1
|
Phadke VK, Gromer DJ, Rebolledo PA, Graciaa DS, Wiley Z, Sherman AC, Scherer EM, Leary M, Girmay T, McCullough MP, Min JY, Capone S, Sommella A, Vitelli A, Retallick J, Seetahal J, Koller M, Tsong R, Neill-Gubitz H, Mulligan MJ, Rouphael NG. Safety and immunogenicity of a ChAd155-vectored rabies vaccine compared with inactivated, purified chick embryo cell rabies vaccine in healthy adults. Vaccine 2024; 42:126441. [PMID: 39418686 DOI: 10.1016/j.vaccine.2024.126441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Rabies is a zoonotic viral encephalitis that is endemic in many countries and confers a high mortality. Licensed vaccines require several doses to ensure efficacy. To investigate a logistically favorable approach, we assessed the safety and immunogenicity of ChAd155-RG, a novel investigational rabies vaccine using a replication-defective chimpanzee adenovirus vector. METHODS We conducted a first-in-human, phase 1, randomized, double-blind, dose-escalation trial comparing ChAd155-RG with a licensed inactivated vaccine (RabAvert) in healthy adults. Participants received either RabAvert at standard dosing or ChAd155-RG at a low dose for one immunization or a high dose for one or two immunizations. To assess safety, we evaluated reactogenicity, unsolicited adverse events, and thrombotic events. To measure immunogenicity, we measured rabies viral neutralizing antibody (VNA) titers and anti-ChAd155 neutralizing antibodies. RESULTS Mild to moderate systemic reactogenicity and transient lymphopenia and neutropenia were more common among recipients of ChAd155-RG compared with those who received RabAvert. No thrombotic events or serious adverse events were reported. Only the groups receiving RabAvert or two doses of high-dose ChAd155-RG achieved 100 % seroconversion, and seroprotection was most durable in the RabAvert group. Most participants had preexisting anti-vector antibodies, which were boosted by ChAd155-RG. Baseline and post-vaccination anti-vector antibody titers were negatively associated with post-vaccination rabies VNA titers. CONCLUSIONS In this phase 1 clinical trial, a novel rabies vaccine using a simian adenovirus vector was safe and tolerable, but generated lower, less durable rabies VNA titers than a standard inactivated rabies virus vaccine, which may be due to preexisting, anti-vector immunity.
Collapse
Affiliation(s)
- Varun K Phadke
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.
| | - Daniel J Gromer
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Paulina A Rebolledo
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel S Graciaa
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Zanthia Wiley
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Amy C Sherman
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Erin M Scherer
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Maranda Leary
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Tigisty Girmay
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Michele P McCullough
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | - Jamie Retallick
- Rabies Laboratory, Kansas State Veterinary Diagnostic Laboratory (KSVDL), Kansas State University, Manhattan, KS, USA
| | - Janine Seetahal
- Rabies Laboratory, Kansas State Veterinary Diagnostic Laboratory (KSVDL), Kansas State University, Manhattan, KS, USA
| | - Mark Koller
- Rabies Laboratory, Kansas State Veterinary Diagnostic Laboratory (KSVDL), Kansas State University, Manhattan, KS, USA
| | | | | | - Mark J Mulligan
- New York University Grossman School of Medicine and New York University Vaccine Center, NY, New York, USA
| | - Nadine G Rouphael
- Hope Clinic, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Vatzia E, Paudyal B, Dema B, Carr BV, Sedaghat-Rostami E, Gubbins S, Sharma B, Moorhouse E, Morris S, Ulaszewska M, MacLoughlin R, Salguero FJ, Gilbert SC, Tchilian E. Aerosol immunization with influenza matrix, nucleoprotein, or both prevents lung disease in pig. NPJ Vaccines 2024; 9:188. [PMID: 39397062 PMCID: PMC11471855 DOI: 10.1038/s41541-024-00989-8] [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: 02/21/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
Current influenza vaccines are strain-specific and require frequent updates to combat new strains, making a broadly protective influenza vaccine (BPIV) highly desirable. A promising strategy is to induce T-cell responses against internal proteins conserved across influenza strains. In this study, pH1N1 pre-exposed pigs were immunized by aerosol using viral vectored vaccines (ChAdOx2 and MVA) expressing matrix (M1) and nucleoprotein (NP). Following H3N2 challenge, all immunizations (M1, NP or NPM1) reduced lung pathology, but M1 alone offered the greatest protection. NP or NPM1 immunization induced both T-cell and antibody responses. M1 immunization generated no detectable antibodies but elicited M1-specific T-cell responses, suggesting T cell-mediated protection. Additionally, a single aerosol immunization with the ChAdOx vaccine encoding M1, NP and neuraminidase reduced lung pathology. These findings provide insights into BPIV development using a relevant large natural host, the pig.
Collapse
Affiliation(s)
| | | | - Barbara Dema
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Susan Morris
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Sarah C Gilbert
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | |
Collapse
|
3
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [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: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
4
|
Wallace R, Bliss CM, Parker AL. The Immune System-A Double-Edged Sword for Adenovirus-Based Therapies. Viruses 2024; 16:973. [PMID: 38932265 PMCID: PMC11209478 DOI: 10.3390/v16060973] [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: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.
Collapse
Affiliation(s)
- Rebecca Wallace
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
| | - Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
5
|
Del Moral-Sánchez I, Wee EG, Xian Y, Lee WH, Allen JD, Torrents de la Peña A, Fróes Rocha R, Ferguson J, León AN, Koekkoek S, Schermer EE, Burger JA, Kumar S, Zwolsman R, Brinkkemper M, Aartse A, Eggink D, Han J, Yuan M, Crispin M, Ozorowski G, Ward AB, Wilson IA, Hanke T, Sliepen K, Sanders RW. Triple tandem trimer immunogens for HIV-1 and influenza nucleic acid-based vaccines. NPJ Vaccines 2024; 9:74. [PMID: 38582771 PMCID: PMC10998906 DOI: 10.1038/s41541-024-00862-8] [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: 09/11/2023] [Accepted: 03/14/2024] [Indexed: 04/08/2024] Open
Abstract
Recombinant native-like HIV-1 envelope glycoprotein (Env) trimers are used in candidate vaccines aimed at inducing broadly neutralizing antibodies. While state-of-the-art SOSIP or single-chain Env designs can be expressed as native-like trimers, undesired monomers, dimers and malformed trimers that elicit non-neutralizing antibodies are also formed, implying that these designs could benefit from further modifications for gene-based vaccination approaches. Here, we describe the triple tandem trimer (TTT) design, in which three Env protomers are genetically linked in a single open reading frame and express as native-like trimers. Viral vectored Env TTT induced similar neutralization titers but with a higher proportion of trimer-specific responses. The TTT design was also applied to generate influenza hemagglutinin (HA) trimers without the need for trimerization domains. Additionally, we used TTT to generate well-folded chimeric Env and HA trimers that harbor protomers from three different strains. In summary, the TTT design is a useful platform for the design of HIV-1 Env and influenza HA immunogens for a multitude of vaccination strategies.
Collapse
Affiliation(s)
- Iván Del Moral-Sánchez
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Edmund G Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yuejiao Xian
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Alba Torrents de la Peña
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Rebeca Fróes Rocha
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - James Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - André N León
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sylvie Koekkoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Edith E Schermer
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Judith A Burger
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Sanjeev Kumar
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Robby Zwolsman
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Mitch Brinkkemper
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Aafke Aartse
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kwinten Sliepen
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Rogier W Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands.
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
6
|
Sáez-Llorens X, Norero X, Mussi-Pinhata MM, Luciani K, de la Cueva IS, Díez-Domingo J, Lopez-Medina E, Epalza C, Brzostek J, Szymański H, Boucher FD, Cetin BS, De Leon T, Dinleyici EC, Gabriel MÁM, Ince T, Macias-Parra M, Langley JM, Martinón-Torres F, Rämet M, Kuchar E, Pinto J, Puthanakit T, Baquero-Artigao F, Gattinara GC, Arribas JMM, Ramos Amador JT, Szenborn L, Tapiero B, Anderson EJ, Campbell JD, Faust SN, Nikic V, Zhou Y, Pu W, Friel D, Dieussaert I, Lopez AG, McPhee R, Stoszek SK, Vanhoutte N. Safety and Immunogenicity of a ChAd155-Vectored Respiratory Syncytial Virus Vaccine in Infants 6-7 Months of age: A Phase 1/2 Randomized Trial. J Infect Dis 2024; 229:95-107. [PMID: 37477875 PMCID: PMC10786261 DOI: 10.1093/infdis/jiad271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/16/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections in infants. This phase 1/2, observer-blind, randomized, controlled study assessed the safety and immunogenicity of an investigational chimpanzee-derived adenoviral vector RSV vaccine (ChAd155-RSV, expressing RSV F, N, and M2-1) in infants. METHODS Healthy 6- to 7-month-olds were 1:1:1-randomized to receive 1 low ChAd155-RSV dose (1.5 × 1010 viral particles) followed by placebo (RSV_1D); 2 high ChAd155-RSV doses (5 × 1010 viral particles) (RSV_2D); or active comparator vaccines/placebo (comparator) on days 1 and 31. Follow-up lasted approximately 2 years. RESULTS Two hundred one infants were vaccinated (RSV_1D: 65; RSV_2D: 71; comparator: 65); 159 were RSV-seronaive at baseline. Most solicited and unsolicited adverse events after ChAd155-RSV occurred at similar or lower rates than after active comparators. In infants who developed RSV infection, there was no evidence of vaccine-associated enhanced respiratory disease (VAERD). RSV-A neutralizing titers and RSV F-binding antibody concentrations were higher post-ChAd155-RSV than postcomparator at days 31, 61, and end of RSV season 1 (mean follow-up, 7 months). High-dose ChAd155-RSV induced stronger responses than low-dose, with further increases post-dose 2. CONCLUSIONS ChAd155-RSV administered to 6- to 7-month-olds had a reactogenicity/safety profile like other childhood vaccines, showed no evidence of VAERD, and induced a humoral immune response. Clinical Trials Registration. NCT03636906.
Collapse
Affiliation(s)
- Xavier Sáez-Llorens
- Department of Infectious Diseases, Hospital del Niño Dr. José Renán Esquivel
- Vaccine Research Department, Centro de Vacunación Internacional
- Sistema Nacional de Investigación
- Secretaria Nacional de Ciencia y Tecnologia, Panama City, Panama
| | - Ximena Norero
- Department of Infectious Diseases, Hospital del Niño Dr. José Renán Esquivel
- Vaccine Research Department, Centro de Vacunación Internacional
| | - Marisa Márcia Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kathia Luciani
- Department of Infectious Diseases, Hospital de Especialidades Pediátricas Omar Torrijos Herrera, Caja de Seguro Social, Panama City, Panama
| | | | - Javier Díez-Domingo
- FISABIO Fundación para el Fomento Investigación Sanitaria y Biomédica de la Comunitat Valenciana, Centro de Investigación Biomédica en Red of Epidemiology and Public Health, Valencia, Spain
| | - Eduardo Lopez-Medina
- Centro de Estudios en Infectología Pediátrica, Department of Pediatrics, Universidad del Valle, Clínica Imbanaco, Grupo Quironsalud, Cali, Colombia
| | - Cristina Epalza
- Pediatric Infectious Diseases Unit, Department of Pediatrics, Hospital Universitario 12 de Octubre, Research and Clinical Trials Unit, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid, Spain
| | - Jerzy Brzostek
- Oddział Dziecięcy, Zespół Opieki Zdrowotnej w Dębicy, Dębica
| | - Henryk Szymański
- Department of Pediatrics, St Hedwig of Silesia Hospital, Trzebnica, Poland
| | - François D Boucher
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
| | - Benhur S Cetin
- Department of Pediatric Infectious Diseases, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Tirza De Leon
- Department of Vaccines, Cevaxin Sede David, Chiriquí, Panama
| | - Ener Cagri Dinleyici
- Department of Pediatrics, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Miguel Ángel Marín Gabriel
- Departamento de Pediatría, Hospital Universitario Puerta de Hierro-Majadahonda, Departamento de Pediatría, Universidad Autónoma de Madrid, Madrid, Spain
| | - Tolga Ince
- Department of Social Pediatrics, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | | | - Joanne M Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK Health and Nova Scotia Health, Halifax, Canada
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases Section, Pediatrics Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela
- Vaccines, Infections and Pediatrics Research Group, Healthcare Research Institute of Santiago de Compostela, Santiago de Compostela
- Centro de Investigación Biomédica en Red of Respiratory Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Mika Rämet
- Vaccine Research Center, Tampere University, Tampere, Finland
| | - Ernest Kuchar
- Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Warsaw, Poland
| | - Jorge Pinto
- Department of Pediatrics, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thanyawee Puthanakit
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Fernando Baquero-Artigao
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales, Hospital Universitario Infantil La Paz, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, ISCIII, Madrid, Spain
| | - Guido Castelli Gattinara
- Centro Vaccinazioni, Dipartimento Pediatrico Universitario Ospedaliero, Istituti di Ricovero e Cura a Carattere Scientifico, Ospedale Pediatrico Bambino Gesù, Lazio, Rome, Italy
| | | | - Jose Tomas Ramos Amador
- Department of Pediatrics, Universidad Complutense–Instituto de Investigación Sanitaria del Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Madrid, Spain
| | - Leszek Szenborn
- Department of Pediatrics and Infectious Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Bruce Tapiero
- Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal, Canada
| | - Evan J Anderson
- Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - James D Campbell
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Saul N Faust
- National Institute for Health and Care Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton National Health Service Foundation Trust, and Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | | | | | - Wenji Pu
- GSK, Biostatistics, Rockville, Maryland
| | | | | | | | | | | | | |
Collapse
|
7
|
Ulaszewska M, Merelie S, Sebastian S, Lambe T. Preclinical immunogenicity of an adenovirus-vectored vaccine for herpes zoster. Hum Vaccin Immunother 2023; 19:2175558. [PMID: 36785938 PMCID: PMC10026912 DOI: 10.1080/21645515.2023.2175558] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Herpes zoster (HZ) results from waning immunity following childhood infection with varicella zoster virus (VZV) but is preventable by vaccination with recombinant HZ vaccine or live HZ vaccine (two doses or one dose, respectively). Vaccine efficacy declines with age, live HZ vaccine is contraindicated in immunosuppressed individuals, and severe local reactogenicity of recombinant HZ vaccine is seen in up to 20% of older adults, indicating a potential need for new vaccines. Nonreplicating chimpanzee adenovirus (ChAd) vectors combine potent immunogenicity with well-established reactogenicity and safety profiles. We evaluated the cellular and humoral immunogenicity of ChAdOx1 encoding VZV envelope glycoprotein E (ChAdOx1-VZVgE) in mice using IFN-γ ELISpot, flow cytometry with intracellular cytokine staining, and ELISA. In outbred CD-1 mice, one dose of ChAdOx1-VZVgE (1 × 107 infectious units) elicited higher gE-specific T cell responses than two doses of recombinant HZ vaccine (1 µg) or one dose of live HZ vaccine (1.3 × 103 plaque-forming units). Antibody responses were higher with two doses of recombinant HZ vaccine than with two doses of ChAdOx1-VZVgE or one dose of live HZ vaccine. ChAdOx1-VZVgE boosted T cell and antibody responses following live HZ vaccine priming. The frequencies of polyfunctional CD4+ and CD8+ T cells expressing more than one cytokine (IFN-γ, TNF-α and IL-2) were higher with ChAdOx1-VZVgE than with the conventional vaccines. Results were similar in young and aged BALB/c mice. These findings support the clinical development of ChAdOx1-VZVgE for prevention of HZ in adults aged 50 years or over, including those who have already received conventional vaccines.
Collapse
Affiliation(s)
- Marta Ulaszewska
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Merelie
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Teresa Lambe
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Dogadov DI, Kyuregyan KK, Alexandra GM, Minosyan AA, Kochkonyan AA, Karlsen AA, Vyshemirsky OI, Karal-Ogly DD, Mikhailov MI. Markers of antroponotic viral infections in vervet monkeys arrived from their natural habitat (Tanzania). Vopr Virusol 2023; 68:394-403. [PMID: 38156576 DOI: 10.36233/0507-4088-188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 12/30/2023]
Abstract
INTRODUCTION Various human viruses have been identified in wild monkeys and in captive primates. Cases of transmission of viruses from wild monkeys to humans and vice versa are known. The aim of this study was to identify markers of anthroponotic viral infections in vervet monkeys (Chlorocebus pygerythrus) arrived from their natural habitat (Tanzania). MATERIALS AND METHODS Fecal samples (n = 56) and blood serum samples (n = 75) obtained from 75 animals, respectively, on days 10 and 23 after admission to the primate center, were tested for the markers of anthroponotic viral infections (Ebola virus, Marburg virus, lymphocytic choriomeningitis, hepatitis C virus, herpes simplex virus (HSV), cytomegalovirus (CMV), Epstein-Barr virus (EBV), parainfluenza types 1 and 3, intestinal adenoviruses, rotaviruses) by enzyme immunoassay (ELISA) and polymerase chain reaction (PCR). RESULTS AND DISCUSSION Among the examined animals, markers of 6 out of 11 tested viral infections were identified. Detection rates of IgG antibodies to HSV-1,2 (15.9%) and CMV (15.9%) were two times as low as IgG antibodies to EBV (31.8%). Among the markers of respiratory viral infections, IgG antibodies to parainfluenza virus type 1 were found (6.8%). 14.3% of the animals had rotavirus antigen, and 94% had simian adenovirus DNA. Markers of hemorrhagic fevers Ebola, Marburg, LCM, hepatitis C, and type 3 parainfluenza were not detected. CONCLUSION When importing monkeys from different regions of the world, an expanded screening for viral infections is needed considering the epidemiological situation both in the country of importation and in the country of destination.
Collapse
Affiliation(s)
- D I Dogadov
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - K K Kyuregyan
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| | - G M Alexandra
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - A A Minosyan
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - A A Kochkonyan
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - A A Karlsen
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| | - O I Vyshemirsky
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - D D Karal-Ogly
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia
| | - M I Mikhailov
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| |
Collapse
|
9
|
Gould S, Wrzesinski C, Stebbings R, Segal L. Report from an ICT 2022 workshop on toxicology for Covid19 vaccines: Industry, regulatory and CRO perspectives. Vaccine 2023; 41:5919-5924. [PMID: 37633751 DOI: 10.1016/j.vaccine.2023.08.040] [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: 05/15/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/28/2023]
Abstract
The Covid pandemic took the world by surprise in late 2019 and the need for rapid development of vaccines became paramount. The challenge was how to accelerate standard vaccine development times as much as possible. With knowledge of the genetic code of SARsCOV2, vaccine manufacturers throughout the world have risen to the challenge and several new vaccines were rapidly developed for emergency use. In March 2020, global Regulatory Authorities met to consider how to start early clinical trials and accept rolling submissions. Before use in clinical trials or any mass vaccination campaigns, the safety of the candidate vaccine needs to be evaluated. Non-clinical toxicology studies are required as an important part of vaccine safety evaluation. The extent of the toxicology evaluation prior to the start of clinical trials depended on several factors, including: the type of the candidate vaccine as well as already available supportive information with the candidate vaccine or similar vaccine types. For vaccine candidates with pre-existing data, this would save valuable time whilst a full toxicology evaluation was completed in parallel. For vaccines with more limited data, toxicology data was required before clinical development could start. This workshop examined the nonclinical toxicology studies for new Covid vaccines from the perspectives of: Vaccine manufacturers with different vaccine technologies, managing global regulatory submissions/responses; CROs, managing the urgency of conducting and reporting studies and supporting new players in the vaccine world; and Regulatory Authorities, in supporting the review process, juggling the need for safety and quality with mounting pressure to approve vaccines.
Collapse
|
10
|
Mancuso DM, Gainor K, Dore KM, Gallagher CA, Cruz K, Beierschmitt A, Malik YS, Ghosh S. Detection and Molecular Characterization of Adenoviruses in Captive and Free-Roaming African Green Monkeys ( Chlorocebus sabaeus): Evidence for Possible Recombination and Cross-Species Transmission. Viruses 2023; 15:1605. [PMID: 37515291 PMCID: PMC10385324 DOI: 10.3390/v15071605] [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: 06/14/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
In the present study, 31 samples (12 fecal, 9 nasal and 10 rectal swabs) from 28/92 (30.43%, 10 captive and 18 free-roaming African green monkeys (AGMs, Chlorocebus sabaeus)) apparently healthy AGMs in the Caribbean Island of St. Kitts tested positive for adenoviruses (AdVs) by DNA-dependent DNA polymerase (pol)-, or hexon-based screening PCR assays. Based on analysis of partial deduced amino acid sequences of Pol- and hexon- of nine AGM AdVs, at least two AdV genetic variants (group-I: seven AdVs with a Simian mastadenovirus-F (SAdV-F)/SAdV-18-like Pol and hexon, and group-II: two AdVs with a SAdV-F/SAdV-18-like Pol and a Human mastadenovirus-F (HAdV-F)/HAdV-40-like hexon) were identified, which was corroborated by analysis of the nearly complete putative Pol, complete hexon, and partial penton base sequences of a representative group-I (strain KNA-08975), and -II (KNA-S6) AdV. SAdV-F-like AdVs were reported for the first time in free-roaming non-human primates (NHPs) and after ~six decades from captive NHPs. Molecular characterization of KNA-S6 (and the other group-II AdV) indicated possible recombination and cross-species transmission events involving SAdV-F-like and HAdV-F-like viruses, corroborating the hypothesis that the evolutionary pathways of HAdVs and SAdVs are intermingled, complicated by recombination and inter-species transmission events, especially between related AdV species, such as HAdV-F and SAdV-F. To our knowledge, this is the first report on detection and molecular characterization of AdVs in AGMs.
Collapse
Affiliation(s)
- Diana M Mancuso
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Kerry Gainor
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Kerry M Dore
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
- National Coordinator, CABI/GEF/UNEP Regional Project-'Preventing the COSTS of Invasive Alien Species in Barbados and OECS Countries' in St. Kitts, Ministry of Environment, Climate Action and Constituency Development, Basseterre 00265, Saint Kitts and Nevis
| | - Christa A Gallagher
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Katalina Cruz
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Amy Beierschmitt
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
- Behavioral Science Foundation, Estridge Estate, Basseterre P.O. Box 428, Saint Kitts and Nevis
| | - Yashpal S Malik
- College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Science University, Ludhiana 141012, India
| | - Souvik Ghosh
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| |
Collapse
|
11
|
Zhang C, Berg A, Joe CCD, Dalby PA, Douglas AD. Lyophilization to enable distribution of ChAdOx1 and ChAdOx2 adenovirus-vectored vaccines without refrigeration. NPJ Vaccines 2023; 8:85. [PMID: 37277337 PMCID: PMC10240132 DOI: 10.1038/s41541-023-00674-2] [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: 01/10/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023] Open
Abstract
Distribution of vaccines which require refrigerated or frozen storage can be challenging and expensive. The adenovirus vector platform has been widely used for COVID-19 vaccines while several further candidate vaccines using the platform are in clinical development. In current liquid formulations, adenoviruses require distribution at 2-8 °C. The development of formulations suitable for ambient temperature distribution would be advantageous. Previous peer-reviewed reports of adenovirus lyophilization are relatively limited. Here, we report the development of a formulation and process for lyophilization of simian adenovirus-vectored vaccines based on the ChAdOx1 platform. We describe the iterative selection of excipients using a design of experiments approach, and iterative cycle improvement to achieve both preservation of potency and satisfactory cake appearance. The resulting method achieved in-process infectivity titre loss of around 50%. After drying, there was negligible further loss over a month at 30 °C. Around 30% of the predrying infectivity remained after a month at 45 °C. This performance is likely to be suitable for 'last leg' distribution at ambient temperature. This work may also facilitate the development of other product presentations using dried simian adenovirus-vectored vaccines.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London, WC1E 6BT, United Kingdom
| | - Adam Berg
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, OX3 7DQ, Oxford, United Kingdom
| | - Carina C D Joe
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, OX3 7DQ, Oxford, United Kingdom
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London, WC1E 6BT, United Kingdom
| | - Alexander D Douglas
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, OX3 7DQ, Oxford, United Kingdom.
| |
Collapse
|
12
|
Saunders JE, Gilbride C, Dowall S, Morris S, Ulaszewska M, Spencer AJ, Rayner E, Graham VA, Kennedy E, Thomas K, Hewson R, Gilbert SC, Belij-Rammerstorfer S, Lambe T. Adenoviral vectored vaccination protects against Crimean-Congo Haemorrhagic Fever disease in a lethal challenge model. EBioMedicine 2023; 90:104523. [PMID: 36933409 PMCID: PMC10025009 DOI: 10.1016/j.ebiom.2023.104523] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage. METHODS In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV. FINDINGS We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease. INTERPRETATION There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV. FUNDING This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1].
Collapse
Affiliation(s)
- Jack E Saunders
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Ciaran Gilbride
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stuart Dowall
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Susan Morris
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Emma Rayner
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Victoria A Graham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Emma Kennedy
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Kelly Thomas
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Roger Hewson
- UK Health Security Agency (UKHSA), Porton Down, Salisbury, Wiltshire, UK
| | - Sarah C Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Belij-Rammerstorfer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Beck A, Dietenberger H, Kunz SN, Mellert K, Möller P. Emergence of SARS-CoV-2 spike protein at the vaccination site. Immun Inflamm Dis 2023; 11:e827. [PMID: 36988249 PMCID: PMC10052447 DOI: 10.1002/iid3.827] [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: 10/10/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The anti-coronavirus disease 2019 (COVID-19) vaccines are of paramount importance in the fight against the COVID-19 pandemic. Both viral vector- and nucleic acid-based vaccines are known to effectively induce protection against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus by generating high antibody titers and effective T-cell responses to the spike protein they encode. Although these vaccines are being applied worldwide and have been extensively investigated, the immunomorphological events at the vaccination site with respect to SARS-CoV-2 spike protein expression have not yet been described. METHODS We had the opportunity to examine the deltoid muscles of three men who died shortly after vaccination for unrelated reasons. We examined the vaccination sites histologically and immunohistochemically with various antibodies. Furthermore we incubated two different cell lines with one vaccine and examined the expression of the spike protein. RESULTS The vaccination sites show a dense lymphohistiocytic interstitial infiltrate which surrounds the small vessels and extends into the perimysium. The spike protein is expressed by histiocytic cells with a dendritic shape that are CD68-positive and CD207-negative, fibrocytes, and very rare S100-positive cells. Interestingly, the skeletal muscle, being constitutively human leukocyte antigen (HLA)-A,B,C-negative, is induced at different levels in each specimen. In a cell culture experiment, we confirmed the ability of fibroblasts and interdigitating dendritic sarcoma cells to express spike protein in vitro after incubation with the Comirnaty vaccine. CONCLUSIONS Histiocytic cells and fibrocytes are the heralds of spike protein synthesis at the vaccination site. The underlying cause of this apparent cell specifity is unknown. This needs to be investigated in future experiments, for example in an animal model.
Collapse
Affiliation(s)
- Annika Beck
- Institute of PathologyUniversity Hospital of UlmUlmGermany
| | | | | | - Kevin Mellert
- Institute of PathologyUniversity Hospital of UlmUlmGermany
| | - Peter Möller
- Institute of PathologyUniversity Hospital of UlmUlmGermany
| |
Collapse
|
14
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
15
|
Vatzia E, Feest K, McNee A, Manjegowda T, Carr BV, Paudyal B, Chrun T, Maze EA, Mccarron A, Morris S, Everett HE, MacLoughlin R, Salguero FJ, Lambe T, Gilbert SC, Tchilian E. Immunization with matrix-, nucleoprotein and neuraminidase protects against H3N2 influenza challenge in pH1N1 pre-exposed pigs. NPJ Vaccines 2023; 8:19. [PMID: 36792640 PMCID: PMC9930017 DOI: 10.1038/s41541-023-00620-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
There is an urgent need for influenza vaccines providing broader protection that may decrease the need for annual immunization of the human population. We investigated the efficacy of heterologous prime boost immunization with chimpanzee adenovirus (ChAdOx2) and modified vaccinia Ankara (MVA) vectored vaccines, expressing conserved influenza virus nucleoprotein (NP), matrix protein 1 (M1) and neuraminidase (NA) in H1N1pdm09 pre-exposed pigs. We compared the efficacy of intra-nasal, aerosol and intra-muscular vaccine delivery against H3N2 influenza challenge. Aerosol prime boost immunization induced strong local lung T cell and antibody responses and abrogated viral shedding and lung pathology following H3N2 challenge. In contrast, intramuscular immunization induced powerful systemic responses and weak local lung responses but also abolished lung pathology and reduced viral shedding. These results provide valuable insights into the development of a broadly protective influenza vaccine in a highly relevant large animal model and will inform future vaccine and clinical trial design.
Collapse
Affiliation(s)
- Eleni Vatzia
- The Pirbright Institute, Pirbright, United Kingdom.
| | | | - Adam McNee
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | | | | | | | | - Amy Mccarron
- The Pirbright Institute, Pirbright, United Kingdom
| | - Susan Morris
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Helen E Everett
- Animal and Plant Health Agency-Weybridge, New Haw, Addlestone, United Kingdom
| | | | - Francisco J Salguero
- United Kingdom Health Security Agency, UKHSA-Porton Down, Salisbury, United Kingdom
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Sarah C Gilbert
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
16
|
A Review on Zoonotic Pathogens Associated with Non-Human Primates: Understanding the Potential Threats to Humans. Microorganisms 2023; 11:microorganisms11020246. [PMID: 36838210 PMCID: PMC9964884 DOI: 10.3390/microorganisms11020246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Non-human primates (NHP) share a close relationship with humans due to a genetic homology of 75-98.5%. NHP and humans have highly similar tissue structures, immunity, physiology, and metabolism and thus often can act as hosts to the same pathogens. Agriculture, meat consumption habits, tourism development, religious beliefs, and biological research have led to more extensive and frequent contact between NHPs and humans. Deadly viruses, such as rabies virus, herpes B virus, Marburg virus, Ebola virus, human immunodeficiency virus, and monkeypox virus can be transferred from NHP to humans. Similarly, herpes simplex virus, influenza virus, and yellow fever virus can be transmitted to NHP from humans. Infectious pathogens, including viruses, bacteria, and parasites, can affect the health of both primates and humans. A vast number of NHP-carrying pathogens exhibit a risk of transmission to humans. Therefore, zoonotic infectious diseases should be evaluated in future research. This article reviews the research evidence, diagnostic methods, prevention, and treatment measures that may be useful in limiting the spread of several common viral pathogens via NHP and providing ideas for preventing zoonotic diseases with epidemic potential.
Collapse
|
17
|
Araújo NM, Rubio IGS, Toneto NPA, Morale MG, Tamura RE. The use of adenoviral vectors in gene therapy and vaccine approaches. Genet Mol Biol 2022; 45:e20220079. [PMID: 36206378 PMCID: PMC9543183 DOI: 10.1590/1678-4685-gmb-2022-0079] [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: 03/06/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Adenovirus was first identified in the 1950s and since then this pathogenic group
of viruses has been explored and transformed into a genetic transfer vehicle.
Modification or deletion of few genes are necessary to transform it into a
conditionally or non-replicative vector, creating a versatile tool capable of
transducing different tissues and inducing high levels of transgene expression.
In the early years of vector development, the application in monogenic diseases
faced several hurdles, including short-term gene expression and even a fatality.
On the other hand, an adenoviral delivery strategy for treatment of cancer was
the first approved gene therapy product. There is an increasing interest in
expressing transgenes with therapeutic potential targeting the cancer hallmarks,
inhibiting metastasis, inducing cancer cell death or modulating the immune
system to attack the tumor cells. Replicative adenovirus as vaccines may be even
older and date to a few years of its discovery, application of non-replicative
adenovirus for vaccination against different microorganisms has been
investigated, but only recently, it demonstrated its full potential being one of
the leading vaccination tools for COVID-19. This is not a new vector nor a new
technology, but the result of decades of careful and intense work in this
field.
Collapse
Affiliation(s)
- Natália Meneses Araújo
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil.
| | - Ileana Gabriela Sanchez Rubio
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | | | - Mirian Galliote Morale
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil. ,Universidade Federal de São Paulo, Laboratório de Ciências
Moleculares da Tireóide, Diadema, SP, Brazil.
| | - Rodrigo Esaki Tamura
- Universidade Federal de São Paulo, Laboratório de Biologia Molecular
do Câncer, São Paulo, SP, Brazil. ,Universidade Federal de São Paulo, Departamento de Ciências
Biológicas, Diadema, SP, Brazil.
| |
Collapse
|
18
|
Jenkin D, Ritchie AJ, Aboagye J, Fedosyuk S, Thorley L, Provstgaad-Morys S, Sanders H, Bellamy D, Makinson R, Xiang ZQ, Bolam E, Tarrant R, Ramos Lopez F, Platt A, Poulton I, Green C, Ertl HCJ, Ewer KJ, Douglas AD. Safety and immunogenicity of a simian-adenovirus-vectored rabies vaccine: an open-label, non-randomised, dose-escalation, first-in-human, single-centre, phase 1 clinical trial. THE LANCET. MICROBE 2022; 3:e663-e671. [PMID: 35907430 PMCID: PMC7614839 DOI: 10.1016/s2666-5247(22)00126-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Rabies kills around 60 000 people each year. ChAdOx2 RabG, a simian adenovirus-vectored rabies vaccine candidate, might have potential to provide low-cost single-dose pre-exposure rabies prophylaxis. This first-in-human study aimed to evaluate its safety and immunogenicity in healthy adults. METHODS We did a single-centre phase 1 study of ChAdOx2 RabG, administered as a single intramuscular dose, with non-randomised open-label dose escalation at the Centre for Clinical Vaccinology and Tropical Medicine, Oxford, UK. Healthy adults were sequentially allocated to groups receiving low (5 × 109 viral particles), middle (2·5 × 1010 viral particles), and high doses (5 x 1010 viral particles) of ChAdOx2 RabG and were followed up to day 56 after vaccination. The primary objective was to assess safety. The secondary objective was to assess immunogenicity with the internationally standardised rabies virus neutralising antibody assay. In an optional follow-up phase 1 year after enrolment, we measured antibody maintenance then administered a licensed rabies vaccine (to simulate post-exposure prophylaxis) and measured recall responses. The trial is registered with ClinicalTrials.gov, NCT04162600, and is now closed to new participants. FINDINGS Between Jan 2 and Oct 28, 2020, 12 adults received low (n=3), middle (n=3), and high doses (n=6) of ChAdOx2 RabG. Participants reported predominantly mild-to-moderate reactogenicity. There were no serious adverse events. Virus neutralising antibody concentrations exceeded the recognised correlate of protection (0·5 IU/mL) in three middle-dose recipients and six high-dose recipients within 56 days of vaccination (median 18·0 IU/mL). The median peak virus neutralising antibody concentrations within 56 days were 0·7 IU/mL (range 0·0-54·0 IU/mL) for the low-dose group, 18·0 IU/mL (0·7-18·0 IU/mL) for the middle-dose group, and 18·0 IU/mL (6·0-486·0 IU/mL) for the high-dose group. Nine participants returned for the additional follow-up after 1 year. Of these nine participants, virus neutralising antibody titres of more than 0·5 IU/mL were maintained in six of seven who had received middle-dose or high-dose ChAdOx2 RabG. Within 7 days of administration of the first dose of a licensed rabies vaccine, nine participants had virus neutralising antibody titres of more than 0·5 IU/mL. INTERPRETATION In this study, ChAdOx2 RabG showed an acceptable safety and tolerability profile and encouraging immunogenicity, supporting further clinical evaluation. FUNDING UK Medical Research Council and Engineering and Physical Sciences Research Council.
Collapse
Affiliation(s)
- Daniel Jenkin
- Jenner Institute, University of Oxford, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | | | | | | | - Luke Thorley
- Jenner Institute, University of Oxford, Oxford, UK
| | | | | | | | | | - Zhi Quan Xiang
- Wistar Institute of Anatomy & Biology, Philadelphia, PA, USA
| | - Emma Bolam
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | - Richard Tarrant
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | - Fernando Ramos Lopez
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Abigail Platt
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Ian Poulton
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Catherine Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | | | - Katie J Ewer
- Jenner Institute, University of Oxford, Oxford, UK
| | | |
Collapse
|
19
|
Chavda VP, Chen Y, Dave J, Chen ZS, Chauhan SC, Yallapu MM, Uversky VN, Bezbaruah R, Patel S, Apostolopoulos V. COVID-19 and vaccination: myths vs science. Expert Rev Vaccines 2022; 21:1603-1620. [PMID: 35980281 DOI: 10.1080/14760584.2022.2114900] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Several vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been developed since the inception of the coronavirus disease 2019 (COVID-19) in December 2019, at unprecedented speed. However, these rapidly developed vaccines raised many questions related to the efficacy and safety of vaccines in different communities across the globe. Various hypotheses regarding COVID-19 and its vaccines were generated, and many of them have also been answered with scientific evidence. Still, there are many myths/misinformation related to COVID-19 and its vaccines, which create hesitancy for COVID-19 vaccination, and must be addressed critically to achieve success in the battle against the pandemic. AREA COVERED The development of anti-SARS-CoV-2 vaccines against COVID-19, their safety and efficacy, and myths/misinformation relating to COVID-19 and vaccines are presented. EXPERT OPINION In this pandemic we have seen a global collaborative effort of researchers, governments, and industry, supported by billions of dollars in funding, have allowed the development of vaccines far more quickly than in the past. Vaccines go through rigorous testing, analysis, and evaluations in clinical settings prior to their approval, even if they are approved for emergency use. Despite the myths, vaccination represents an important strategy to get back to normality.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad
| | - Yangmin Chen
- Peter J. Tobin College of Business, St. John's University, Queens, NY 11439, USA
| | - Jayant Dave
- Department of Pharmaceutical Quality Assurance, L.M. College of Pharmacy, Ahmedabad
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institure, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh -786004, Assam, India
| | - Sandip Patel
- Department of Pharmacology, L.M. College of Pharmacy, Ahmedabad
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Immunology and Translational Research Group, Victoria University, Melbourne, VIC, 3030, Australia.,Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC, 3021, Australia
| |
Collapse
|
20
|
Lu ZH, Li J, Dmitriev IP, Kashentseva EA, Curiel DT. Efficient Genome Editing Achieved via Plug-and-Play Adenovirus Piggyback Transport of Cas9/gRNA Complex on Viral Capsid Surface. ACS NANO 2022; 16:10443-10455. [PMID: 35749339 PMCID: PMC9330763 DOI: 10.1021/acsnano.2c00909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The capacity to efficiently deliver the gene-editing enzyme complex to target cells is favored over other forms of gene delivery as it offers one-time hit-and-run gene editing, thus improving precision and safety and reducing potential immunogenicity against edited cells in clinical applications. Here we performed a proof-of-mechanism study and demonstrated that a simian adenoviral vector for DNA delivery can be repurposed as a robust intracellular delivery platform for a functional Cas9/guide RNA (gRNA) complex to recipient cells. In this system, the clinically relevant adenovirus was genetically engineered with a plug-and-display technology based on SpyTag003/SpyCatcher003 coupling chemistry. Under physiological conditions, an off-the-shelf mixture of viral vector with SpyTag003 incorporated into surface capsid proteins and Cas9 fused with SpyCatcher003 led to a rapid titration reaction yielding adenovirus carrying Cas9SpyCatcher003 on the virus surface. The Cas9 fusion protein-conjugated viruses in the presence of a reporter gRNA delivered gene-editing functions to cells with an efficiency comparable to that of a commercial CRISPR/Cas9 transfection reagent. Our data fully validate the adenoviral "piggyback" approach to deliver an intracellularly acting enzyme cargo and, thus, warrant the prospect of engineering tissue-targeted adenovirus carrying Cas9/gRNA for in vivo gene editing.
Collapse
Affiliation(s)
- Zhi Hong Lu
- Department of Radiation Oncology,
Biologic Therapeutics Center, Washington
University School of Medicine, 660 South Euclid Avenue, Campus Box 8224, St. Louis, Missouri 63110, United States
| | - Jie Li
- Department of Radiation Oncology,
Biologic Therapeutics Center, Washington
University School of Medicine, 660 South Euclid Avenue, Campus Box 8224, St. Louis, Missouri 63110, United States
| | - Igor P. Dmitriev
- Department of Radiation Oncology,
Biologic Therapeutics Center, Washington
University School of Medicine, 660 South Euclid Avenue, Campus Box 8224, St. Louis, Missouri 63110, United States
| | - Elena A. Kashentseva
- Department of Radiation Oncology,
Biologic Therapeutics Center, Washington
University School of Medicine, 660 South Euclid Avenue, Campus Box 8224, St. Louis, Missouri 63110, United States
| | - David T. Curiel
- Department of Radiation Oncology,
Biologic Therapeutics Center, Washington
University School of Medicine, 660 South Euclid Avenue, Campus Box 8224, St. Louis, Missouri 63110, United States
| |
Collapse
|
21
|
Immunity after COVID-19 Recovery and Vaccination: Similarities and Differences. Vaccines (Basel) 2022; 10:vaccines10071068. [PMID: 35891232 PMCID: PMC9322013 DOI: 10.3390/vaccines10071068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is associated with a robust immune response. The development of systemic inflammation leads to a hyperinflammatory state due to cytokine release syndrome during severe COVID-19. The emergence of many new SARS-CoV-2 variants across the world deteriorates the protective antiviral immunity induced after infection or vaccination. The innate immune response to SARS-CoV-2 is crucial for determining the fate of COVID-19 symptomatology. T cell-mediated immunity is the main factor of the antiviral immune response; moreover, SARS-CoV-2 infection initiates a rapid B-cell response. In this paper, we present the current state of knowledge on immunity after COVID-19 infection and vaccination. We discuss the mechanisms of immune response to various types of vaccines (nucleoside-modified, adenovirus-vectored, inactivated virus vaccines and recombinant protein adjuvanted formulations). This includes specific aspects of vaccination in selected patient populations with altered immune activity (the elderly, children, pregnant women, solid organ transplant recipients, patients with systemic rheumatic diseases or malignancies). We also present diagnostic and research tools available to study the anti-SARS-CoV-2 cellular and humoral immune responses.
Collapse
|
22
|
Sluder AE, Raju Paul S, Moise L, Dold C, Richard G, Silva-Reyes L, Baeten LA, Scholzen A, Reeves PM, Pollard AJ, Garritsen A, Bowen RA, De Groot AS, Rollier C, Poznansky MC. Evaluation of a Human T Cell-Targeted Multi-Epitope Vaccine for Q Fever in Animal Models of Coxiella burnetii Immunity. Front Immunol 2022; 13:901372. [PMID: 35651616 PMCID: PMC9149306 DOI: 10.3389/fimmu.2022.901372] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-mediated immunity plays a central role in the control and clearance of intracellular Coxiella burnetii infection, which can cause Q fever. Therefore, we aimed to develop a novel T cell-targeted vaccine that induces pathogen-specific cell-mediated immunity to protect against Q fever in humans while avoiding the reactogenicity of the current inactivated whole cell vaccine. Human HLA class II T cell epitopes from C. burnetii were previously identified and selected by immunoinformatic predictions of HLA binding, conservation in multiple C. burnetii isolates, and low potential for cross-reactivity with the human proteome or microbiome. Epitopes were selected for vaccine inclusion based on long-lived human T cell recall responses to corresponding peptides in individuals that had been naturally exposed to the bacterium during a 2007-2010 Q fever outbreak in the Netherlands. Multiple viral vector-based candidate vaccines were generated that express concatemers of selected epitope sequences arranged to minimize potential junctional neo-epitopes. The vaccine candidates caused no antigen-specific reactogenicity in a sensitized guinea pig model. A subset of the vaccine epitope peptides elicited antigenic recall responses in splenocytes from C57BL/6 mice previously infected with C. burnetii. However, immunogenicity of the vaccine candidates in C57BL/6 mice was dominated by a single epitope and this was insufficient to confer protection against an infection challenge, highlighting the limitations of assessing human-targeted vaccine candidates in murine models. The viral vector-based vaccine candidates induced antigen-specific T cell responses to a broader array of epitopes in cynomolgus macaques, establishing a foundation for future vaccine efficacy studies in this large animal model of C. burnetii infection.
Collapse
Affiliation(s)
- Ann E Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | - Susan Raju Paul
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | | | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Laurie A Baeten
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Patrick M Reeves
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | - Richard A Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Christine Rollier
- Oxford Vaccine Group, Department of Paediatrics, The National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
23
|
Park H, Park MS, Seok JH, You J, Kim J, Kim J, Park MS. Insights into the immune responses of SARS-CoV-2 in relation to COVID-19 vaccines. J Microbiol 2022; 60:308-320. [PMID: 35235179 PMCID: PMC8890016 DOI: 10.1007/s12275-022-1598-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022]
Abstract
The three types of approved coronavirus disease 2019 (COVID-19) vaccines that have been emergency-use listed (EUL) by the World Health Organization are mRNA vaccines, adenovirus-vectored vaccines, and inactivated vaccines. Canonical vaccine developments usually take years or decades to be completed to commercialization; however, the EUL vaccines being used in the current situation comprise several COVID-19 vaccine candidates applied in studies and clinical settings across the world. The extraordinary circumstances of the COVID-19 pandemic have necessitated the emergency authorization of these EUL vaccines, which have been rapidly developed. Although the benefits of the EUL vaccines outweigh their adverse effects, there have been reports of rare but fatal cases directly associated with COVID-19 vaccinations. Thus, a reassessment of the immunological rationale underlying EUL vaccines in relation to COVID-19 caused by SARSCOV-2 virus infection is now required. In this review, we discuss the manifestations of COVID-19, immunologically projected effects of EUL vaccines, reported immune responses, informed issues related to COVID-19 vaccination, and the potential strategies for future vaccine use against antigenic variants.
Collapse
Affiliation(s)
- Heedo Park
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Mee Sook Park
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jong Hyeon Seok
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jaehwan You
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jineui Kim
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jeonghun Kim
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Chung Mong-Koo Vaccine Innovation Center, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
24
|
Booty MG, Hlavaty KA, Stockmann A, Ozay EI, Smith C, Tian L, How E, Subramanya D, Venkitaraman A, Yee C, Pryor O, Volk K, Blagovic K, Vicente-Suarez I, Yarar D, Myint M, Merino A, Chow J, Abdeljawad T, An H, Liu S, Mao S, Heimann M, Talarico L, Jacques MK, Chong E, Pomerance L, Gonzalez JT, von Andrian UH, Jensen KF, Langer R, Knoetgen H, Trumpfheller C, Umaña P, Bernstein H, Sharei A, Loughhead SM. Microfluidic Squeezing Enables MHC Class I Antigen Presentation by Diverse Immune Cells to Elicit CD8 + T Cell Responses with Antitumor Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:929-940. [PMID: 35091434 PMCID: PMC9012083 DOI: 10.4049/jimmunol.2100656] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/07/2021] [Indexed: 12/30/2022]
Abstract
CD8+ T cell responses are the foundation of the recent clinical success of immunotherapy in oncologic indications. Although checkpoint inhibitors have enhanced the activity of existing CD8+ T cell responses, therapeutic approaches to generate Ag-specific CD8+ T cell responses have had limited success. Here, we demonstrate that cytosolic delivery of Ag through microfluidic squeezing enables MHC class I presentation to CD8+ T cells by diverse cell types. In murine dendritic cells (DCs), squeezed DCs were ∼1000-fold more potent at eliciting CD8+ T cell responses than DCs cross-presenting the same amount of protein Ag. The approach also enabled engineering of less conventional APCs, such as T cells, for effective priming of CD8+ T cells in vitro and in vivo. Mixtures of immune cells, such as murine splenocytes, also elicited CD8+ T cell responses in vivo when squeezed with Ag. We demonstrate that squeezing enables effective MHC class I presentation by human DCs, T cells, B cells, and PBMCs and that, in clinical scale formats, the system can squeeze up to 2 billion cells per minute. Using the human papillomavirus 16 (HPV16) murine model, TC-1, we demonstrate that squeezed B cells, T cells, and unfractionated splenocytes elicit antitumor immunity and correlate with an influx of HPV-specific CD8+ T cells such that >80% of CD8s in the tumor were HPV specific. Together, these findings demonstrate the potential of cytosolic Ag delivery to drive robust CD8+ T cell responses and illustrate the potential for an autologous cell-based vaccine with minimal turnaround time for patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Harry An
- SQZ Biotechnologies, Watertown, MA
| | - Sophia Liu
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Shirley Mao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Megan Heimann
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | | | | | | | | | | | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA
- Center for Immune Imaging at Harvard Medical School, Boston, MA
| | - Klavs F Jensen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
- David Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Hendrik Knoetgen
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Basel, Switzerland; and
| | - Christine Trumpfheller
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development, Schlieren, Switzerland
| | | | | | | |
Collapse
|
25
|
Joe CCD, Jiang J, Linke T, Li Y, Fedosyuk S, Gupta G, Berg A, Segireddy RR, Mainwaring D, Joshi A, Cashen P, Rees B, Chopra N, Nestola P, Humphreys J, Davies S, Smith N, Bruce S, Verbart D, Bormans D, Knevelman C, Woodyer M, Davies L, Cooper L, Kapanidou M, Bleckwenn N, Pappas D, Lambe T, Smith DC, Green CM, Venkat R, Ritchie AJ, Gilbert SC, Turner R, Douglas AD. Manufacturing a chimpanzee adenovirus-vectored SARS-CoV-2 vaccine to meet global needs. Biotechnol Bioeng 2022; 119:48-58. [PMID: 34585736 PMCID: PMC8653296 DOI: 10.1002/bit.27945] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022]
Abstract
Manufacturing has been the key factor limiting rollout of vaccination during the COVID-19 pandemic, requiring rapid development and large-scale implementation of novel manufacturing technologies. ChAdOx1 nCoV-19 (AZD1222, Vaxzevria) is an efficacious vaccine against SARS-CoV-2, based upon an adenovirus vector. We describe the development of a process for the production of this vaccine and others based upon the same platform, including novel features to facilitate very large-scale production. We discuss the process economics and the "distributed manufacturing" approach we have taken to provide the vaccine at globally-relevant scale and with international security of supply. Together, these approaches have enabled the largest viral vector manufacturing campaign to date, providing a substantial proportion of global COVID-19 vaccine supply at low cost.
Collapse
Affiliation(s)
- Carina C. D. Joe
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Jinlin Jiang
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Thomas Linke
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Yuanyuan Li
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Sofiya Fedosyuk
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Gaurav Gupta
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Adam Berg
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nicole Bleckwenn
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Daniel Pappas
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Teresa Lambe
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | | | - Catherine M. Green
- Clinical Biomanufacturing Facility, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Raghavan Venkat
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Adam J. Ritchie
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Sarah C. Gilbert
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Richard Turner
- Purification Process Sciences, Biopharmaceuticals DevelopmentR&D, AstraZenecaCambridgeUK
| | | |
Collapse
|
26
|
Islam KU, A-Elgadir TME, Afaq S, Ahmad T, Iqbal J. Molecular and Clinical Aspects of COVID-19 Vaccines and Other Therapeutic Interventions Apropos Emerging Variants of Concern. Front Pharmacol 2021; 12:778219. [PMID: 35002711 PMCID: PMC8734653 DOI: 10.3389/fphar.2021.778219] [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: 09/16/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has overwhelmed the healthcare and economy of the world, with emerging new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posing an everlasting threat to humanity. While most COVID-19 vaccines provide adequate protective immunological response against the original SARS-CoV-2 variant, there is a pressing need to understand their biological and clinical responses. Recent evidence suggests that some of the new variants of SARS-CoV-2 evade the protection conferred by the existing vaccines, which may impede the ongoing efforts to expedite the vaccination programs worldwide. These concerns have also highlighted the importance of a pan-COVID-19 vaccine, which is currently in the making. Thus, it is imperative to have a better molecular and clinical understanding of the various COVID-19 vaccines and their immunological trajectory against any emerging variant of concerns (VOCs) in particular to break this vicious cycle. Furthermore, other treatment regimens based on cellular therapies and monoclonal antibodies should be explored systematically as an alternative and readily available option considering the possibility of the emergence of more virulent SARS-CoV-2 mutants. In this review, we shed light on the various molecular mechanisms and clinical responses of COVID-19 vaccines. Importantly, we review the recent findings of their long-term immune protection and efficacy against emerging VOCs. Considering that other targeted and effective treatments will complement vaccine therapy, we provide a comprehensive understanding of the role of cell-based therapies, monoclonal antibodies, and immunomodulatory agents as alternative and readily available treatment modalities against any emerging SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Khursheed Ul Islam
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | | | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Jawed Iqbal
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
27
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
28
|
Vatzia E, Allen ER, Manjegowda T, Morris S, McNee A, Martini V, Kaliath R, Ulaszewska M, Boyd A, Paudyal B, Carr VB, Chrun T, Maze E, MacLoughlin R, van Diemen PM, Everett HE, Lambe T, Gilbert SC, Tchilian E. Respiratory and Intramuscular Immunization With ChAdOx2-NPM1-NA Induces Distinct Immune Responses in H1N1pdm09 Pre-Exposed Pigs. Front Immunol 2021; 12:763912. [PMID: 34804053 PMCID: PMC8595216 DOI: 10.3389/fimmu.2021.763912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/13/2021] [Indexed: 01/12/2023] Open
Abstract
There is a critical need to develop superior influenza vaccines that provide broader protection. Influenza vaccines are traditionally tested in naive animals, although humans are exposed to influenza in the first years of their lives, but the impact of prior influenza exposure on vaccine immune responses has not been well studied. Pigs are an important natural host for influenza, are a source of pandemic viruses, and are an excellent model for human influenza. Here, we investigated the immunogenicity of the ChAdOx2 viral vectored vaccine, expressing influenza nucleoprotein, matrix protein 1, and neuraminidase in H1N1pdm09 pre-exposed pigs. We evaluated the importance of the route of administration by comparing intranasal, aerosol, and intramuscular immunizations. Aerosol delivery boosted the local lung T-cell and antibody responses, while intramuscular immunization boosted peripheral blood immunity. These results will inform how best to deliver vaccines in order to harness optimal protective immunity.
Collapse
Affiliation(s)
- Eleni Vatzia
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Elizabeth R Allen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Tanuja Manjegowda
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Susan Morris
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Adam McNee
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica Martini
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Reshma Kaliath
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amy Boyd
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Basudev Paudyal
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica B Carr
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Tiphany Chrun
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Emmanuel Maze
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Helen E Everett
- Animal and Plant Health Agency (APHA)-Weybridge, Addlestone, United Kingdom
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah C Gilbert
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Elma Tchilian
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| |
Collapse
|
29
|
Othman M, Baker AT, Gupalo E, Elsebaie A, Bliss CM, Rondina MT, Lillicrap D, Parker AL. To clot or not to clot? Ad is the question-Insights on mechanisms related to vaccine-induced thrombotic thrombocytopenia. J Thromb Haemost 2021; 19:2845-2856. [PMID: 34351057 PMCID: PMC8420166 DOI: 10.1111/jth.15485] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023]
Abstract
Vaccine-induced immune thrombotic thrombocytopenia (VITT) has caused global concern. VITT is characterized by thrombosis and thrombocytopenia following COVID-19 vaccinations with the AstraZeneca ChAdOx1 nCov-19 and the Janssen Ad26.COV2.S vaccines. Patients present with thrombosis, severe thrombocytopenia developing 5-24 days following first dose of vaccine, with elevated D-dimer, and PF4 antibodies, signifying platelet activation. As of June 1, 2021, more than 1.93 billion COVID-19 vaccine doses had been administered worldwide. Currently, 467 VITT cases (0.000024%) have been reported across the UK, Europe, Canada, and Australia. Guidance on diagnosis and management of VITT has been reported but the pathogenic mechanism is yet to be fully elucidated. Here, we propose and discuss potential mechanisms in relation to adenovirus induction of VITT. We provide insights and clues into areas warranting investigation into the mechanistic basis of VITT, highlighting the unanswered questions. Further research is required to help solidify a pathogenic model for this condition.
Collapse
Affiliation(s)
- Maha Othman
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
- School of Baccalaureate NursingSt. Lawrence CollegeKingstonOntarioCanada
| | - Alexander T. Baker
- Center for Individualized MedicineMayo ClinicScottsdaleArizonaUSA
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| | - Elena Gupalo
- National Medical Research Center for CardiologyMoscowRussia
| | - Abdelrahman Elsebaie
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
| | - Carly M. Bliss
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| | - Matthew T. Rondina
- Departments of Internal Medicine and Pathology, and the Molecular Medicine ProgramUniversity of Utah HealthSalt Lake CityUtahUSA
- Department of Internal Medicine and GRECCGeorge E. Wahlen VAMCSalt Lake CityUtahUSA
| | - David Lillicrap
- Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - Alan L. Parker
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| |
Collapse
|
30
|
Berg A, Wright D, Dulal P, Stedman A, Fedosyuk S, Francis MJ, Charleston B, Warimwe GM, Douglas AD. Stability of Chimpanzee Adenovirus Vectored Vaccines (ChAdOx1 and ChAdOx2) in Liquid and Lyophilised Formulations. Vaccines (Basel) 2021; 9:1249. [PMID: 34835180 PMCID: PMC8623940 DOI: 10.3390/vaccines9111249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/04/2022] Open
Abstract
Adenovirus vectored vaccines have entered global use during the COVID-19 pandemic, and are in development for multiple other human and veterinary applications. An attraction of the technology is the suitability of the vaccines for storage at 2-8 °C for months. Widely used COVID-19 vaccine ChAdOx1 nCoV-19 (University of Oxford/AstraZeneca) is based on a species E simian adenovirus. Species E simian serotypes have been used in a wide range of other development programs, but the stability of such vectors has not been extensively described in the peer-reviewed literature. Here, we explore the stability of two candidate vaccines based on two species E serotypes: a Rift Valley fever vaccine based upon the ChAdOx1 vector (Y25 serotype) used in ChAdOx1 nCoV-19, and a rabies vaccine based upon a ChAdOx2 vector (AdC68 serotype). We describe each vector's stability in liquid and lyophilised formulations using in vitro and in vivo potency measurements. Our data support the suitability of liquid formulations of these vectors for storage at 2-8 °C for up to 1 year, and potentially for nonrefrigerated storage for a brief period during last-leg distribution (perhaps 1-3 days at 20 °C-the precise definition of acceptable last-leg storage conditions would require further product-specific data). Depending upon the level of inprocess potency loss that is economically acceptable, and the level of instorage loss that is compatible with maintenance of acceptable end-of-storage potency, a previously reported lyophilised formulation may enable longer term storage at 20 °C or storage for a number of days at 30 °C.
Collapse
Affiliation(s)
- Adam Berg
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Daniel Wright
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Pawan Dulal
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Anna Stedman
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
| | - Sofiya Fedosyuk
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Michael J. Francis
- BioVacc Consulting Ltd., The Red House, 10 Market Square, Amersham HP7 0DQ, UK;
| | - Bryan Charleston
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
| | - George M. Warimwe
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
- KEMRI-Wellcome Trust Research Programme, Kilifi P.O. Box 230-80108, Kenya
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford OX3 7LG, UK
| | - Alexander D. Douglas
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| |
Collapse
|
31
|
Cari L, Alhosseini MN, Fiore P, Pierno S, Pacor S, Bergamo A, Sava G, Nocentini G. Cardiovascular, neurological, and pulmonary events following vaccination with the BNT162b2, ChAdOx1 nCoV-19, and Ad26.COV2.S vaccines: An analysis of European data. J Autoimmun 2021; 125:102742. [PMID: 34710832 PMCID: PMC8547775 DOI: 10.1016/j.jaut.2021.102742] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/07/2021] [Accepted: 10/19/2021] [Indexed: 12/21/2022]
Abstract
The ChAdOx1 nCoV-19 (ChA) (AstraZeneca) and Ad26.COV2.S (AD26) (Janssen) vaccines are virus-based coronavirus disease 2019 (COVID-19) vaccines used worldwide. In spring 2021, venous blood clots and thrombocytopenia were described in some vaccine recipients. We evaluated the frequency of severe adverse events (SAEs) documented in the EudraVigilance European database in young adult (18–64 years old) and older (≥65 years old) vaccine recipients up to 23 June 2021 and related them to coagulation disorders and arterial, cardiac, and nervous system events. Comparison between the frequency of SAEs and SAE-related deaths in ChA and AD26 vs. BNT162b2 COVID-19 (BNT) (Pfizer/BioNTech) vaccine recipients demonstrated: 1) ChA and AD26 recipients than BNT recipients had higher frequencies of not only SAEs caused by venous blood clots and hemorrhage, but also thromboembolic disease and arterial events, including myocardial infarction and stroke; 2) a corresponding higher frequency of SAE-related deaths. The frequency was higher in both young adults and older adults. Comparison between the frequency of SAEs and SAE-related deaths in AD26 vs. ChA recipients demonstrated in AD26 recipients: 1) lower frequency of thrombocytopenia; 2) lower frequency of SAEs in young adult recipients; 3) higher frequency of SAEs in older recipients. Interestingly, most of the venous thrombotic SAEs associated with ChA and AD26 vaccines were not associated with thrombocytopenia, suggesting that TTS (thrombosis with thrombocytopenia syndrome) is not the only type of thrombosis observed following virus-based vaccines. In conclusion, both virus-based COVID-19 vaccines show more SAEs than BNT, but the frequency of the SAE type in the different age groups differs, suggesting that the mechanisms responsible of SAEs overlap only partly.
Collapse
Affiliation(s)
- Luigi Cari
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, 06129, Perugia, Italy
| | - Mahdieh Naghavi Alhosseini
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, 06129, Perugia, Italy
| | - Paolo Fiore
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, 06129, Perugia, Italy
| | - Sabata Pierno
- University of Bari, Department of Pharmacy-Drug Sciences, Section of Pharmacology, 70125, Bari, Italy
| | - Sabrina Pacor
- University of Trieste, Department of Life Sciences, 34127, Trieste, Italy
| | - Alberta Bergamo
- University of Trieste, Department of Life Sciences, 34127, Trieste, Italy
| | - Gianni Sava
- University of Trieste, Department of Life Sciences, 34127, Trieste, Italy
| | - Giuseppe Nocentini
- University of Perugia, Department of Medicine and Surgery, Section of Pharmacology, 06129, Perugia, Italy.
| |
Collapse
|
32
|
Monagle P, Ng AP, Linden M, Ignjatovic V, Farley A, Taoudi S, Pasricha SR, Torresi J. Vaccine-induced immune thrombosis and thrombocytopenia syndrome following adenovirus-vectored severe acute respiratory syndrome coronavirus 2 vaccination: a novel hypothesis regarding mechanisms and implications for future vaccine development. Immunol Cell Biol 2021; 99:1006-1010. [PMID: 34664303 PMCID: PMC8652900 DOI: 10.1111/imcb.12505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023]
Abstract
We hypothesize that thrombosis with thrombocytopenia syndrome recently described after administration of adenovirus‐vectored vaccines for severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) occurs as a result of the unique properties of the adenovirus vectors, which can have widespread biodistribution throughout the body. The antigen is delivered to megakaryocyte cells, which act as part of the primary immune system and distribute the antigen within progeny platelets, also a key component of the immune system. The interaction of the antigen induces preformed antiplatelet factor 4 (PF4) antibodies to bind to PF4–heparan sulfate complexes in the absence of exogenous heparin, at sites where the heparan sulfate concentration in the vascular glycocalyx is optimal for complex formation, causing thrombosis and thrombocytopenia as observed clinically. This hypothesis is testable in cell culture and animal models, and potentially in vivo, and if proven correct has significant implications for vaccine development and our understanding of the links between the coagulation and immune systems.
Collapse
Affiliation(s)
- Paul Monagle
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia.,Department of Clinical Haematology, Royal Children's Hospital, Melbourne, VIC, Australia.,Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Ashley P Ng
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Matthew Linden
- School of Biomedical Science, University of Western Australia, Crawley, WA, Australia
| | - Vera Ignjatovic
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,Haematology Research, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Alison Farley
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Samir Taoudi
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Sant Rayn Pasricha
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Department of Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, VIC, Australia
| |
Collapse
|
33
|
Azzarone B, Veneziani I, Moretta L, Maggi E. Pathogenic Mechanisms of Vaccine-Induced Immune Thrombotic Thrombocytopenia in People Receiving Anti-COVID-19 Adenoviral-Based Vaccines: A Proposal. Front Immunol 2021; 12:728513. [PMID: 34484238 PMCID: PMC8415022 DOI: 10.3389/fimmu.2021.728513] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022] Open
Abstract
VITT is a rare, life-threatening syndrome characterized by thrombotic symptoms in combination with thrombocytopenia, which may occur in individuals receiving the first administration of adenoviral non replicating vectors (AVV) anti Covid19 vaccines. Vaccine-induced immune thrombotic thrombocytopenia (VITT) is characterized by high levels of serum IgG that bind PF4/polyanion complexes, thus triggering platelet activation. Therefore, identification of the fine pathophysiological mechanism by which vaccine components trigger platelet activation is mandatory. Herein, we propose a multistep mechanism involving both the AVV and the neo-synthetized Spike protein. The former can: i) spread rapidly into blood stream, ii), promote the early production of high levels of IL-6, iii) interact with erythrocytes, platelets, mast cells and endothelia, iv) favor the presence of extracellular DNA at the site of injection, v) activate platelets and mast cells to release PF4 and heparin. Moreover, AVV infection of mast cells may trigger aberrant inflammatory and immune responses in people affected by the mast cell activation syndrome (MCAS). The pre-existence of natural antibodies binding PF4/heparin complexes may amplify platelet activation and thrombotic events. Finally, neosynthesized Covid 19 Spike protein interacting with its ACE2 receptor on endothelia, platelets and leucocyte may trigger further thrombotic events unleashing the WITT syndrome.
Collapse
Affiliation(s)
- Bruno Azzarone
- Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Irene Veneziani
- Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Enrico Maggi
- Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
34
|
Ehianeta T, Mzee SAS, Adebisi MK, Ehianeta O. Recent SARS-CoV-2 Outlook and Implications in a COVID-19 Vaccination Era. INFECTIOUS MICROBES & DISEASES 2021; 3:125-133. [PMID: 38630122 PMCID: PMC8454280 DOI: 10.1097/im9.0000000000000072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/27/2021] [Accepted: 08/08/2021] [Indexed: 11/26/2022]
Abstract
While repurposed drugs came in handy earlier in the wake of the coronavirus disease 2019 (COVID-19) pandemic, vaccination has been considered a more sustainable approach. The recent spikes have been linked to "double," "triple," and even multi-mutant variants, thus renewing calls for deeper structural and functional insights of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a lead to rationale design of therapeutics, vaccines, and point-of-care diagnostics. There is a repertoire of findings from the earliest SARS-CoV-2 molecular mimicry to evade host immunity cum host immune responses to the role of the viral glycocalyx in modulating the susceptibility and severity of infection through attraction and repulsive interactions. Recently, molecular studies of some viral components that aid infection in the face of vaccination seem unending. In addition, the wave of infections and the attendant case fatality ratios have necessitated the need for emergency use authorizations for COVID-19 vaccines and in vitro diagnostics. This review provides key updates of SARS-CoV-2, current antigenic and formulation strategies, with emergency use authorizations considerations for future vaccine candidates and diagnostics. We also premise that despite the difficulty in modeling and analyzing glycans, understanding and exploiting their roles in the SARS-CoV-2 architecture is fundamental to glycan-based COVID-19 vaccines devoid of inconsistent clinical outcomes.
Collapse
Affiliation(s)
- Teddy Ehianeta
- Institute of Biological Chemistry, “Academia Sinica,” Taipei, Taiwan, China
| | | | | | | |
Collapse
|
35
|
Mendonça SA, Lorincz R, Boucher P, Curiel DT. Adenoviral vector vaccine platforms in the SARS-CoV-2 pandemic. NPJ Vaccines 2021; 6:97. [PMID: 34354082 PMCID: PMC8342436 DOI: 10.1038/s41541-021-00356-x] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Adenoviral vectors have been explored as vaccine agents for a range of infectious diseases, and their ability to induce a potent and balanced immune response made them logical candidates to apply to the COVID-19 pandemic. The unique molecular characteristics of these vectors enabled the rapid development of vaccines with advanced designs capable of overcoming the biological challenges faced by early adenoviral vector systems. These successes and the urgency of the COVID-19 situation have resulted in a flurry of candidate adenoviral vector vaccines for COVID-19 from both academia and industry. These vaccines represent some of the lead candidates currently supported by Operation Warp Speed and other government agencies for rapid translational development. This review details adenoviral vector COVID-19 vaccines currently in human clinical trials and provides an overview of the new technologies employed in their design. As these vaccines have formed a cornerstone of the COVID-19 global vaccination campaign, this review provides a full consideration of the impact and development of this emerging platform.
Collapse
Affiliation(s)
- Samir Andrade Mendonça
- Washington University in Saint Louis, School of Medicine, Biologic Therapeutics Center, Radiation Oncology Department. 660 South Euclid Avenue, St. Louis, MO, USA
| | - Reka Lorincz
- Washington University in Saint Louis, School of Medicine, Biologic Therapeutics Center, Radiation Oncology Department. 660 South Euclid Avenue, St. Louis, MO, USA
| | - Paul Boucher
- Washington University in Saint Louis, School of Medicine, Biologic Therapeutics Center, Radiation Oncology Department. 660 South Euclid Avenue, St. Louis, MO, USA
| | - David T Curiel
- Washington University in Saint Louis, School of Medicine, Biologic Therapeutics Center, Radiation Oncology Department. 660 South Euclid Avenue, St. Louis, MO, USA.
| |
Collapse
|
36
|
Alhashimi M, Elkashif A, Sayedahmed EE, Mittal SK. Nonhuman Adenoviral Vector-Based Platforms and Their Utility in Designing Next Generation of Vaccines for Infectious Diseases. Viruses 2021; 13:1493. [PMID: 34452358 PMCID: PMC8402644 DOI: 10.3390/v13081493] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Several human adenoviral (Ad) vectors have been developed for vaccine delivery owing to their numerous advantages, including the feasibility of different vector designs, the robustness of elicited immune responses, safety, and scalability. To expand the repertoire of Ad vectors for receptor usage and circumvention of Ad vector immunity, the use of less prevalent human Ad types or nonhuman Ads were explored for vector design. Notably, many nonhuman Ad vectors have shown great promise in preclinical and clinical studies as vectors for vaccine delivery. This review describes the key features of several nonhuman Ad vector platforms and their implications in developing effective vaccines against infectious diseases.
Collapse
Affiliation(s)
| | | | | | - Suresh K. Mittal
- Immunology and Infectious Disease, and Purdue University Center for Cancer Research, Department of Comparative Pathobiology, Purdue Institute for Inflammation, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907-2027, USA; (M.A.); (A.E.); (E.E.S.)
| |
Collapse
|
37
|
Costanzo M, De Giglio MAR, Roviello GN. Anti-Coronavirus Vaccines: Past Investigations on SARS-CoV-1 and MERS-CoV, the Approved Vaccines from BioNTech/Pfizer, Moderna, Oxford/AstraZeneca and others under Development Against SARS-CoV-2 Infection. Curr Med Chem 2021; 29:4-18. [PMID: 34355678 DOI: 10.2174/0929867328666210521164809] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 11/22/2022]
Abstract
The aim of this review article is to summarize the knowledge available to date on prophylaxis achievements to fight against Coronavirus. This work will give an overview of what is reported in the most recent literature on vaccines (under investigation or already developed like BNT162b2, mRNA-1273, and ChAdOx1-S) effective against the most pathogenic Coronaviruses (SARS-CoV-1, MERS-CoV-1, and SARS-CoV-2), with of course particular attention paid to those under development or already in use to combat the current COVID-19 (COronaVIrus Disease 19) pandemic. Our main objective is to make a contribution to the comprehension, additionally at a molecular level, of what is currently ready for anti-SARS-CoV-2 prophylactic intervention, as well as to provide the reader with an overall picture of the most innovative approaches for the development of vaccines that could be of general utility in the fight against the most pathogenic Coronaviruses.
Collapse
Affiliation(s)
- Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples 'Federico II', Via S. Pansini 5, I-80131 Naples, Italy
| | | | - Giovanni N Roviello
- Istituto di Biostrutture e Bioimmagini IBB - CNR, Via Mezzocannone 16; I-80134 Naples, Italy
| |
Collapse
|
38
|
Koch T, Fathi A, Addo MM. The COVID-19 Vaccine Landscape. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:549-573. [PMID: 33973199 DOI: 10.1007/978-3-030-63761-3_31] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The history of vaccine development spans centuries. At first, whole pathogens were used as vaccine agents, either inactivated or attenuated, to reduce virulence in humans. Safety and tolerability were increased by including only specific proteins as antigens and using cell culture methods, while novel vaccine strategies, like nucleic acid- or vector-based vaccines, hold high promise for the future. Vaccines have generally not been employed as the primary tools in outbreak response, but this might change since advances in medical technology in the last decades have made the concept of developing vaccines against novel pathogens a realistic strategy. Wandering the uncharted territory of a novel pathogen, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we can learn from other human Betacoronaviridae that emerged in the last decades, SARS-CoV-1 and MERS-CoV. We can identify the most likely target structures of immunity, establish animal models that emulate human disease and immunity as closely as possible, and learn about complex mechanisms of immune interaction such as cross-reactivity or antibody-dependent enhancement (ADE). However, significant knowledge gaps remain. What are the correlates of protection? How do we best induce immunity in vulnerable populations like the elderly? Will the immunity induced by vaccination (or by natural infection) wane over time? To date, at least 149 vaccine candidates against SARS-CoV-2 are under development. At the time of writing, at least 17 candidates have already progressed past preclinical studies (in vitro models and in vivo animal experiments) into clinical development. This chapter will provide an overview of this rapidly developing field.
Collapse
Affiliation(s)
- Till Koch
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany. .,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany.
| | - Anahita Fathi
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M Addo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lubeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
39
|
Watanabe Y, Mendonça L, Allen ER, Howe A, Lee M, Allen JD, Chawla H, Pulido D, Donnellan F, Davies H, Ulaszewska M, Belij-Rammerstorfer S, Morris S, Krebs AS, Dejnirattisai W, Mongkolsapaya J, Supasa P, Screaton GR, Green CM, Lambe T, Zhang P, Gilbert SC, Crispin M. Native-like SARS-CoV-2 Spike Glycoprotein Expressed by ChAdOx1 nCoV-19/AZD1222 Vaccine. ACS CENTRAL SCIENCE 2021; 7:594-602. [PMID: 34056089 PMCID: PMC8043200 DOI: 10.1021/acscentsci.1c00080] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Indexed: 05/08/2023]
Abstract
Vaccine development against the SARS-CoV-2 virus focuses on the principal target of the neutralizing immune response, the spike (S) glycoprotein. Adenovirus-vectored vaccines offer an effective platform for the delivery of viral antigen, but it is important for the generation of neutralizing antibodies that they produce appropriately processed and assembled viral antigen that mimics that observed on the SARS-CoV-2 virus. Here, we describe the structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine. We demonstrate native-like post-translational processing and assembly, and reveal the expression of S proteins on the surface of cells adopting the trimeric prefusion conformation. The data presented here confirm the use of ChAdOx1 adenovirus vectors as a leading platform technology for SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Yasunori Watanabe
- School
of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.
- Oxford
Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, U.K.
| | - Luiza Mendonça
- Division
of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.
| | - Elizabeth R. Allen
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Andrew Howe
- Electron
Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K.
| | - Mercede Lee
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
| | - Joel D. Allen
- School
of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.
| | - Himanshi Chawla
- School
of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.
| | - David Pulido
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Francesca Donnellan
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Hannah Davies
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Marta Ulaszewska
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Sandra Belij-Rammerstorfer
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
- NIHR Oxford
Biomedical Research Centre, Oxford, U.K.
| | - Susan Morris
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
| | - Anna-Sophia Krebs
- Division
of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.
| | - Wanwisa Dejnirattisai
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
| | - Juthathip Mongkolsapaya
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
- Dengue
Hemorrhagic Fever Research Unit, Office for Research and Development,
Faculty of Medicine, Siriraj Hospital, Mahidol
University, Bangkok, Thailand
- Chinese
Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, U.K.
| | - Piyada Supasa
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
| | - Gavin R. Screaton
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
- Division
of Medical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, U.K.
| | - Catherine M. Green
- The
Wellcome Centre for Human Genetics, University
of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.
| | - Teresa Lambe
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
- NIHR Oxford
Biomedical Research Centre, Oxford, U.K.
| | - Peijun Zhang
- Division
of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.
- Electron
Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K.
| | - Sarah C. Gilbert
- The
Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.
- NIHR Oxford
Biomedical Research Centre, Oxford, U.K.
| | - Max Crispin
- School
of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.
| |
Collapse
|
40
|
Wherry EJ, Jaffee EM, Warren N, D’Souza G, Ribas A. How Did We Get a COVID-19 Vaccine in Less Than 1 Year? Clin Cancer Res 2021; 27:2136-2138. [PMID: 33542081 PMCID: PMC8052930 DOI: 10.1158/1078-0432.ccr-21-0079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 11/16/2022]
Abstract
The successful development of COVID-19 vaccines within an unprecedented short time needs to be followed by rapid vaccine uptake, in particular, in high-risk populations such as patients with cancer. It is important for the scientific research community and cancer physicians to convey the knowledge behind the COVID-19 vaccine development and contribute to build the required trust on their use.
Collapse
Affiliation(s)
- E. John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth M. Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Nicholas Warren
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | - Gypsyamber D’Souza
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Antoni Ribas
- Department of Medicine and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
41
|
Heinz FX, Stiasny K. Profiles of current COVID-19 vaccines. Wien Klin Wochenschr 2021; 133:271-283. [PMID: 33725201 PMCID: PMC7962631 DOI: 10.1007/s00508-021-01835-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Franz X Heinz
- Center for Virology, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria.
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria
| |
Collapse
|
42
|
Chauhan N, Soni S, Gupta A, Aslam M, Jain U. Interpretative immune targets and contemporary position for vaccine development against SARS-CoV-2: A systematic review. J Med Virol 2021; 93:1967-1982. [PMID: 33270225 PMCID: PMC7753271 DOI: 10.1002/jmv.26709] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022]
Abstract
The year 2020 started with the emergence of novel coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which causes COVID-19 infection. Soon after the first evidence was reported in Wuhan, China, the World Health Organization declared global public health emergency and imminent need to understand the pathogenicity of the virus was required in limited time. Once the genome sequence of the virus was delineated, scientists across the world started working on the development of vaccines. Although, some laboratories have been using previously developed vaccine platforms from severe acute respiratory syndrome coronavirus (SARS) and middle east respiratory syndrome-related coronavirus and apply them in COVID-19 vaccines due to genetic similarities between coronaviruses. We have conducted a literature review to assess the background and current status of COVID-19 vaccines. The worldwide implementation and strategies for COVID-19 vaccine development are summarized from studies reported in years 2015-2020. While discussing the vaccine candidates, we have also explained interpretative immune responses of SARS-CoV-2 infection. There are several vaccine candidates at preclinical and clinical stages; however, only 42 vaccines are under clinical trials. Therefore, more industry collaborations and financial supports to COVID-19 studies are needed for mass-scale vaccine development. To develop effective vaccine platforms against SARS-CoV-2, the genetic resemblance with other coronaviruses are being evaluated which may further promote fast-track trials on previously developed SARS-CoV vaccines.
Collapse
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT)Amity University Uttar Pradesh (AUUP)NoidaIndia
| | - Shringika Soni
- Amity Institute of Nanotechnology (AINT)Amity University Uttar Pradesh (AUUP)NoidaIndia
| | - Abhinandan Gupta
- Amity Institute of Nanotechnology (AINT)Amity University Uttar Pradesh (AUUP)NoidaIndia
| | - Mohammad Aslam
- Rahat Hospital and Research Centre, Noor Mahal, AVAS VikasRampurIndia
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT)Amity University Uttar Pradesh (AUUP)NoidaIndia
| |
Collapse
|
43
|
Folegatti PM, Flaxman A, Jenkin D, Makinson R, Kingham-Page L, Bellamy D, Ramos Lopez F, Sheridan J, Poulton I, Aboagye J, Tran N, Mitton C, Roberts R, Lawrie AM, Hill AVS, Ewer KJ, Gilbert S. Safety and Immunogenicity of Adenovirus and Poxvirus Vectored Vaccines against a Mycobacterium Avium Complex Subspecies. Vaccines (Basel) 2021; 9:262. [PMID: 33809415 PMCID: PMC8000717 DOI: 10.3390/vaccines9030262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 11/20/2022] Open
Abstract
Heterologous prime-boost strategies are known to substantially increase immune responses in viral vectored vaccines. Here we report on safety and immunogenicity of the poxvirus Modified Vaccinia Ankara (MVA) vectored vaccine expressing four Mycobacterium avium subspecies paratuberculosis antigens as a single dose or as a booster vaccine following a simian adenovirus (ChAdOx2) prime. We demonstrate that a heterologous prime-boost schedule is well tolerated and induced T-cell immune responses.
Collapse
Affiliation(s)
- Pedro M. Folegatti
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (A.F.); (D.J.); (R.M.); (L.K.-P.); (D.B.); (F.R.L.); (J.S.); (I.P.); (J.A.); (N.T.); (C.M.); (R.R.); (A.M.L.); (A.V.S.H.); (K.J.E.); (S.G.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Almuqrin A, Davidson AD, Williamson MK, Lewis PA, Heesom KJ, Morris S, Gilbert SC, Matthews DA. SARS-CoV-2 vaccine ChAdOx1 nCoV-19 infection of human cell lines reveals low levels of viral backbone gene transcription alongside very high levels of SARS-CoV-2 S glycoprotein gene transcription. Genome Med 2021; 13:43. [PMID: 33722288 PMCID: PMC7958140 DOI: 10.1186/s13073-021-00859-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND ChAdOx1 nCoV-19 is a recombinant adenovirus vaccine against SARS-CoV-2 that has passed phase III clinical trials and is now in use across the globe. Although replication-defective in normal cells, 28 kbp of adenovirus genes is delivered to the cell nucleus alongside the SARS-CoV-2 S glycoprotein gene. METHODS We used direct RNA sequencing to analyse transcript expression from the ChAdOx1 nCoV-19 genome in human MRC-5 and A549 cell lines that are non-permissive for vector replication alongside the replication permissive cell line, HEK293. In addition, we used quantitative proteomics to study over time the proteome and phosphoproteome of A549 and MRC5 cells infected with the ChAdOx1 nCoV-19 vaccine. RESULTS The expected SARS-CoV-2 S coding transcript dominated in all cell lines. We also detected rare S transcripts with aberrant splice patterns or polyadenylation site usage. Adenovirus vector transcripts were almost absent in MRC-5 cells, but in A549 cells, there was a broader repertoire of adenoviral gene expression at very low levels. Proteomically, in addition to S glycoprotein, we detected multiple adenovirus proteins in A549 cells compared to just one in MRC5 cells. CONCLUSIONS Overall, the ChAdOx1 nCoV-19 vaccine's transcriptomic and proteomic repertoire in cell culture is as expected. The combined transcriptomic and proteomics approaches provide a detailed insight into the behaviour of this important class of vaccine using state-of-the-art techniques and illustrate the potential of this technique to inform future viral vaccine vector design.
Collapse
Affiliation(s)
- Abdulaziz Almuqrin
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
- Department of Clinical Laboratory Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Andrew D Davidson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Maia Kavanagh Williamson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Philip A Lewis
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Kate J Heesom
- Proteomics Facility, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Susan Morris
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sarah C Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - David A Matthews
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
45
|
Yoo JH. What We Do Know and Do Not Yet Know about COVID-19 Vaccines as of the Beginning of the Year 2021. J Korean Med Sci 2021; 36:e54. [PMID: 33559409 PMCID: PMC7870421 DOI: 10.3346/jkms.2021.36.e54] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which started at the end of 2019 and has spread worldwide, has remained unabated in 2021. Since non-pharmaceutical interventions including social distancing are facing limitations in controlling COVID-19, additional absolute means to change the trend are necessary. To this end, coronavirus-specific antiviral drugs and vaccines are urgently needed, but for now, the priority is to promote herd immunity through extensive nationwide vaccination campaign. In addition to the vaccines based on the conventional technology such inactivated or killed virus or protein subunit vaccines, several vaccines on the new technological platforms, for example, nucleic acids-based vaccines delivered by viral carriers, nanoparticles, or plasmids as a medium were introduced in this pandemic. In addition to achieving sufficient herd immunity with vaccination, the development of antiviral treatments that work specifically against COVID-19 will also be necessary to terminate the epidemic completely.
Collapse
Affiliation(s)
- Jin Hong Yoo
- Division of Infectious Diseases, Department of Internal Medicine, Bucheon St. Mary's Hospital, Bucheon, Korea
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
46
|
Mehmood I, Ijaz M, Ahmad S, Ahmed T, Bari A, Abro A, Allemailem KS, Almatroudi A, Tahir ul Qamar M. SARS-CoV-2: An Update on Genomics, Risk Assessment, Potential Therapeutics and Vaccine Development. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041626. [PMID: 33567746 PMCID: PMC7915969 DOI: 10.3390/ijerph18041626] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a great threat to public health, being a causative pathogen of a deadly coronavirus disease (COVID-19). It has spread to more than 200 countries and infected millions of individuals globally. Although SARS-CoV-2 has structural/genomic similarities with the previously reported SARS-CoV and MERS-CoV, the specific mutations in its genome make it a novel virus. Available therapeutic strategies failed to control this virus. Despite strict standard operating procedures (SOPs), SARS-CoV-2 has spread globally and it is mutating gradually as well. Diligent efforts, special care, and awareness are needed to reduce transmission among susceptible masses particularly elder people, children, and health care workers. In this review, we highlighted the basic genome organization and structure of SARS-CoV-2. Its transmission dynamics, symptoms, and associated risk factors are discussed. This review also presents the latest mutations identified in its genome, the potential therapeutic options being used, and a brief explanation of vaccine development efforts against COVID-19. The effort will not only help readers to understand the deadly SARS-CoV-2 virus but also provide updated information to researchers for their research work.
Collapse
Affiliation(s)
- Iqra Mehmood
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Munazza Ijaz
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (I.M.); (M.I.)
| | - Sajjad Ahmad
- Department of Microbiology and Pharmacy, Abasyn University, Peshawar 25000, Pakistan;
| | - Temoor Ahmed
- Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Amna Bari
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| | - Asma Abro
- Department of Biotechnology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87100, Pakistan;
| | - Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
- Correspondence: (A.A.); (M.T.u.Q.)
| | - Muhammad Tahir ul Qamar
- College of Life Science and Technology, Guangxi University, Nanning 530004, China
- Correspondence: (A.A.); (M.T.u.Q.)
| |
Collapse
|
47
|
Watanabe Y, Mendonça L, Allen ER, Howe A, Lee M, Allen JD, Chawla H, Pulido D, Donnellan F, Davies H, Ulaszewska M, Belij-Rammerstorfer S, Morris S, Krebs AS, Dejnirattisai W, Mongkolsapaya J, Supasa P, Screaton GR, Green CM, Lambe T, Zhang P, Gilbert SC, Crispin M. Native-like SARS-CoV-2 spike glycoprotein expressed by ChAdOx1 nCoV-19/AZD1222 vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.15.426463. [PMID: 33501433 PMCID: PMC7836103 DOI: 10.1101/2021.01.15.426463] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Vaccine development against the SARS-CoV-2 virus focuses on the principal target of the neutralizing immune response, the spike (S) glycoprotein. Adenovirus-vectored vaccines offer an effective platform for the delivery of viral antigen, but it is important for the generation of neutralizing antibodies that they produce appropriately processed and assembled viral antigen that mimics that observed on the SARS-CoV-2 virus. Here, we describe the structure, conformation and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine. We demonstrate native-like post-translational processing and assembly, and reveal the expression of S proteins on the surface of cells adopting the trimeric prefusion conformation. The data presented here confirms the use of ChAdOx1 adenovirus vectors as a leading platform technology for SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Yasunori Watanabe
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Luiza Mendonça
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford, OX3 7BN, UK
| | - Elizabeth R. Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Howe
- Electron Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Mercede Lee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Joel D. Allen
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Himanshi Chawla
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - David Pulido
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Francesca Donnellan
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah Davies
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Belij-Rammerstorfer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Susan Morris
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Anna-Sophia Krebs
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford, OX3 7BN, UK
| | - Wanwisa Dejnirattisai
- The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Juthathip Mongkolsapaya
- The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Chinese Academy of Medical Science(CAMS) Oxford Institute (COI), University of Oxford, Oxford, U.K
| | - Piyada Supasa
- The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Gavin R. Screaton
- The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- Division of Medical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Catherine M. Green
- The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Peijun Zhang
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford, OX3 7BN, UK
- Electron Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Sarah C. Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
48
|
Haidere MF, Ratan ZA, Nowroz S, Zaman SB, Jung YJ, Hosseinzadeh H, Cho JY. COVID-19 Vaccine: Critical Questions with Complicated Answers. Biomol Ther (Seoul) 2021; 29:1-10. [PMID: 33372165 PMCID: PMC7771841 DOI: 10.4062/biomolther.2020.178] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
COVID-19 has caused extensive human casualties with significant economic impacts around the globe, and has imposed new challenges on health systems worldwide. Over the past decade, SARS, Ebola, and Zika also led to significant concerns among the scientific community. Interestingly, the SARS and Zika epidemics ended before vaccine development; however, the scholarly community and the pharmaceutical companies responded very quickly at that time. Similarly, when the genetic sequence of SARSCoV-2 was revealed, global vaccine companies and scientists have stepped forward to develop a vaccine, triggering a race toward vaccine development that the whole world is relying on. Similarly, an effective and safe vaccine could play a pivotal role in eradicating COVID-19. However, few important questions regarding SARS-CoV-2 vaccine development are explored in this review.
Collapse
Affiliation(s)
| | - Zubair Ahmed Ratan
- School of Health & Society, University of Wollongong, NSW 2500, Australia
- Department of Biomedical Engineering, Khulna University of Engineering and Technology, Khulna 9203, Bangladesh
| | - Senjuti Nowroz
- Department of Chemistry, University of Dhaka, Dhaka 1000, Bangladesh
| | - Sojib Bin Zaman
- Department of Medicine, School of Clinical Sciences, Monash University, Victoria 3800, Australia
| | - You-Jung Jung
- Biological Resources Utilization Department, National Institute of Biological Resources, Incheon 22689, Republic of Korea
| | | | - Jae Youl Cho
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
49
|
Russell RL, Pelka P, Mark BL. Frontrunners in the race to develop a SARS-CoV-2 vaccine. Can J Microbiol 2020; 67:189-212. [PMID: 33264067 DOI: 10.1139/cjm-2020-0465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Numerous studies continue to be published on the COVID-19 pandemic that is being caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Given the rapidly evolving global response to SARS-CoV-2, here we primarily review the leading COVID-19 vaccine strategies that are currently in Phase III clinical trials. Nonreplicating viral vector strategies, inactivated virus, recombinant protein subunit vaccines, and nucleic acid vaccine platforms are all being pursued in an effort to combat the infection. Preclinical and clinal trial results of these efforts are examined as well as the characteristics of each vaccine strategy from the humoral and cellular immune responses they stimulate, effects of any adjuvants used, and the potential risks associated with immunization such as antibody-dependent enhancement. A number of promising advancements have been made toward the development of multiple vaccine candidates. Preliminary data now emerging from phase III clinical trials show encouraging results for the protective efficacy and safety of at least 3 frontrunning candidates. There is hope that one or more will emerge as potent weapons to protect against SARS-CoV-2.
Collapse
Affiliation(s)
- Raquel L Russell
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Peter Pelka
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Brian L Mark
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
50
|
Hayman B, Pagliusi S. Emerging vaccine manufacturers are innovating for the next decade. Vaccine X 2020; 5:100066. [PMID: 32462140 PMCID: PMC7242873 DOI: 10.1016/j.jvacx.2020.100066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/31/2022] Open
Abstract
The Developing Countries Vaccine Manufacturers Network (DCVMN) is a public health-driven alliance consisting of 43 vaccine manufacturers from 14 countries and territories, operating under the mandate to protect all people against known and emerging infectious diseases, by improving the availability of high-quality vaccines globally. The Network provides a platform for organizations to come together regularly to share technical information, best practices and future prospects. DCVMN members are playing an increasingly important role in public health supplying over 50% of the doses of vaccines procured by UNICEF globally. To evaluate the progress made by Network's members, a survey consisting of 9 questions covering three important components of the manufacturers in the network was created, focusing on company dedicated human resources, vaccine production, and research and development efforts. Results show that more vaccines from more manufacturers are achieving WHO Pre-qualification, with areas of focus including the contributions to a Polio-free world, ending cholera, and tackling re-emerging diseases, such as measles. An increase by 50% of number of manufacturing companies holding WHO prequalified vaccines was observed from 2013 to 2019, strengthening open competitiveness for global vaccines supply. Notably, Network members have 181 vaccine projects in the research and development pipeline, highlighting novel vaccines against mosquito-borne diseases, such as dengue, chikungunya and Zika, novel human papillomavirus and pneumococcal conjugated vaccines. This report summarizes the progressive efforts of DCVMN members to contribute to reducing the burden of infectious diseases globally and details their commitment to vaccine innovation, particularly in the past five years, in the context of how vaccine innovations of today will shape the fight against infectious diseases tomorrow.
Collapse
Affiliation(s)
- Benoit Hayman
- DCVMN International, Route de Crassier 7, 1262 Nyon, Switzerland
| | - Sonia Pagliusi
- DCVMN International, Route de Crassier 7, 1262 Nyon, Switzerland
| |
Collapse
|