1
|
Bugden S, Mital S, Nguyen HV. Cost-effectiveness of nirsevimab and maternal RSVpreF for preventing respiratory syncytial virus disease in infants across Canada. BMC Med 2025; 23:102. [PMID: 39984979 PMCID: PMC11846277 DOI: 10.1186/s12916-025-03928-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Nirsevimab, a long-acting monoclonal antibody, and RSVpreF, a maternal vaccine, are newly approved respiratory syncytial virus (RSV) prophylactics for infants in Canada. Both have the potential to expand prevention efforts, but there is limited evidence regarding their cost-effectiveness and how it varies across the country, despite disparate hospitalisation rates and resource use among different populations. METHODS We developed a decision tree model to follow twelve monthly birth cohorts through their first year of life, incorporating risk differentiation based on Canadian region, prematurity, and comorbidities. The model tracked medically attended infections, including hospitalisations, intensive care unit admissions, and outpatient visits, comparing costs (in 2024 Canadian dollars) and effectiveness (in quality-adjusted life years (QALYs)) of nine different immunisation strategies compared to no intervention. The analysis was conducted from both healthcare and societal perspectives. We conducted threshold price analyses, varying the price-per-dose of each product to determine the threshold prices at which expanded coverage becomes cost-effective. RESULTS At base case prices, the optimal strategy varies by region, but in all cases, the optimal strategy is both cost-saving and more effective than no intervention. In southern Canada, it is optimal to immunise only palivizumab-eligible infants (those born very prematurely or with high-risk comorbidities) with nirsevimab, resulting in cost savings of $4.14 and QALY gains of 0.000022 QALY per infant compared to no intervention. In the Northwest Territories, it is best to expand protection with nirsevimab to include all preterm infants (cost savings of $28.68 and QALY gains of 0.00007 per infant). In Nunavik and Nunavut, immunising all infants under 6 months and all infants under twelve months with nirsevimab are the best strategies, respectively (cost savings of $399.61 and QALY gains of 0.000821 per infant in Nunavik, and cost savings of $1067.03 and QALY gains of 0.000884 per infant in Nunavut). Universal, country-wide immunisation with nirsevimab would require a price-per-dose of under $112 to become the most cost-effective prevention strategy. CONCLUSIONS The optimal strategy for preventing respiratory syncytial virus disease in Canadian infants depends on product price and regional risk level and resource use. Canadian policy should account for these factors.
Collapse
Affiliation(s)
- Samara Bugden
- School of Pharmacy, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada.
| | - Shweta Mital
- College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Avenue West, Winnipeg, MB, R3E 0T5, Canada
| | - Hai V Nguyen
- School of Pharmacy, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| |
Collapse
|
2
|
Mao X, Hua X, Wu C, Ge X, Zhang J, Wu X, Kubiak RJ, Wählby Hamrén U, Villafana T, Christou G, Green J, Takas T, Jin Y. A phase I, randomized, placebo-controlled trial to evaluate the pharmacokinetics, safety, and tolerability of nirsevimab in healthy Chinese adults. Clin Transl Sci 2025; 18:e70095. [PMID: 39736101 DOI: 10.1111/cts.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/18/2024] [Accepted: 11/07/2024] [Indexed: 01/01/2025] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection (LRTI) in infants worldwide. Nirsevimab, an extended half-life monoclonal antibody against RSV, is approved in China for the prevention of RSV lower respiratory tract disease in infants; however, global nirsevimab trials did not enroll Chinese infants. To inform the investigation of nirsevimab for the prevention of RSV LRTI in Chinese infants, this Phase I, randomized, placebo-controlled trial evaluated the pharmacokinetics (PK) and safety of nirsevimab in healthy Chinese adults. Participants were randomized 3:1 to a single 300 mg intramuscular dose of nirsevimab or placebo and were followed through 150 days post-dose. Serum nirsevimab concentrations were measured and PK parameters of maximum serum concentration (Cmax), time to maximum concentration (tmax), and area under the concentration-time curve from time 0 to Day 150 (AUC0-150) were estimated. Treatment emergent adverse events (AEs), clinical laboratory data, and vital signs were evaluated. Overall, 24 participants were randomized to nirsevimab (n = 18) or placebo (n = 6). Nirsevimab geometric mean (coefficient of variation [%CV]) Cmax was 46.9 (21.7) μg/mL, median (range) tmax was 7.0 (4.9, 29.9) days, and geometric mean (%CV) AUC0-150 was 4210.6 (13.6) μg·day/mL. Treatment-emergent AEs (all Grade 1 or Grade 2 in severity) were reported in 5/18 (27.8%) nirsevimab recipients and 2/6 (33.3%) placebo recipients. No serious AEs, new onset chronic disease, or deaths were reported. Overall, safety and PK outcomes were consistent with those observed in healthy adults in the USA, with no new safety concerns.
Collapse
Affiliation(s)
- Xiaomeng Mao
- Clinical Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohan Hua
- Clinical Development, Respiratory & Immunology, R&D China, AstraZeneca, Shanghai, China
| | - Chengyi Wu
- Biometrics, R&D China, AstraZeneca, Shanghai, China
| | - Xiaoyun Ge
- Clinical Safety, R&D China, AstraZeneca, Shanghai, China
| | - Jie Zhang
- Development Operations, R&D China, AstraZeneca, Shanghai, China
| | - Xiaojie Wu
- Clinical Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Robert J Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Georgios Christou
- Patient Safety, Chief Medical Office, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Jannine Green
- Global Clinical Operations, R&D, AstraZeneca, Macclesfield, UK
| | - Therese Takas
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Yuwen Jin
- Clinical Pharmacology, R&D China, AstraZeneca, Shanghai, China
| |
Collapse
|
3
|
Duan Y, Liu Z, Zang N, Cong B, Shi Y, Xu L, Jiang M, Wang P, Zou J, Zhang H, Feng Z, Feng L, Ren L, Liu E, Li Y, Zhang Y, Xie Z. Landscape of respiratory syncytial virus. Chin Med J (Engl) 2024; 137:2953-2978. [PMID: 39501814 PMCID: PMC11706595 DOI: 10.1097/cm9.0000000000003354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Indexed: 01/11/2025] Open
Abstract
ABSTRACT Respiratory syncytial virus (RSV) is an enveloped, negative-sense, single-stranded RNA virus of the Orthopneumovirus genus of the Pneumoviridae family in the order Mononegavirales. RSV can cause acute upper and lower respiratory tract infections, sometimes with extrapulmonary complications. The disease burden of RSV infection is enormous, mainly affecting infants and older adults aged 75 years or above. Currently, treatment options for RSV are largely supportive. Prevention strategies remain a critical focus, with efforts centered on vaccine development and the use of prophylactic monoclonal antibodies. To date, three RSV vaccines have been approved for active immunization among individuals aged 60 years and above. For children who are not eligible for these vaccines, passive immunization is recommended. A newly approved prophylactic monoclonal antibody, Nirsevimab, which offers enhanced neutralizing activity and an extended half-life, provides exceptional protection for high-risk infants and young children. This review provides a comprehensive and detailed exploration of RSV's virology, immunology, pathogenesis, epidemiology, clinical manifestations, treatment options, and prevention strategies.
Collapse
Affiliation(s)
- Yuping Duan
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Zimeng Liu
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
| | - Na Zang
- Department of Respiratory Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Key Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing 400014, China
| | - Bingbing Cong
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuqing Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Medical Virology and Viral Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Lili Xu
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| | - Mingyue Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Peixin Wang
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
| | - Jing Zou
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Han Zhang
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ziheng Feng
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lili Ren
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Enmei Liu
- Department of Respiratory Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Key Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing 400014, China
| | - You Li
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
- Changzhou Third People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yan Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Medical Virology and Viral Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Zhengde Xie
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| |
Collapse
|
4
|
Bonavia A, Levi M, Rouha H, Badarau A, Terstappen J, Watson S, Anderson AB, White JT, Ananworanich J, Taylor D, Radivojevic A, Shaffer M, Stamm LM, Dunne MW. RSM01, a novel respiratory syncytial virus monoclonal antibody: preclinical characterization and results of a first-in-human, randomised clinical trial. BMC Infect Dis 2024; 24:1378. [PMID: 39627701 PMCID: PMC11616243 DOI: 10.1186/s12879-024-10120-w] [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: 06/14/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract disease among infants and young children worldwide, especially in low- and middle-income countries (LMICs). RSM01 is a novel, highly potent, half-life-extended anti-RSV monoclonal antibody (mAb) candidate primarily being developed for LMICs. Here we present the preclinical characterisation and results of a phase 1 trial of RSM01. METHODS Preclinical characterisation of RSM01 was conducted using in-vitro neutralization assays and cotton rat models. In the first-in-human, double-blind, phase 1 trial, 56 healthy adults were randomised 6:1 within dose cohorts to receive a single dose of RSM01 (n = 48) or placebo (n = 8): 300 mg intravenously (IV), 300 mg intramuscularly (IM) or 1000 mg IV (parallel cohorts), 3000 mg IV, and an expansion cohort of 600 mg IM. Systemic solicited adverse events (AEs) were assessed through day 7; unsolicited AEs were collected through day 151. Pharmacokinetics and anti-drug antibodies (ADA) to RSM01 were assessed using immunoassays. A population pharmacokinetics model predicted paediatric pharmacokinetics parameters using allometric scaling and age-specific population weight statistics of North American and African infants. RESULTS RSM01 exhibited highly potent neutralizing activity in the single ng/mL range (0.7-6.4) against diverse RSV-A and RSV-B isolates in vitro. RSM01 also demonstrated prophylactic efficacy in cotton rat models with both RSV subtypes. In the phase 1 clinical trial, the most common unsolicited AEs were COVID-19 (2/48), headache (2/48), and nausea (2/48), all in RSM01-treated participants. The only systemic solicited AEs reported were headache (5/48) and tiredness (2/48) in participants receiving RSM01. No serious AEs or deaths were reported. The half-life of RSM01 was 78 days with dose-proportional increases in Tmax and AUClast after IV administration. Among RSM01-treated participants, 2/48 were ADA positive at baseline, and 1/48 seroconverted to ADA-positive post-baseline. CONCLUSIONS RSM01 is a highly potent, half-life-extended, RSV-neutralising mAb candidate that was shown to be well tolerated in healthy adults. The rate of ADA to RSM01 was low. The long half-life of RSM01 and pharmacokinetics profile support further development of RSM01 as a potential single dose per season prophylaxis to prevent RSV disease in infants. TRIAL REGISTRATION Clinicaltrials.gov NCT05118386, Nov 12, 2021.
Collapse
Affiliation(s)
- Aurelio Bonavia
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Micha Levi
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA.
| | | | | | - Jonne Terstappen
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Shayne Watson
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | - Joleen T White
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | | | | | - Michael Shaffer
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Luisa M Stamm
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Michael W Dunne
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| |
Collapse
|
5
|
Lee YZ, Han J, Zhang YN, Ward G, Braz Gomes K, Auclair S, Stanfield RL, He L, Wilson IA, Zhu J. Rational design of uncleaved prefusion-closed trimer vaccines for human respiratory syncytial virus and metapneumovirus. Nat Commun 2024; 15:9939. [PMID: 39550381 PMCID: PMC11569192 DOI: 10.1038/s41467-024-54287-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause human respiratory diseases and are major targets for vaccine development. In this study, we design uncleaved prefusion-closed (UFC) trimers for the fusion protein (F) of both viruses by examining mutations critical to F metastability. For RSV, we assess four previous prefusion F designs, including the first and second generations of DS-Cav1, SC-TM, and 847A. We then identify key mutations that can maintain prefusion F in a native-like, closed trimeric form (up to 76%) without introducing any interprotomer disulfide bond. For hMPV, we develop a stable UFC trimer with a truncated F2-F1 linkage and an interprotomer disulfide bond. Dozens of UFC constructs are characterized by negative-stain electron microscopy (nsEM), x-ray crystallography (11 RSV-F structures and one hMPV-F structure), and antigenic profiling. Using an optimized RSV-F UFC trimer as bait, we identify three potent RSV neutralizing antibodies (NAbs) from a phage-displayed human antibody library, with a public NAb lineage targeting sites Ø and V and two cross-pneumovirus NAbs recognizing site III. In mouse immunization, rationally designed RSV-F and hMPV-F UFC trimers induce robust antibody responses with high neutralizing titers. Our study provides a foundation for future prefusion F-based RSV and hMPV vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jerome Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Uvax Bio, LLC, Newark, DE, 19702, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
6
|
Siu W. Prévention par immunisation passive de l’infection au virus respiratoire syncytial chez les nourrissons. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2024; 70:e187-e191. [PMID: 39638406 PMCID: PMC11634255 DOI: 10.46747/cfp.701112e187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Objectif Assister les médecins de famille dans leurs discussions avec les patients concernant les immunisations contre le virus respiratoire syncytial (VRS). Sources de l’information Les renseignements sont tirés : d’une recherche documentaire sur le fardeau de la maladie causée par le VRS chez les nourrissons; d’études observationnelles; d’essais randomisés contrôlés; des données probantes présentées par des agences d’examen; des monographies des produits; et des déclarations du Comité consultatif national de l’immunisation. Message principal Trois options sont maintenant disponibles pour prévenir la maladie sévère causée par le VRS chez les nourrissons : l’anticorps monoclonal palivizumab, l’anticorps monoclonal à action prolongée nirsevimab et le nouveau vaccin VRSpreF administré durant la grossesse. Seulement un petit nombre de nourrissons à risque élevé d’une infection grave au VRS sont admissibles à recevoir le palivizumab. Le nirsevimab a reçu l’autorisation de Santé Canada pour tous les nourrissons, et le VRSpreF est homologué pour toutes les femmes enceintes et les personnes enceintes. Il y a de nombreux facteurs à prendre en considération dans l’utilisation de chacun des produits, notamment : l’autorisation; la disponibilité; le moment de l’administration; l’état de santé et les déterminants sociaux de la santé du nourrisson; l’efficacité et l’efficience; l’innocuité; la préférence du patient; et le coût. Le Comité consultatif national de l’immunisation a récemment publié des conseils sur l’utilisation des produits immunisants contre le VRS chez les nourrissons. Conclusion Les médecins de famille devraient être au fait des indications, des bienfaits relatifs et des recommandations relatifs aux 3 produits d’immunisation dans le but d’avoir des discussions éclairées avec les patients, en tenant compte des préférences et des circonstances de la femme enceinte, de la personne enceinte ou du parent et du nourrisson.
Collapse
Affiliation(s)
- Winnie Siu
- Conseillère médicale principale au Centre de surveillance et des programmes d’immunisation à l’Agence de la santé publique du Canada à Ottawa (Ontario) et professeure adjointe à l’École d’épidémiologie et de santé publique à l’Université d’Ottawa
| |
Collapse
|
7
|
Siu W. Approach to prevention of respiratory syncytial virus disease in infants by passive immunization. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2024; 70:697-700. [PMID: 39638376 PMCID: PMC11634253 DOI: 10.46747/cfp.701112697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
OBJECTIVE To support family physicians in discussing respiratory syncytial virus (RSV) immunizations with patients. SOURCES OF INFORMATION Information was obtained through a literature review on the burden of RSV disease in infants; observational studies; randomized controlled trials; evidence presented by review agencies; product monographs; and National Advisory Committee on Immunization statements. MAIN MESSAGE There are now 3 options available for preventing severe RSV disease in infants: the monoclonal antibody palivizumab, the long-acting monoclonal antibody nirsevimab, and the new RSVpreF vaccine administered during pregnancy. Only a small number of infants at high risk of severe RSV disease are eligible for palivizumab. Nirsevimab has received Health Canada authorization for all infants and RSVpreF has received authorization for all pregnant women and pregnant people. There are multiple considerations for the use of each product, including authorization; availability; timing of administration; health status and social determinants of health of the infant; efficacy and effectiveness; safety; patient preference; and cost. The National Advisory Committee on Immunization has recently issued guidance on the use of RSV immunization products for infants. CONCLUSION Family doctors should be aware of the indications, relative benefits, and recommendations for the 3 RSV immunization products to have informed discussions with patients, taking into account the preferences and circumstances of the pregnant woman or pregnant person or of the parent and infant.
Collapse
Affiliation(s)
- Winnie Siu
- Senior Medical Advisor in the Centre for Immunization Surveillance and Programs at the Public Health Agency of Canada in Ottawa, Ont, and Adjunct Professor in the School of Epidemiology and Public Health at the University of Ottawa
| |
Collapse
|
8
|
Domachowske J, Hamrén UW, Banu I, Baronio R, Basavaraju B, Koen A, Leach A, Mankad VS, Pannaraj PS, Soler-Palacin P, Takas T, Mori M, Villafana T. Safety and Pharmacokinetics of Nirsevimab in Immunocompromised Children. Pediatrics 2024; 154:e2024066508. [PMID: 39257380 DOI: 10.1542/peds.2024-066508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Immunocompromised children may have increased risk for severe respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI), potentially leading to prolonged hospitalization, intensive care, and death. The open-label phase II MUSIC trial evaluated the safety and pharmacokinetics of nirsevimab, an extended half-life monoclonal antibody against RSV, in immunocompromised children aged ≤24 months. METHODS Participants received a single intramuscular injection of nirsevimab (first RSV season: 50 mg if <5 kg/100 mg if ≥5 kg; second season: 200 mg). Safety, antidrug antibodies, and pharmacokinetics were evaluated to day 361. RESULTS Participants (n = 100) had ≥1 immunocompromising conditions: primary immunodeficiency (n = 33), previous transplantation (n = 16), HIV infection (n = 8) or treatment with high-dose systemic corticosteroids (n = 29), immunosuppressive chemotherapy (n = 20), or other immunosuppressive therapies (n = 15). Six children experienced eight treatment-related adverse events (none categorized as serious). Three deaths occurred, all were unrelated to treatment. Eleven children, developed antidrug antibodies, with minimal effects on pharmacokinetics and no apparent impact on safety. Nirsevimab serum concentrations at day 151 were similar to those effective in preventing medically attended RSV LRTI in healthy infants. Fourteen children had increased nirsevimab clearance. No protocol-defined medically attended RSV LRTIs occured through day 151. CONCLUSIONS Among immunocompromised children aged ≤24 months, nirsevimab was well tolerated with no safety concerns and serum concentrations were supportive of efficacy. A subset of children with increased nirsevimab clearance, had conditions potentially associated with protein loss; however, the impact on efficacy is unknown.
Collapse
Affiliation(s)
- Joseph Domachowske
- State University of New York Upstate Medical University Syracuse, New York
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Irfana Banu
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Roberta Baronio
- Biometrics, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Bhanu Basavaraju
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Anthonet Koen
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (Wits-VIDA), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amanda Leach
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Vaishali S Mankad
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Durham, North Carolina
| | - Pia S Pannaraj
- University of California San Diego, San Diego, California
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, and Vall d'Hebron Research Institute, Barcelona, Catalonia, Spain
| | - Therese Takas
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Masaaki Mori
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tonya Villafana
- Clinical Development, Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
9
|
Banoun H. Analysis of Beyfortus ® (Nirsevimab) Immunization Campaign: Effectiveness, Biases, and ADE Risks in RSV Prevention. Curr Issues Mol Biol 2024; 46:10369-10395. [PMID: 39329969 PMCID: PMC11431526 DOI: 10.3390/cimb46090617] [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: 07/27/2024] [Revised: 08/27/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Respiratory infections with respiratory syncytial virus (RSV) account for an important part of hospital admissions for acute respiratory infections. Nirsevimab has been developed to reduce the hospital burden of RSV infections. Compared with the product previously used, it has a stronger binding capacity to RSV F protein and a high affinity for FcRn (neonatal receptor for the Fc fragment of IgG), which extends its lifespan. Nirsevimab has been shown to be highly effective in reducing hospitalization rates of RSV infections but a large or unknown number of treated subjects have been excluded in clinical and post-marketing studies. However, analysis of these studies cannot exclude that, in rare cases, nirsevimab facilitates and worsens RSV infection (or other respiratory infections). This could be attributable to antibody-dependent enhancement (ADE) which has been observed with RSV F protein antibodies in inactivated vaccine trials. This risk has been incompletely assessed in pre-clinical and clinical trials (incomplete exploration of nirsevimab effector functions and pharmacokinetics). ADE by disruption of the immune system (not studied and due to FcRn binding) could explain why there is no reduction in all-cause hospital admissions in treated age groups. Given the high price of nirsevimab, the cost-effectiveness of mass immunization campaigns may therefore be debated from an economic as well as a scientific point of view.
Collapse
|
10
|
Tulika T, Ruso-Julve F, Ahmadi S, Ljungars A, Rivera-de-Torre E, Wade J, Fernández-Quintero ML, Jenkins TP, Belfakir SB, Ross GMS, Boyens-Thiele L, Buell AK, Sakya SA, Sørensen CV, Bohn MF, Ledsgaard L, Voldborg BG, Francavilla C, Schlothauer T, Lomonte B, Andersen JT, Laustsen AH. Engineering of pH-dependent antigen binding properties for toxin-targeting IgG1 antibodies using light-chain shuffling. Structure 2024; 32:1404-1418.e7. [PMID: 39146931 PMCID: PMC11385703 DOI: 10.1016/j.str.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/07/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Immunoglobulin G (IgG) antibodies that bind their cognate antigen in a pH-dependent manner (acid-switched antibodies) can release their bound antigen for degradation in the acidic environment of endosomes, while the IgGs are rescued by the neonatal Fc receptor (FcRn). Thus, such IgGs can neutralize multiple antigens over time and therefore be used at lower doses than their non-pH-responsive counterparts. Here, we show that light-chain shuffling combined with phage display technology can be used to discover IgG1 antibodies with increased pH-dependent antigen binding properties, using the snake venom toxins, myotoxin II and α-cobratoxin, as examples. We reveal differences in how the selected IgG1s engage their antigens and human FcRn and show how these differences translate into distinct cellular handling properties related to their pH-dependent antigen binding phenotypes and Fc-engineering for improved FcRn binding. Our study showcases the complexity of engineering pH-dependent antigen binding IgG1s and demonstrates the effects on cellular antibody-antigen recycling.
Collapse
Affiliation(s)
- Tulika Tulika
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Fulgencio Ruso-Julve
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Jack Wade
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | | | - Timothy P Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Selma B Belfakir
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; VenomAid Diagnostics ApS, Lyngby, Denmark
| | | | - Lars Boyens-Thiele
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Alexander K Buell
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Siri A Sakya
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | - Christoffer V Sørensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Markus-Frederik Bohn
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Line Ledsgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Bjørn G Voldborg
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Chiara Francavilla
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiologia, Universidad de Costa Rica, San Jose, Costa Rica
| | - Jan Terje Andersen
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway.
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
11
|
Sevendal ATK, Hurley S, Bartlett AW, Rawlinson W, Walker GJ. Systematic Review of the Efficacy and Safety of RSV-Specific Monoclonal Antibodies and Antivirals in Development. Rev Med Virol 2024; 34:e2576. [PMID: 39209729 DOI: 10.1002/rmv.2576] [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: 03/26/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of acute respiratory infection amongst all ages, causing a significant global health burden. Preventative and therapeutic options for RSV infection have long been under development, and recently, several widely-publicised vaccines targeting older adult and maternal populations have become available. Promising monoclonal antibody (mAb) and antiviral (AV) therapies are also progressing in clinical trials, with the prophylactic mAb nirsevimab recently approved for clinical use in infant populations. A systematic review on current progress in this area is lacking. We performed a systematic literature search (PubMed, Embase, Web of Science, ClinicalTrials.gov, EudraCT, ANZCTR-searched Nov 29th, 2023) to identify studies on all RSV-specific mAbs and AV therapies that has undergone human clinical trials since year 2000. Data extraction focused on outcomes related to the therapeutic efficacy and safety of the intervention on trial, and all studies were graded against the OCEBM Levels of Evidence Table. Results from 59 studies were extracted, covering efficacy and safety data on six mAbs (motavizumab, motavizumab-YTE, nirsevimab, ALX-0171, suptavumab, clesrovimab) and 12 AV therapies (ALN-RSV01, RSV604, presatovir, MDT-637, lumicitabine, IFN-α1b, rilematovir, enzaplatovir, AK0529, sisunatovir, PC786, EDP-938). Of the mAbs reviewed, nirsevimab and clesrovimab hold considerable promise. The timeline for RSV-specific AV availability is less advanced, although EDP-938 and AK0529 have reported promising phase 2 efficacy and safety data. Moving forward, passive immunisation and treatment options for RSV infection will play a significant role in reducing the health burden of RSV, complementing recent advancements in vaccine development. TRIAL REGISTRATION: PROSPERO registration: CRD42022376633.
Collapse
Affiliation(s)
- Andrea T K Sevendal
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Siobhan Hurley
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
| | - Adam W Bartlett
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital Network, Sydney, Australia
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - William Rawlinson
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Gregory J Walker
- Virology Research Laboratory, Serology and Virology Division (SAViD), NSW Health Pathology, Prince of Wales Hospital, Sydney, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
12
|
Mankad VS, Leach A, Chang Y, Wählby Hamrén U, Kiazand A, Kubiak RJ, Takas T, Villafana T, Shroff M. Comprehensive Summary of Safety Data on Nirsevimab in Infants and Children from All Pivotal Randomized Clinical Trials. Pathogens 2024; 13:503. [PMID: 38921800 PMCID: PMC11206492 DOI: 10.3390/pathogens13060503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Nirsevimab is approved in the US for the prevention of respiratory syncytial virus (RSV) lower respiratory tract disease in neonates and infants during their first RSV season and in children aged ≤24 months who remain vulnerable to severe RSV disease through their second RSV season. We summarize a pre-specified analysis of nirsevimab safety data from three randomized controlled trials: Phase 2b (NCT02878330; healthy infants born ≥29 to <35 weeks' gestational age [wGA]); Phase 3 MELODY (NCT03979313; healthy infants born ≥35 wGA); and Phase 2/3 MEDLEY (NCT03959488; infants with congenital heart disease [CHD] and/or chronic lung disease of prematurity [CLD] or born ≤35 wGA). METHODS Participants (randomized 2:1) received a single intramuscular dose of nirsevimab or comparator (placebo, Phase 2b/MELODY; 5× once-monthly palivizumab, MEDLEY) before their first RSV season (recipients < 5 kg, nirsevimab 50 mg; ≥5 kg, nirsevimab 100 mg). In MEDLEY, children with CHD/CLD continued to a second RSV season: first-season nirsevimab recipients received nirsevimab 200 mg; first-season palivizumab recipients were re-randomized 1:1 to receive nirsevimab 200 mg or 5× once-monthly palivizumab. RESULTS The incidence, severity, and nature of AEs were similar across treatments (nirsevimab, n = 3184; placebo, n = 1284; palivizumab, n = 304). Most AEs were mild to moderate in severity, with ≥98% unrelated to treatment. AEs of special interest occurred infrequently (<1%): no anaphylaxis or thrombocytopenia were treatment-related, and no immune complex disease was reported. Deaths (incidence < 1.0%) were all unrelated to treatment. CONCLUSIONS A single dose per season of nirsevimab for the prevention of RSV disease had a favorable safety profile, irrespective of wGA or comorbidities.
Collapse
Affiliation(s)
- Vaishali S. Mankad
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Durham, NC 27703, USA;
| | - Amanda Leach
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (A.L.); (Y.C.); (T.T.); (T.V.)
| | - Yue Chang
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (A.L.); (Y.C.); (T.T.); (T.V.)
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, SE-43183 Gothenburg, Sweden;
| | - Alexandre Kiazand
- Patient Safety, Chief Medical Office, Oncology R&D, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Robert J. Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Therese Takas
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (A.L.); (Y.C.); (T.T.); (T.V.)
| | - Tonya Villafana
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (A.L.); (Y.C.); (T.T.); (T.V.)
| | - Manish Shroff
- Patient Safety, Chief Medical Office, Oncology R&D, AstraZeneca, Waltham, MA 02451, USA
| |
Collapse
|
13
|
Eto T, Okubo Y, Momose A, Tamura H, Zheng R, Callendret B, Bastian A, Comeaux C. A Randomized, Double-Blind, Placebo-Controlled, Phase 1 Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of Single Vaccination of Ad26.RSV.preF-Based Regimen in Japanese Adults Aged 60 Years and Older. Influenza Other Respir Viruses 2024; 18:e13336. [PMID: 38880785 PMCID: PMC11180550 DOI: 10.1111/irv.13336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is increasingly recognized as a significant cause of lower respiratory tract disease (LRTD) in older adults. The Ad26.RSV.preF/RSV preF protein vaccine demonstrated protective efficacy against RSV related LRTD in a Phase 2b study in the United States. Hence, Ad26.RSV.preF/RSV preF protein vaccine candidate was evaluated in the Japanese older adult population. METHODS This Phase 1 study evaluated safety, reactogenicity, and immunogenicity of Ad26.RSV.preF/RSV preF protein vaccine at dose level of 1 × 1011 vp/150 μg in Japanese healthy adult aged ≥60 years. The study included a screening Phase, vaccination, 28-day follow up Phase, a 182-day follow-up period, and final visit on Day 183. A total of 36 participants were randomized in a 2:1 ratio to receive Ad26.RSV.preF/RSV preF protein vaccine (n = 24) or placebo (n = 12). After study intervention administration, the safety and immunogenicity analysis were performed as per planned schedule. Immune responses including virus-neutralizing and preF-specific binding antibodies were measured on Days 1, 15, 29, and 183. RESULTS There were no deaths, SAEs, or AEs leading to discontinuation reported during the study. The Ad26.RSV.preF/RSV preF protein vaccine had acceptable safety and tolerability profile with no safety concern in Japanese older adults. The Ad26.RSV.preF/RSV preF protein vaccine induced RSV-specific humoral immunity, with increase in antibody titers on Days 15 and 29 compared with baseline which was well maintained until Day 183. CONCLUSIONS A single dose of Ad26.RSV.preF/RSV preF protein vaccine had an acceptable safety and tolerability profile and induced RSV-specific humoral immunity in Japanese healthy adults. TRIAL REGISTRATION NCT number: NCT04354480; Clinical Registry number: CR108768.
Collapse
|
14
|
Barsan Kaya T, Öztunalı Ç, Aydemir Ö, Sürmeli Onay Ö, Tekin AN. A Tragic Consequence of Respiratory Syncytial Virus Infection. Clin Pediatr (Phila) 2024; 63:171-175. [PMID: 37804143 DOI: 10.1177/00099228231204445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Affiliation(s)
- Tuğba Barsan Kaya
- Department of Neonatology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Çiğdem Öztunalı
- Department of Pediatric Radiology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Özge Aydemir
- Department of Neonatology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Özge Sürmeli Onay
- Department of Neonatology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Ayşe Neslihan Tekin
- Department of Neonatology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| |
Collapse
|
15
|
Loaiza RA, Ramírez RA, Sepúlveda-Alfaro J, Ramírez MA, Andrade CA, Soto JA, González PA, Bueno SM, Kalergis AM. A molecular perspective for the development of antibodies against the human respiratory syncytial virus. Antiviral Res 2024; 222:105783. [PMID: 38145755 DOI: 10.1016/j.antiviral.2023.105783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/27/2023]
Abstract
The human respiratory syncytial virus (hRSV) is the leading etiologic agent causing respiratory infections in infants, children, older adults, and patients with comorbidities. Sixty-seven years have passed since the discovery of hRSV, and only a few successful mitigation or treatment tools have been developed against this virus. One of these is immunotherapy with monoclonal antibodies against structural proteins of the virus, such as Palivizumab, the first prophylactic approach approved by the Food and Drug Administration (FDA) of the USA. In this article, we discuss different strategies for the prevention and treatment of hRSV infection, focusing on the molecular mechanisms against each target that underly the rational design of antibodies against hRSV. At the same time, we describe the latest results regarding currently approved therapies against hRSV and the challenges associated with developing new candidates.
Collapse
Affiliation(s)
- Ricardo A Loaiza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Robinson A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Mario A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
16
|
Sun BW, Zhang PP, Wang ZH, Yao X, He ML, Bai RT, Che H, Lin J, Xie T, Hui Z, Ye XY, Wang LW. Prevention and Potential Treatment Strategies for Respiratory Syncytial Virus. Molecules 2024; 29:598. [PMID: 38338343 PMCID: PMC10856762 DOI: 10.3390/molecules29030598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a significant viral pathogen that causes respiratory infections in infants, the elderly, and immunocompromised individuals. RSV-related illnesses impose a substantial economic burden worldwide annually. The molecular structure, function, and in vivo interaction mechanisms of RSV have received more comprehensive attention in recent times, and significant progress has been made in developing inhibitors targeting various stages of the RSV replication cycle. These include fusion inhibitors, RSV polymerase inhibitors, and nucleoprotein inhibitors, as well as FDA-approved RSV prophylactic drugs palivizumab and nirsevimab. The research community is hopeful that these developments might provide easier access to knowledge and might spark new ideas for research programs.
Collapse
Affiliation(s)
- Bo-Wen Sun
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Peng-Peng Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zong-Hao Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xia Yao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Meng-Lan He
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Rui-Ting Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Che
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jing Lin
- Drug Discovery, Hangzhou Haolu Pharma Co., Hangzhou 311121, China;
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Li-Wei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
17
|
Gong X, Luo E, Fan L, Zhang W, Yang Y, Du Y, Yang X, Xing S. Clinical research on RSV prevention in children and pregnant women: progress and perspectives. Front Immunol 2024; 14:1329426. [PMID: 38327765 PMCID: PMC10847284 DOI: 10.3389/fimmu.2023.1329426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a significant causative agent of bronchitis and pneumonia in infants and children. The identification and structural analysis of the surface fusion glycoprotein of RSV represents a pivotal advancement in the development of RSV prevention. This review provides a comprehensive summary of RSV monoclonal antibody (mAb) and vaccine clinical trials registered on ClinicalTrials.gov, emphasizing on the classification, name, target, phase, clinical outcomes, and safety data of RSV vaccination in newborns, infants and children. We also discuss the characteristics of the types of RSV vaccines for maternal immunity and summarize the current clinical research progress of RSV vaccination in pregnant women and their protective efficacy in infants. This review will provide new ideas for the development of RSV prevention for children in the future.
Collapse
Affiliation(s)
- Xuejia Gong
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Erdan Luo
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Fan
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wanggang Zhang
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Yang
- Medical Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuhua Du
- Obstetrical Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Yang
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shasha Xing
- Good Clinical Practice Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
18
|
Zou G, Cao S, Gao Z, Yie J, Wu JZ. Current state and challenges in respiratory syncytial virus drug discovery and development. Antiviral Res 2024; 221:105791. [PMID: 38160942 DOI: 10.1016/j.antiviral.2023.105791] [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: 11/21/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections (LRTI) in young children and elderly people worldwide. Recent significant progress in our understanding of the structure and function of RSV proteins has led to the discovery of several clinical candidates targeting RSV fusion and replication. These include both the development of novel small molecule interventions and the isolation of potent monoclonal antibodies. In this review, we summarize the state-of-the-art of RSV drug discovery, with a focus on the characteristics of the candidates that reached the clinical stage of development. We also discuss the lessons learned from failed and discontinued clinical developments and highlight the challenges that remain for development of RSV therapies.
Collapse
Affiliation(s)
- Gang Zou
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China.
| | - Sushan Cao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Zhao Gao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Junming Yie
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Jim Zhen Wu
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| |
Collapse
|
19
|
Phuah JY, Maas BM, Tang A, Zhang Y, Caro L, Railkar RA, Swanson MD, Cao Y, Li H, Roadcap B, Catchpole AP, Aliprantis AO, Vora KA. Quantification of clesrovimab, an investigational, half-life extended, anti-respiratory syncytial virus protein F human monoclonal antibody in the nasal epithelial lining fluid of healthy adults. Biomed Pharmacother 2023; 169:115851. [PMID: 37976891 DOI: 10.1016/j.biopha.2023.115851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Clesrovimab (MK-1654) is an investigational, half-life extended human monoclonal antibody (mAb) against RSV F glycoprotein in clinical trials as a prophylactic agent against RSV infection for infants. METHODS This adult study measured clesrovimab concentrations in the serum and nasal epithelial lining fluid (ELF) to establish the partitioning of the antibody after dosing. Clesrovimab concentrations in the nasal ELF were normalized for sampling dilution using urea concentrations from ELF and serum. Furthermore, in vitro RSV neutralization of human nasal ELF following dosing was also measured to examine the activity of clesrovimab in the nasal compartment. FINDINGS mAbs with YTE mutations are reported in literature to partition ∼1-2 % of serum antibodies into nasal mucosa. Nasal: serum ratios of 1:69-1:30 were observed for clesrovimab in two separate adult human trials after urea normalization, translating to 1.4-3.3 % of serum concentrations. The nasal PK and estimates of peripheral volume of distribution correlated with higher extravascular distribution of clesrovimab. These higher concentration of the antibody in the nasal ELF corroborated with the nasal sample's ability to neutralize RSV ex vivo. An overall trend of decreased viral plaque AUC was also noted with increasing availability of clesrovimab in the nasal ELF from a human RSV challenge study. INTERPRETATION Along with its extended half-life, the higher penetration of clesrovimab into the nasal epithelial lining fluid and the associated local increase in RSV neutralization activity could offer infants better protection against RSV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yu Cao
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abelson D, Barajas J, Stuart L, Kim D, Marimuthu A, Hu C, Yamamoto B, Ailor E, Whaley KJ, Vu H, Agans KN, Borisevich V, Deer DJ, Dobias NS, Woolsey C, Prasad AN, Peel JE, Lawrence WS, Cross RW, Geisbert TW, Fenton KA, Zeitlin L. Long-term Prophylaxis Against Aerosolized Marburg Virus in Nonhuman Primates With an Afucosylated Monoclonal Antibody. J Infect Dis 2023; 228:S701-S711. [PMID: 37474248 PMCID: PMC11009508 DOI: 10.1093/infdis/jiad278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Marburg virus (MARV) causes a hemorrhagic fever disease in human and nonhuman primates with high levels of morbidity and mortality. Concerns about weaponization of aerosolized MARV have spurred the development of nonhuman primate (NHP) models of aerosol exposure. To address the potential threat of aerosol exposure, a monoclonal antibody that binds MARV glycoprotein was tested, MR186YTE, for its efficacy as a prophylactic. MR186YTE was administered intramuscularly to NHPs at 15 or 5 mg/kg 1 month prior to MARV aerosol challenge. Seventy-five percent (3/4) of the 15 mg/kg dose group and 50% (2/4) of the 5 mg/kg dose group survived. Serum analyses showed that the NHP dosed with 15 mg/kg that succumbed to infection developed an antidrug antibody response and therefore had no detectable MR186YTE at the time of challenge. These results suggest that intramuscular dosing of mAbs may be a clinically useful prophylaxis for MARV aerosol exposure.
Collapse
Affiliation(s)
- Dafna Abelson
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Lauren Stuart
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | - Do Kim
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Chris Hu
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Eric Ailor
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| | | | - Hong Vu
- Integrated Biotherapeutics, Rockville, Maryland, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jennifer E Peel
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - William S Lawrence
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc, San Diego, California, USA
| |
Collapse
|
21
|
Brady T, Cayatte C, Roe TL, Speer SD, Ji H, Machiesky L, Zhang T, Wilkins D, Tuffy KM, Kelly EJ. Fc-mediated functions of nirsevimab complement direct respiratory syncytial virus neutralization but are not required for optimal prophylactic protection. Front Immunol 2023; 14:1283120. [PMID: 37901217 PMCID: PMC10600457 DOI: 10.3389/fimmu.2023.1283120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Nirsevimab is an extended half-life (M252Y/S254T/T256E [YTE]-modified) monoclonal antibody to the pre-fusion conformation of the respiratory syncytial virus (RSV) Fusion protein, with established efficacy in preventing RSV-associated lower respiratory tract infection in infants for the duration of a typical RSV season. Previous studies suggest that nirsevimab confers protection via direct virus neutralization. Here we use preclinical models to explore whether fragment crystallizable (Fc)-mediated effector functions contribute to nirsevimab-mediated protection. Methods Nirsevimab, MEDI8897* (i.e., nirsevimab without the YTE modification), and MEDI8897*-TM (i.e., MEDI8897* without Fc effector functions) binding to Fc γ receptors (FcγRs) was evaluated using surface plasmon resonance. Antibody-dependent neutrophil phagocytosis (ADNP), antibody-dependent cellular phagocytosis (ADCP), antibody-dependent complement deposition (ADCD), and antibody-dependent cellular cytotoxicity (ADCC) were assessed through in vitro and ex vivo serological analyses. A cotton rat challenge study was performed with MEDI8897* and MEDI8897*-TM to explore whether Fc effector functions contribute to protection from RSV. Results Nirsevimab and MEDI8897* exhibited binding to a range of FcγRs, with expected reductions in FcγR binding affinities observed for MEDI8897*-TM. Nirsevimab exhibited in vitro ADNP, ADCP, ADCD, and ADCC activity above background levels, and similar ADNP, ADCP, and ADCD activity to palivizumab. Nirsevimab administration increased ex vivo ADNP, ADCP, and ADCD activity in participant serum from the MELODY study (NCT03979313). However, ADCC levels remained similar between nirsevimab and placebo. MEDI8897* and MEDI8897*-TM exhibited similar dose-dependent reduction in lung and nasal turbinate RSV titers in the cotton rat model. Conclusion Nirsevimab possesses Fc effector activity comparable with the current standard of care, palivizumab. However, despite possessing the capacity for Fc effector activity, data from RSV challenge experiments illustrate that nirsevimab-mediated protection is primarily dependent on direct virus neutralization.
Collapse
Affiliation(s)
- Tyler Brady
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Corinne Cayatte
- Early Oncology ICA, Oncology R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Tiffany L. Roe
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Scott D. Speer
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Hong Ji
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - LeeAnn Machiesky
- Process and Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Tianhui Zhang
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Deidre Wilkins
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Kevin M. Tuffy
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Elizabeth J. Kelly
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
22
|
Domachowske JB, Chang Y, Atanasova V, Cabañas F, Furuno K, Nguyen KA, Banu I, Kubiak RJ, Leach A, Mankad VS, Shroff M, Takas T, Villafana T, Wählby Hamrén U. Safety of Re-dosing Nirsevimab Prior to RSV Season 2 in Children With Heart or Lung Disease. J Pediatric Infect Dis Soc 2023; 12:477-480. [PMID: 37466917 PMCID: PMC10469583 DOI: 10.1093/jpids/piad052] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 07/20/2023]
Abstract
In children with congenital heart disease and/or chronic lung disease entering their second respiratory syncytial virus (RSV) season, 200 mg nirsevimab had a similar safety profile to that of palivizumab and resulted in nirsevimab serum exposures associated with efficacy in healthy infants, supporting efficacy in this population at risk of severe RSV disease.
Collapse
Affiliation(s)
- Joseph B Domachowske
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Yue Chang
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Fernando Cabañas
- Department of Paediatrics-Neonatology, Quironsalud Madrid University Hospital and Quironsalud San José Hospital, Madrid, Spain
| | - Kenji Furuno
- Department of General Pediatrics and Interdisciplinary Medicine, Fukuoka Children’s Hospital, Fukuoka, Japan
| | - Kim A Nguyen
- Hospices Civils de Lyon, Hôpital Femme-Mere-Enfant, Service de Réanimation Néonatale et de Neonatologie (NICU) et CIC 1407, Lyon, France
| | - Irfana Banu
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Robert J Kubiak
- Clinical Pharmacology and Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Amanda Leach
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Vaishali S Mankad
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Durham, North Carolina, USA
| | - Manish Shroff
- Patient Safety, Chief Medical Office, R&D, AstraZeneca, Waltham, Massachusetts, USA
| | - Therese Takas
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Tonya Villafana
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
23
|
Ahani B, Tuffy KM, Aksyuk AA, Wilkins D, Abram ME, Dagan R, Domachowske JB, Guest JD, Ji H, Kushnir A, Leach A, Madhi SA, Mankad VS, Simões EAF, Sparklin B, Speer SD, Stanley AM, Tabor DE, Hamrén UW, Kelly EJ, Villafana T. Molecular and phenotypic characteristics of RSV infections in infants during two nirsevimab randomized clinical trials. Nat Commun 2023; 14:4347. [PMID: 37468530 PMCID: PMC10356750 DOI: 10.1038/s41467-023-40057-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Nirsevimab is a monoclonal antibody that binds to the respiratory syncytial virus (RSV) fusion protein. During the Phase 2b (NCT02878330) and MELODY (NCT03979313) clinical trials, infants received one dose of nirsevimab or placebo before their first RSV season. In this pre-specified analysis, isolates from RSV infections were subtyped, sequenced and analyzed for nirsevimab binding site substitutions; subsequently, recombinant RSVs were engineered for microneutralization susceptibility testing. Here we show that the frequency of infections caused by subtypes A and B is similar across and within the two trials. In addition, RSV A had one and RSV B had 10 fusion protein substitutions occurring at >5% frequency. Notably, RSV B binding site substitutions were rare, except for the highly prevalent I206M:Q209R, which increases nirsevimab susceptibility; RSV B isolates from two participants had binding site substitutions that reduce nirsevimab susceptibility. Overall, >99% of isolates from the Phase 2b and MELODY trials retained susceptibility to nirsevimab.
Collapse
Affiliation(s)
- Bahar Ahani
- Bioinformatics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kevin M Tuffy
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Michael E Abram
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ron Dagan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences of the Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Johnathan D Guest
- Virology and Vaccine Discovery, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Hong Ji
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anna Kushnir
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vaishali S Mankad
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - Eric A F Simões
- University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Benjamin Sparklin
- Bioinformatics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Scott D Speer
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ann Marie Stanley
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - David E Tabor
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
24
|
Abstract
IgGs are essential soluble components of the adaptive immune response that evolved to protect the body from infection. Compared with other immunoglobulins, the role of IgGs is distinguished and enhanced by their high circulating levels, long half-life and ability to transfer from mother to offspring, properties that are conferred by interactions with neonatal Fc receptor (FcRn). FcRn binds to the Fc portion of IgGs in a pH-dependent manner and protects them from intracellular degradation. It also allows their transport across polarized cells that separate tissue compartments, such as the endothelium and epithelium. Further, it is becoming apparent that FcRn functions to potentiate cellular immune responses when IgGs, bound to their antigens, form IgG immune complexes. Besides the protective role of IgG, IgG autoantibodies are associated with numerous pathological conditions. As such, FcRn blockade is a novel and effective strategy to reduce circulating levels of pathogenic IgG autoantibodies and curtail IgG-mediated diseases, with several FcRn-blocking strategies on the path to therapeutic use. Here, we describe the current state of knowledge of FcRn-IgG immunobiology, with an emphasis on the functional and pathological aspects, and an overview of FcRn-targeted therapy development.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa K Kozicky
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit K Gandhi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Diseases Center, Boston, MA, USA.
| |
Collapse
|
25
|
Wilkins D, Yuan Y, Chang Y, Aksyuk AA, Núñez BS, Wählby-Hamrén U, Zhang T, Abram ME, Leach A, Villafana T, Esser MT. Durability of neutralizing RSV antibodies following nirsevimab administration and elicitation of the natural immune response to RSV infection in infants. Nat Med 2023; 29:1172-1179. [PMID: 37095249 PMCID: PMC10202809 DOI: 10.1038/s41591-023-02316-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Nirsevimab is an extended half-life monoclonal antibody specific for the prefusion conformation of the respiratory syncytial virus (RSV) F protein, which has been studied in preterm and full-term infants in the phase 2b and phase 3 MELODY trials. We analyzed serum samples collected from 2,143 infants during these studies to characterize baseline levels of RSV-specific immunoglobulin G antibodies and neutralizing antibodies (NAbs), duration of RSV NAb levels following nirsevimab administration, the risk of RSV exposure during the first year of life and the infant's adaptive immune response to RSV following nirsevimab administration. Baseline RSV antibody levels varied widely; consistent with reports that maternal antibodies are transferred late in the third trimester, preterm infants had lower baseline RSV antibody levels than full-term infants. Nirsevimab recipients had RSV NAb levels >140-fold higher than baseline at day 31 and remained >50-fold higher at day 151 and >7-fold higher at day 361. Similar seroresponse rates to the postfusion form of RSV F protein in nirsevimab recipients (68-69%) compared with placebo recipients (63-70%; not statistically significant) suggest that while nirsevimab protects from RSV disease, it still allows an active immune response. In summary, nirsevimab provided sustained, high levels of NAb throughout an infant's first RSV season and prevented RSV disease while allowing the development of an immune response to RSV.
Collapse
Affiliation(s)
- Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Yuan Yuan
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yue Chang
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Anastasia A Aksyuk
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Beatriz Seoane Núñez
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Madrid, Spain
| | - Ulrika Wählby-Hamrén
- Clinical Pharmacology & Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tianhui Zhang
- Data Sciences and Quantitative Biology, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Michael E Abram
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mark T Esser
- Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
26
|
Diethelm-Varela B, Soto JA, Riedel CA, Bueno SM, Kalergis AM. New Developments and Challenges in Antibody-Based Therapies for the Respiratory Syncytial Virus. Infect Drug Resist 2023; 16:2061-2074. [PMID: 37063935 PMCID: PMC10094422 DOI: 10.2147/idr.s379660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023] Open
Abstract
Since the discovery of the human respiratory syncytial virus (hRSV), multiple research efforts have been conducted to develop vaccines and treatments capable of reducing the risk of severe disease, hospitalization, long-term sequelae, and death from this pathogen in susceptible populations. In this sense, therapies specifically directed against hRSV are mainly based on monoclonal and polyclonal antibodies such as intravenous IgG (IVIG)-RSV and the monoclonal antibody palivizumab. However, these therapies are associated with significant limitations, including the need for the recruitment of a high number of convalescent volunteers who donate blood to procure IVIG-RSV and the costs associated with the need for repeated administrations of palivizumab. These limitations render this product not cost-effective for populations other than high-risk patients. These problems have underscored that it is still necessary to identify new safe and effective therapies for human use. However, these new therapies must benefit from a comparatively cheap production cost and the opportunity to be available to the high-risk population and anyone who requires treatment. Here, we review the different antibodies used to prevent the pathology caused by hRSV infection, highlighting therapies currently approved for human use and their clinical value. Also, the new, most promising candidates based on preclinical studies and clinical trial results are revised.
Collapse
Affiliation(s)
- Benjamín Diethelm-Varela
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Simões EAF, Madhi SA, Muller WJ, Atanasova V, Bosheva M, Cabañas F, Baca Cots M, Domachowske JB, Garcia-Garcia ML, Grantina I, Nguyen KA, Zar HJ, Berglind A, Cummings C, Griffin MP, Takas T, Yuan Y, Wählby Hamrén U, Leach A, Villafana T. Efficacy of nirsevimab against respiratory syncytial virus lower respiratory tract infections in preterm and term infants, and pharmacokinetic extrapolation to infants with congenital heart disease and chronic lung disease: a pooled analysis of randomised controlled trials. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:180-189. [PMID: 36634694 PMCID: PMC9940918 DOI: 10.1016/s2352-4642(22)00321-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND In a phase 2b trial and the phase 3 MELODY trial, nirsevimab, an extended half-life, monoclonal antibody against respiratory syncytial virus (RSV), protected healthy infants born preterm or at full term against medically attended RSV lower respiratory tract infection (LRTI). In the MEDLEY phase 2-3 trial in infants at higher risk for severe RSV infection, nirsevimab showed a similar safety profile to that of palivizumab. The aim of the current analysis was to assess the efficacy of nirsevimab using a weight-banded dosing regimen in infants born between 29 weeks gestational age and full term. METHODS Infants enrolled in the phase 2b and MELODY trials were randomised (2:1) to receive a single intramuscular injection of nirsevimab (infants weighing <5 kg received 50 mg; those weighing ≥5 kg received 100 mg) or placebo before the RSV season. Infants in MEDLEY were randomised (2:1) to receive one dose of nirsevimab (infants weighing <5 kg received 50 mg; those weighing ≥5 kg received 100 mg) followed by four monthly placebo doses, or five once-a-month intramuscular doses of palivizumab. We report a prespecified pooled efficacy analysis assessing the weight-banded dosing regimen proposed on the basis of the phase 2b and MELODY trials, in addition to extrapolated efficacy in infants with chronic lung disease, congenital heart disease, or extreme preterm birth (<29 weeks' gestational age) based on pharmacokinetic data from the phase 2-3 MEDLEY safety trial. For the pooled efficacy analysis, the primary endpoint was incidence of medically attended RSV LRTI through 150 days post-dose. The secondary efficacy endpoint was number of admissions to hospital for medically attended RSV LRTI. The incidence of very severe RSV LRTI was an exploratory endpoint, defined as cases of hospital admission for medically attended RSV LRTI that required supplemental oxygen or intravenous fluids. We also did a prespecified exploratory analysis of medically attended LRTI of any cause (in the investigator's judgement) and hospital admission for respiratory illness of any cause (defined as any upper respiratory tract infection or LRTI leading to hospital admission). Post hoc exploratory analyses of outpatient visits and antibiotic use were also done. Nirsevimab serum concentrations in MEDLEY were assessed using population pharmacokinetic methods and the pooled data from the phase 2b and MELODY trials. An exposure target was defined on the basis of an exposure-response analysis. To successfully demonstrate extrapolation, more than 80% of infants in MEDLEY had to achieve serum nirsevimab exposures at or above the predicted efficacious target. FINDINGS Overall, 2350 infants (1564 in the nirsevimab group and 786 in the placebo group) in the phase 2b and MELODY trials were included in the pooled analysis. Nirsevimab showed efficacy versus placebo with respect to the primary endpoint of medically attended RSV LRTI (19 [1%] nirsevimab recipients vs 51 [6%] placebo recipients; relative risk reduction [RRR] 79·5% [95% CI 65·9-87·7]). Consistent efficacy was shown for additional endpoints of RSV LRTI hospital admission (nine [1%] nirsevimab recipients vs 21 [3%] placebo recipients; 77·3% [50·3-89·7]) and very severe RSV (five [<1%] vs 18 [2%]; 86·0% [62·5-94·8]). Nirsevimab recipients had fewer hospital admissions for any-cause respiratory illness (RRR 43·8% [18·8-61·1]), any-cause medically attended LRTI (35·4% [21·5-46·9]), LRTI outpatient visits (41·9% [25·7-54·6]), and antibiotic prescriptions (23·6% [3·8-39·3]). Among infants with chronic lung disease, congenital heart disease, or extreme preterm birth in MEDLEY, nirsevimab serum exposures were similar to those found in the pooled data; exposures were above the target in more than 80% of the overall MEDLEY trial population (94%), including infants with chronic lung disease (94%) or congenital heart disease (80%) and those born extremely preterm (94%). INTERPRETATION A single dose of nirsevimab protected healthy infants born at term or preterm from medically attended RSV LRTI, associated hospital admission, and severe RSV. Pharmacokinetic data support efficacy extrapolation to infants with chronic lung disease, congenital heart disease, or extreme prematurity. Together, these data suggest that nirsevimab has the potential to change the landscape of infant RSV disease by reducing a major cause of infant morbidity and the consequent burden on caregivers, clinicians, and health-care providers. FUNDING AstraZeneca and Sanofi.
Collapse
Affiliation(s)
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - William J Muller
- Ann & Robert H Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Miroslava Bosheva
- University Multiprofile Hospital for Active Treatment Sv Georgi Medical University, Plovdiv, Bulgaria
| | | | | | | | | | | | - Kim A Nguyen
- Hospices Civils de Lyon, Neonatal Intensive Care Units and CIC 1407, Lyon, France
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross Children's Hospital, Cape Town, South Africa; SA-MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Anna Berglind
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Celeste Cummings
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - M Pamela Griffin
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Therese Takas
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yuan Yuan
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
28
|
Jenkins VA, Hoet B, Hochrein H, De Moerlooze L. The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein. Vaccines (Basel) 2023; 11:382. [PMID: 36851260 PMCID: PMC9963583 DOI: 10.3390/vaccines11020382] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of paediatric respiratory tract infection and causes a significant health burden in older adults. Natural immunity to RSV is incomplete, permitting recurrent symptomatic infection over an individual's lifespan. When combined with immunosenescence, this increases older adults' susceptibility to more severe disease symptoms. As RSV prophylaxis is currently limited to infants, older adults represent an important target population for RSV vaccine development. The relationship between RSV and our immune systems is complex, and these interactions require deeper understanding to tailor an effective vaccine candidate towards older adults. To date, vaccine candidates targeting RSV antigens, including pre-F, F, G (A), G (B), M2-1, and N, have shown efficacy against RSV infection in older adults in clinical trial settings. Although vaccine candidates have demonstrated robust neutralising IgG and cellular responses, it is important that research continues to investigate the RSV immune response in order to further understand how the choice of antigenic target site may impact vaccine effectiveness. In this article, we discuss the Phase 3 vaccine candidates being tested in older adults and review the hurdles that must be overcome to achieve effective protection against RSV.
Collapse
|
29
|
Haraya K, Tachibana T. Translational Approach for Predicting Human Pharmacokinetics of Engineered Therapeutic Monoclonal Antibodies with Increased FcRn-Binding Mutations. BioDrugs 2023; 37:99-108. [PMID: 36449140 PMCID: PMC9709760 DOI: 10.1007/s40259-022-00566-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Recently, increasing FcRn binding by Fc engineering has become a promising approach for prolonging the half-life of therapeutic monoclonal antibodies (mAbs). This study is the first to investigate the optimization of an allometric scaling approach for engineered mAbs based on cynomolgus monkey data to predict human pharmacokinetics. METHODS Linear two-compartmental model parameters (clearance [CL]; volume of distribution in the central compartment [Vc]; inter-compartmental clearance [Q]; volume of distribution in the peripheral compartment [Vp]) after the intravenous (IV) injection of engineered mAbs (M252Y/S254T/T256E or M428L/N434S mutations) in cynomolgus monkeys and humans were collected from published data. We explored the optimal exponent for allometric scaling to predict parameters in humans based on cynomolgus monkey data. Moreover, the plasma concentration-time profile of engineered mAbs after IV injection in humans was predicted using parameters estimated based on an optimized exponent. RESULTS For engineered mAbs, a significant positive correlation between cynomolgus monkeys and humans was observed for CL, but not for other parameters. Whereas conventional exponents (CL: 0.8, Q: 0.75, Vc: 1.0, Vp: 0.95) previously established for normal mAbs showed poor prediction accuracy for CL and Q of engineered mAbs, the newly optimized exponents (CL: 0.55, Q: 0.6, Vc: 0.95, Vp: 0.95) achieved superior predictability for engineered mAbs. Moreover, the optimized exponents accurately predicted plasma mAb concentration-time profiles after IV injection of engineered mAbs in humans. CONCLUSIONS We found that engineered mAbs require specially optimized exponents to accurately predict pharmacokinetic parameters and plasma concentration-time profiles after IV injections in humans based on cynomolgus monkey data. This optimized approach can contribute to a more accurate prediction of human pharmacokinetics in the development of engineered mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan.
| | - Tatsuhiko Tachibana
- Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| |
Collapse
|
30
|
Mashiyama F, Hashimoto K, Norito S, Okabe H, Sato A, Kume Y, Maeda R, Sato M, Sato M, Kyozuka H, Fujimori K, Nishigori H, Shinoki K, Yasumura S, Sakuma H, Hosoya M. Neutralizing and Epitope-Specific Antibodies against Respiratory Syncytial Virus in Maternal and Cord Blood Paired Samples. Viruses 2022; 14:2702. [PMID: 36560707 PMCID: PMC9784505 DOI: 10.3390/v14122702] [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: 10/28/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Only a few qualitative studies of neutralizing antibody titers (NATs) against respiratory syncytial virus (RSV) have focused on epitope-specific antibody (ESA) levels. Here, NATs against RSV in sera were measured using the blood of 412 mothers and cord blood (CB) of 95 of the 412 mother-child pairs. ESA levels against sites zero (Ø) and IIa of the F protein of RSV were measured in 87 of the 95 mother-child pairs. The median gestational age was 39 weeks. The NATs and ESA levels in CB were slightly higher than those in maternal blood (MB). The NATs for RSV subtype A (RSV-A) in MB and CB showed a positive correlation (r = 0.75). The ESA levels against sites Ø and IIa in MB and CB showed positive correlations, r = 0.76 and r = 0.69, respectively. In MB, the NATs and ESA levels against RSV were positively correlated, more significantly against site Ø (RSV-A: r = 0.70, RSV-B: r = 0.48) than against site IIa (RSV-A: r = 0.19, RSV-B: r = 0.31). Sufficient amounts of ESAs against sites Ø and IIa of RSV were transferred from mothers to term infants. ESA levels against site Ø contribute to NATs.
Collapse
Affiliation(s)
- Fumi Mashiyama
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
- Pediatrics, Hoshi General Hospital, Koriyama 960-1295, Japan
| | - Koichi Hashimoto
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
| | - Sakurako Norito
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hisao Okabe
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Akiko Sato
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
| | - Yohei Kume
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Ryo Maeda
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Maki Sato
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Masatoki Sato
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hyo Kyozuka
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Keiya Fujimori
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
- Department of Obstetrics and Gynecology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hidekazu Nishigori
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
- Fukushima Medical Center for Children and Women, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kosei Shinoki
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
| | - Seiji Yasumura
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
- Department of Public Health, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hiroko Sakuma
- Pediatrics, Hoshi General Hospital, Koriyama 960-1295, Japan
| | - Mitsuaki Hosoya
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
- Fukushima Regional Center for the Japan Environmental and Children’s Study, Fukushima 960-1295, Japan
| |
Collapse
|
31
|
Qiu X, Xu S, Lu Y, Luo Z, Yan Y, Wang C, Ji J. Development of mRNA vaccines against respiratory syncytial virus (RSV). Cytokine Growth Factor Rev 2022; 68:37-53. [PMID: 36280532 DOI: 10.1016/j.cytogfr.2022.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 02/06/2023]
Abstract
Respiratory syncytial virus (RSV) is a single-stranded negative-sense RNA virus that is the primary etiologic pathogen of bronchitis and pneumonia in infants and the elderly. Currently, no preventative vaccine has been approved for RSV infection. However, advances in the characterization, and structural resolution, of the RSV surface fusion glycoprotein have revolutionized RSV vaccine development by providing a new target for preventive interventions. In general, six different approaches have been adopted in the development of preventative RSV therapeutics, namely, particle-based vaccines, vector-based vaccines, live-attenuated or chimeric vaccines, subunit vaccines, mRNA vaccines, and monoclonal antibodies. Among these preventive interventions, MVA-BN-RSV, RSVpreF3, RSVpreF, Ad26. RSV.preF, nirsevimab, clesrovimab and mRNA-1345 is being tested in phase 3 clinical trials, and displays the most promising in infant or elderly populations. Accompanied by the huge success of mRNA vaccines in COVID-19, mRNA vaccines have been rapidly developed, with many having entered clinical studies, in which they have demonstrated encouraging results and acceptable safety profiles. In fact, Moderna has received FDA approval, granting fast-track designation for an investigational single-dose mRNA-1345 vaccine against RSV in adults over 60 years of age. Hence, mRNA vaccines may represent a new, more successful, chapter in the continued battle to develop effective preventative measures against RSV. This review discusses the structure, life cycle, and brief history of RSV, while also presenting the current advancements in RSV preventatives, with a focus on the latest progress in RSV mRNA vaccine development. Finally, future prospects for this field are presented.
Collapse
Affiliation(s)
- Xirui Qiu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyan Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Lu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yangtian Yan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chuyue Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
32
|
Issmail L, Möser C, Jäger C, Altattan B, Ramsbeck D, Kleinschmidt M, Buchholz M, Smith D, Grunwald T. Prefusion-specific antibody-derived peptides trivalently presented on DNA-nanoscaffolds as an innovative strategy against RSV entry. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.994843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human respiratory syncytial virus (RSV) is the primary cause of acute lower respiratory tract infections in children and the elderly worldwide, for which neither a vaccine nor an effective therapy is approved. The entry of RSV into the host cell is mediated by stepwise structural changes in the surface RSV fusion (RSV-F) glycoprotein. Recent progress in structural and functional studies of RSV-F glycoprotein revealed conformation-dependent neutralizing epitopes which have become attractive targets for vaccine and therapeutic development. As RSV-F is present on viral surface in a trimeric form, a trivalent binding interaction between a candidate fusion inhibitor and the respective epitopes on each of the three monomers is expected to prevent viral infection at higher potency than a monovalent or bivalent inhibitor. Here we demonstrate a novel RSV entry inhibitory approach by implementing a trimeric DNA nanostructure as a template to display up to three linear peptide moieties that simultaneously target an epitope on the surface of the prefusion RSV-F protein. In order to design synthetic binding peptides that can be coupled to the DNA nanostructure, the prefusion RSV-F-specific monoclonal antibody (D25) was selected. Complementarity-determining region 3 (CDR3) derived peptides underwent truncation and alanine-scanning mutagenesis analysis, followed by systematic sequence modifications using non-canonical amino acids. The most effective peptide candidate was used as a binding moiety to functionalize the DNA nanostructure. The designed DNA-peptide construct was able to block RSV infection on cells more efficiently than the monomeric peptides, however a more moderate reduction of viral load was observed in the lungs of infected mice upon intranasal application, likely due to dissociation or absorption of the underlying DNA structure by cells in the lungs. Taken together, our results point towards the inhibitory potential of a novel trimeric DNA-peptide based approach against RSV and open the possibility to apply this platform to target other viral infections.
Collapse
|
33
|
Challenges in Maximizing Impacts of Preventive Strategies against Respiratory Syncytial Virus (RSV) Disease in Young Children. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:293-300. [PMID: 35782467 PMCID: PMC9235255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract illness in infants and young children. It causes substantial morbidity and mortality in young children and older adults. As few therapeutic and prophylaxis options against RSV illness are currently available, there is a great need for effective RSV vaccines and immune-prophylaxis. Encouragingly, multiple vaccines and immuno-prophylaxis aiming to protect pediatric populations have shown promising progress in clinical trials. The three major preventive strategies include RSV F-protein-based vaccines for pregnant women, extended half-life monoclonal antibodies for neonates, and live-attenuated vaccines for infants. Each preventive strategy has its own merits and challenges yet to be overcome. Challenges also exist in maximizing vaccine impacts in the post-implementation era. This perspectives piece focuses on RSV preventive strategies in young children and highlights the remaining questions in current development of RSV immunization products and design of immunization programs.
Collapse
|
34
|
Orito Y, Otani N, Matsumoto Y, Fujimoto K, Oshima N, Maas BM, Caro L, Aliprantis AO, Cox KS, Tokumaru O, Kodama M, Kudo H, Imai H, Uemura N. A Phase 1 Study to Evaluate Safety, Pharmacokinetics, and Pharmacodynamics of Respiratory Syncytial Virus (RSV) Neutralizing Monoclonal Antibody MK-1654 in Healthy Japanese Adults. Clin Transl Sci 2022; 15:1753-1763. [PMID: 35506164 PMCID: PMC9283748 DOI: 10.1111/cts.13290] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 12/05/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among all infants worldwide and remains a significant cause of morbidity and mortality. To address this unmet medical need, MK‐1654, a half‐life extended RSV neutralizing monoclonal antibody, is in clinical development for the prevention of RSV disease in infants. This was a phase I, randomized, placebo‐controlled, single‐site, double‐blind trial of MK‐1654 in 44 healthy Japanese adults. The safety, tolerability, pharmacokinetics, antidrug antibodies (ADAs), and serum neutralizing antibody (SNA) titers against RSV were evaluated for 1 year after a single intramuscular (i.m.) or intravenous (i.v.) dose of MK‐1654 or placebo in five groups (100 mg i.m., 300 mg i.m., 300 mg i.v., 1000 mg i.v., or placebo). MK‐1654 was generally well‐tolerated in Japanese adults. There were no serious drug‐related adverse events (AEs) reported in any MK‐1654 recipient and no discontinuations due to any AEs in the study. The half‐life of MK‐1654 ranged from 76 to 91 days across dosing groups. Estimated bioavailability was 86% for 100 mg i.m. and 77% for 300 mg i.m. One participant out of 33 (3.0%) developed detectable ADA with no apparent associated AEs. The RSV SNA titers increased in a dose‐dependent manner among participants who received MK‐1654. These data support the development of MK‐1654 for use in Japanese infants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Antonios O Aliprantis
- Merck and Co., Inc., Kenilworth, NJ, USA.,Present address: Flagship Pioneering, Boston, MA, USA
| | - Kara S Cox
- Merck and Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | |
Collapse
|
35
|
Haraya K, Tsutsui H, Komori Y, Tachibana T. Recent Advances in Translational Pharmacokinetics and Pharmacodynamics Prediction of Therapeutic Antibodies Using Modeling and Simulation. Pharmaceuticals (Basel) 2022; 15:ph15050508. [PMID: 35631335 PMCID: PMC9145563 DOI: 10.3390/ph15050508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have been a promising therapeutic approach for several diseases and a wide variety of mAbs are being evaluated in clinical trials. To accelerate clinical development and improve the probability of success, pharmacokinetics and pharmacodynamics (PKPD) in humans must be predicted before clinical trials can begin. Traditionally, empirical-approach-based PKPD prediction has been applied for a long time. Recently, modeling and simulation (M&S) methods have also become valuable for quantitatively predicting PKPD in humans. Although several models (e.g., the compartment model, Michaelis–Menten model, target-mediated drug disposition model, and physiologically based pharmacokinetic model) have been established and used to predict the PKPD of mAbs in humans, more complex mechanistic models, such as the quantitative systemics pharmacology model, have been recently developed. This review summarizes the recent advances and future direction of M&S-based approaches to the quantitative prediction of human PKPD for mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
- Correspondence:
| | - Haruka Tsutsui
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
| | - Yasunori Komori
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| |
Collapse
|
36
|
Protein and Peptide Substances in the Treatment of Respiratory Syncytial Infection: Current State. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072263. [PMID: 35408661 PMCID: PMC9000545 DOI: 10.3390/molecules27072263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 12/05/2022]
Abstract
Respiratory syncytial virus infection (RSVI) is an acute medical and social problem in many countries globally. Infection is most dangerous for infants under one year old and the elderly. Despite its epidemiological relevance, only two drugs are registered for clinical use against RSVI: ribavirin (approved in a limited number of countries due to side effects) and palivizumab (Synagis), which is intended only for the prevention, but not the treatment, of infection. Currently, various research groups are searching for new drugs against RSV, with three main areas of research: small molecules, polymeric drugs (proteins and peptides), and plant extracts. This review is devoted to currently developed protein and peptide anti-RSV drugs.
Collapse
|
37
|
Loo YM, McTamney PM, Arends RH, Abram ME, Aksyuk AA, Diallo S, Flores DJ, Kelly EJ, Ren K, Roque R, Rosenthal K, Streicher K, Tuffy KM, Bond NJ, Cornwell O, Bouquet J, Cheng LI, Dunyak J, Huang Y, Rosenbaum AI, Reddy VP, Andersen H, Carnahan RH, Crowe JE, Kuehne AI, Herbert AS, Dye JM, Bright H, Kallewaard NL, Pangalos MN, Esser MT. The SARS-CoV-2 monoclonal antibody combination, AZD7442, is protective in nonhuman primates and has an extended half-life in humans. Sci Transl Med 2022; 14:eabl8124. [PMID: 35076282 PMCID: PMC8939769 DOI: 10.1126/scitranslmed.abl8124] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022]
Abstract
Despite the success of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines, there remains a need for more prevention and treatment options for individuals remaining at risk of coronavirus disease 2019 (COVID-19). Monoclonal antibodies (mAbs) against the viral spike protein have potential to both prevent and treat COVID-19 and reduce the risk of severe disease and death. Here, we describe AZD7442, a combination of two mAbs, AZD8895 (tixagevimab) and AZD1061 (cilgavimab), that simultaneously bind to distinct, nonoverlapping epitopes on the spike protein receptor binding domain to neutralize SARS-CoV-2. Initially isolated from individuals with prior SARS-CoV-2 infection, the two mAbs were designed to extend their half-lives and reduce effector functions. The AZD7442 mAbs individually prevent the spike protein from binding to angiotensin-converting enzyme 2 receptor, blocking virus cell entry, and neutralize all tested SARS-CoV-2 variants of concern. In a nonhuman primate model of SARS-CoV-2 infection, prophylactic AZD7442 administration prevented infection, whereas therapeutic administration accelerated virus clearance from the lung. In an ongoing phase 1 study in healthy participants (NCT04507256), a 300-mg intramuscular injection of AZD7442 provided SARS-CoV-2 serum geometric mean neutralizing titers greater than 10-fold above those of convalescent serum for at least 3 months, which remained threefold above those of convalescent serum at 9 months after AZD7442 administration. About 1 to 2% of serum AZD7442 was detected in nasal mucosa, a site of SARS-CoV-2 infection. Extrapolation of the time course of serum AZD7442 concentration suggests AZD7442 may provide up to 12 months of protection and benefit individuals at high-risk of COVID-19.
Collapse
Affiliation(s)
- Yueh-Ming Loo
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Patrick M. McTamney
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Rosalinda H. Arends
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Michael E. Abram
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Anastasia A. Aksyuk
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Seme Diallo
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Daniel J. Flores
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Elizabeth J. Kelly
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Kuishu Ren
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Richard Roque
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Kim Rosenthal
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Katie Streicher
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Kevin M. Tuffy
- Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Nicholas J. Bond
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| | - Owen Cornwell
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| | - Jerome Bouquet
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, San Francisco, CA, 94080, USA
| | - Lily I. Cheng
- Oncology Safety Pathology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - James Dunyak
- Clinical Pharmacology and Pharmacometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, San Francisco, CA, 94080, USA
| | - Anton I. Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, San Francisco, CA, 94080, USA
| | - Venkatesh Pilla Reddy
- Clinical Pharmacology and Pharmacometrics, BioPharmaceuticals R&D, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| | | | - Robert H. Carnahan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James E. Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | - Helen Bright
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Nicole L. Kallewaard
- Virology and Vaccine Discovery, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 20878, USA
| | | | - Mark T. Esser
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| |
Collapse
|
38
|
Abstract
Antibodies have been used to prevent or treat viral infections since the nineteenth century, but the full potential to use passive immunization for infectious diseases has yet to be realized. The advent of efficient methods for isolating broad and potently neutralizing human monoclonal antibodies is enabling us to develop antibodies with unprecedented activities. The discovery of IgG Fc region modifications that extend antibody half-life in humans to three months or more suggests that antibodies could become the principal tool with which we manage future viral epidemics. Antibodies for members of most virus families that cause severe disease in humans have been isolated, and many of them are in clinical development, an area that has accelerated during the effort to prevent or treat COVID-19 (coronavirus disease 2019). Broad and potently neutralizing antibodies are also important research reagents for identification of protective epitopes that can be engineered into active vaccines through structure-based reverse vaccinology. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James E Crowe
- Vanderbilt Vaccine Center, Department of Pediatrics, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| |
Collapse
|
39
|
Zhang Z, Ma P, Ahmed R, Wang J, Akin D, Soto F, Liu BF, Li P, Demirci U. Advanced Point-of-Care Testing Technologies for Human Acute Respiratory Virus Detection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2103646. [PMID: 34623709 DOI: 10.1002/adma.202103646] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/25/2021] [Indexed: 04/14/2023]
Abstract
The ever-growing global threats to human life caused by the human acute respiratory virus (RV) infections have cost billions of lives, created a significant economic burden, and shaped society for centuries. The timely response to emerging RVs could save human lives and reduce the medical care burden. The development of RV detection technologies is essential for potentially preventing RV pandemic and epidemics. However, commonly used detection technologies lack sensitivity, specificity, and speed, thus often failing to provide the rapid turnaround times. To address this problem, new technologies are devised to address the performance inadequacies of the traditional methods. These emerging technologies offer improvements in convenience, speed, flexibility, and portability of point-of-care test (POCT). Herein, recent developments in POCT are comprehensively reviewed for eight typical acute respiratory viruses. This review discusses the challenges and opportunities of various recognition and detection strategies and discusses these according to their detection principles, including nucleic acid amplification, optical POCT, electrochemistry, lateral flow assays, microfluidics, enzyme-linked immunosorbent assays, and microarrays. The importance of limits of detection, throughput, portability, and specificity when testing clinical samples in resource-limited settings is emphasized. Finally, the evaluation of commercial POCT kits for both essential RV diagnosis and clinical-oriented practices is included.
Collapse
Affiliation(s)
- Zhaowei Zhang
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, National Reference Laboratory for Agricultural Testing (Biotoxin), Key Laboratory of Biology and Genetic Improvement of Oil Crops, Key Laboratory of Detection for Mycotoxins, Ministry of Agriculture and Rural Affairs, Wuhan, 430062, P. R. China
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Peng Ma
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Rajib Ahmed
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Jie Wang
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Demir Akin
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Fernando Soto
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Peiwu Li
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, National Reference Laboratory for Agricultural Testing (Biotoxin), Key Laboratory of Biology and Genetic Improvement of Oil Crops, Key Laboratory of Detection for Mycotoxins, Ministry of Agriculture and Rural Affairs, Wuhan, 430062, P. R. China
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| |
Collapse
|
40
|
Grevys A, Frick R, Mester S, Flem-Karlsen K, Nilsen J, Foss S, Sand KMK, Emrich T, Fischer JAA, Greiff V, Sandlie I, Schlothauer T, Andersen JT. Antibody variable sequences have a pronounced effect on cellular transport and plasma half-life. iScience 2022; 25:103746. [PMID: 35118359 PMCID: PMC8800109 DOI: 10.1016/j.isci.2022.103746] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 11/15/2022] Open
Abstract
Monoclonal IgG antibodies are the fastest growing class of biologics, but large differences exist in their plasma half-life in humans. Thus, to design IgG antibodies with favorable pharmacokinetics, it is crucial to identify the determinants of such differences. Here, we demonstrate that the variable region sequences of IgG antibodies greatly affect cellular uptake and subsequent recycling and rescue from intracellular degradation by endothelial cells. When the variable sequences are masked by the cognate antigen, it influences both their transport behavior and binding to the neonatal Fc receptor (FcRn), a key regulator of IgG plasma half-life. Furthermore, we show how charge patch differences in the variable domains modulate both binding and transport properties and that a short plasma half-life, due to unfavorable charge patches, may partly be overcome by Fc-engineering for improved FcRn binding. IgG variable region sequences greatly affect cellular uptake and recycling Variable region charge patches affect FcRn binding and transport The presence of cognate antigen modulates cellular transport and FcRn binding Fc-engineering for improved FcRn binding can overcome unfavorable charge patches
Collapse
Affiliation(s)
- Algirdas Grevys
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
- Corresponding author
| | - Rahel Frick
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Simone Mester
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Karine Flem-Karlsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Jeannette Nilsen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Stian Foss
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Thomas Emrich
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | | | - Victor Greiff
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, 0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Tilman Schlothauer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Corresponding author
| |
Collapse
|
41
|
Bergeron HC, Tripp RA. Breakthrough therapy designation of nirsevimab for the prevention of lower respiratory tract illness caused by respiratory syncytial virus infections (RSV). Expert Opin Investig Drugs 2021; 31:23-29. [PMID: 34937485 DOI: 10.1080/13543784.2022.2020248] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Respiratory syncytial virus (RSV) is a leading cause of serious lower respiratory tract infection (LRTI) in infants and young children. Palivizumab is an RSV-specific prophylactic for use in high-risk infants but treatment requires monthly injections and only modestly reduces hospitalization. Thus, new immunoprophylactic candidates are under development. Nirsevimab (MEDI8897) is a monoclonal antibody with an extended half-life developed to protect infants for an entire RSV season with a single dose. AREAS COVERED This review summarizes clinical trial data on nirsevimab. The authors introduce RSV and surface viral proteins involved in infection, then discuss the development and achievements of nirsevimab in clinical trials concluding with expert opinion. Information was compiled from PubMed, clinicaltrials.gov, and press releases from AstraZeneca and Sanofi. EXPERT OPINION Nirsevimab (MEDI8897) is an RSV F protein monoclonal antibody and the next-generation RSV medicine having an extended half-life developed for the prevention of LRTI caused by RSV. Nirsevimab will supplant the current standard of care for RSV prevention. Importantly, nirsevimab requires a single dose to last the entire RSV season and may be given to term, preterm, and high-risk infants. However, even with nirsevimab approval there remains a need for an efficacious RSV vaccine and treatments.
Collapse
Affiliation(s)
- Harrison C Bergeron
- Department of Infectious Diseases College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Ralph A Tripp
- Department of Infectious Diseases College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
42
|
Maas BM, Lommerse J, Plock N, Railkar RA, Cheung SYA, Caro L, Chen J, Liu W, Zhang Y, Huang Q, Gao W, Qin L, Meng J, Witjes H, Schindler E, Guiastrennec B, Bellanti F, Spellman DS, Roadcap B, Kalinova M, Fok-Seang J, Catchpole AP, Espeseth AS, Stoch SA, Lai E, Vora KA, Aliprantis AO, Sachs JR. Forward and reverse translational approaches to predict efficacy of neutralizing respiratory syncytial virus (RSV) antibody prophylaxis. EBioMedicine 2021; 73:103651. [PMID: 34775220 PMCID: PMC8603022 DOI: 10.1016/j.ebiom.2021.103651] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/29/2021] [Accepted: 10/12/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Neutralizing mAbs can prevent communicable viral diseases. MK-1654 is a respiratory syncytial virus (RSV) F glycoprotein neutralizing monoclonal antibody (mAb) under development to prevent RSV infection in infants. Development and validation of methods to predict efficacious doses of neutralizing antibodies across patient populations exposed to a time-varying force of infection (i.e., seasonal variation) are necessary. METHODS Five decades of clinical trial literature were leveraged to build a model-based meta-analysis (MBMA) describing the relationship between RSV serum neutralizing activity (SNA) and clinical endpoints. The MBMA was validated by backward translation to animal challenge experiments and forward translation to predict results of a recent RSV mAb trial. MBMA predictions were evaluated against a human trial of 70 participants who received either placebo or one of four dose-levels of MK-1654 and were challenged with RSV [NCT04086472]. The MBMA was used to perform clinical trial simulations and predict efficacy of MK-1654 in the infant target population. FINDINGS The MBMA established a quantitative relationship between RSV SNA and clinical endpoints. This relationship was quantitatively consistent with animal model challenge experiments and results of a recently published clinical trial. Additionally, SNA elicited by increasing doses of MK-1654 in humans reduced RSV symptomatic infection rates with a quantitative relationship that approximated the MBMA. The MBMA indicated a high probability that a single dose of ≥ 75 mg of MK-1654 will result in prophylactic efficacy (> 75% for 5 months) in infants. INTERPRETATION An MBMA approach can predict efficacy of neutralizing antibodies against RSV and potentially other respiratory pathogens.
Collapse
Affiliation(s)
- Brian M Maas
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Jos Lommerse
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | - Nele Plock
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | - Radha A Railkar
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - S Y Amy Cheung
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | - Luzelena Caro
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Jingxian Chen
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Wen Liu
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Ying Zhang
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Qinlei Huang
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Wei Gao
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Li Qin
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | - Jie Meng
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | - Han Witjes
- Certara, 100 Overlook Center STE 101, Princeton, NJ 08540, USA
| | | | | | | | - Daniel S Spellman
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Brad Roadcap
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | | | | | | | - Amy S Espeseth
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - S Aubrey Stoch
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Eseng Lai
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | - Kalpit A Vora
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA
| | | | - Jeffrey R Sachs
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033, USA.
| |
Collapse
|
43
|
Shang Z, Tan S, Ma D. Respiratory syncytial virus: from pathogenesis to potential therapeutic strategies. Int J Biol Sci 2021; 17:4073-4091. [PMID: 34671221 PMCID: PMC8495404 DOI: 10.7150/ijbs.64762] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/18/2021] [Indexed: 01/23/2023] Open
Abstract
Respiratory syncytial virus (RSV) is one of the most important viral pathogens causing respiratory tract infection in infants, the elderly and people with poor immune function, which causes a huge disease burden worldwide every year. It has been more than 60 years since RSV was discovered, and the palivizumab monoclonal antibody, the only approved specific treatment, is limited to use for passive immunoprophylaxis in high-risk infants; no other intervention has been approved to date. However, in the past decade, substantial progress has been made in characterizing the structure and function of RSV components, their interactions with host surface molecules, and the host innate and adaptive immune response to infection. In addition, basic and important findings have also piqued widespread interest among researchers and pharmaceutical companies searching for effective interventions for RSV infection. A large number of promising monoclonal antibodies and inhibitors have been screened, and new vaccine candidates have been designed for clinical evaluation. In this review, we first briefly introduce the structural composition, host cell surface receptors and life cycle of RSV virions. Then, we discuss the latest findings related to the pathogenesis of RSV. We also focus on the latest clinical progress in the prevention and treatment of RSV infection through the development of monoclonal antibodies, vaccines and small-molecule inhibitors. Finally, we look forward to the prospects and challenges of future RSV research and clinical intervention.
Collapse
Affiliation(s)
- Zifang Shang
- Institute of Pediatrics, Shenzhen Children's Hospital, 518026 Shenzhen, Guangdong Province, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101Beijing, China
| | - Shuguang Tan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101Beijing, China
| | - Dongli Ma
- Institute of Pediatrics, Shenzhen Children's Hospital, 518026 Shenzhen, Guangdong Province, China
| |
Collapse
|
44
|
Ananworanich J, Heaton PM. Bringing Preventive RSV Monoclonal Antibodies to Infants in Low- and Middle-Income Countries: Challenges and Opportunities. Vaccines (Basel) 2021; 9:961. [PMID: 34579198 PMCID: PMC8473431 DOI: 10.3390/vaccines9090961] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections (LRTIs) in infants. Most deaths occur in infants under 3 months old, and those living in low and middle-income countries (LMICs). There are no maternal or infant RSV vaccines currently approved. An RSV monoclonal antibody (mAb) could fill the gap until vaccines are available. It could also be used when a vaccine is not given, or when there is insufficient time to vaccinate and generate an antibody response. The only currently approved RSV mAb, palivizumab, is too costly and needs monthly administration, which is not possible in LMICs. It is imperative that a safe, effective, and affordable mAb to prevent severe RSV LRTI be developed for infants in LMICs. Next generation, half-life extended mAbs in clinical development, such as nirsevimab, show promise in protecting infants against RSV LRTI. Given that a single dose could cover an entire 5-month season, there is an opportunity to make RSV mAbs affordable for LMICs by investing in improvements in manufacturing efficiency. The challenges of using RSV mAbs in LMICs are the complexities of integrating them into existing healthcare delivery programs and surveillance systems, both of which are needed to define seasonal patterns, and monitor for escape mutants. Collaboration with key stakeholders such as the World Health Organization and Gavi, the Vaccine Alliance, will be essential for achieving this goal.
Collapse
Affiliation(s)
- Jintanat Ananworanich
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA 02139, USA;
- Amsterdam Medical Center, Department of Global Health, University of Amsterdam, 1105 BP Amsterdam, The Netherlands
| | - Penny M. Heaton
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA 02139, USA;
| |
Collapse
|
45
|
Esposito S, Abu-Raya B, Bonanni P, Cahn-Sellem F, Flanagan KL, Martinon Torres F, Mejias A, Nadel S, Safadi MAP, Simon A. Coadministration of Anti-Viral Monoclonal Antibodies With Routine Pediatric Vaccines and Implications for Nirsevimab Use: A White Paper. Front Immunol 2021; 12:708939. [PMID: 34456918 PMCID: PMC8386277 DOI: 10.3389/fimmu.2021.708939] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/15/2021] [Indexed: 02/01/2023] Open
Abstract
Routine childhood vaccinations are key for the protection of children from a variety of serious and potentially fatal diseases. Current pediatric vaccine schedules mainly cover active vaccines. Active vaccination in infants is a highly effective approach against several infectious diseases; however, thus far, for some important viral pathogens, including respiratory syncytial virus (RSV), vaccine development and license by healthcare authorities have not been accomplished. Nirsevimab is a human-derived, highly potent monoclonal antibody (mAb) with an extended half-life for RSV prophylaxis in all infants. In this manuscript, we consider the potential implications for the introduction of an anti-viral mAb, such as nirsevimab, into the routine pediatric vaccine schedule, as well as considerations for coadministration. Specifically, we present evidence on the general mechanism of action of anti-viral mAbs and experience with palivizumab, the only approved mAb for the prevention of RSV infection in preterm infants, infants with chronic lung disease of prematurity and certain infants with hemodynamically significant heart disease. Palivizumab has been used for over two decades in infants who also receive routine vaccinations without any alerts concerning the safety and efficacy of coadministration. Immunization guidelines (Advisory Committee on Immunization Practices, Joint Committee on Vaccination and Immunization, National Advisory Committee on Immunization, Centers for Disease Control and Prevention, American Academy of Pediatrics, The Association of the Scientific Medical Societies in Germany) support coadministration of palivizumab with routine pediatric vaccines, noting that immunobiologics, such as palivizumab, do not interfere with the immune response to licensed live or inactivated active vaccines. Based on the mechanism of action of the new generation of anti-viral mAbs, such as nirsevimab, which is highly specific targeting viral antigenic sites, it is unlikely that it could interfere with the immune response to other vaccines. Taken together, we anticipate that nirsevimab could be concomitantly administered to infants with routine pediatric vaccines during the same clinic visit.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Bahaa Abu-Raya
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Paolo Bonanni
- Specialization Medical School of Hygiene, Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Katie L. Flanagan
- Tasmanian Vaccine Trial Centre, Launceston General Hospital, Launceston, TAS, Australia
- School of Medicine, University of Tasmania, Launceston, TAS, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC, Australia
| | - Federico Martinon Torres
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- Genetics, Vaccines and Pediatrics Research Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Universidad de Santiago, Santiago de Compostela, Spain
| | - Asuncion Mejias
- Division of Infectious Diseases, Department of Pediatrics, Center for Vaccines and Immunity Nationwide Children’s Hospital-The Ohio State University College of Medicine, Columbus, OH, United States
- Department of Pharmacology and Pediatrics, Malaga Medical School, Malaga University, Malaga, Spain
| | | | - Marco A. P. Safadi
- Department of Pediatrics, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Arne Simon
- Klinik für Pädiatrische Onkologie und Hämatologie Universitätsklinikum des Saarlandes, Homburg, Germany
| |
Collapse
|
46
|
Whaley KJ, Zeitlin L. Emerging antibody-based products for infectious diseases: Planning for metric ton manufacturing. Hum Vaccin Immunother 2021; 18:1930847. [PMID: 34259613 PMCID: PMC9103258 DOI: 10.1080/21645515.2021.1930847] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
This review focuses on the emerging monoclonal antibody market for infectious diseases and the metric ton scale manufacturing requirements to meet global demand. Increasing access to existing antibody-based products coupled with the unmet need in infectious disease will likely exceed the current existing global manufacturing capacity. Further, the large numbers of individuals infected during epidemics such as the ongoing COVID-19 pandemic emphasizes the need to plan for metric ton manufacturing of monoclonal antibodies by expanding infrastructure and exploring alternative production systems.
Collapse
Affiliation(s)
- Kevin J Whaley
- Mapp Biopharmaceutical, San Diego, CA, USA.,ZabBio, San Diego, CA, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, San Diego, CA, USA.,ZabBio, San Diego, CA, USA
| |
Collapse
|
47
|
Narkhede YB, Gonzalez KJ, Strauch EM. Targeting Viral Surface Proteins through Structure-Based Design. Viruses 2021; 13:v13071320. [PMID: 34372526 PMCID: PMC8310314 DOI: 10.3390/v13071320] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/28/2022] Open
Abstract
The emergence of novel viral infections of zoonotic origin and mutations of existing human pathogenic viruses represent a serious concern for public health. It warrants the establishment of better interventions and protective therapies to combat the virus and prevent its spread. Surface glycoproteins catalyzing the fusion of viral particles and host cells have proven to be an excellent target for antivirals as well as vaccines. This review focuses on recent advances for computational structure-based design of antivirals and vaccines targeting viral fusion machinery to control seasonal and emerging respiratory viruses.
Collapse
Affiliation(s)
- Yogesh B Narkhede
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA;
| | - Karen J Gonzalez
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA;
| | - Eva-Maria Strauch
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA;
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA;
- Correspondence:
| |
Collapse
|
48
|
The burden of Respiratory Syncytial Virus (RSV) infection in the Middle East and North Africa (MENA) region across age groups: A systematic review. Vaccine 2021; 39:3803-3813. [PMID: 34099329 DOI: 10.1016/j.vaccine.2021.05.076] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/02/2021] [Accepted: 05/21/2021] [Indexed: 11/22/2022]
Abstract
Respiratory Syncytial Virus (RSV) is a common respiratory virus that generally causes a mild illness in children and adults or severe symptoms with complications in infants and the elderly, particularly in the presence of underlying comorbidities. While epidemiological data about this virus are available globally, data from the Middle East and North Africa (MENA) region are still scarce. For this reason, we conducted a systematic review to determine the burden of RSV disease in the MENA region by searching the available literature up until September 2018. A total of 1242 studies were retrieved of which 90 were included in the review. Most of the included studies were conducted in subjects aged 0-18 years with the majority being in children below 3 years of age, while only 2 studies included exclusively adults above 18 years of age. RSV infection rates varied greatly between different studies on hospitalized subjects and ranged between 4% and 82%, while the range was smaller in studies on outpatient subjects (between 6% and 36%). When calculating the RSV infection rates in the hospitalized subjects with different inclusion criteria, we found that it was 19%, 70%, and 33% among subjects admitted with Acute Respiratory Infections (ARIs), Acute Lower Respiratory Infections (ALRIs), and bronchiolitis, respectively. RSV infections were most common during the winter season. With regards to complications, intensive care unit admissions ranged between 1% and 15%, while the need for mechanical ventilation ranged between 1% and 10%. The overall RSV related mortality rate across all age groups in studies included in our review was 1.9%. This review identifies several limitations in the existing data and under-representation of the adult population. Future studies should be providing more evidence on the RSV burden in adults and children with comorbidities in order to better assess the potential impact of future preventive strategies in the MENA region.
Collapse
|
49
|
Kumar D, Gauthami S, Bayry J, Kaveri SV, Hegde NR. Antibody Therapy: From Diphtheria to Cancer, COVID-19, and Beyond. Monoclon Antib Immunodiagn Immunother 2021; 40:36-49. [PMID: 33900819 DOI: 10.1089/mab.2021.0004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The dawn of the 20th century saw the formative years of developments in immunology. In particular, immunochemistry, specifically pertaining to antibodies, was extensively studied. These studies laid the foundations for employing antibodies in a variety of ways. Not surprisingly, antibodies have been used for applications ranging from biomedical research to disease diagnostics and therapeutics to evaluation of immune responses during natural infection and those elicited by vaccines. Despite recent advancements in cellular immunology and the excitement of T cell therapy, use of antibodies represents a large proportion of immunotherapeutic approaches as well as clinical interventions. Polyclonal antibodies in the form of plasma or sera continue to be used to treat a number of diseases, including autoimmune disorders, cancers, and infectious diseases. Historically, antisera to toxins have been the longest serving biotherapeutics. In addition, intravenous immunoglobulins (IVIg) have been extensively used to treat not only immunodeficiency conditions but also autoimmune disorders. Beyond the simplistic suppositions of their action, the IVIg have also unraveled the immune regulatory and homeostatic ramifications of their use. The advent of monoclonal antibodies (MAbs), on the other hand, has provided a clear pathway for their development as drug molecules. MAbs have found a clear place in the treatment of cancers and extending lives and have been used in a variety of other conditions. In this review, we capture the important developments in the therapeutic applications of antibodies to alleviate disease, with a focus on some of the recent developments.
Collapse
Affiliation(s)
| | - Sulgey Gauthami
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Indian Institute of Technology Palakkad, Palakkad, Kerala, India
| | - Srinivas V Kaveri
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Centre National de la Recherche Scientifique (CNRS) Bureau India, IFI, New Delhi, India
| | | |
Collapse
|
50
|
Rodriguez-Fernandez R, Mejias A, Ramilo O. Monoclonal Antibodies for Prevention of Respiratory Syncytial Virus Infection. Pediatr Infect Dis J 2021; 40:S35-S39. [PMID: 34042909 DOI: 10.1097/inf.0000000000003121] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of hospitalizations in infants worldwide. Palivizumab, a humanized monoclonal antibody against the RSV F protein, is the only licensed agent for prevention of severe RSV infection in high-risk infants. Palivizumab is administered intramuscularly, every month during the RSV season, usually 5 doses are required. In recent years, the resolution of the structure of the RSV F protein, with identification of potent neutralizing epitopes, and new technologies for production of monoclonal antibodies (mAbs) have facilitated the development of new alternative strategies for the prevention of RSV infections. One promising approach is a new generation of mAbs directed to new neutralizing epitopes and with prolonged half life. These enhanced mAbs are expected to provide adequate protection during the complete RSV season with a single intramuscular (IM) dose. The long-term goal of this approach is to provide passive immunization for the prevention of RSV lower respiratory tract infection to all infants (preterm and full term) in the first months of life before their initial exposure to RSV.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/history
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Clinical Trials as Topic
- Epitopes/drug effects
- Half-Life
- History, 20th Century
- History, 21st Century
- Humans
- Immunization, Passive/methods
- Infant
- Infant, Newborn
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus, Human/drug effects
- Viral Fusion Proteins
Collapse
Affiliation(s)
- Rosa Rodriguez-Fernandez
- From the Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, Hospital Infantil Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón Madrid, Spain
| | - Asuncion Mejias
- From the Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Octavio Ramilo
- From the Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|