1
|
Sanderson J, Aboagye J, Makinson R, Rapi K, Provstgaard-Morys S, Stockdale L, Alison Lawrie, Lanigan I, Halim N, Douiri A, Greenlay E, Malek R, Gray E, West L, El Oulidi F, Cross PI, Stallibrass M, Gilbert SC, Hill AVS, Ewer KJ. A phase 1b clinical trial to determine the safety, tolerability and immunogenicity of simian adenovirus and poxvirus vectored vaccines against a Mycobacterium avium complex subspecies in patients with active Crohn's disease. EBioMedicine 2025; 113:105570. [PMID: 39922068 PMCID: PMC11848756 DOI: 10.1016/j.ebiom.2025.105570] [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: 12/18/2023] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Crohn's Disease (CD) is a chronic, debilitating condition hypothesised to be associated with Mycobacterium avium ssp paratuberculosis (MAP) infection. It is the causative pathogen of the granulomatous inflammatory enteritis in ruminants, Johne's Disease. A developing treatment approach is utilising heterologous prime-boost viral vectored vaccines. We report a Phase 1b dose-escalation trial to determine the safety, tolerability and immunogenicity of candidate recombinant ChAdOx2 and MVA vectored vaccines against MAP in patients with CD. METHODS 28 patients with mild to moderate CD, aged 18-50, were randomly allocated into 5 groups. Group 1 and 2 were vaccinated with ChAdOx2 HAV, Groups 3 and 4 with MVA HAV and Group 5 with both vaccines in a prime-boost regimen. A 112-day follow-up period assessed safety and tolerability by recording adverse events (AEs) and serious adverse events (SAEs). Secondary objectives of immunogenicity were assessed by ELISpot (enzyme-linked immunosorbent spot) and clinical response by Crohn's Disease Activity Index (CDAI) and Simple Endoscopic Score for Crohn's Disease (SES-CD). FINDINGS 28 participants received either a single dose of ChAdOx2 HAV (n = 12), a single dose of MVA HAV (n = 6) or a prime dose of ChAdOx2 HAV (n = 10) followed by an MVA HAV (n = 9) boost. Solicited AEs were 196 in all participants, one AE was graded as severe but resolved within 24 h. The majority of solicited AEs were graded as mild (149/196; 76%, 95% CI 69%-82%) or moderate (45/196; 23%, 95% CI 17%-29%). ELISpot responses increased in Groups 1 and 2 and significantly more after boosting with MVA HAV. INTERPRETATION Candidate vaccines ChAdOx2 HAV and MVA HAV were safe, well-tolerated and immunogenic in patients with active CD. A heterologous prime-boost schedule induces a T cell-mediated immune response. Further studies are required to determine the efficacy and optimal regime of the vaccines. FUNDING HAV Vaccines Limited funded the trial and acted as trial sponsor. The Sponsor was involved in protocol development, trial conduct, including data monitoring and analysis, and the preparation of this manuscript in line with the Medicines for Human Use (Clinical Trials) Regulations 2004 and amendments.
Collapse
Affiliation(s)
- Jeremy Sanderson
- Guy's and St Thomas' Hospitals NHS Foundation Trust, Great Maze Pond, London SE1 9RT, UK
| | - Jeremy Aboagye
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Rebecca Makinson
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Katerina Rapi
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | | | - Lisa Stockdale
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Alison Lawrie
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Isabelle Lanigan
- Guy's and St Thomas' Hospitals NHS Foundation Trust, Great Maze Pond, London SE1 9RT, UK
| | - Nishat Halim
- Guy's and St Thomas' Hospitals NHS Foundation Trust, Great Maze Pond, London SE1 9RT, UK
| | - Abdel Douiri
- Kings College London, Great Maze Pond, London SE1 1UL, UK
| | - Emily Greenlay
- Kings College London, Great Maze Pond, London SE1 1UL, UK
| | - Rayka Malek
- Kings College London, Great Maze Pond, London SE1 1UL, UK
| | - Emma Gray
- Kings College London, Great Maze Pond, London SE1 1UL, UK
| | - Lindsey West
- Kings College London, Great Maze Pond, London SE1 1UL, UK
| | - Fatima El Oulidi
- HAV Vaccines Limited, Np-105, Icentre, Howard Way, Newport Pagnell, Milton Keynes MK16 9PY, UK
| | - Paul Ian Cross
- HAV Vaccines Limited, Np-105, Icentre, Howard Way, Newport Pagnell, Milton Keynes MK16 9PY, UK.
| | - Michael Stallibrass
- HAV Vaccines Limited, Np-105, Icentre, Howard Way, Newport Pagnell, Milton Keynes MK16 9PY, UK
| | - Sarah C Gilbert
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Adrian V S Hill
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| | - Katie J Ewer
- The Jenner Institute, ORCRB, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
2
|
Colombatti Olivieri MA, Cuerda MX, Moyano RD, Gravisaco MJ, Pinedo MFA, Delgado FO, Calamante G, Mundo S, de la Paz Santangelo M, Romano MI, Alonso MN, Del Medico Zajac MP. Superior protection against paratuberculosis by a heterologous prime-boost immunization in a murine model. Vaccine 2024; 42:126055. [PMID: 38880691 DOI: 10.1016/j.vaccine.2024.06.022] [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: 07/14/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.
Collapse
MESH Headings
- Animals
- Mycobacterium avium subsp. paratuberculosis/immunology
- Immunization, Secondary/methods
- Mice
- Paratuberculosis/prevention & control
- Paratuberculosis/immunology
- Immunoglobulin G/blood
- Cytokines/metabolism
- Female
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Adjuvants, Immunologic/administration & dosage
- Disease Models, Animal
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Mice, Inbred BALB C
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Immunity, Cellular/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Freund's Adjuvant/administration & dosage
- Freund's Adjuvant/immunology
Collapse
Affiliation(s)
| | - María Ximena Cuerda
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Roberto Damián Moyano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María José Gravisaco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Fiorella Alvarado Pinedo
- Centro de Diagnóstico e Investigaciones Veterinarias (CEDIVE) de la Facultad de Ciencias Veterinarias - Universidad de La Plata, Chascomús, Buenos Aires 7130, Argentina
| | - Fernando Oscar Delgado
- Instituto de Patobiologia Veterinaria (IPV), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Gabriela Calamante
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Silvia Mundo
- Cátedra de Inmunología de la Facultad de Ciencias Veterinarias - Universidad de Buenos Aires, Ciudad de Buenos Aires 1427, Argentina
| | - María de la Paz Santangelo
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Isabel Romano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Natalia Alonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina.
| | - María Paula Del Medico Zajac
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| |
Collapse
|
3
|
Petherbridge L, Davis C, Robinson A, Evans T, Sebastian S. Pre-Clinical Development of an Adenovirus Vector Based RSV and Shingles Vaccine Candidate. Vaccines (Basel) 2023; 11:1679. [PMID: 38006010 PMCID: PMC10674764 DOI: 10.3390/vaccines11111679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/24/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection and shingles are two viral diseases that affect older adults, and a combined vaccine to protect against both could be beneficial. RSV infection causes hospitalisations and significant morbidity in both children and adults and can be fatal in the elderly. The RSV fusion (F) envelope glycoprotein induces a strong RSV-neutralising antibody response and is the target of protective immunity in the first RSV vaccine for older adults, recently approved by the FDA. An initial childhood infection with the varicella zoster virus (VZV) results in chickenpox disease, but reactivation in older adults can cause shingles. This reactivation in sensory and autonomic neurons is characterized by a skin-blistering rash that can be accompanied by prolonged pain. The approved protein-in-adjuvant shingles vaccine induces VZV glycoprotein E (gE)-fspecific antibody and CD4+ T cell responses and is highly effective. Here we report the evaluation of RSV/shingles combination vaccine candidates based on non-replicating chimpanzee adenovirus (ChAd) vectors. We confirmed the cellular and humoral immunogenicity of the vaccine vectors in mice using T cell and antibody assays. We also carried out an RSV challenge study in cotton rats which demonstrated protective efficacy following a homologous prime-boost regimen with our preferred vaccine candidate.
Collapse
Affiliation(s)
| | | | | | | | - Sarah Sebastian
- Vaccitech Ltd., Harwell OX11 0DF, UK; (L.P.); (A.R.); (T.E.)
| |
Collapse
|
4
|
Dold C, Marsay L, Wang N, Silva-Reyes L, Clutterbuck E, Paterson GK, Sharkey K, Wyllie D, Beernink PT, Hill AV, Pollard AJ, Rollier CS. An adenoviral-vectored vaccine confers seroprotection against capsular group B meningococcal disease. Sci Transl Med 2023; 15:eade3901. [PMID: 37343082 DOI: 10.1126/scitranslmed.ade3901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/30/2023] [Indexed: 06/23/2023]
Abstract
Adenoviral-vectored vaccines are licensed for prevention of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ebola virus, but, for bacterial proteins, expression in a eukaryotic cell may affect the antigen's localization and conformation or lead to unwanted glycosylation. Here, we investigated the potential use of an adenoviral-vectored vaccine platform for capsular group B meningococcus (MenB). Vector-based candidate vaccines expressing MenB antigen factor H binding protein (fHbp) were generated, and immunogenicity was assessed in mouse models, including the functional antibody response by serum bactericidal assay (SBA) using human complement. All adenovirus-based vaccine candidates induced high antigen-specific antibody and T cell responses. A single dose induced functional serum bactericidal responses with titers superior or equal to those induced by two doses of protein-based comparators, as well as longer persistence and a similar breadth. The fHbp transgene was further optimized for human use by incorporating a mutation abrogating binding to the human complement inhibitor factor H. The resulting vaccine candidate induced high and persistent SBA responses in transgenic mice expressing human factor H. The optimized transgene was inserted into the clinically relevant ChAdOx1 backbone, and this vaccine has now progressed to clinical development. The results of this preclinical vaccine development study underline the potential of vaccines based on genetic material to induce functional antibody responses against bacterial outer membrane proteins.
Collapse
Affiliation(s)
- Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Leanne Marsay
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Nelson Wang
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Elizabeth Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Gavin K Paterson
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Kelsey Sharkey
- Division of Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David Wyllie
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Peter T Beernink
- Division of Infectious Diseases and Global Health, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adrian V Hill
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, CCVTM, Churchill Lane, Oxford OX3 7LE, UK
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
5
|
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: 16] [Impact Index Per Article: 8.0] [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
|
6
|
Fernandes Q, Inchakalody VP, Merhi M, Mestiri S, Taib N, Moustafa Abo El-Ella D, Bedhiafi T, Raza A, Al-Zaidan L, Mohsen MO, Yousuf Al-Nesf MA, Hssain AA, Yassine HM, Bachmann MF, Uddin S, Dermime S. Emerging COVID-19 variants and their impact on SARS-CoV-2 diagnosis, therapeutics and vaccines. Ann Med 2022; 54:524-540. [PMID: 35132910 PMCID: PMC8843115 DOI: 10.1080/07853890.2022.2031274] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
The emergence of novel and evolving variants of SARS-CoV-2 has fostered the need for change in the form of newer and more adaptive diagnostic methods for the detection of SARS-CoV-2 infections. On the other hand, developing rapid and sensitive diagnostic technologies is now more challenging due to emerging variants and varying symptoms exhibited among the infected individuals. In addition to this, vaccines remain the major mainstay of prevention and protection against infection. Novel vaccines and drugs are constantly being developed to unleash an immune response for the robust targeting of SARS-CoV-2 and its associated variants. In this review, we provide an updated perspective on the current challenges posed by the emergence of novel SARS-CoV-2 mutants/variants and the evolution of diagnostic techniques to enable their detection. In addition, we also discuss the development, formulation, working mechanisms, advantages, and drawbacks of some of the most used vaccines/therapeutic drugs and their subsequent immunological impact.Key messageThe emergence of novel variants of the SARS-CoV-2 in the past couple of months, highlights one of the primary challenges in the diagnostics, treatment, as well as vaccine development against the virus.Advancements in SARS-CoV-2 detection include nucleic acid based, antigen and immuno- assay-based and antibody-based detection methodologies for efficient, robust, and quick testing; while advancements in COVID-19 preventive and therapeutic strategies include novel antiviral and immunomodulatory drugs and SARS-CoV-2 targeted vaccines.The varied COVID-19 vaccine platforms and the immune responses induced by each one of them as well as their ability to battle post-vaccination infections have all been discussed in this review.
Collapse
Affiliation(s)
- Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
| | - Varghese Philipose Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Dina Moustafa Abo El-Ella
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Takwa Bedhiafi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Lobna Al-Zaidan
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mona O. Mohsen
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Department of Biomedical Research, Immunology RIA, University of Bern, Bern, Switzerland
| | | | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Martin F. Bachmann
- Department of Biomedical Research, Immunology RIA, University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
7
|
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
|
8
|
Development of Viral-Vectored Vaccines and Virus Replicon Particle-Based Neutralisation Assay against Mayaro Virus. Int J Mol Sci 2022; 23:ijms23084105. [PMID: 35456923 PMCID: PMC9026931 DOI: 10.3390/ijms23084105] [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: 03/14/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 02/07/2023] Open
Abstract
Mayaro virus (MAYV) is an emerging alphavirus causing acute febrile illness associated with chronic polyarthralgia. Although MAYV is currently restricted to tropical regions in South America around the Amazon basin, it has the potential to spread globally by Aedes species mosquitoes. In addition, there are currently no specific therapeutics or licenced vaccines against MAYV infection. We have previously shown that an adenovirus based Mayaro vaccine (ChAdOx1 May) was able to provide full protection against MAYV challenge in vaccinated A129 mice and induced high neutralising antibody titres. In this study, we have constructed a replication deficient simian adenovirus (ChAdOx2) and a Modified Ankara Virus (MVA) based vaccine expressing the MAYV structural cassette (sMAYV) similar to ChAdOx1 May, and characterised recombinant MAYV E2 glycoprotein expressed in a mammalian system for immune monitoring. We demonstrate that ChAdOx2 May was able to induce high antibody titres similar to ChAdOx1 May, and MVA May was shown to be an effective boosting strategy following prime vaccination with ChAdOx1 or ChAdOx2 May. In order to measure MAYV neutralising ability, we have developed a virus replicon particle-based neutralisation assay which effectively detected neutralising antibodies against MAYV. In summary, our study indicates the potential for further clinical development of the viral vectored MAYV vaccines against MAYV infections.
Collapse
|
9
|
Mohamed N, Hamad MA, Ghaleb AH, Esmat G, Elsabahy M. Applications of nanoengineered therapeutics and vaccines: special emphasis on COVID-19. IMMUNOMODULATORY EFFECTS OF NANOMATERIALS 2022. [PMCID: PMC9212255 DOI: 10.1016/b978-0-323-90604-3.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nanomedicine provides innovative strategies that had significantly improved drug and gene delivery and allowed control over the engineering of therapeutics, diagnostics, vaccines, and other medical devices, for a diversity of medical applications. This review focuses on the current attempts to develop potent nanoengineered vaccines and therapeutics against coronaviruses, and the recent fabrication strategies and design principles to control acute infections from the escalating SARS-CoV-2 pandemic. Nanomedical approaches provide versatile platforms that can be utilized to enhance the overall potency, safety, and stability of vaccines, thus augmenting the desired immune response. Their modulable conformational features of size, shape, surface charge, antigen display, and composition allow for precise tuning and optimization of the nanoconstructs for the management of a variety of diseases and pathological conditions. The ability to control the release of their encapsulated cargoes and the possibility of surface decoration with various moieties support the construction of multifunctional nanomaterials that ultimately boost and prolong the immune response elicited and/or therapeutic effects, selectively at the diseased tissues and target sites.
Collapse
|
10
|
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
|
11
|
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
|
12
|
Fathi A, Addo MM, Dahlke C. Sex Differences in Immunity: Implications for the Development of Novel Vaccines Against Emerging Pathogens. Front Immunol 2021; 11:601170. [PMID: 33488596 PMCID: PMC7820860 DOI: 10.3389/fimmu.2020.601170] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Vaccines are one of the greatest public health achievements and have saved millions of lives. They represent a key countermeasure to limit epidemics caused by emerging infectious diseases. The Ebola virus disease crisis in West Africa dramatically revealed the need for a rapid and strategic development of vaccines to effectively control outbreaks. Seven years later, in light of the SARS-CoV-2 pandemic, this need has never been as urgent as it is today. Vaccine development and implementation of clinical trials have been greatly accelerated, but still lack strategic design and evaluation. Responses to vaccination can vary widely across individuals based on factors like age, microbiome, co-morbidities and sex. The latter aspect has received more and more attention in recent years and a growing body of data provide evidence that sex-specific effects may lead to different outcomes of vaccine safety and efficacy. As these differences might have a significant impact on the resulting optimal vaccine regimen, sex-based differences should already be considered and investigated in pre-clinical and clinical trials. In this Review, we will highlight the clinical observations of sex-specific differences in response to vaccination, delineate sex differences in immune mechanisms, and will discuss the possible resulting implications for development of vaccine candidates against emerging infections. As multiple vaccine candidates against COVID-19 that target the same antigen are tested, vaccine development may undergo a decisive change, since we now have the opportunity to better understand mechanisms that influence vaccine-induced reactogenicity and effectiveness of different vaccines.
Collapse
Affiliation(s)
- Anahita Fathi
- University Medical Center Hamburg-Eppendorf, 1st Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M. Addo
- University Medical Center Hamburg-Eppendorf, 1st Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Christine Dahlke
- University Medical Center Hamburg-Eppendorf, 1st Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
13
|
Lundstrom K. Application of Viral Vectors for Vaccine Development with a Special Emphasis on COVID-19. Viruses 2020; 12:E1324. [PMID: 33218001 PMCID: PMC7698750 DOI: 10.3390/v12111324] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Viral vectors can generate high levels of recombinant protein expression providing the basis for modern vaccine development. A large number of different viral vector expression systems have been utilized for targeting viral surface proteins and tumor-associated antigens. Immunization studies in preclinical animal models have evaluated the elicited humoral and cellular responses and the possible protection against challenges with lethal doses of infectious pathogens or tumor cells. Several vaccine candidates for both infectious diseases and various cancers have been subjected to a number of clinical trials. Human immunization trials have confirmed safe application of viral vectors, generation of neutralizing antibodies and protection against challenges with lethal doses. A special emphasis is placed on COVID-19 vaccines based on viral vectors. Likewise, the flexibility and advantages of applying viral particles, RNA replicons and DNA replicon vectors of self-replicating RNA viruses for vaccine development are presented.
Collapse
|
14
|
Abstract
The current COVID-19 pandemic has substantially accelerated the demands for efficient vaccines. A wide spectrum of approaches includes live attenuated and inactivated viruses, protein subunits and peptides, viral vector-based delivery, DNA plasmids, and synthetic mRNA. Preclinical studies have demonstrated robust immune responses, reduced viral loads and protection against challenges with SARS-CoV-2 in rodents and primates. Vaccine candidates based on all delivery systems mentioned above have been subjected to clinical trials in healthy volunteers. Phase I clinical trials have demonstrated in preliminary findings good safety and tolerability. Evaluation of immune responses in a small number of individuals has demonstrated similar or superior levels of neutralizing antibodies in comparison to immunogenicity detected in COVID-19 patients. Both adenovirus- and mRNA-based vaccines have entered phase II and study protocols for phase III trials with 30,000 participants have been finalized.
Collapse
|
15
|
Folegatti PM, Ewer KJ, Aley PK, Angus B, Becker S, Belij-Rammerstorfer S, Bellamy D, Bibi S, Bittaye M, Clutterbuck EA, Dold C, Faust SN, Finn A, Flaxman AL, Hallis B, Heath P, Jenkin D, Lazarus R, Makinson R, Minassian AM, Pollock KM, Ramasamy M, Robinson H, Snape M, Tarrant R, Voysey M, Green C, Douglas AD, Hill AVS, Lambe T, Gilbert SC, Pollard AJ. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020; 396:467-478. [PMID: 32702298 PMCID: PMC7445431 DOI: 10.1016/s0140-6736(20)31604-4] [Citation(s) in RCA: 1751] [Impact Index Per Article: 350.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) might be curtailed by vaccination. We assessed the safety, reactogenicity, and immunogenicity of a viral vectored coronavirus vaccine that expresses the spike protein of SARS-CoV-2. METHODS We did a phase 1/2, single-blind, randomised controlled trial in five trial sites in the UK of a chimpanzee adenovirus-vectored vaccine (ChAdOx1 nCoV-19) expressing the SARS-CoV-2 spike protein compared with a meningococcal conjugate vaccine (MenACWY) as control. Healthy adults aged 18-55 years with no history of laboratory confirmed SARS-CoV-2 infection or of COVID-19-like symptoms were randomly assigned (1:1) to receive ChAdOx1 nCoV-19 at a dose of 5 × 1010 viral particles or MenACWY as a single intramuscular injection. A protocol amendment in two of the five sites allowed prophylactic paracetamol to be administered before vaccination. Ten participants assigned to a non-randomised, unblinded ChAdOx1 nCoV-19 prime-boost group received a two-dose schedule, with the booster vaccine administered 28 days after the first dose. Humoral responses at baseline and following vaccination were assessed using a standardised total IgG ELISA against trimeric SARS-CoV-2 spike protein, a muliplexed immunoassay, three live SARS-CoV-2 neutralisation assays (a 50% plaque reduction neutralisation assay [PRNT50]; a microneutralisation assay [MNA50, MNA80, and MNA90]; and Marburg VN), and a pseudovirus neutralisation assay. Cellular responses were assessed using an ex-vivo interferon-γ enzyme-linked immunospot assay. The co-primary outcomes are to assess efficacy, as measured by cases of symptomatic virologically confirmed COVID-19, and safety, as measured by the occurrence of serious adverse events. Analyses were done by group allocation in participants who received the vaccine. Safety was assessed over 28 days after vaccination. Here, we report the preliminary findings on safety, reactogenicity, and cellular and humoral immune responses. The study is ongoing, and was registered at ISRCTN, 15281137, and ClinicalTrials.gov, NCT04324606. FINDINGS Between April 23 and May 21, 2020, 1077 participants were enrolled and assigned to receive either ChAdOx1 nCoV-19 (n=543) or MenACWY (n=534), ten of whom were enrolled in the non-randomised ChAdOx1 nCoV-19 prime-boost group. Local and systemic reactions were more common in the ChAdOx1 nCoV-19 group and many were reduced by use of prophylactic paracetamol, including pain, feeling feverish, chills, muscle ache, headache, and malaise (all p<0·05). There were no serious adverse events related to ChAdOx1 nCoV-19. In the ChAdOx1 nCoV-19 group, spike-specific T-cell responses peaked on day 14 (median 856 spot-forming cells per million peripheral blood mononuclear cells, IQR 493-1802; n=43). Anti-spike IgG responses rose by day 28 (median 157 ELISA units [EU], 96-317; n=127), and were boosted following a second dose (639 EU, 360-792; n=10). Neutralising antibody responses against SARS-CoV-2 were detected in 32 (91%) of 35 participants after a single dose when measured in MNA80 and in 35 (100%) participants when measured in PRNT50. After a booster dose, all participants had neutralising activity (nine of nine in MNA80 at day 42 and ten of ten in Marburg VN on day 56). Neutralising antibody responses correlated strongly with antibody levels measured by ELISA (R2=0·67 by Marburg VN; p<0·001). INTERPRETATION ChAdOx1 nCoV-19 showed an acceptable safety profile, and homologous boosting increased antibody responses. These results, together with the induction of both humoral and cellular immune responses, support large-scale evaluation of this candidate vaccine in an ongoing phase 3 programme. FUNDING UK Research and Innovation, Coalition for Epidemic Preparedness Innovations, National Institute for Health Research (NIHR), NIHR Oxford Biomedical Research Centre, Thames Valley and South Midland's NIHR Clinical Research Network, and the German Center for Infection Research (DZIF), Partner site Gießen-Marburg-Langen.
Collapse
Affiliation(s)
- Pedro M Folegatti
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Stephan Becker
- Institute of Virology, Philipps University of Marburg, Marburg, Germany
| | - Sandra Belij-Rammerstorfer
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Duncan Bellamy
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Mustapha Bittaye
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Adam Finn
- School of Population Health Sciences, University of Bristol, Bristol, UK
| | - Amy L Flaxman
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Bassam Hallis
- National Infection Service, Public Health England, Salisbury, UK
| | - Paul Heath
- Vaccine Institute, St George's University, London, UK
| | - Daniel Jenkin
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Rajeka Lazarus
- Department of Microbiology, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Rebecca Makinson
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M Minassian
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katrina M Pollock
- NIHR Imperial Clinical Research Facility, Imperial College London, London, UK
| | - Maheshi Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Matthew Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Richard Tarrant
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Catherine Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | - Alexander D Douglas
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
16
|
Honap S, Johnston E, Agrawal G, Al-Hakim B, Hermon-Taylor J, Sanderson J. Anti- Mycobacterium paratuberculosis (MAP) therapy for Crohn's disease: an overview and update. Frontline Gastroenterol 2020; 12:397-403. [PMID: 35401965 PMCID: PMC8989010 DOI: 10.1136/flgastro-2020-101471] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/28/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023] Open
Abstract
The role of Mycobacterium avium subspecies paratuberculosis (MAP) in the pathogenesis of Crohn's disease (CD) has been strongly debated for many years. MAP is the known aetiological agent of Johne's disease, a chronic enteritis affecting livestock. At present, due to the paucity of high-quality data, anti-MAP therapy (AMT) is not featured in international guidelines as a treatment for CD. Although the much-quoted randomised trial of AMT did not show sustained benefits over placebo, questions have been raised regarding trial design, antibiotic dosing and the formulation used. There are several lines of evidence supporting the CD and MAP association with uncontrolled and controlled trials demonstrating effectiveness, including a retrospective review of cases treated at our own institution. Here, we provide an overview of the evidence supporting and refuting AMT in CD before focussing on updates of the current research in the field, including the ongoing trials with the novel RHB-104 formulation and the MAP vaccine trial. While controversial, gastroenterologists are often asked about long-term combination antibiotic therapy for CD. There has been broadcast and social media coverage surrounding this, particularly with regard to current trials. Although patients should not be deterred from treatments of proven effectiveness, this review aims to help with commonly asked questions and highlights our own approach for the use of anti-MAP in specific circumstances.
Collapse
Affiliation(s)
- Sailish Honap
- IBD Centre, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | - Emma Johnston
- Department of Gastroenterology, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Gaurav Agrawal
- IBD Centre, Guy's and Saint Thomas' NHS Foundation Trust, London, UK,Department of Nutritional Sciences, King's College London, London, UK
| | - Bahij Al-Hakim
- IBD Centre, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | | | - Jeremy Sanderson
- IBD Centre, Guy's and Saint Thomas' NHS Foundation Trust, London, UK,Department of Nutritional Sciences, King's College London, London, UK
| |
Collapse
|
17
|
Bots ST, Hoeben RC. Non-Human Primate-Derived Adenoviruses for Future Use as Oncolytic Agents? Int J Mol Sci 2020; 21:ijms21144821. [PMID: 32650405 PMCID: PMC7404033 DOI: 10.3390/ijms21144821] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
Non-human primate (NHP)-derived adenoviruses have formed a valuable alternative for the use of human adenoviruses in vaccine development and gene therapy strategies by virtue of the low seroprevalence of neutralizing immunity in the human population. The more recent use of several human adenoviruses as oncolytic agents has exhibited excellent safety profiles and firm evidence of clinical efficacy. This proffers the question whether NHP-derived adenoviruses could also be employed for viral oncolysis in human patients. While vaccine vectors are conventionally made as replication-defective vectors, in oncolytic applications replication-competent viruses are used. The data on NHP-derived adenoviral vectors obtained from vaccination studies can only partially support the suitability of NHP-derived adenoviruses for use in oncolytic virus therapy. In addition, the use of NHP-derived adenoviruses in humans might be received warily given the recent zoonotic infections with influenza viruses and coronaviruses. In this review, we discuss the similarities and differences between human- and NHP-derived adenoviruses in view of their use as oncolytic agents. These include their genome organization, receptor use, replication and cell lysis, modulation of the host’s immune responses, as well as their pathogenicity in humans. Together, the data should facilitate a rational and data-supported decision on the suitability of NHP-derived adenoviruses for prospective use in oncolytic virus therapy.
Collapse
|
18
|
Folegatti PM, Bittaye M, Flaxman A, Lopez FR, Bellamy D, Kupke A, Mair C, Makinson R, Sheridan J, Rohde C, Halwe S, Jeong Y, Park YS, Kim JO, Song M, Boyd A, Tran N, Silman D, Poulton I, Datoo M, Marshall J, Themistocleous Y, Lawrie A, Roberts R, Berrie E, Becker S, Lambe T, Hill A, Ewer K, Gilbert S. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral-vectored vaccine: a dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial. THE LANCET. INFECTIOUS DISEASES 2020; 20:816-826. [PMID: 32325038 PMCID: PMC7172901 DOI: 10.1016/s1473-3099(20)30160-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cases of Middle East respiratory syndrome coronavirus (MERS-CoV) infection continue to rise in the Arabian Peninsula 7 years after it was first described in Saudi Arabia. MERS-CoV poses a significant risk to public health security because of an absence of currently available effective countermeasures. We aimed to assess the safety and immunogenicity of the candidate simian adenovirus-vectored vaccine expressing the full-length spike surface glycoprotein, ChAdOx1 MERS, in humans. METHODS This dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial was done at the Centre for Clinical Vaccinology and Tropical Medicine (Oxford, UK) and included healthy people aged 18-50 years with negative pre-vaccination tests for HIV antibodies, hepatitis B surface antigen, and hepatitis C antibodies (and a negative urinary pregnancy test for women). Participants received a single intramuscular injection of ChAdOx1 MERS at three different doses: the low-dose group received 5 × 109 viral particles, the intermediate-dose group received 2·5 × 1010 viral particles, and the high-dose group received 5 × 1010 viral particles. The primary objective was to assess safety and tolerability of ChAdOx1 MERS, measured by the occurrence of solicited, unsolicited, and serious adverse events after vaccination. The secondary objective was to assess the cellular and humoral immunogenicity of ChAdOx1 MERS, measured by interferon-γ-linked enzyme-linked immunospot, ELISA, and virus neutralising assays after vaccination. Participants were followed up for up to 12 months. This study is registered with ClinicalTrials.gov, NCT03399578. FINDINGS Between March 14 and Aug 15, 2018, 24 participants were enrolled: six were assigned to the low-dose group, nine to the intermediate-dose group, and nine to the high-dose group. All participants were available for follow-up at 6 months, but five (one in the low-dose group, one in the intermediate-dose group, and three in the high-dose group) were lost to follow-up at 12 months. A single dose of ChAdOx1 MERS was safe at doses up to 5 × 1010 viral particles with no vaccine-related serious adverse events reported by 12 months. One serious adverse event reported was deemed to be not related to ChAdOx1 MERS. 92 (74% [95% CI 66-81]) of 124 solicited adverse events were mild, 31 (25% [18-33]) were moderate, and all were self-limiting. Unsolicited adverse events in the 28 days following vaccination considered to be possibly, probably, or definitely related to ChAdOx1 MERS were predominantly mild in nature and resolved within the follow-up period of 12 months. The proportion of moderate and severe adverse events was significantly higher in the high-dose group than in the intermediate-dose group (relative risk 5·83 [95% CI 2·11-17·42], p<0·0001) Laboratory adverse events considered to be at least possibly related to the study intervention were self-limiting and predominantly mild in severity. A significant increase from baseline in T-cell (p<0·003) and IgG (p<0·0001) responses to the MERS-CoV spike antigen was observed at all doses. Neutralising antibodies against live MERS-CoV were observed in four (44% [95% CI 19-73]) of nine participants in the high-dose group 28 days after vaccination, and 19 (79% [58-93]) of 24 participants had antibodies capable of neutralisation in a pseudotyped virus neutralisation assay. INTERPRETATION ChAdOx1 MERS was safe and well tolerated at all tested doses. A single dose was able to elicit both humoral and cellular responses against MERS-CoV. The results of this first-in-human clinical trial support clinical development progression into field phase 1b and 2 trials. FUNDING UK Department of Health and Social Care, using UK Aid funding, managed by the UK National Institute for Health Research.
Collapse
Affiliation(s)
- Pedro M Folegatti
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mustapha Bittaye
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Amy Flaxman
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Fernando Ramos Lopez
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Duncan Bellamy
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra Kupke
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, Thematic Translational Unit Emerging Infections, Marburg, Germany
| | - Catherine Mair
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rebecca Makinson
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jonathan Sheridan
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Cornelius Rohde
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, Thematic Translational Unit Emerging Infections, Marburg, Germany
| | - Sandro Halwe
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, Thematic Translational Unit Emerging Infections, Marburg, Germany
| | - Yuji Jeong
- International Vaccine Institute, Science Unit, Seoul, South Korea
| | - Young-Shin Park
- International Vaccine Institute, Science Unit, Seoul, South Korea
| | - Jae-Ouk Kim
- International Vaccine Institute, Science Unit, Seoul, South Korea
| | - Manki Song
- International Vaccine Institute, Science Unit, Seoul, South Korea
| | - Amy Boyd
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nguyen Tran
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel Silman
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ian Poulton
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mehreen Datoo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Julia Marshall
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Yrene Themistocleous
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alison Lawrie
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rachel Roberts
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eleanor Berrie
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stephan Becker
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, Thematic Translational Unit Emerging Infections, Marburg, Germany
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Ewer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|