1
|
Jang SO, Lee JH, Chung YJ, Oh HS, Shin M, Kim SO, Hong SP. Chimeric adenovirus-based herpes zoster vaccine with the tPA signal peptide elicits a robust T-cell immune response. Virology 2024; 600:110243. [PMID: 39288613 DOI: 10.1016/j.virol.2024.110243] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Herpes zoster (HZ), or shingles, is caused by reactivation of the varicella-zoster virus (VZV), which remains latent in the sensory ganglia until immunity wanes with age. The representative HZ vaccine, Shingrix is efficacious but causes side effects due to vaccine adjuvants. Therefore, the development of highly efficacious vaccines with minimal side effects is required. We developed chimeric adenovirus vector (ChimAd)-based HZ vaccine candidates encoding the VZV glycoprotein E (gE). These candidates include ChimAd-tPAgE, in which the signal peptide is replaced with tissue plasminogen activator (tPA), and ChimAd-WTgE, which retains the original signal peptide. C57BL/6 mice were immunized with VZV-vaccine candidates, and cellular and humoral immune responses were evaluated using interferon-γ ELISPOT and ELISA. The ChimAd-based HZ vaccines induced high levels of gE-specific antibodies and cell-mediated immunity. ChimAd-tPAgE (optimal dose: 1 × 107 IFU) elicited a more robust gE-specific T-cell response than Shingrix and Zostavax, showing potential as HZ prophylactic vaccines.
Collapse
Affiliation(s)
- Sun Ok Jang
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae Hyun Lee
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Yong-Ju Chung
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyun-Seok Oh
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Myeongcheol Shin
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soo-Ok Kim
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sun Pyo Hong
- R&D Center, GeneMatrix, Inc., Seongnam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
2
|
Shrwani KJ, Mahallawi WH, Mohana AI, Algaissi A, Dhayhi N, Sharwani NJ, Gadour E, Aldossari SM, Asiri H, Kameli N, Asiri AY, Asiri AM, Sherwani AJ, Cunliffe N, Zhang Q. Mucosal immunity in upper and lower respiratory tract to MERS-CoV. Front Immunol 2024; 15:1358885. [PMID: 39281686 PMCID: PMC11392799 DOI: 10.3389/fimmu.2024.1358885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/15/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Middle East respiratory syndrome coronavirus (MERS-CoV) has emerged as a deadly pathogen with a mortality rate of up to 36.2%. MERS-CoV can cause severe respiratory tract disease and multiorgan failure. Therefore, therapeutic vaccines are urgently needed. This intensive review explores the human immune responses and their immunological mechanisms during MERS-CoV infection in the mucosa of the upper and lower respiratory tracts (URT and LRT, respectively). Objective The aim of this study is to provide a valuable, informative, and critical summary of the protective immune mechanisms against MERS-CoV infection in the URT/LRT for the purpose of preventing and controlling MERS-CoV disease and designing effective therapeutic vaccines. Methods In this review, we focus on the immune potential of the respiratory tract following MERS-CoV infection. We searched PubMed, Embase, Web of Science, Cochrane, Scopus, and Google Scholar using the following terms: "MERS-CoV", "B cells", "T cells", "cytokines", "chemokines", "cytotoxic", and "upper and lower respiratory tracts". Results We found and included 152 studies in this review. We report that the cellular innate immune response, including macrophages, dendritic cells, and natural killer cells, produces antiviral substances such as interferons and interleukins to prevent the virus from spreading. In the adaptive and humoral immune responses, CD4+ helper T cells, CD8+ cytotoxic T cells, B cells, and plasma cells protect against MERS-CoV infection in URT and LRT. Conclusion The human nasopharynx-associated lymphoid tissue (NALT) and bronchus-associated lymphoid tissue (BALT) could successfully limit the spread of several respiratory pathogens. However, in the case of MERS-CoV infection, limited research has been conducted in humans with regard to immunopathogenesis and mucosal immune responses due to the lack of relevant tissues. A better understanding of the immune mechanisms of the URT and LRT is vital for the design and development of effective MERS-CoV vaccines.
Collapse
Affiliation(s)
- Khalid J Shrwani
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Public Health Authority, Saudi Center for Disease Prevention and Control (SCDC), Jazan, Saudi Arabia
| | - Waleed H Mahallawi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Abdulrhman I Mohana
- Department of Antimicrobial Resistance, Public Health Authority, Riyadh, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Emerging and Endemic Infectious Diseases Research Unit, Health Sciences Research Center, Jazan University, Jazan, Saudi Arabia
| | - Nabil Dhayhi
- Department of Pediatrics, King Fahad Central Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nouf J Sharwani
- Department of Surgery, Mohammed bin Nasser Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Eyad Gadour
- Department of Gastroenterology and Hepatology, King Abdulaziz National Guard Hospital, Ahsa, Saudi Arabia
- Department of Medicine, Faculty of Medicine, Zamzam University College, Khartoum, Sudan
| | - Saeed M Aldossari
- Medical Laboratory Technology Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Hasan Asiri
- Medical Laboratory Department, Prince Mohammed bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Nader Kameli
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ayad Y Asiri
- Intensive Care Unit Department, Al Inma Medical Group, Al Hayat National Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah M Asiri
- Preventive Medicine Assistant Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Alaa J Sherwani
- Department of Pediatrics, Abu-Arish General Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nigel Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Qibo Zhang
- Academic and Research Departments, Section of Immunology, School of Biosciences, University of Surrey, Surrey, United Kingdom
| |
Collapse
|
3
|
Willison AG, Pawlitzki M, Lunn MP, Willison HJ, Hartung HP, Meuth SG. SARS-CoV-2 Vaccination and Neuroimmunological Disease: A Review. JAMA Neurol 2024; 81:179-186. [PMID: 38227318 DOI: 10.1001/jamaneurol.2023.5208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Importance The temporal association between the occurrence of neurological diseases, many autoimmune diseases, and vaccination against SARS-CoV-2 has been topically interesting and remains hotly debated both in the medical literature and the clinic. Given the very low incidences of these events both naturally occurring and in relation to vaccination, it is challenging to determine with certainty whether there is any causative association and most certainly what the pathophysiology of that causation could be. Observations Data from international cohorts including millions of vaccinated individuals suggest that there is a probable association between the adenovirus-vectored vaccines and Guillain-Barré syndrome (GBS). Further associations between other SARS-CoV-2 vaccines and GBS or Bell palsy have not been clearly demonstrated in large cohort studies, but the possible rare occurrence of Bell palsy following messenger RNA vaccination is a topic of interest. It is also yet to be clearly demonstrated that any other neurological diseases, such as central nervous system demyelinating disease or myasthenia gravis, have any causative association with vaccination against SARS-CoV-2 using any vaccine type, although it is possible that vaccination may rarely trigger a relapse or worsen symptoms or first presentation in already-diagnosed or susceptible individuals. Conclusions and Relevance The associated risk between SARS-CoV-2 vaccination and GBS, and possibly Bell palsy, is slight, and this should not change the recommendation for individuals to be vaccinated. The same advice should be given to those with preexisting neurological autoimmune disease.
Collapse
Affiliation(s)
- Alice Grizzel Willison
- Department of Neurology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Marc Pawlitzki
- Department of Neurology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Peter Lunn
- Centre for Neuromuscular Disease, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
- Department of Neuromuscular Disease, Institute of Neurology, University College London, London, United Kingdom
| | - Hugh John Willison
- College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Hans-Peter Hartung
- Department of Neurology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Brain and Mind Center, University of Sydney, Sydney, Australia
- Department of Neurology, Palacky University, Olomouc, Czech Republic
| | - Sven Günther Meuth
- Department of Neurology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
4
|
Gholap AD, Gupta J, Kamandar P, Bhowmik DD, Rojekar S, Faiyazuddin M, Hatvate NT, Mohanto S, Ahmed MG, Subramaniyan V, Kumarasamy V. Harnessing Nanovaccines for Effective Immunization─A Special Concern on COVID-19: Facts, Fidelity, and Future Prospective. ACS Biomater Sci Eng 2024; 10:271-297. [PMID: 38096426 DOI: 10.1021/acsbiomaterials.3c01247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Nanotechnology has emerged as a transformative pathway in vaccine research and delivery. Nanovaccines, encompassing lipid and nonlipid formulations, exhibit considerable advantages over traditional vaccine techniques, including enhanced antigen stability, heightened immunogenicity, targeted distribution, and the potential for codelivery with adjuvants or immune modulators. This review provides a comprehensive overview of the latest advancements and applications of lipid and non-lipid-based nanovaccines in current vaccination strategies for immunization. The review commences by outlining the fundamental concepts underlying lipid and nonlipid nanovaccine design before delving into the diverse components and production processes employed in their development. Subsequently, a comparative analysis of various nanocarriers is presented, elucidating their distinct physicochemical characteristics and impact on the immune response, along with preclinical and clinical studies. The discussion also highlights how nanotechnology enables the possibility of personalized and combined vaccination techniques, facilitating the creation of tailored nanovaccines to meet the individual patient needs. The ethical aspects concerning the use of nanovaccines, as well as potential safety concerns and public perception, are also addressed. The study underscores the gaps and challenges that must be overcome before adopting nanovaccines in clinical practice. This comprehensive analysis offers vital new insights into lipid and nonlipid nanovaccine status. It emphasizes the significance of continuous research, collaboration among interdisciplinary experts, and regulatory measures to fully unlock the potential of nanotechnology in enhancing immunization and ensuring a healthier, more resilient society.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Juhi Gupta
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Pallavi Kamandar
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Deblina D Bhowmik
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Md Faiyazuddin
- Department of Pharmaceutics, School of Pharmacy, Al-Karim University, Katihar 854106, Bihar, India
| | - Navnath T Hatvate
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Barker RA, Buttery PC. Disease-specific interventions: The use of cell and gene therapies for Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:171-191. [PMID: 39341654 DOI: 10.1016/b978-0-323-90120-8.00003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Approaches to repair the brain around the loss of the nigrostriatal dopaminergic pathways in Parkinson disease (PD) are not new and have been attempted over many years. However, of late, the situation has moved forward in two main ways. In the case of cell therapies, the ability to make large numbers of authentic midbrain dopaminergic neuroblasts from human pluripotent stem cell sources has turned what was an interesting avenue of research into a major area of investment and trialing, by academics in conjunction with Pharma. In the case of gene therapies, their use around dopamine replacement has waned, as the interest in using them for disease modification targeting PD-specific pathways has grown. In this chapter, we discuss all these developments and the current status of cell and gene therapies for PD.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - Philip C Buttery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Gupta S, Pellett S. Recent Developments in Vaccine Design: From Live Vaccines to Recombinant Toxin Vaccines. Toxins (Basel) 2023; 15:563. [PMID: 37755989 PMCID: PMC10536331 DOI: 10.3390/toxins15090563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
Vaccines are one of the most effective strategies to prevent pathogen-induced illness in humans. The earliest vaccines were based on live inoculations with low doses of live or related pathogens, which carried a relatively high risk of developing the disease they were meant to prevent. The introduction of attenuated and killed pathogens as vaccines dramatically reduced these risks; however, attenuated live vaccines still carry a risk of reversion to a pathogenic strain capable of causing disease. This risk is completely eliminated with recombinant protein or subunit vaccines, which are atoxic and non-infectious. However, these vaccines require adjuvants and often significant optimization to induce robust T-cell responses and long-lasting immune memory. Some pathogens produce protein toxins that cause or contribute to disease. To protect against the effects of such toxins, chemically inactivated toxoid vaccines have been found to be effective. Toxoid vaccines are successfully used today at a global scale to protect against tetanus and diphtheria. Recent developments for toxoid vaccines are investigating the possibilities of utilizing recombinant protein toxins mutated to eliminate biologic activity instead of chemically inactivated toxins. Finally, one of the most contemporary approaches toward vaccine design utilizes messenger RNA (mRNA) as a vaccine candidate. This approach was used globally to protect against coronavirus disease during the COVID-19 pandemic that began in 2019, due to its advantages of quick production and scale-up, and effectiveness in eliciting a neutralizing antibody response. Nonetheless, mRNA vaccines require specialized storage and transport conditions, posing challenges for low- and middle-income countries. Among multiple available technologies for vaccine design and formulation, which technology is most appropriate? This review focuses on the considerable developments that have been made in utilizing diverse vaccine technologies with a focus on vaccines targeting bacterial toxins. We describe how advancements in vaccine technology, combined with a deeper understanding of pathogen-host interactions, offer exciting and promising avenues for the development of new and improved vaccines.
Collapse
Affiliation(s)
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| |
Collapse
|
9
|
Pandit NB, Fulmali PR, Chandrayan P, Chauhan K, Bhil DL, Rasania MN. How safe is COVID-19 vaccination among pregnant women and its outcome - A hospital-based retrospective study in Indian population. J Family Med Prim Care 2023; 12:2140-2145. [PMID: 38024942 PMCID: PMC10657075 DOI: 10.4103/jfmpc.jfmpc_333_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Although getting the Covid infection is equal for every person, during pregnancy, the women's immunity is a little lower than usual, so they are more prone to infection. That is why they should be taken care of with more precautions. A vaccine is the best weapon to fight such infection. Covishield and Covaxin are the two vaccines first introduce in country India including for pregnant women. The safety of the vaccine was a big concern as one of them is a newer type of vaccine. The current study was planned with objectives to understand the safety aspect of Covid19 vaccine on pregnancy outcome and Adverse events following immunization (AEFI) following vaccination. Materials and Methods This was a hospital-based retrospective cohort study. The sample size was all the pregnant women who delivered a baby from July 2021 to April 2022 at the tertiary care hospital in Vadodara. These women were retrospectively assessed for the status of vaccination based on the record and other information related to ANC from the record. Total of 1974 women were eligible for study after inclusion-exclusion criteria. The collected data was analysed. Result Of the 1974 pregnant women, 531 (27%) took any of one covid19 vaccine and 1443 (73%) did not take vaccine. There were 511 (96%) women opted for Covishield vaccine and 20 (4%) women who opted for Covaxin. Of 531 women who took vaccination, 46% women had AEFI. The risk of low birth weight (LBW) baby was 40% among vaccinated v/s 39% among non-vaccinated and congenital malformation was 0.6% among vaccinated v/s 1% among non-vaccinated women. On the contrary, the risk of premature birth was 8% among the vaccinated group v/s 13% among the non-vaccinated group and NICU admission following delivery was 8% among the vaccinated group v/s 12% among the non-vaccinated group. Conclusion AEFI among pregnant women were found less compared to the general population. The study also revealed that both Covishield and Covaxin are found safe for pregnancy outcomes and can be given to pregnant women during any trimester of pregnancy message for a family physician.
Collapse
Affiliation(s)
- Niraj B. Pandit
- Department of Community Medicine, SBKS MI and RC, Sumandeep Vidypeeth, Vadodara, Gujarat, India
| | - Purvi R. Fulmali
- Department of Community Medicine Healthy Mother Healthy Child Project, Sumandeep Vidyapeeth, Vadodara, Gujarat, India
| | - Piyusha Chandrayan
- Department of Obstetrics and Gynaecology, Dhiraj Hospital, SBKS MIRC, Sumandeep Vidyapeeth, Vadodara, Gujarat, India
| | - Kishor Chauhan
- Department of Obstetrics and Gynaecology, Dhiraj Hospital, SBKS MIRC, Sumandeep Vidyapeeth, Vadodara, Gujarat, India
| | - Dipika L. Bhil
- Department of Paediatrics, Dhiraj Hospital, SBKS MIRC, Sumandeep Vidyapeeth, Vadodara, Gujarat, India
| | - Manish N. Rasania
- Department of Paediatrics, Dhiraj Hospital, SBKS MIRC, Sumandeep Vidyapeeth, Vadodara, Gujarat, India
| |
Collapse
|
10
|
Li Z, Lei Z, Cai Y, Cheng DB, Sun T. MicroRNA therapeutics and nucleic acid nano-delivery systems in bacterial infection: a review. J Mater Chem B 2023; 11:7804-7833. [PMID: 37539650 DOI: 10.1039/d3tb00694h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Bacteria that have worked with humans for thousands of years pose a major threat to human health even today, as drug resistance has become a prominent problem. Compared to conventional drug therapy, nucleic acid-based therapies are a promising and potential therapeutic strategy for diseases in which nucleic acids are delivered through a nucleic acid delivery system to regulate gene expression in specific cells, offering the possibility of curing intractable diseases that are difficult to treat at this stage. Among the many nucleic acid therapeutic ideas, microRNA, a class of small nucleic acids with special properties, has made great strides in biology and medicine in just over two decades, showing promise in preclinical drug development. In this review, we introduce recent advances in nucleic acid delivery systems and their clinical applications, highlighting the potential of nucleic acid therapies, especially miRNAs extracted from traditional herbs, in combination with the existing set of nucleic acid therapeutic systems, to potentially open up a new line of thought in the treatment of cancer, viruses, and especially bacterial infectious diseases.
Collapse
Affiliation(s)
- Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Yilun Cai
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
11
|
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
|
12
|
Feitsma EA, Janssen YF, Boersma HH, van Sleen Y, van Baarle D, Alleva DG, Lancaster TM, Sathiyaseelan T, Murikipudi S, Delpero AR, Scully MM, Ragupathy R, Kotha S, Haworth JR, Shah NJ, Rao V, Nagre S, Ronca SE, Green FM, Aminetzah A, Sollie F, Kruijff S, Brom M, van Dam GM, Zion TC. A randomized phase I/II safety and immunogenicity study of the Montanide-adjuvanted SARS-CoV-2 spike protein-RBD-Fc vaccine, AKS-452. Vaccine 2023; 41:2184-2197. [PMID: 36842886 PMCID: PMC9946892 DOI: 10.1016/j.vaccine.2023.02.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND Previous interim data from a phase I study of AKS-452, a subunit vaccine comprising an Fc fusion of the respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein receptor binding domain (SP/RBD) emulsified in the water-in-oil adjuvant, Montanide™ ISA 720, suggested a good safety and immunogenicity profile in healthy adults. This phase I study was completed and two dosing regimens were further evaluated in this phase II study. METHODS This phase II randomized, open-labelled, parallel group study was conducted at a single site in The Netherlands with 52 healthy adults (18 - 72 years) receiving AKS-452 subcutaneously at one 90 µg dose (cohort 1, 26 subjects) or two 45 µg doses 28 days apart (cohort 2, 26 subjects). Serum samples were collected at the first dose (day 0) and at days 28, 56, 90, and 180. Safety and immunogenicity endpoints were assessed, along with induction of IgG isotypes, cross-reactive immunity against viral variants, and IFN-γ T cell responses. RESULTS All AEs were mild/moderate (grades 1 or 2), and no SAEs were attributable to AKS-452. Seroconversion rates reached 100% in both cohorts, although cohort 2 showed greater geometric mean IgG titers that were stable through day 180 and associated with enhanced potencies of SP/RBD-ACE2 binding inhibition and live virus neutralization. AKS-452-induced IgG titers strongly bound mutant SP/RBD from several SARS-CoV-2 variants (including Omicrons) that were predominantly of the favorable IgG1/3 isotype and IFN-γ-producing T cell phenotype. CONCLUSION These favorable safety and immunogenicity profiles of the candidate vaccine as demonstrated in this phase II study are consistent with those of the phase I study (ClinicalTrials.gov: NCT04681092) and suggest that a total of 90 µg received in 2 doses may offer a greater duration of cross-reactive neutralizing titers than when given in a single dose.
Collapse
Affiliation(s)
- Eline A Feitsma
- Department of Surgery, University Medical Center Groningen (UMCG), Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Yester F Janssen
- Department of Nuclear Medicine and Molecular Imaging, UMCG, the Netherlands
| | - Hendrikus H Boersma
- Department of Nuclear Medicine and Molecular Imaging, UMCG, the Netherlands; Department of Clinical Pharmacy and Pharmacology, UMCG, the Netherlands
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, UMCG, the Netherlands
| | - Debbie van Baarle
- Department of Rheumatology and Clinical Immunology, UMCG, the Netherlands
| | - David G Alleva
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Thomas M Lancaster
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | | | - Sylaja Murikipudi
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Andrea R Delpero
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Melanie M Scully
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Ramya Ragupathy
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sravya Kotha
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Jeffrey R Haworth
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Nishit J Shah
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Vidhya Rao
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Shashikant Nagre
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Shannon E Ronca
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Freedom M Green
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Ari Aminetzah
- TRACER BV, L.J. Zielstraweg 1, 9766 GX Groningen, the Netherlands
| | - Frans Sollie
- ICON, van Swietenlaan 6, 9728 NZ Groningen, the Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Center Groningen (UMCG), Hanzeplein 1, 9700 RB Groningen, the Netherlands; Department of Nuclear Medicine and Molecular Imaging, UMCG, the Netherlands
| | - Maarten Brom
- TRACER BV, L.J. Zielstraweg 1, 9766 GX Groningen, the Netherlands
| | - Gooitzen M van Dam
- Department of Nuclear Medicine and Molecular Imaging, UMCG, the Netherlands; TRACER BV, L.J. Zielstraweg 1, 9766 GX Groningen, the Netherlands
| | - Todd C Zion
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States.
| |
Collapse
|
13
|
Chavda VP, Bezbaruah R, Valu D, Patel B, Kumar A, Prasad S, Kakoti BB, Kaushik A, Jesawadawala M. Adenoviral Vector-Based Vaccine Platform for COVID-19: Current Status. Vaccines (Basel) 2023; 11:432. [PMID: 36851309 PMCID: PMC9965371 DOI: 10.3390/vaccines11020432] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus disease (COVID-19) breakout had an unimaginable worldwide effect in the 21st century, claiming millions of lives and putting a huge burden on the global economy. The potential developments in vaccine technologies following the determination of the genetic sequence of SARS-CoV-2 and the increasing global efforts to bring potential vaccines and therapeutics into the market for emergency use have provided a small bright spot to this tragic event. Several intriguing vaccine candidates have been developed using recombinant technology, genetic engineering, and other vaccine development technologies. In the last decade, a vast amount of the vaccine development process has diversified towards the usage of viral vector-based vaccines. The immune response elicited by such vaccines is comparatively higher than other approved vaccine candidates that require a booster dose to provide sufficient immune protection. The non-replicating adenoviral vectors are promising vaccine carriers for infectious diseases due to better yield, cGMP-friendly manufacturing processes, safety, better efficacy, manageable shipping, and storage procedures. As of April 2022, the WHO has approved a total of 10 vaccines around the world for COVID-19 (33 vaccines approved by at least one country), among which three candidates are adenoviral vector-based vaccines. This review sheds light on the developmental summary of all the adenoviral vector-based vaccines that are under emergency use authorization (EUA) or in the different stages of development for COVID-19 management.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Disha Valu
- Drug Product Development Laboratory, Biopharma Division, Intas Pharmaceutical Ltd., Moraiya, Ahmedabad 382213, Gujarat, India
| | - Bindra Patel
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Anup Kumar
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Sanjay Prasad
- Cell and Gene Therapy Drug Product Development Laboratory, Biopharma Division, Intas Pharmaceutical Ltd., Moraiya, Ahmedabad 382213, Gujarat, India
| | - Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health Systems Engineering, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805-8531, USA
| | - Mariya Jesawadawala
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
14
|
Sunagar R, Prasad SD, Ella R, Vadrevu KM. Preclinical evaluation of safety and immunogenicity of a primary series intranasal COVID-19 vaccine candidate (BBV154) and humoral immunogenicity evaluation of a heterologous prime-boost strategy with COVAXIN (BBV152). Front Immunol 2022; 13:1063679. [PMID: 36569867 PMCID: PMC9773076 DOI: 10.3389/fimmu.2022.1063679] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Most if not all vaccine candidates developed to combat COVID-19 due to SARS-CoV-2 infection are administered parenterally. As SARS-CoV-2 is transmitted through infectious respiratory fluids, vaccine-induced mucosal immunity could provide an important contribution to control this pandemic. ChAd-SARS-CoV-2-S (BBV154), a replication-defective chimpanzee adenovirus (ChAd)-vectored intranasal (IN) COVID-19 vaccine candidate, encodes a prefusion-stabilized version of the SARS-CoV-2 spike protein containing two proline substitutions in the S2 subunit. We performed preclinical evaluations of BBV154 in mice, rats, hamsters and rabbits. Repeated dose toxicity studies presented excellent safety profiles in terms of pathology and biochemical analysis. IN administration of BBV154 elicited robust mucosal and systemic humoral immune responses coupled with Th1 cell-mediated immune responses. BBV154 IN vaccination also elicited potent variant (omicron) cross neutralization antibodies. Assessment of anti-vector (ChAd36) neutralizing antibodies following repeated doses of BBV154 IN administration showed insignificant titers of ChAd36 neutralizing antibodies. However, the immune sera derived from the same animals displayed significantly higher levels of SARS-CoV-2 virus neutralization (p<0.003). We also evaluated the safety and immunogenicity of heterologous prime-boost vaccination with intramuscular (IM) COVAXIN-prime followed by BBV154 IN administration. COVAXIN priming followed by BBV154 IN-booster showed an acceptable reactogenicity profile comparable to the homologous COVAXIN/COVAXIN or BBV154/BBV154 vaccination model. Heterologous vaccination of COVAXIN-prime and BBV154 booster also elicited superior (p<0.005) and cross variant (omicron) protective immune responses (p<0.013) compared with the homologous COVAXIN/COVAXIN schedule. BBV154 has successfully completed both homologous and heterologous combination schedules of human phase 3 clinical trials and received the restricted emergency use approval (in those aged above 18 years) from the Drugs Controller General of India (DCGI).
Collapse
|
15
|
Adenovirus vector system: construction, history and therapeutic applications. Biotechniques 2022; 73:297-305. [DOI: 10.2144/btn-2022-0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Since the isolation of adenovirus (AdV) in 1953, AdVs have been used as vectors for various therapeutic purposes, such as gene therapy in cancers and other malignancies, vaccine development and delivery of CRISPR-Cas9 machinery. Over the years, several AdV vector modifications have been introduced, including fiber switching, incorporation of ligands in the viral capsid and hexon modification of the fiber, to improve the efficiency of AdV as a vector. CRISPR-Cas9 has recently been used for these modifications and is also used in other adeno-associated viruses. These modifications further allow the production of AdV libraries that display random peptides for the production of cancer-targeting AdV vectors. This review focuses on the common methods of AdV construction, changes in AdV tropism for the improvement of therapeutic efficiency and the role of AdV vectors in gene therapy, vaccine development and CRISPR-Cas9 delivery.
Collapse
|
16
|
Gazal S, Sharma N, Gazal S, Tikoo M, Shikha D, Badroo GA, Rashid M, Lee SJ. Nipah and Hendra Viruses: Deadly Zoonotic Paramyxoviruses with the Potential to Cause the Next Pandemic. Pathogens 2022; 11:pathogens11121419. [PMID: 36558753 PMCID: PMC9784551 DOI: 10.3390/pathogens11121419] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Nipah and Hendra viruses are deadly zoonotic paramyxoviruses with a case fatality rate of upto 75%. The viruses belong to the genus henipavirus in the family Paramyxoviridae, a family of negative-sense single-stranded RNA viruses. The natural reservoirs of NiV and HeV are bats (flying foxes) in which the virus infection is asymptomatic. The intermediate hosts for NiV and HeV are swine and equine, respectively. In humans, NiV infections result in severe and often fatal respiratory and neurological manifestations. The Nipah virus was first identified in Malaysia and Singapore following an outbreak of encephalitis in pig farmers and subsequent outbreaks have been reported in Bangladesh and India almost every year. Due to its extreme pathogenicity, pandemic potential, and lack of established antiviral therapeutics and vaccines, research on henipaviruses is highly warranted so as to develop antivirals or vaccines that could aid in the prevention and control of future outbreaks.
Collapse
Affiliation(s)
- Sabahat Gazal
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Neelesh Sharma
- Division of Veterinary Medicine, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
- Correspondence: (N.S.); (S.-J.L.)
| | - Sundus Gazal
- Division of Veterinary Microbiology, College of Veterinary Sciences, Guru Angad Dev Veterinary and Animal Science University, Ludhiana 141004, Punjab, India
| | - Mehak Tikoo
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Deep Shikha
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Gulzar Ahmed Badroo
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Mohd Rashid
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Sung-Jin Lee
- Department of Applied Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
- Correspondence: (N.S.); (S.-J.L.)
| |
Collapse
|
17
|
Isaac AB, Karolina W, Temitope AA, Anuska R, Joanne E, Deborah A, Bianca OC, Filip T, Zofia P, Oluwasegun OI, Oluwaferanmi O, Grace BT. PROSPECTS OF LASSA FEVER CANDIDATE VACCINES. Afr J Infect Dis 2022; 16:46-58. [PMID: 36124324 PMCID: PMC9480887 DOI: 10.21010/ajid.v16i2s.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/09/2022] Open
Abstract
Background Lassa fever is an acute viral haemorrhagic disease caused by the Lassa virus (LASV). It is endemic in West Africa and infects about 300,000 people each year, leading to approximately 5000 deaths annually. The development of the LASV vaccine has been listed as a priority by the World Health Organization since 2018. Considering the accelerated development and availability of vaccines against COVID-19, we set out to assess the prospects of LASV vaccines and the progress made so far. Materials and Methods We reviewed the progress made on twenty-six vaccine candidates listed by Salami et al. (2019) and searched for new vaccine candidates through Google Scholar, PubMed, and DOAJ from June to July 2021. We searched the articles published in English using keywords that included "vaccine" AND "Lassa fever" OR "Lassa virus" in the title/abstract. Results Thirty-four candidate vaccines were identified - 26 already listed in the review by Salami et al. and an additional 8, which were developed over the last seven years. 30 vaccines are still in the pre-clinical stage while 4 of them are currently undergoing clinical trials. The most promising candidates in 2019 were vesicular stomatitis virus-vectored vaccine and live-attenuated MV/LASV vaccine; both had progressed to clinical trials. Conclusions Despite the focus on COVID-19 vaccines since 2020, LASV vaccine is under development and continues to make impressive progress, hence more emphasis should be put into exploring further clinical studies related to the most promising types of vaccines identified.
Collapse
Affiliation(s)
- Ademusire Babatunde Isaac
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Wieczorek Karolina
- Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria,Queen Mary University of London Barts and The London School of Medicine and Dentistry, United Kingdom,Corresponding author’s E-Mail:
| | - Alonge Aishat Temitope
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Rajen Anuska
- Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria,Queen Mary University of London Barts and The London School of Medicine and Dentistry, United Kingdom
| | - Egbe Joanne
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Adebambo Deborah
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Offorbuike Chiamaka Bianca
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Trojan Filip
- University College London, Medical School, London, United Kingdom
| | - Przypaśniak Zofia
- Queen Mary University of London Barts and The London School of Medicine and Dentistry, United Kingdom
| | - Oduguwa Ifeoluwa Oluwasegun
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Omitoyin Oluwaferanmi
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| | - Balogun Toluwalogo Grace
- College of Medicine, University of Ibadan, Ibadan, Nigeria,Polygeia (Global Health Student Think Tank), Ibadan Branch, Nigeria
| |
Collapse
|
18
|
Fathi A, Dahlke C, Krähling V, Kupke A, Okba NMA, Raadsen MP, Heidepriem J, Müller MA, Paris G, Lassen S, Klüver M, Volz A, Koch T, Ly ML, Friedrich M, Fux R, Tscherne A, Kalodimou G, Schmiedel S, Corman VM, Hesterkamp T, Drosten C, Loeffler FF, Haagmans BL, Sutter G, Becker S, Addo MM. Increased neutralization and IgG epitope identification after MVA-MERS-S booster vaccination against Middle East respiratory syndrome. Nat Commun 2022; 13:4182. [PMID: 35853863 PMCID: PMC9295877 DOI: 10.1038/s41467-022-31557-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
Vaccine development is essential for pandemic preparedness. We previously conducted a Phase 1 clinical trial of the vector vaccine candidate MVA-MERS-S against the Middle East respiratory syndrome coronavirus (MERS-CoV), expressing its full spike glycoprotein (MERS-CoV-S), as a homologous two-dose regimen (Days 0 and 28). Here, we evaluate the safety (primary objective) and immunogenicity (secondary and exploratory objectives: magnitude and characterization of vaccine-induced humoral responses) of a third vaccination with MVA-MERS-S in a subgroup of trial participants one year after primary immunization. MVA-MERS-S booster vaccination is safe and well-tolerated. Both binding and neutralizing anti-MERS-CoV antibody titers increase substantially in all participants and exceed maximum titers observed after primary immunization more than 10-fold. We identify four immunogenic IgG epitopes, located in the receptor-binding domain (RBD, n = 1) and the S2 subunit (n = 3) of MERS-CoV-S. The level of baseline anti-human coronavirus antibody titers does not impact the generation of anti-MERS-CoV antibody responses. Our data support the rationale of a booster vaccination with MVA-MERS-S and encourage further investigation in larger trials. Trial registration: Clinicaltrials.gov NCT03615911.
Collapse
Affiliation(s)
- Anahita Fathi
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - Christine Dahlke
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Verena Krähling
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Alexandra Kupke
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Nisreen M A Okba
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Matthijs P Raadsen
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Jasmin Heidepriem
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Marcel A Müller
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Grigori Paris
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Susan Lassen
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Michael Klüver
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Asisa Volz
- University of Veterinary Medicine Hanover, Institute of Virology, Hanover, Germany
- German Center for Infection Research, partner site Hanover-Brunswick, Hanover, Germany
| | - Till Koch
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - My L Ly
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Monika Friedrich
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Robert Fux
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Alina Tscherne
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Georgia Kalodimou
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Stefan Schmiedel
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, First Department of Medicine, Division of Infectious Diseases, Hamburg, Germany
| | - Victor M Corman
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Thomas Hesterkamp
- German Center for Infection Research, Translational Project Management Office, Brunswick, Germany
| | - Christian Drosten
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
- German Center for Infection Research, partner site Berlin, Berlin, Germany
| | - Felix F Loeffler
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, Germany
| | - Bart L Haagmans
- Erasmus Medical Center, Department of Viroscience, Rotterdam, the Netherlands
| | - Gerd Sutter
- LMU University of Munich, Institute of Infectious Diseases and Zoonoses, Munich, Germany
- German Center for Infection Research, partner site Munich, Munich, Germany
| | - Stephan Becker
- Philipps University Marburg, Institute of Virology, Marburg, Germany
- German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Marylyn M Addo
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany.
- Bernhard-Nocht-Institute for Tropical Medicine, Department for Clinical Immunology of Infectious Diseases, Hamburg, Germany.
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
19
|
Abstract
Cancer is one of the leading causes of death in the world, which is the second after heart diseases. Adenoviruses (Ads) have become the promise of new therapeutic strategy for cancer treatment. The objective of this review is to discuss current advances in the applications of adenoviral vectors in cancer therapy. Adenoviral vectors can be engineered in different ways so as to change the tumor microenvironment from cold tumor to hot tumor, including; 1. by modifying Ads to deliver transgenes that codes for tumor suppressor gene (p53) and other proteins whose expression result in cell cycle arrest 2. Ads can also be modified to express tumor specific antigens, cytokines, and other immune-modulatory molecules. The other strategy to use Ads in cancer therapy is to use oncolytic adenoviruses, which directly kills tumor cells. Gendicine and Advexin are replication-defective recombinant human p53 adenoviral vectors that have been shown to be effective against several types of cancer. Gendicine was approved for treatment of squamous cell carcinoma of the head and neck by the Chinese Food and Drug Administration (FDA) agency in 2003 as a first-ever gene therapy product. Oncorine and ONYX-015 are oncolytic adenoviral vectors that have been shown to be effective against some types of cancer. The Chiness FDA agency has also approved Oncorin for the treatment of head and neck cancer. Ads that were engineered to express immune-stimulatory cytokines and other immune-modulatory molecules such as TNF-α, IL-2, BiTE, CD40L, 4-1BBL, GM-CSF, and IFN have shown promising outcome in treatment of cancer. Ads can also improve therapeutic efficacy of immune checkpoint inhibitors and adoptive cell therapy (Chimeric Antigen Receptor T Cells). In addition, different replication-deficient adenoviral vectors (Ad5-CEA, Ad5-PSA, Ad-E6E7, ChAdOx1-MVA and Ad-transduced Dendritic cells) that were tested as anticancer vaccines have been demonstrated to induce strong antitumor immune response. However, the use of adenoviral vectors in gene therapy is limited by several factors such as pre-existing immunity to adenoviral vectors and high immunogenicity of the viruses. Thus, innovative strategies must be continually developed so as to overcome the obstacles of using adenoviral vectors in gene therapy.
Collapse
Affiliation(s)
- Sintayehu Tsegaye Tseha
- Lecturer of Biomedical Sciences, Department of Biology, College of Natural and Computational Sciences, Arba Minch University, Arba Minch, Ethiopia
- Department of Microbial, Cellular and Molecular Biology, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
20
|
Coughlan L, Kremer EJ, Shayakhmetov DM. Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther 2022; 30:1822-1849. [PMID: 35092844 PMCID: PMC8801892 DOI: 10.1016/j.ymthe.2022.01.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Zoonotic viruses continually pose a pandemic threat. Infection of humans with viruses for which we typically have little or no prior immunity can result in epidemics with high morbidity and mortality. These epidemics can have public health and economic impact and can exacerbate civil unrest or political instability. Changes in human behavior in the past few decades-increased global travel, farming intensification, the exotic animal trade, and the impact of global warming on animal migratory patterns, habitats, and ecosystems-contribute to the increased frequency of cross-species transmission events. Investing in the pre-clinical advancement of vaccine candidates against diverse emerging viral threats is crucial for pandemic preparedness. Replication-defective adenoviral (Ad) vectors have demonstrated their utility as an outbreak-responsive vaccine platform during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Ad vectors are easy to engineer; are amenable to rapid, inexpensive manufacturing; are relatively safe and immunogenic in humans; and, importantly, do not require specialized cold-chain storage, making them an ideal platform for equitable global distribution or stockpiling. In this review, we discuss the progress in applying Ad-based vaccines against emerging viruses and summarize their global safety profile, as reflected by their widespread geographic use during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS 5535, Montpellier, France.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
21
|
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
|
22
|
Janssen YF, Feitsma EA, Boersma HH, Alleva DG, Lancaster TM, Sathiyaseelan T, Murikipudi S, Delpero AR, Scully MM, Ragupathy R, Kotha S, Haworth JR, Shah NJ, Rao V, Nagre S, Ronca SE, Green FM, Aminetzah A, Sollie F, Kruijff S, Brom M, van Dam GM, Zion TC. Phase I interim results of a phase I/II study of the IgG-Fc fusion COVID-19 subunit vaccine, AKS-452. Vaccine 2022; 40:1253-1260. [PMID: 35115195 PMCID: PMC8802018 DOI: 10.1016/j.vaccine.2022.01.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023]
Abstract
To address the coronavirus disease 2019 (COVID-19) pandemic caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a recombinant subunit vaccine, AKS-452, is being developed comprising an Fc fusion protein of the SARS-CoV-2 viral spike protein receptor binding domain (SP/RBD) antigen and human IgG1 Fc emulsified in the water-in-oil adjuvant, Montanide™ ISA 720. A single-center, open-label, phase I dose-finding and safety study was conducted with 60 healthy adults (18–65 years) receiving one or two doses 28 days apart of 22.5 µg, 45 µg, or 90 µg of AKS-452 (i.e., six cohorts, N = 10 subjects per cohort). Primary endpoints were safety and reactogenicity and secondary endpoints were immunogenicity assessments. No AEs ≥ 3, no SAEs attributable to AKS-452, and no SARS-CoV-2 viral infections occurred during the study. Seroconversion rates of anti-SARS-CoV-2 SP/RBD IgG titers in the 22.5, 45, and 90 µg cohorts at day 28 were 70%, 90%, and 100%, respectively, which all increased to 100% at day 56 (except 89% for the single-dose 22.5 µg cohort). All IgG titers were Th1-isotype skewed and efficiently bound mutant SP/RBD from several SARS-CoV-2 variants with strong neutralization potencies of live virus infection of cells (including alpha and delta variants). The favorable safety and immunogenicity profiles of this phase I study (ClinicalTrials.gov: NCT04681092) support phase II initiation of this room-temperature stable vaccine that can be rapidly and inexpensively manufactured to serve vaccination at a global scale without the need of a complex distribution or cold chain.
Collapse
|
23
|
Sterilizing Immunity against COVID-19: Developing Helper T cells I and II activating vaccines is imperative. Biomed Pharmacother 2021; 144:112282. [PMID: 34624675 PMCID: PMC8486642 DOI: 10.1016/j.biopha.2021.112282] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 01/04/2023] Open
Abstract
Six months after the publication of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) sequence, a record number of vaccine candidates were listed, and quite a number of them have since been approved for emergency use against the novel coronavirus disease 2019 (COVID-19). This unprecedented pharmaceutical feat did not only show commitment, creativity and collaboration of the scientific community, but also provided a swift solution that prevented global healthcare system breakdown. Notwithstanding, the available data show that most of the approved COVID-19 vaccines protect only a proportion of recipients against severe disease but do not prevent clinical manifestation of COVID-19. There is therefore the need to probe further to establish whether these vaccines can induce sterilizing immunity, otherwise, COVID-19 vaccination would have to become a regular phenomenon. The emergence of SARS-CoV-2 variants could further affect the capability of the available COVID-19 vaccines to prevent infection and protect recipients from a severe form of the disease. These notwithstanding, data about which vaccine(s), if any, can confer sterilizing immunity are unavailable. Here, we discuss the immune responses to viral infection with emphasis on COVID-19, and the specific adaptive immune response to SARS-CoV-2 and how it can be harnessed to develop COVID-19 vaccines capable of conferring sterilizing immunity. We further propose factors that could be considered in the development of COVID-19 vaccines capable of stimulating sterilizing immunity. Also, an old, but effective vaccine development technology that can be applied in the development of COVID-19 vaccines with sterilizing immunity potential is reviewed.
Collapse
|
24
|
Alleva DG, Delpero AR, Scully MM, Murikipudi S, Ragupathy R, Greaves EK, Sathiyaseelan T, Haworth JR, Shah NJ, Rao V, Nagre S, Lancaster TM, Webb SS, Jasa AI, Ronca SE, Green FM, Elyard HA, Yee J, Klein J, Karnes L, Sollie F, Zion TC. Development of an IgG-Fc fusion COVID-19 subunit vaccine, AKS-452. Vaccine 2021; 39:6601-6613. [PMID: 34642088 PMCID: PMC8491978 DOI: 10.1016/j.vaccine.2021.09.077] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
AKS-452 is a biologically-engineered vaccine comprising an Fc fusion protein of the SARS-CoV-2 viral spike protein receptor binding domain antigen (Ag) and human IgG1 Fc (SP/RBD-Fc) in clinical development for the induction and augmentation of neutralizing IgG titers against SARS-CoV-2 viral infection to address the COVID-19 pandemic. The Fc moiety is designed to enhance immunogenicity by increasing uptake via Fc-receptors (FcγR) on Ag-presenting cells (APCs) and prolonging exposure due to neonatal Fc receptor (FcRn) recycling. AKS-452 induced approximately 20-fold greater neutralizing IgG titers in mice relative to those induced by SP/RBD without the Fc moiety and induced comparable long-term neutralizing titers with a single dose vs. two doses. To further enhance immunogenicity, AKS-452 was evaluated in formulations containing a panel of adjuvants in which the water-in-oil adjuvant, Montanide™ ISA 720, enhanced neutralizing IgG titers by approximately 7-fold after one and two doses in mice, including the neutralization of live SARS-CoV-2 virus infection of VERO-E6 cells. Furthermore, ISA 720-adjuvanted AKS-452 was immunogenic in rabbits and non-human primates (NHPs) and protected from infection and clinical symptoms with live SARS-CoV-2 virus in NHPs (USA-WA1/2020 viral strain) and the K18 human ACE2-trangenic (K18-huACE2-Tg) mouse (South African B.1.351 viral variant). These preclinical studies support the initiation of Phase I clinical studies with adjuvanted AKS-452 with the expectation that this room-temperature stable, Fc-fusion subunit vaccine can be rapidly and inexpensively manufactured to provide billions of doses per year especially in regions where the cold-chain is difficult to maintain.
Collapse
Affiliation(s)
- David G Alleva
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Andrea R Delpero
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Melanie M Scully
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sylaja Murikipudi
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Ramya Ragupathy
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Emma K Greaves
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | | | - Jeffrey R Haworth
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Nishit J Shah
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Vidhya Rao
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Shashikant Nagre
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Thomas M Lancaster
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States
| | - Sarah S Webb
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Allison I Jasa
- Biomere Biomedical Research Models, 57 Union St., Worcester, MA 01608, United States
| | - Shannon E Ronca
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Freedom M Green
- Feigin ABSL-3 Facility, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX 77030, United States
| | - Hanne Andersen Elyard
- BIOQUAL, Inc., 9600 Medical Center Drive, Suite 101, Rockville, MD 20850-3336, United States
| | - JoAnn Yee
- Primate Assay Laboratory, CA National Primate Research Center, University of California, Davis, CA 95616, United States
| | - Jeffrey Klein
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Larry Karnes
- Sinclair Research Center, 562 State Road DD, Auxvasse, MO 65231, United States
| | - Frans Sollie
- Pharmaceutical Research Associates Group B.V., Amerikaweg 18, 9407 TK Assen, Netherlands
| | - Todd C Zion
- Akston Biosciences Corporation., 100 Cummings Center, Suite 454C, Beverly, MA 01915, United States.
| |
Collapse
|
25
|
Dulal P, Gharaei R, Berg A, Walters AA, Hawkins N, Claridge TDW, Kowal K, Neill S, Ritchie AJ, Ashfield R, Hill AVS, Tronci G, Russell SJ, Douglas AD. Characterisation of factors contributing to the performance of nonwoven fibrous matrices as substrates for adenovirus vectored vaccine stabilisation. Sci Rep 2021; 11:20877. [PMID: 34686689 PMCID: PMC8536692 DOI: 10.1038/s41598-021-00065-4] [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: 05/24/2021] [Accepted: 09/23/2021] [Indexed: 11/09/2022] Open
Abstract
Adenovirus vectors offer a platform technology for vaccine development. The value of the platform has been proven during the COVID-19 pandemic. Although good stability at 2-8 °C is an advantage of the platform, non-cold-chain distribution would have substantial advantages, in particular in low-income countries. We have previously reported a novel, potentially less expensive thermostabilisation approach using a combination of simple sugars and glass micro-fibrous matrix, achieving excellent recovery of adenovirus-vectored vaccines after storage at temperatures as high as 45 °C. This matrix is, however, prone to fragmentation and so not suitable for clinical translation. Here, we report an investigation of alternative fibrous matrices which might be suitable for clinical use. A number of commercially-available matrices permitted good protein recovery, quality of sugar glass and moisture content of the dried product but did not achieve the thermostabilisation performance of the original glass fibre matrix. We therefore further investigated physical and chemical characteristics of the glass fibre matrix and its components, finding that the polyvinyl alcohol present in the glass fibre matrix assists vaccine stability. This finding enabled us to identify a potentially biocompatible matrix with encouraging performance. We discuss remaining challenges for transfer of the technology into clinical use, including reliability of process performance.
Collapse
Affiliation(s)
- Pawan Dulal
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK.
| | - Robabeh Gharaei
- grid.9909.90000 0004 1936 8403Clothworkers’ Centre for Textile Materials Innovation for Healthcare, University of Leeds, Leeds, LS2 9JT UK
| | - Adam Berg
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Adam A. Walters
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Nicholas Hawkins
- grid.4991.50000 0004 1936 8948Oxford Silk Group, ABRG, Department of Zoology, University of Oxford, Oxford, OX2 3RE UK
| | - Tim D. W. Claridge
- grid.4991.50000 0004 1936 8948Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA UK
| | - Katarzyna Kowal
- grid.436666.7Nonwovens Innovation and Research Institute Ltd, 169 Meanwood Road, Leeds, LS7 1SR UK
| | - Steven Neill
- grid.436666.7Nonwovens Innovation and Research Institute Ltd, 169 Meanwood Road, Leeds, LS7 1SR UK
| | - Adam J. Ritchie
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Rebecca Ashfield
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Adrian V. S. Hill
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Giuseppe Tronci
- grid.9909.90000 0004 1936 8403Clothworkers’ Centre for Textile Materials Innovation for Healthcare, University of Leeds, Leeds, LS2 9JT UK
| | - Stephen J. Russell
- grid.9909.90000 0004 1936 8403Clothworkers’ Centre for Textile Materials Innovation for Healthcare, University of Leeds, Leeds, LS2 9JT UK ,grid.436666.7Nonwovens Innovation and Research Institute Ltd, 169 Meanwood Road, Leeds, LS7 1SR UK
| | - Alexander D. Douglas
- grid.270683.80000 0004 0641 4511Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN UK
| |
Collapse
|
26
|
Szóstek-Mioduchowska A, Kordowitzki P. Shedding Light on the Possible Link between ADAMTS13 and Vaccine-Induced Thrombotic Thrombocytopenia. Cells 2021; 10:cells10102785. [PMID: 34685765 PMCID: PMC8535032 DOI: 10.3390/cells10102785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/26/2022] Open
Abstract
Several recent reports have highlighted the onset of vaccine-induced thrombotic thrombocytopaenia (VITT) in some recipients (approximately 1 case out of 100k exposures) of the ChAdOx1 nCoV-19 vaccine (AstraZeneca). Although the underlying events leading to this blood-clotting phenomenon has yet to be elucidated, several critical observations present a compelling potential mechanism. Thrombus formation requires the von Willebrand (VWF) protein to be in ultra-large multimeric state. The conservation of this state is controlled by the ADAMTS13 enzyme, whose proteolytic activity reduces the size of VWF multimers, keeping blood clotting at bay. However, ADAMTS13 cannot act on VWF that is bound to platelet factor 4 (PF4). As such, it is of particular interest to note that a common feature between subjects presenting with VITT is high titres of antibodies against PF4. This raises the possibility that these antibodies preserve the stability of ultra-large VWF complexes, leading to the formation of endothelium-anchored VWF strings, which are capable of recruiting circulating platelets and causing uncontrolled thrombosis in terminal capillaries. Here, we share our viewpoint about the current understanding of the VITT pathogenesis involving the prevention of ADAMTS13's activity on VWF by PF4 antibody-mediated stabilisation/ protection of the PF4-VWF complex.
Collapse
Affiliation(s)
- Anna Szóstek-Mioduchowska
- Department for Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-243 Olsztyn, Poland;
| | - Paweł Kordowitzki
- Department for Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-243 Olsztyn, Poland;
- Faculty of Biology and Veterinary Medicine, Nicolaus Copernicus University, Gagarina Street 1, 87-100 Torun, Poland
- Correspondence: ; Tel.: +48-89-539-31-28
| |
Collapse
|
27
|
Rojas JM, Sevilla N, Martín V. A New Look at Vaccine Strategies Against PPRV Focused on Adenoviral Candidates. Front Vet Sci 2021; 8:729879. [PMID: 34568477 PMCID: PMC8455998 DOI: 10.3389/fvets.2021.729879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is a virus that mainly infects goats and sheep causing significant economic loss in Africa and Asia, but also posing a serious threat to Europe, as recent outbreaks in Georgia (2016) and Bulgaria (2018) have been reported. In order to carry out the eradication of PPRV, an objective set for 2030 by the Office International des Epizooties (OIE) and the Food and Agriculture Organization of the United Nations (FAO), close collaboration between governments, pharmaceutical companies, farmers and researchers, among others, is needed. Today, more than ever, as seen in the response to the SARS-CoV2 pandemic that we are currently experiencing, these goals are feasible. We summarize in this review the current vaccination approaches against PPRV in the field, discussing their advantages and shortfalls, as well as the development and generation of new vaccination strategies, focusing on the potential use of adenovirus as vaccine platform against PPRV and more broadly against other ruminant pathogens.
Collapse
Affiliation(s)
| | | | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA-CSIC), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
28
|
Cao Q, Wu S, Xiao C, Chen S, Chi X, Cui X, Tang H, Su W, Zheng Y, Zhong J, Li Z, Li F, Chen H, Hou L, Wang H, Wen W. Integrated single-cell analysis revealed immune dynamics during Ad5-nCoV immunization. Cell Discov 2021; 7:64. [PMID: 34373443 PMCID: PMC8352953 DOI: 10.1038/s41421-021-00300-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), driven by SARS-CoV-2, is a severe infectious disease that has become a global health threat. Vaccines are among the most effective public health tools for combating COVID-19. Immune status is critical for evaluating the safety and response to the vaccine, however, the evolution of the immune response during immunization remains poorly understood. Single-cell RNA sequencing (scRNA-seq) represents a powerful tool for dissecting multicellular behavior and discovering therapeutic antibodies. Herein, by performing scRNA/V(D)J-seq on peripheral blood mononuclear cells from four COVID-19 vaccine trial participants longitudinally during immunization, we revealed enhanced cellular immunity with concerted and cell type-specific IFN responses as well as boosted humoral immunity with SARS-CoV-2-specific antibodies. Based on the CDR3 sequence and germline enrichment, we were able to identify several potential binding antibodies. We synthesized, expressed and tested 21 clones from the identified lineages. Among them, one monoclonal antibody (P3V6-1) exhibited relatively high affinity with the extracellular domain of Spike protein, which might be a promising therapeutic reagent for COVID-19. Overall, our findings provide insights for assessing vaccine through the novel scRNA/V(D)J-seq approach, which might facilitate the development of more potent, durable and safe prophylactic vaccines.
Collapse
Affiliation(s)
- Qiqi Cao
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuzhen Chen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China.,National Center for Liver Cancer, Second Military Medical University / Naval Medical University, Shanghai, China
| | | | - Xiuliang Cui
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China.,National Center for Liver Cancer, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Hao Tang
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Second Military Medical University / Naval Medical University, Shanghai, China.,Department of Critical Care, Wuhan Huoshenshan Hospital, Wuhan, Hubei, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingfeng Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiayong Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaomin Li
- HuaAn McAb Biotech Company, Hangzhou, Zhejiang, China
| | - Fang Li
- HuaAn McAb Biotech Company, Hangzhou, Zhejiang, China
| | - Haijia Chen
- Guangzhou SALIAI Stemcell Science and Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China.
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China. .,National Center for Liver Cancer, Second Military Medical University / Naval Medical University, Shanghai, China. .,Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wen Wen
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China. .,National Center for Liver Cancer, Second Military Medical University / Naval Medical University, Shanghai, China. .,Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University / Naval Medical University, Shanghai, China.
| |
Collapse
|
29
|
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
|
30
|
Cupovic J, Ring SS, Onder L, Colston JM, Lütge M, Cheng HW, De Martin A, Provine NM, Flatz L, Oxenius A, Scandella E, Krebs P, Engeler D, Klenerman P, Ludewig B. Adenovirus vector vaccination reprograms pulmonary fibroblastic niches to support protective inflating memory CD8 + T cells. Nat Immunol 2021; 22:1042-1051. [PMID: 34267375 PMCID: PMC7611414 DOI: 10.1038/s41590-021-00969-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.
Collapse
Affiliation(s)
- Jovana Cupovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Sandra S Ring
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Julia M Colston
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nicholas M Provine
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Elke Scandella
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Berne, Berne, Switzerland
| | - Daniel Engeler
- Department of Urology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Abstract
Hendra virus (HeV) and Nipah virus (NiV) are bat-borne zoonotic para-myxoviruses identified in the mid- to late 1990s in outbreaks of severe disease in livestock and people in Australia and Malaysia, respectively. HeV repeatedly re-emerges in Australia while NiV continues to cause outbreaks in South Asia (Bangladesh and India), and these viruses have remained transboundary threats. In people and several mammalian species, HeV and NiV infections present as a severe systemic and often fatal neurologic and/or respiratory disease. NiV stands out as a potential pandemic threat because of its associated high case-fatality rates and capacity for human-to-human transmission. The development of effective vaccines, suitable for people and livestock, against HeV and NiV has been a research focus. Here, we review the progress made in NiV and HeV vaccine development, with an emphasis on those approaches that have been tested in established animal challenge models of NiV and HeV infection and disease.
Collapse
Affiliation(s)
- Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA;
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA;
| |
Collapse
|
32
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
33
|
Daussy CF, Pied N, Wodrich H. Understanding Post Entry Sorting of Adenovirus Capsids; A Chance to Change Vaccine Vector Properties. Viruses 2021; 13:1221. [PMID: 34202573 PMCID: PMC8310329 DOI: 10.3390/v13071221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.
Collapse
Affiliation(s)
| | | | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR 5234, University of Bordeaux, 146 rue Leo Saignat, CEDEX, 33076 Bordeaux, France; (C.F.D.); (N.P.)
| |
Collapse
|
34
|
Spencer AJ, McKay PF, Belij-Rammerstorfer S, Ulaszewska M, Bissett CD, Hu K, Samnuan K, Blakney AK, Wright D, Sharpe HR, Gilbride C, Truby A, Allen ER, Gilbert SC, Shattock RJ, Lambe T. Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice. Nat Commun 2021; 12:2893. [PMID: 34001897 PMCID: PMC8129084 DOI: 10.1038/s41467-021-23173-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/19/2021] [Indexed: 01/08/2023] Open
Abstract
Several vaccines have demonstrated efficacy against SARS-CoV-2 mediated disease, yet there is limited data on the immune response induced by heterologous vaccination regimens using alternate vaccine modalities. Here, we present a detailed description of the immune response, in mice, following vaccination with a self-amplifying RNA (saRNA) vaccine and an adenoviral vectored vaccine (ChAdOx1 nCoV-19/AZD1222) against SARS-CoV-2. We demonstrate that antibody responses are higher in two-dose heterologous vaccination regimens than single-dose regimens. Neutralising titres after heterologous prime-boost were at least comparable or higher than the titres measured after homologous prime boost vaccination with viral vectors. Importantly, the cellular immune response after a heterologous regimen is dominated by cytotoxic T cells and Th1+ CD4 T cells, which is superior to the response induced in homologous vaccination regimens in mice. These results underpin the need for clinical trials to investigate the immunogenicity of heterologous regimens with alternate vaccine technologies.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- ChAdOx1 nCoV-19
- Immunization, Secondary
- Immunogenicity, Vaccine
- Mice
- RNA, Viral/administration & dosage
- RNA, Viral/genetics
- RNA, Viral/immunology
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Th1 Cells/immunology
- Vaccination/methods
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Alexandra J Spencer
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK.
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Cameron D Bissett
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Karnyart Samnuan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Anna K Blakney
- Department of Infectious Disease, Imperial College London, London, UK
| | - Daniel Wright
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Ciaran Gilbride
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Adam Truby
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
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
|
36
|
Adamaki M, Zoumpourlis V. Immunotherapy as a Precision Medicine Tool for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:E173. [PMID: 33419051 PMCID: PMC7825410 DOI: 10.3390/cancers13020173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed type of cancer among Caucasian males over the age of 60 and is characterized by remarkable heterogeneity and clinical behavior, ranging from decades of indolence to highly lethal disease. Despite the significant progress in PCa systemic therapy, therapeutic response is usually transient, and invasive disease is associated with high mortality rates. Immunotherapy has emerged as an efficacious and non-toxic treatment alternative that perfectly fits the rationale of precision medicine, as it aims to treat patients on the basis of patient-specific, immune-targeted molecular traits, so as to achieve the maximum clinical benefit. Antibodies acting as immune checkpoint inhibitors and vaccines entailing tumor-specific antigens seem to be the most promising immunotherapeutic strategies in offering a significant survival advantage. Even though patients with localized disease and favorable prognostic characteristics seem to be the ones that markedly benefit from such interventions, there is substantial evidence to suggest that the survival benefit may also be extended to patients with more advanced disease. The identification of biomarkers that can be immunologically targeted in patients with disease progression is potentially amenable in this process and in achieving significant advances in the decision for precision treatment of PCa.
Collapse
Affiliation(s)
- Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| | | |
Collapse
|
37
|
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
|
38
|
Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020. [DOI: 10.3390/vaccines8040783
expr 839529059 + 832255227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
|
39
|
Lukacs NW, Malinczak CA. Harnessing Cellular Immunity for Vaccination against Respiratory Viruses. Vaccines (Basel) 2020; 8:783. [PMID: 33371275 PMCID: PMC7766447 DOI: 10.3390/vaccines8040783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Severe respiratory viral infections, such as influenza, metapneumovirus (HMPV), respiratory syncytial virus (RSV), rhinovirus (RV), and coronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), cause significant mortality and morbidity worldwide. These viruses have been identified as important causative agents of acute respiratory disease in infants, the elderly, and immunocompromised individuals. Clinical signs of infection range from mild upper respiratory illness to more serious lower respiratory illness, including bronchiolitis and pneumonia. Additionally, these illnesses can have long-lasting impact on patient health well beyond resolution of the viral infection. Aside from influenza, there are currently no licensed vaccines against these viruses. However, several research groups have tested various vaccine candidates, including those that utilize attenuated virus, virus-like particles (VLPs), protein subunits, and nanoparticles, as well as recent RNA vaccines, with several of these approaches showing promise. Historically, vaccine candidates have advanced, dependent upon the ability to activate the humoral immune response, specifically leading to strong B cell responses and neutralizing antibody production. More recently, it has been recognized that the cellular immune response is also critical in proper resolution of viral infection and protection against detrimental immunopathology associated with severe disease and therefore, must also be considered when analyzing the efficacy and safety of vaccine candidates. These candidates would ideally result in robust CD4+ and CD8+ T cell responses as well as high-affinity neutralizing antibody. This review will aim to summarize established and new approaches that are being examined to harness the cellular immune response during respiratory viral vaccination.
Collapse
Affiliation(s)
- Nicholas W. Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
40
|
Sasso E, D'Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol 2020; 50:101430. [PMID: 33262065 DOI: 10.1016/j.smim.2020.101430] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of 'viral vectors') has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for 'personalised' cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Nouscom srl, Via di Castel Romano 100, 00128 Rome, Italy; Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy.
| | | | - Nicola Zambrano
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| | | | | | - Alfredo Nicosia
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
41
|
Ura T, Yamashita A, Mizuki N, Okuda K, Shimada M. New vaccine production platforms used in developing SARS-CoV-2 vaccine candidates. Vaccine 2020; 39:197-201. [PMID: 33279318 PMCID: PMC7685034 DOI: 10.1016/j.vaccine.2020.11.054] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
The threat of the current coronavirus disease pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is accelerating the development of potential vaccines. Candidate vaccines have been generated using existing technologies that have been applied for developing vaccines against other infectious diseases. Two new types of platforms, mRNA- and viral vector-based vaccines, have been gaining attention owing to the rapid advancement in their methodologies. In clinical trials, setting appropriate immunological endpoints plays a key role in evaluating the efficacy and safety of candidate vaccines. Updated information about immunological features from individuals who have or have not been exposed to SARS-CoV-2 continues to guide effective vaccine development strategies. This review highlights key strategies for generating candidate SARS-CoV-2 vaccines and considerations for vaccine development and clinical trials.
Collapse
Affiliation(s)
- Takehiro Ura
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Nobuhisa Mizuki
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Kenji Okuda
- Department of Molecular Biodefense Research, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Masaru Shimada
- Department of Molecular Biodefense Research, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
42
|
Jain S, Batra H, Yadav P, Chand S. COVID-19 Vaccines Currently under Preclinical and Clinical Studies, and Associated Antiviral Immune Response. Vaccines (Basel) 2020; 8:vaccines8040649. [PMID: 33153096 PMCID: PMC7711779 DOI: 10.3390/vaccines8040649] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/17/2020] [Accepted: 10/29/2020] [Indexed: 01/10/2023] Open
Abstract
With a death toll of over one million worldwide, the COVID-19 pandemic caused by SARS-CoV-2 has become the most devastating humanitarian catastrophe in recent decades. The fear of acquiring infection and spreading to vulnerable people has severely impacted society's socio-economic status. To put an end to this growing number of infections and deaths as well as to switch from restricted to everyday living, an effective vaccine is desperately needed. As a result, enormous efforts have been made globally to develop numerous vaccine candidates in a matter of months. Currently, over 30 vaccine candidates are under assessment in clinical trials, with several undergoing preclinical studies. Here, we reviewed the major vaccine candidates based on the specific vaccine platform utilized to develop them. We also discussed the immune responses generated by these candidates in humans and preclinical models to determine vaccine safety, immunogenicity, and efficacy. Finally, immune responses induced in recovered COVID-19 patients and their possible vaccine development implications were also briefly reviewed.
Collapse
Affiliation(s)
- Swati Jain
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (S.J.); (H.B.)
| | - Himanshu Batra
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (S.J.); (H.B.)
| | - Poonam Yadav
- CHI Health, Department of Pulmonary Medicine, Creighton University Medical Center, Omaha, NE 68131, USA;
| | - Subhash Chand
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-8017
| |
Collapse
|
43
|
COVID-19: Current Developments and Further Opportunities in Drug Delivery and Therapeutics. Pharmaceutics 2020; 12:pharmaceutics12100945. [PMID: 33023033 PMCID: PMC7601382 DOI: 10.3390/pharmaceutics12100945] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 has affected people from all age groups, races and ethnicities. Given that many infected individuals are asymptomatic, they transmit the disease to others unknowingly, which has resulted in the spread of infection at an alarming rate. This review aims to provide an overview of the pathophysiology, preventive measures to reduce the disease spread, therapies currently in use, an update on vaccine development and opportunities for vaccine delivery. The World Health Organization has advised several precautions including social distancing, hand washing and the use of PPE including gloves and face masks for minimizing the spread of SARS-CoV-2 infection. At present, several antiviral therapies previously approved for other infections are being repositioned to study their efficacy against SARS-CoV-2. In addition, some medicines (i.e., remdesivir, chloroquine, hydroxychloroquine) have received emergency use authorisation from the FDA. Plasma therapy has also been authorised for emergency use for the treatment of COVID-19 on a smaller scale. However, no vaccine has been approved so far against this virus. Nevertheless, several potential vaccine targets have been reported, and development of different types of vaccines including DNA, mRNA, viral vector, inactivated, subunit and vaccine-like particles is in process. It is concluded that a suitable candidate delivered through an advanced drug delivery approach would effectively boost the immune system against this coronavirus.
Collapse
|
44
|
Jeyanathan M, Afkhami S, Smaill F, Miller MS, Lichty BD, Xing Z. Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol 2020; 20:615-632. [PMID: 32887954 PMCID: PMC7472682 DOI: 10.1038/s41577-020-00434-6] [Citation(s) in RCA: 659] [Impact Index Per Article: 131.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the most formidable challenge to humanity in a century. It is widely believed that prepandemic normalcy will never return until a safe and effective vaccine strategy becomes available and a global vaccination programme is implemented successfully. Here, we discuss the immunological principles that need to be taken into consideration in the development of COVID-19 vaccine strategies. On the basis of these principles, we examine the current COVID-19 vaccine candidates, their strengths and potential shortfalls, and make inferences about their chances of success. Finally, we discuss the scientific and practical challenges that will be faced in the process of developing a successful vaccine and the ways in which COVID-19 vaccine strategies may evolve over the next few years.
Collapse
MESH Headings
- Antibodies, Viral/biosynthesis
- Betacoronavirus/drug effects
- Betacoronavirus/immunology
- Betacoronavirus/pathogenicity
- COVID-19
- COVID-19 Vaccines
- Clinical Trials as Topic
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Genetic Vectors/chemistry
- Genetic Vectors/immunology
- Humans
- Immunity, Herd/drug effects
- Immunity, Innate/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Pandemics/prevention & control
- Patient Safety
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- SARS-CoV-2
- Severe Acute Respiratory Syndrome/epidemiology
- Severe Acute Respiratory Syndrome/immunology
- Severe Acute Respiratory Syndrome/prevention & control
- Severe Acute Respiratory Syndrome/virology
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
- Vaccines, Virus-Like Particle
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Fiona Smaill
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Brian D Lichty
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
45
|
|
46
|
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in a global pandemic, prompting unprecedented efforts to contain the virus. Many developed countries have implemented widespread testing and have rapidly mobilized research programmes to develop vaccines and therapeutics. However, these approaches may be impractical in Africa, where the infrastructure for testing is poorly developed and owing to the limited manufacturing capacity to produce pharmaceuticals. Furthermore, a large burden of HIV-1 and tuberculosis in Africa could exacerbate the severity of infection and may affect vaccine immunogenicity. This Review discusses global efforts to develop diagnostics, therapeutics and vaccines, with these considerations in mind. We also highlight vaccine and diagnostic production platforms that are being developed in Africa and that could be translated into clinical development through appropriate partnerships for manufacture. The COVID-19 pandemic has prompted unparalleled progress in the development of vaccines and therapeutics in many countries, but it has also highlighted the vulnerability of resource-limited countries in Africa. Margolin and colleagues review global efforts to develop SARS-CoV-2 diagnostics, therapeutics and vaccines, with a focus on the opportunities and challenges in Africa.
Collapse
|
47
|
Alturki SO, Alturki SO, Connors J, Cusimano G, Kutzler MA, Izmirly AM, Haddad EK. The 2020 Pandemic: Current SARS-CoV-2 Vaccine Development. Front Immunol 2020; 11:1880. [PMID: 32973779 PMCID: PMC7466534 DOI: 10.3389/fimmu.2020.01880] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/13/2020] [Indexed: 01/12/2023] Open
Abstract
Coronaviruses are enveloped viruses with a positive-sense single-stranded RNA genome infecting animals and humans. Coronaviruses have been described more than 70 years ago and contain many species. Severe Acute Respiratory Syndrome (SARS) and Middle East Respiratory Syndrome (MERS) are lethal species caused by human coronaviruses (HCoVs). Currently, a novel strain of HCoVs, named Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (Covid-19). SARS-CoV-2 was first identified in December 2019 in Wuhan, the capital city of the Hubei province of China, and has since spread worldwide causing an outbreak in more than 200 countries. The SARS-CoV-2 outbreak was declared a pandemic on March 11th, 2020 and a public health emergency of international concern (PHEIC) in late January 2020 by the World Health Organization (WHO). SARS-CoV-2 infects the respiratory tract causing flu-like symptoms and, in some, may cause severe illness like pneumonia and multi-organ failure leading to death. Today, Covid-19 cases almost reaching 9 million, with more than 450 thousand deaths. There is an urgent demand for developing a vaccine since no effective therapies or vaccines have been approved to this day to prevent or minimize the spread of the infection. In this review, we summarized the furthest vaccines in the clinical pipeline.
Collapse
Affiliation(s)
- Sana O. Alturki
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sawsan O. Alturki
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jennifer Connors
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Gina Cusimano
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Michele A. Kutzler
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Abdullah M. Izmirly
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Elias K. Haddad
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
48
|
Sharpe HR, Gilbride C, Allen E, Belij-Rammerstorfer S, Bissett C, Ewer K, Lambe T. The early landscape of coronavirus disease 2019 vaccine development in the UK and rest of the world. Immunology 2020; 160:223-232. [PMID: 32460358 PMCID: PMC7283842 DOI: 10.1111/imm.13222] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Since the first World Health Organization notification on 31 December 2019, coronavirus disease 2019 (COVID‐19), the respiratory disease caused by the coronavirus severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2), has been responsible for over four million confirmed infections and almost 300 000 deaths worldwide. The pandemic has led to over half of the world's population living under lockdown conditions. To allow normal life to resume, public health interventions will be needed to prevent further waves of infections as lockdown measures are lifted. As one of the most effective countermeasures against infectious diseases, an efficacious vaccine is considered crucial to containing the COVID‐19 pandemic. Following the publication of the genome sequence of SARS‐CoV‐2, vaccine development has accelerated at an unprecedented pace across the world. Here we review the different platforms employed to develop vaccines, the standard timelines of development and how they can be condensed in a pandemic situation. We focus on vaccine development in the UK and vaccines that have entered clinical trials around the world.
Collapse
Affiliation(s)
- Hannah R Sharpe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ciaran Gilbride
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Cameron Bissett
- 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
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
49
|
Sebastian S, Flaxman A, Cha KM, Ulaszewska M, Gilbride C, Sharpe H, Wright E, Spencer AJ, Dowall S, Hewson R, Gilbert S, Lambe T. A Multi-Filovirus Vaccine Candidate: Co-Expression of Ebola, Sudan, and Marburg Antigens in a Single Vector. Vaccines (Basel) 2020; 8:E241. [PMID: 32455764 PMCID: PMC7349952 DOI: 10.3390/vaccines8020241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leaving individuals susceptible to disease after exposure to related virus species. This is a significant hurdle in the field of vaccine development, in which broadly protective vaccines represent an unmet need. This is particularly evident for filoviruses, as there are multiple family members that can cause lethal haemorrhagic fever, including Zaire ebolavirus, Sudan ebolavirus, and Marburg virus. In an attempt to address this need, both pre-clinical and clinical studies previously used mixed or co-administered monovalent vaccines to prevent filovirus mediated disease. However, these multi-vaccine and multi-dose vaccination regimens do not represent a practical immunisation scheme when considering the target endemic areas. We describe here the development of a single multi-pathogen filovirus vaccine candidate based on a replication-deficient simian adenoviral vector. Our vaccine candidate encodes three different filovirus glycoproteins in one vector and induces strong cellular and humoral immunity to all three viral glycoproteins after a single vaccination. Crucially, it was found to be protective in a stringent Zaire ebolavirus challenge in guinea pigs in a one-shot vaccination regimen. This trivalent filovirus vaccine offers a tenable vaccine product that could be rapidly translated to the clinic to prevent filovirus-mediated viral haemorrhagic fever.
Collapse
Affiliation(s)
- Sarah Sebastian
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
- Current address: Vaccitech Ltd., Oxford Science Park, Oxford OX4 4GE, UK
| | - Amy Flaxman
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Kuan M. Cha
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Marta Ulaszewska
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Ciaran Gilbride
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Hannah Sharpe
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Edward Wright
- School of Life Sciences, University of Sussex, Falmer BN1 9QG, UK;
| | - Alexandra J. Spencer
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Stuart Dowall
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK; (S.D.); (R.H.)
| | - Roger Hewson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK; (S.D.); (R.H.)
| | - Sarah Gilbert
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Teresa Lambe
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| |
Collapse
|
50
|
Ricobaraza A, Gonzalez-Aparicio M, Mora-Jimenez L, Lumbreras S, Hernandez-Alcoceba R. High-Capacity Adenoviral Vectors: Expanding the Scope of Gene Therapy. Int J Mol Sci 2020; 21:E3643. [PMID: 32455640 PMCID: PMC7279171 DOI: 10.3390/ijms21103643] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
The adaptation of adenoviruses as gene delivery tools has resulted in the development of high-capacity adenoviral vectors (HC-AdVs), also known, helper-dependent or "gutless". Compared with earlier generations (E1/E3-deleted vectors), HC-AdVs retain relevant features such as genetic stability, remarkable efficacy of in vivo transduction, and production at high titers. More importantly, the lack of viral coding sequences in the genomes of HC-AdVs extends the cloning capacity up to 37 Kb, and allows long-term episomal persistence of transgenes in non-dividing cells. These properties open a wide repertoire of therapeutic opportunities in the fields of gene supplementation and gene correction, which have been explored at the preclinical level over the past two decades. During this time, production methods have been optimized to obtain the yield, purity, and reliability required for clinical implementation. Better understanding of inflammatory responses and the implementation of methods to control them have increased the safety of these vectors. We will review the most significant achievements that are turning an interesting research tool into a sound vector platform, which could contribute to overcome current limitations in the gene therapy field.
Collapse
Affiliation(s)
| | | | | | | | - Ruben Hernandez-Alcoceba
- Gene Therapy Program. University of Navarra-CIMA. Navarra Institute of Health Research, 31008 Pamplona, Spain; (A.R.); (M.G.-A.); (L.M.-J.); (S.L.)
| |
Collapse
|