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
6
|
Warner BM, Yates JGE, Vendramelli R, Truong T, Meilleur C, Chan L, Leacy A, Pham PH, Pei Y, Susta L, Wootton SK, Kobasa D. Intranasal vaccination with an NDV-vectored SARS-CoV-2 vaccine protects against Delta and Omicron challenges. NPJ Vaccines 2024; 9:90. [PMID: 38782986 PMCID: PMC11116387 DOI: 10.1038/s41541-024-00870-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 03/29/2024] [Indexed: 05/25/2024] Open
Abstract
The rapid development and deployment of vaccines following the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been estimated to have saved millions of lives. Despite their immense success, there remains a need for next-generation vaccination approaches for SARS-CoV-2 and future emerging coronaviruses and other respiratory viruses. Here we utilized a Newcastle Disease virus (NDV) vectored vaccine expressing the ancestral SARS-CoV-2 spike protein in a pre-fusion stabilized chimeric conformation (NDV-PFS). When delivered intranasally, NDV-PFS protected both Syrian hamsters and K18 mice against Delta and Omicron SARS-CoV-2 variants of concern. Additionally, intranasal vaccination induced robust, durable protection that was extended to 6 months post-vaccination. Overall, our data provide evidence that NDV-vectored vaccines represent a viable next-generation mucosal vaccination approach.
Collapse
Affiliation(s)
- Bryce M Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Jacob G E Yates
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada
| | - Robert Vendramelli
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Courtney Meilleur
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Lily Chan
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada
| | - Alexander Leacy
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada
| | - Phuc H Pham
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada.
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, N1G 2W1, Canada.
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada.
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
7
|
Chiuppesi F, Zaia JA, Gutierrez-Franco MA, Ortega-Francisco S, Ly M, Kha M, Kim T, Dempsey S, Kar S, Grifoni A, Sette A, Wussow F, Diamond DJ. Synthetic modified vaccinia Ankara vaccines confer cross-reactive and protective immunity against mpox virus. COMMUNICATIONS MEDICINE 2024; 4:19. [PMID: 38366141 PMCID: PMC10873322 DOI: 10.1038/s43856-024-00443-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Although the mpox global health emergency caused by mpox virus (MPXV) clade IIb.1 has ended, mpox cases are still reported due to low vaccination coverage and waning immunity. COH04S1 is a clinically evaluated, multiantigen COVID-19 vaccine candidate built on a fully synthetic platform of the highly attenuated modified vaccinia Ankara (MVA) vector, representing the only FDA-approved smallpox/mpox vaccine JYNNEOS. Given the potential threat of MPXV resurgence and need for vaccine alternatives, we aimed to assess the capacity COH04S1 and its synthetic MVA (sMVA) backbone to confer MPXV-specific immunity. METHODS We evaluated orthopoxvirus-specific and MPXV cross-reactive immune responses in samples collected during a Phase 1 clinical trial of COH04S1 and in non-human primates (NHP) vaccinated with COH04S1 or its sMVA backbone. MPXV cross-reactive immune responses in COH04S1-vaccinated healthy adults were compared to responses measured in healthy subjects vaccinated with JYNNEOS. Additionally, we evaluated the protective efficacy of COH04S1 and sMVA against mpox in mpox-susceptible CAST/EiJ mice. RESULTS COH04S1-vaccinated individuals develop robust orthopoxvirus-specific humoral and cellular responses, including cross-reactive antibodies to MPXV-specific virion proteins as well as MPXV cross-neutralizing antibodies in 45% of the subjects. In addition, NHP vaccinated with COH04S1 or sMVA show similar MPXV cross-reactive antibody responses. Moreover, MPXV cross-reactive humoral responses elicited by COH04S1 are comparable to those measured in JYNNEOS-vaccinated subjects. Finally, we show that mice vaccinated with COH04S1 or sMVA are protected from lung infection following challenge with MPXV clade IIb.1. CONCLUSIONS These results demonstrate the capacity of sMVA vaccines to elicit cross-reactive and protective orthopox-specific immunity against MPXV, suggesting that COH04S1 and sMVA could be developed as bivalent or monovalent mpox vaccine alternatives against MPXV.
Collapse
Affiliation(s)
- Flavia Chiuppesi
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA.
| | - John A Zaia
- Center for Gene Therapy, City of Hope National Medical Center, Duarte, CA, USA
| | - Miguel-Angel Gutierrez-Franco
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sandra Ortega-Francisco
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Minh Ly
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Mindy Kha
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Taehyun Kim
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Shannon Dempsey
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Felix Wussow
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Don J Diamond
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
8
|
Yu H, Guan F, Miller H, Lei J, Liu C. The role of SARS-CoV-2 nucleocapsid protein in antiviral immunity and vaccine development. Emerg Microbes Infect 2023; 12:e2164219. [PMID: 36583642 PMCID: PMC9980416 DOI: 10.1080/22221751.2022.2164219] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACTThe coronavirus disease 2019 (COVID-19) has caused enormous health risks and global economic disruption. This disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The SARS-CoV-2 nucleocapsid protein is a structural protein involved in viral replication and assembly. There is accumulating evidence indicating that the nucleocapsid protein is multi-functional, playing a key role in the pathogenesis of COVID-19 and antiviral immunity against SARS-CoV-2. Here, we summarize its potential application in the prevention of COVID-19, which is based on its role in inflammation, cell death, antiviral innate immunity, and antiviral adaptive immunity.
Collapse
Affiliation(s)
- Haiyun Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China, Chaohong Liu
| |
Collapse
|
9
|
Abdelnabi R, Pérez P, Astorgano D, Albericio G, Kerstens W, Thibaut HJ, Coelmont L, Weynand B, Labiod N, Delgado R, Montenegro D, Puentes E, Rodríguez E, Neyts J, Dallmeier K, Esteban M, García-Arriaza J. Optimized vaccine candidate MVA-S(3P) fully protects against SARS-CoV-2 infection in hamsters. Front Immunol 2023; 14:1163159. [PMID: 37920464 PMCID: PMC10619667 DOI: 10.3389/fimmu.2023.1163159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
The development of novel optimized vaccines against coronavirus disease 2019 (COVID-19) that are capable of controlling the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic and the appearance of different variants of concern (VoC) is needed to fully prevent the transmission of the virus. In the present study, we describe the enhanced immunogenicity and efficacy elicited in hamsters by a modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein [termed MVA-S(3P)]. Hamsters vaccinated with one or two doses of MVA-S(3P) developed high titers of S-binding IgG antibodies and neutralizing antibodies against the ancestral Wuhan SARS-CoV-2 virus and VoC beta, gamma, and delta, as well as against omicron, although with a somewhat lower neutralization activity. After SARS-CoV-2 challenge, vaccinated hamsters did not lose body weight as compared to matched placebo (MVA-WT) controls. Consistently, vaccinated hamsters exhibited significantly reduced viral RNA in the lungs and nasal washes, and no infectious virus was detected in the lungs in comparison to controls. Furthermore, almost no lung histopathology was detected in MVA-S(3P)-vaccinated hamsters, which also showed significantly reduced levels of proinflammatory cytokines in the lungs compared to unvaccinated hamsters. These results reinforce the use of MVA-S(3P) as a vaccine candidate against COVID-19 in clinical trials.
Collapse
Affiliation(s)
- Rana Abdelnabi
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, KU Leuven, Leuven, Belgium
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Winnie Kerstens
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Hendrik Jan Thibaut
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Lotte Coelmont
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, KU Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Imaging and Pathology, Translational Cell and Tissue Research, Division of Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Nuria Labiod
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Medical School, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | | | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, KU Leuven, Leuven, Belgium
| | - Kai Dallmeier
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, KU Leuven, Leuven, Belgium
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
10
|
Cao J, Gu H, Zhang X, Yun H, Li J, Si CY, Zhang J, Wang H. Intranasal inoculation of female BALB/c mice with replication-deficient human adenovirus type 5 expressing SARS-CoV-2 nucleocapsid protein aggravates lung pathology upon re-encountering the antigen. Virus Res 2023; 335:199201. [PMID: 37595663 PMCID: PMC10470087 DOI: 10.1016/j.virusres.2023.199201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/27/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Preclinical studies indicate that SARS-CoV-2 nucleocapsid (N)-based vaccines, along with other viral protein(s), confer protection in various animal models against infection by SARS-CoV-2 ancestral virus and variants of concern. However, the optimal vaccination procedure and the role of N-specific host adaptive immune responses remain elusive. Here, we report that intranasal inoculation with replication-deficient human adenovirus type 5 expressing SARS-CoV-2 N protein (Ad5-N) conferred no protection in the lung of female BALB/c mice upon re-encountering the antigen, either by 10-fold Ad5-N re-exposure or sublethal infection of mouse-adapted SARS-CoV-2. By contrast, this procedure led to aggravated lung pathology with more necroptotic CD3+ T cells and Ly6G+ granulocytes, which was associated with the accumulation of IFN-γ-expressing antigen-experienced CD4+ and CD8+ T cells. These findings pre-caution the clinical application of this vaccination procedure. Furthermore, our data suggest that excessive host adaptive immune responses against N protein contributes to COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Junxia Cao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hongjing Gu
- Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xueting Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hongfang Yun
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jiarong Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China; University of South China, Hengyang Medical School, Hengyang 421001, China
| | - Chuan-Yimu Si
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China; University of South China, Hengyang Medical School, Hengyang 421001, China; Anhui Medical University, Hefei 230032, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Hui Wang
- Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|
11
|
Chiuppesi F, Ortega-Francisco S, Gutierrez MA, Li J, Ly M, Faircloth K, Mack-Onyeike J, La Rosa C, Thomas S, Zhou Q, Drake J, Slape C, Fernando P, Rida W, Kaltcheva T, Grifoni A, Sette A, Patterson A, Dempsey S, Ball B, Ali H, Salhotra A, Stein A, Nathwani N, Rosenzweig M, Nikolaenko L, Al Malki MM, Dickter J, Nanayakkara DD, Puing A, Forman SJ, Taplitz RA, Zaia JA, Nakamura R, Wussow F, Diamond DJ, Dadwal SS. Stimulation of Potent Humoral and Cellular Immunity via Synthetic Dual-Antigen MVA-Based COVID-19 Vaccine COH04S1 in Cancer Patients Post Hematopoietic Cell Transplantation and Cellular Therapy. Vaccines (Basel) 2023; 11:1492. [PMID: 37766168 PMCID: PMC10538048 DOI: 10.3390/vaccines11091492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Hematopoietic cell transplantation (HCT) and chimeric antigen receptor (CAR)-T cell patients are immunocompromised, remain at high risk following SARS-CoV-2 infection, and are less likely than immunocompetent individuals to respond to vaccination. As part of the safety lead-in portion of a phase 2 clinical trial in patients post HCT/CAR-T for hematological malignancies (HM), we tested the immunogenicity of the synthetic modified vaccinia Ankara-based COVID-19 vaccine COH04S1 co-expressing spike (S) and nucleocapsid (N) antigens. Thirteen patients were vaccinated 3-12 months post HCT/CAR-T with two to four doses of COH04S1. SARS-CoV-2 antigen-specific humoral and cellular immune responses, including neutralizing antibodies to ancestral virus and variants of concern (VOC), were measured up to six months post vaccination and compared to immune responses in historical cohorts of naïve healthy volunteers (HV) vaccinated with COH04S1 and naïve healthcare workers (HCW) vaccinated with the FDA-approved mRNA vaccine Comirnaty® (Pfizer, New York, NY, USA). After one or two COH04S1 vaccine doses, HCT/CAR-T recipients showed a significant increase in S- and N-specific binding antibody titers and neutralizing antibodies with potent activity against SARS-CoV-2 ancestral virus and VOC, including the highly immune evasive Omicron XBB.1.5 variant. Furthermore, vaccination with COH04S1 resulted in a significant increase in S- and N-specific T cells, predominantly CD4+ T lymphocytes. Elevated S- and N-specific immune responses continued to persist at six months post vaccination. Furthermore, both humoral and cellular immune responses in COH04S1-vaccinated HCT/CAR-T patients were superior or comparable to those measured in COH04S1-vaccinated HV or Comirnaty®-vaccinated HCW. These results demonstrate robust stimulation of SARS-CoV-2 S- and N-specific immune responses including cross-reactive neutralizing antibodies by COH04S1 in HM patients post HCT/CAR-T, supporting further testing of COH04S1 in immunocompromised populations.
Collapse
Affiliation(s)
- Flavia Chiuppesi
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Sandra Ortega-Francisco
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Miguel-Angel Gutierrez
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Jing Li
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Minh Ly
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Katelyn Faircloth
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Jada Mack-Onyeike
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Corinna La Rosa
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Sandra Thomas
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Qiao Zhou
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Jennifer Drake
- Clinical Trials Office, City of Hope National Medical Center, Duarte, CA 91010, USA; (J.D.); (C.S.); (P.F.)
| | - Cynthia Slape
- Clinical Trials Office, City of Hope National Medical Center, Duarte, CA 91010, USA; (J.D.); (C.S.); (P.F.)
| | - Paolo Fernando
- Clinical Trials Office, City of Hope National Medical Center, Duarte, CA 91010, USA; (J.D.); (C.S.); (P.F.)
| | - Wasima Rida
- Independent Researcher, Arlington, VA 22205, USA;
| | - Teodora Kaltcheva
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, University of California San Diego, La Jolla, CA 92037, USA; (A.G.); (A.S.)
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute of Allergy and Immunology, University of California San Diego, La Jolla, CA 92037, USA; (A.G.); (A.S.)
| | - Angela Patterson
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Shannon Dempsey
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Brian Ball
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Haris Ali
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Amandeep Salhotra
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Anthony Stein
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Nitya Nathwani
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Michael Rosenzweig
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Liana Nikolaenko
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Monzr M. Al Malki
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Jana Dickter
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Deepa D. Nanayakkara
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Alfredo Puing
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Stephen J. Forman
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Randy A. Taplitz
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.A.T.); (S.S.D.)
- Department of Medicine, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - John A. Zaia
- Center for Gene Therapy, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Ryotaro Nakamura
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Felix Wussow
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Don J. Diamond
- Department of Hematology and HCT, Hematologic Malignancies Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (F.C.); (S.O.-F.); (M.-A.G.); (J.L.); (K.F.); (J.M.-O.); (C.L.R.); (S.T.); (Q.Z.); (T.K.); (A.P.); (S.D.); (B.B.); (H.A.); (A.S.); (A.S.); (N.N.); (M.R.); (L.N.); (M.M.A.M.); (J.D.); (D.D.N.); (S.J.F.); (R.N.); (F.W.)
| | - Sanjeet S. Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.A.T.); (S.S.D.)
- Department of Medicine, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
12
|
Perdiguero B, Pérez P, Marcos-Villar L, Albericio G, Astorgano D, Álvarez E, Sin L, Elena Gómez C, García-Arriaza J, Esteban M. Highly attenuated poxvirus-based vaccines against emerging viral diseases. J Mol Biol 2023:168173. [PMID: 37301278 DOI: 10.1016/j.jmb.2023.168173] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Although one member of the poxvirus family, variola virus, has caused one of the most devastating human infections worldwide, smallpox, the knowledge gained over the last 30 years on the molecular, virological and immunological mechanisms of these viruses has allowed the use of members of this family as vectors for the generation of recombinant vaccines against numerous pathogens. In this review, we cover different aspects of the history and biology of poxviruses with emphasis on their application as vaccines, from first- to fourth-generation, against smallpox, monkeypox, emerging viral diseases highlighted by the World Health Organization (COVID-19, Crimean-Congo haemorrhagic fever, Ebola and Marburg virus diseases, Lassa fever, Middle East respiratory syndrome and severe acute respiratory syndrome, Nipah and other henipaviral diseases, Rift Valley fever and Zika), as well as against one of the most concerning prevalent virus, the Human Immunodeficiency Virus, the causative agent of AcquiredImmunodeficiency Syndrome. We discuss the implications in human health of the 2022 monkeypox epidemic affecting many countries, and the rapid prophylactic and therapeutic measures adopted to control virus dissemination within the human population. We also describe the preclinical and clinical evaluation of the Modified Vaccinia virus Ankara and New York vaccinia virus poxviral strains expressing heterologous antigens from the viral diseases listed above. Finally, we report different approaches to improve the immunogenicity and efficacy of poxvirus-based vaccine candidates, such as deletion of immunomodulatory genes, insertion of host-range genes and enhanced transcription of foreign genes through modified viral promoters. Some future prospects are also highlighted.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| |
Collapse
|
13
|
Nguyen THO, Rowntree LC, Allen LF, Chua BY, Kedzierski L, Lim C, Lasica M, Tennakoon GS, Saunders NR, Crane M, Chee L, Seymour JF, Anderson MA, Whitechurch A, Clemens EB, Zhang W, Chang SY, Habel JR, Jia X, McQuilten HA, Minervina AA, Pogorelyy MV, Chaurasia P, Petersen J, Menon T, Hensen L, Neil JA, Mordant FL, Tan HX, Cabug AF, Wheatley AK, Kent SJ, Subbarao K, Karapanagiotidis T, Huang H, Vo LK, Cain NL, Nicholson S, Krammer F, Gibney G, James F, Trevillyan JM, Trubiano JA, Mitchell J, Christensen B, Bond KA, Williamson DA, Rossjohn J, Crawford JC, Thomas PG, Thursky KA, Slavin MA, Tam CS, Teh BW, Kedzierska K. Robust SARS-CoV-2 T cell responses with common TCRαβ motifs toward COVID-19 vaccines in patients with hematological malignancy impacting B cells. Cell Rep Med 2023; 4:101017. [PMID: 37030296 PMCID: PMC10040362 DOI: 10.1016/j.xcrm.2023.101017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Immunocompromised hematology patients are vulnerable to severe COVID-19 and respond poorly to vaccination. Relative deficits in immunity are, however, unclear, especially after 3 vaccine doses. We evaluated immune responses in hematology patients across three COVID-19 vaccination doses. Seropositivity was low after a first dose of BNT162b2 and ChAdOx1 (∼26%), increased to 59%-75% after a second dose, and increased to 85% after a third dose. While prototypical antibody-secreting cells (ASCs) and T follicular helper (Tfh) cell responses were elicited in healthy participants, hematology patients showed prolonged ASCs and skewed Tfh2/17 responses. Importantly, vaccine-induced expansions of spike-specific and peptide-HLA tetramer-specific CD4+/CD8+ T cells, together with their T cell receptor (TCR) repertoires, were robust in hematology patients, irrespective of B cell numbers, and comparable to healthy participants. Vaccinated patients with breakthrough infections developed higher antibody responses, while T cell responses were comparable to healthy groups. COVID-19 vaccination induces robust T cell immunity in hematology patients of varying diseases and treatments irrespective of B cell numbers and antibody response.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Hokkaido 060-0808, Japan
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Chhay Lim
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Masa Lasica
- Department of Haematology, St Vincent's Hospital, Fitzroy, VIC 3065, Australia; Department of Haematology, Eastern Health, Box Hill, VIC 3128, Australia
| | - G Surekha Tennakoon
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Natalie R Saunders
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Megan Crane
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Lynette Chee
- Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - John F Seymour
- Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Mary Ann Anderson
- Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Ashley Whitechurch
- Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - E Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - So Young Chang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Anastasia A Minervina
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mikhail V Pogorelyy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Priyanka Chaurasia
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Tejas Menon
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jessica A Neil
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira F Cabug
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC 3010, Australia; Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Theo Karapanagiotidis
- Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Han Huang
- Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lynn K Vo
- Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Natalie L Cain
- Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Grace Gibney
- Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Fiona James
- Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Janine M Trevillyan
- Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia; Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jason A Trubiano
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Jeni Mitchell
- Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Britt Christensen
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia; Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Katherine A Bond
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Microbiology, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Deborah A Williamson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Victorian Infectious Diseases Reference Laboratory (VIDRL), at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, CF14 4XN Cardiff, UK
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Karin A Thursky
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Constantine S Tam
- Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Hokkaido 060-0808, Japan.
| |
Collapse
|
14
|
Wussow F, Kha M, Kim T, Ly M, Yll-Pico M, Kar S, Lewis MG, Chiuppesi F, Diamond DJ. Synthetic multiantigen MVA vaccine COH04S1 and variant-specific derivatives protect Syrian hamsters from SARS-CoV-2 Omicron subvariants. NPJ Vaccines 2023; 8:41. [PMID: 36928589 PMCID: PMC10018591 DOI: 10.1038/s41541-023-00640-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Emerging SARS-CoV-2 Omicron subvariants continue to disrupt COVID-19 vaccine efficacy through multiple immune mechanisms including neutralizing antibody evasion. We developed COH04S1, a synthetic modified vaccinia Ankara vector that co-expresses Wuhan-Hu-1-based spike and nucleocapsid antigens. COH04S1 demonstrated efficacy against ancestral virus and Beta and Delta variants in animal models and was safe and immunogenic in a Phase 1 clinical trial. Here, we report efficacy of COH04S1 and analogous Omicron BA.1- and Beta-specific vaccines to protect Syrian hamsters from Omicron subvariants. Despite eliciting strain-specific antibody responses, all three vaccines protect hamsters from weight loss, lower respiratory tract infection, and lung pathology following challenge with Omicron BA.1 or BA.2.12.1. While the BA.1-specifc vaccine affords consistently improved efficacy compared to COH04S1 to protect against homologous challenge with BA.1, all three vaccines confer similar protection against heterologous challenge with BA.2.12.1. These results demonstrate efficacy of COH04S1 and variant-specific derivatives to confer cross-protective immunity against SARS-CoV-2 Omicron subvariants.
Collapse
Affiliation(s)
- Felix Wussow
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| | - Mindy Kha
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Taehyun Kim
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Minh Ly
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Marcal Yll-Pico
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | | | | | - Flavia Chiuppesi
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Don J Diamond
- Department of Hematology and Transplant Center, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
15
|
Pothast CR, Dijkland RC, Thaler M, Hagedoorn RS, Kester MGD, Wouters AK, Hiemstra PS, van Hemert MJ, Gras S, Falkenburg JHF, Heemskerk MHM. SARS-CoV-2-specific CD4 + and CD8 + T cell responses can originate from cross-reactive CMV-specific T cells. eLife 2022; 11:82050. [PMID: 36408799 PMCID: PMC9822249 DOI: 10.7554/elife.82050] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022] Open
Abstract
Detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specific CD4+ and CD8+ T cells in SARS-CoV-2-unexposed donors has been explained by the presence of T cells primed by other coronaviruses. However, based on the relatively high frequency and prevalence of cross-reactive T cells, we hypothesized cytomegalovirus (CMV) may induce these cross-reactive T cells. Stimulation of pre-pandemic cryo-preserved peripheral blood mononuclear cells (PBMCs) with SARS-CoV-2 peptides revealed that frequencies of SARS-CoV-2-specific T cells were higher in CMV-seropositive donors. Characterization of these T cells demonstrated that membrane-specific CD4+ and spike-specific CD8+ T cells originate from cross-reactive CMV-specific T cells. Spike-specific CD8+ T cells recognize SARS-CoV-2 spike peptide FVSNGTHWF (FVS) and dissimilar CMV pp65 peptide IPSINVHHY (IPS) presented by HLA-B*35:01. These dual IPS/FVS-reactive CD8+ T cells were found in multiple donors as well as severe COVID-19 patients and shared a common T cell receptor (TCR), illustrating that IPS/FVS-cross-reactivity is caused by a public TCR. In conclusion, CMV-specific T cells cross-react with SARS-CoV-2, despite low sequence homology between the two viruses, and may contribute to the pre-existing immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Cilia R Pothast
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Romy C Dijkland
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Melissa Thaler
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Michel GD Kester
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Anne K Wouters
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical CenterLeidenNetherlands
| | - Martijn J van Hemert
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe UniversityVictoriaAustralia,Department of Biochemistry and Molecular Biology, Monash UniversityClaytonAustralia
| | | | | |
Collapse
|