1
|
Kuitunen I, Backman K, Gärdström E, Renko M. Monoclonal antibody therapies in respiratory syncytial virus prophylaxis-An umbrella review. Pediatr Pulmonol 2024; 59:2374-2380. [PMID: 38751021 DOI: 10.1002/ppul.27041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/19/2024] [Indexed: 11/18/2024]
Abstract
The aim was to gather an overview of the evidence of monoclonal antibody therapies in respiratory syncytial virus (RSV) prophylaxis in children. For this umbrella review of previous meta-analyses, we searched PubMed, Scopus, and Web of Science databases in August 2023. We included systematic reviews with meta-analyses of randomized controlled trials. One author extracted the data, and another author validated the data. We extracted all analyses focusing on clinical outcomes and comparing prophylaxis to placebo. Risk of bias in systematic reviews (ROBIS) tool was used to analyze the risk of bias in the included reviews. AMSTAR-2 was used to evaluate the quality of the included reviews. After screening of 323 abstracts and 22 full reports, a total of seven systematic reviews with meta-analyses were included. The risk of bias was rated to be low in four and high in three of these. Three reviews were of moderate quality, one low, and three very low quality according to AMSTAR-2 classification. Six of the meta-analyses were direct comparisons, and one was a network meta-analysis. Five of the reviews focused on palivizumab, one in nirsevimab, and one included all monoclonal antibodies. Six reviews focused on preterm neonates and consisted of 42 comparisons to placebo of which 21 showed efficacy, and 21 had no evidence of a difference. The positive effect was seen in all reviews against RSV infection and RSV hospitalization in palivizumab and nirsevimab groups compared to placebo. None of the mortality comparisons showed efficacy. One meta-analysis focused on cystic fibrosis patients and had eight comparisons, but it was underpowered to detect any results. The overall evidence from previous meta-analyses shows that monoclonal antibodies are effective in preventing RSV infections and RSV hospitalizations in preterm infants. However, no effect on mortality was seen in these studies. Evidence certainty has been ranked mainly from low to moderate.
Collapse
Affiliation(s)
- Ilari Kuitunen
- Institute of Clinical Medicine and Department of Pediatrics, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Department of Pediatrics, Kuopio, Finland
| | - Katri Backman
- Institute of Clinical Medicine and Department of Pediatrics, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Department of Pediatrics, Kuopio, Finland
| | | | - Marjo Renko
- Institute of Clinical Medicine and Department of Pediatrics, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Department of Pediatrics, Kuopio, Finland
| |
Collapse
|
2
|
Jones AL, Campbell MJ, Abernathy B, Neubert S, Hager A, Collier H, Ramsey EZ, Simon A, Schachtner S, Natarajan S. Improvement in Palivizumab Administration Prior to Discharge for Hospitalized Infants with Hemodynamically Significant Congenital Heart Disease: A Quality Improvement Initiative. Pediatr Cardiol 2024; 45:1415-1423. [PMID: 37145121 PMCID: PMC10625646 DOI: 10.1007/s00246-023-03163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 05/06/2023]
Abstract
In this quality improvement initiative, we aimed to increase provider adherence with palivizumab administration guidelines for hospitalized infants with hemodynamically significant congenital heart disease. We included 470 infants over four respiratory syncytial virus (RSV) seasons from 11/2017 to 03/2021 (baseline season: 11/2017-03/2018). Interventions included the following: education, including palivizumab in the sign-out template, identifying a pharmacy expert, and a text alert (seasons 1 and 2: 11/2018-03/2020) that was replaced by an electronic health record (EHR) best practice alert (BPA) in season 3 (11/2020-03/2021). The text alert and BPA prompted providers to add "Need for RSV immunoprophylaxis" to the EHR problem list. The outcome metric was the percentage of eligible patients administered palivizumab prior to discharge. The process metric was the percentage of eligible patients with "Need for RSV immunoprophylaxis" on the EHR problem list. The balancing metric was the percentage of palivizumab doses administered to ineligible patients. A statistical process control P-chart was used to analyze the outcome metric. The mean percentage of eligible patients who received palivizumab prior to hospital discharge increased significantly from 70.1% (82/117) to 90.0% (86/96) in season 1 and to 97.9% (140/143) in season 3. Palivizumab guideline adherence was as high or higher for those with "Need for RSV immunoprophylaxis" on the problem list than for those without it in most time periods. The percentage of inappropriate palivizumab doses decreased from 5.7% (n = 5) at baseline to 4.4% (n = 4) in season 1 and 0.0% (n = 0) in season 3. Through this initiative, we improved adherence with palivizumab administration guidelines for eligible infants prior to hospital discharge.
Collapse
Affiliation(s)
- Andrea L Jones
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelmen School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| | - Matthew J Campbell
- Division of Cardiology, Department of Cardiovascular Medicine, Nemours Children's Hospital, Wilmington, DE, 19803, USA
| | | | - Stephanie Neubert
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelmen School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Alyssa Hager
- Department of Pharmacy Services, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Hailey Collier
- Department of Pharmacy Services, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Evan Zachary Ramsey
- Department of Pharmacy Services, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Anna Simon
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelmen School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Susan Schachtner
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelmen School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Shobha Natarajan
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelmen School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Bonneux B, Jacoby E, Ceconi M, Stobbelaar K, Delputte P, Herschke F. Direct-acting antivirals for RSV treatment, a review. Antiviral Res 2024; 229:105948. [PMID: 38972604 DOI: 10.1016/j.antiviral.2024.105948] [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: 04/19/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024]
Abstract
Respiratory syncytial virus (RSV) causes respiratory disease and complications in infants, the elderly and the immunocompromised. While three vaccines and two prophylactic monoclonal antibodies are now available, only one antiviral, ribavirin, is currently approved for treatment. This review aims to summarize the current state of treatments directly targeting RSV. Two major viral processes are attractive for RSV-specific antiviral drug discovery and development as they play essential roles in the viral cycle: the entry/fusion process carried out by the fusion protein and the replication/transcription process carried out by the polymerase complex constituted of the L, P, N and M2-1 proteins. For each viral target resistance mutations to small molecules of different chemotypes seem to delineate definite binding pockets in the fusion proteins and in the large proteins. Elucidating the mechanism of action of these inhibitors thus helps to understand how the fusion and polymerase complexes execute their functions. While many inhibitors have been studied, few are currently in clinical development for RSV treatment: one is in phase III, three in phase II and two in phase I. Progression was halted for many others because of strategic decisions, low enrollment, safety, but also lack of efficacy. Lessons can be learnt from the halted programs to increase the success rate of the treatments currently in development.
Collapse
Affiliation(s)
- Brecht Bonneux
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium; Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Edgar Jacoby
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Martina Ceconi
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium
| | - Kim Stobbelaar
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610, Wilrijk, Belgium.
| | | |
Collapse
|
4
|
Alharbi AS, Al-Hindi MY, Alqwaiee M, Al-Shamrani A, Alharbi S, Yousef A, Alshammary A, Miqdad A, Said Y, Alnemri A, Alahmadi T, Almudeer AH. Saudi Initiative of Bronchiolitis Diagnosis, Management, and Prevention 2024 updated consensus on the prevention of respiratory syncytial virus. Ann Thorac Med 2024; 19:190-200. [PMID: 39144535 PMCID: PMC11321529 DOI: 10.4103/atm.atm_69_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 08/16/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of bronchiolitis among children under 5 years of age worldwide, accounting for a prevalence of 25%-88% in Saudi Arabia. Although no effective treatment for the virus exists, passive immunoprophylaxis reduced RSV hospitalizations in high-risk children. With recent advances in immunization, the Saudi Initiative of Bronchiolitis Diagnosis, Management, and Prevention panel screened recent relevant international guidelines, locally published data, and expert consensus to update guidelines for RSV prevention, taking into consideration the resources, timing, varying health profiles, and RSV burden in Saudi Arabia. The panel updated its recommendations to include immunization of infants, mothers, and older adults. Practical guidelines were prepared to facilitate the administration of the short-acting and newly developed long-acting RSV monoclonal antibodies (mAb) during the regular follow-ups of high-risk infants in specialized clinics. In addition, long-acting mAb was highlighted as all-infant protection in the routine immunization calendar.
Collapse
Affiliation(s)
- Adel S. Alharbi
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Mohammed Y Al-Hindi
- Department of Pediatric, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
- Research and Development, King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- Department of Pediatrics, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Mansour Alqwaiee
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Abdullah Al-Shamrani
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Saleh Alharbi
- Department of Pediatrics, Umm Al Qura University, Makkah, Saudi Arabia
| | - Abdullah Yousef
- Department of Pediatrics, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Aisha Alshammary
- Department of Pediatrics, Alyammamah Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Abeer Miqdad
- Department of Pediatrics, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Yazan Said
- Department of Pediatrics, King Fahad Specialist Hospital, Ministry of Health, Dammam, Saudi Arabia
| | - Abdulrahman Alnemri
- Department of Pediatrics, College of Medicine, Pediatrics Department, King Saud University, Riyadh, Saudi Arabia
| | - Turki Alahmadi
- Department of Pediatrics, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ali Husein Almudeer
- Department of Pediatrics, College of Medicine, Pediatrics Department, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Bonneux B, Shareef A, Tcherniuk S, Anson B, de Bruyn S, Verheyen N, Thys K, Conceição-Neto N, Van Ginderen M, Kwanten L, Ysebaert N, Vranckx L, Peeters E, Lanckacker E, Gallup JM, Sitthicharoenchai P, Alnajjar S, Ackermann MR, Adhikary S, Bhaumik A, Patrick A, Fung A, Sutto-Ortiz P, Decroly E, Mason SW, Lançois D, Deval J, Jin Z, Eléouët JF, Fearns R, Koul A, Roymans D, Rigaux P, Herschke F. JNJ-7184, a respiratory syncytial virus inhibitor targeting the connector domain of the viral polymerase. Antiviral Res 2024; 227:105907. [PMID: 38772503 DOI: 10.1016/j.antiviral.2024.105907] [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: 04/07/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 05/23/2024]
Abstract
Respiratory syncytial virus (RSV) can cause pulmonary complications in infants, elderly and immunocompromised patients. While two vaccines and two prophylactic monoclonal antibodies are now available, treatment options are still needed. JNJ-7184 is a non-nucleoside inhibitor of the RSV-Large (L) polymerase, displaying potent inhibition of both RSV-A and -B strains. Resistance selection and hydrogen-deuterium exchange experiments suggest JNJ-7184 binds RSV-L in the connector domain. JNJ-7184 prevents RSV replication and transcription by inhibiting initiation or early elongation. JNJ-7184 is effective in air-liquid interface cultures and therapeutically in neonatal lambs, acting to drastically reverse the appearance of lung pathology.
Collapse
Affiliation(s)
- Brecht Bonneux
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsbaan 1, 2610 Wilrijk, Belgium; Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Afzaal Shareef
- Department of Virology, Immunology & Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Sergey Tcherniuk
- Unité de Virologie et Immunologie Moléculaires (VIM, UMR892), INRAE, Université Paris-Saclay, 78352 Jouy-en-Josas, France
| | - Brandon Anson
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Suzanne de Bruyn
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Nick Verheyen
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Kim Thys
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | | | - Leen Kwanten
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Nina Ysebaert
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Luc Vranckx
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Elien Peeters
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Ellen Lanckacker
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | | | | | | | - Suraj Adhikary
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Anusarka Bhaumik
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Aaron Patrick
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Amy Fung
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Priscila Sutto-Ortiz
- AFMB, Aix-Marseille University, CNRS UMR 7257, 163 Avenue de Luminy, Marseille, France
| | - Etienne Decroly
- AFMB, Aix-Marseille University, CNRS UMR 7257, 163 Avenue de Luminy, Marseille, France
| | - Stephen W Mason
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | | | - Jerome Deval
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Zhinan Jin
- Janssen Research & Development LLC, Spring House (PA 19477) And Brisbane (CA 94005), USA
| | - Jean-François Eléouët
- Unité de Virologie et Immunologie Moléculaires (VIM, UMR892), INRAE, Université Paris-Saclay, 78352 Jouy-en-Josas, France
| | - Rachel Fearns
- Department of Virology, Immunology & Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Anil Koul
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium.
| | - Dirk Roymans
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Peter Rigaux
- Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | |
Collapse
|
6
|
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
|
7
|
Oraby A, Bilawchuk L, West FG, Marchant DJ. Structure-Based Discovery of Allosteric Inhibitors Targeting a New Druggable Site in the Respiratory Syncytial Virus Polymerase. ACS OMEGA 2024; 9:22213-22229. [PMID: 38799318 PMCID: PMC11112712 DOI: 10.1021/acsomega.4c01207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 05/29/2024]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory infections for which effective treatment options remain limited. Herein, we employed a computational structure-based design strategy aimed at identifying potential targets for a new class of allosteric inhibitors. Our investigation led to the discovery of a previously undisclosed allosteric binding site within the RSV polymerase, the large (L) protein. This discovery was achieved through a combination of virtual screening and molecular dynamics simulations. Subsequently, we identified two inhibitors, 6a and 10b, which both exhibited promising antiviral activity in the low micromolar range. Resistance profiling revealed a distinctive pattern in how RSV evaded treatment with this class of inhibitors. This pattern strongly suggested that this class of small molecules was targeting a new binding site in the RSV L protein, aligning with the computational predictions made in our study. This study paves the way for the development of more potent inhibitors for combating RSV infections by targeting a new druggable pocket within the RdRp which does not overlap with previously known resistance sites.
Collapse
Affiliation(s)
- Ahmed
K. Oraby
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department
of Pharmaceutical Organic Chemistry, College of Pharmaceutical Sciences
and Drug Manufacturing, Misr University
for Science and Technology, 6th
of October City P.O. Box 77,Egypt
| | - Leanne Bilawchuk
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Frederick G. West
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - David J. Marchant
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
8
|
Gonzales T, Bergamasco A, Cristarella T, Goyer C, Wojdyla M, Oladapo A, Sawicky J, Yee J, Moride Y. Effectiveness and Safety of Palivizumab for the Prevention of Serious Lower Respiratory Tract Infection Caused by Respiratory Syncytial Virus: A Systematic Review. Am J Perinatol 2024; 41:e1107-e1115. [PMID: 36452969 PMCID: PMC11108679 DOI: 10.1055/a-1990-2633] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022]
Abstract
OBJECTIVE Palivizumab is a humanized monoclonal antibody approved for the prevention of serious lower respiratory tract infection (LRTI) caused by respiratory syncytial virus (RSV) in infants and young children at high risk of RSV disease. This systematic review summarized evidence on the effectiveness and safety of palivizumab when used in approved populations. STUDY DESIGN A systematic review of Phase III trials and observational studies was conducted according to the population, intervention, comparator, outcome, timing, setting (PICOTS) approach (PROSPERO, CRD42021281380). Target populations consisted of infants with a history of premature birth (≤35-week gestational age) and children aged <2 years with bronchopulmonary dysplasia (BPD) or with hemodynamically significant congenital heart disease (hs-CHD). Outcomes of interest included RSV-related hospitalization, admission to intensive care unit (ICU), requirement for mechanical ventilation, treatment-related adverse events (AEs), and RSV-related deaths. Information sources were literature search (Ovid MEDLINE and Embase), pragmatic searches, and snowballing (covering the period up to 07 September 2021). RESULTS A total of 60 sources were included (5 Phase III trials and 55 observational studies). RSV-related hospitalization rates following palivizumab prophylaxis in Phase III trials were 1.8% in premature infants and 7.9% in children with BPD, which were significantly lower than rates in placebo arms. In the real-world setting, similar hospitalization rates were found (0.7-4.0% in premature infants [16 studies] and 0-5.5% in patients with BPD [10 studies]) with ICU admission reported in 0 to 33.3% of patients hospitalized for RSV. In Phase III trials, RSV-related mortality rates were 0.2 and 0.3%, while AEs occurred in 11% of premature and/or BPD patients and 7.2% of hs-CHD patients, consisting mainly of injection site reaction, fever, and diarrhea. Similar results were found in observational studies. CONCLUSION This systematic review supports the effectiveness and safety of palivizumab in the indicated populations. KEY POINTS · Systematic review supports the positive benefit-risk profile of palivizumab in the indicated populations.. · Real-world safety and effectiveness of palivizumab are consistent with Phase III trials results.. · Palivizumab reduces RSV-related hospitalizations, ICU admissions, and need for mechanical ventilation..
Collapse
Affiliation(s)
- Tara Gonzales
- SOBI Inc, Specialty Care North America, Waltham, Massachusetts
| | | | | | - Camille Goyer
- YOLARX Consultants Inc, 3550 Côte-des-Neiges Road, Montréal, QC, Canada
| | - Matthew Wojdyla
- SOBI Inc, Specialty Care North America, Waltham, Massachusetts
| | - Abiola Oladapo
- SOBI Inc, Specialty Care North America, Waltham, Massachusetts
| | - John Sawicky
- SOBI Inc, Specialty Care North America, Waltham, Massachusetts
| | - John Yee
- SOBI Inc, Specialty Care North America, Waltham, Massachusetts
| | - Yola Moride
- YOLARX Consultants SAS, 101, rue de Sèvres, Paris Cedex 6, France
| |
Collapse
|
9
|
Ordóñez JE, Huertas VM. Cost-utility analysis of palivizumab for preventing respiratory syncytial virus in preterm neonates and infants in Colombia. BMC Infect Dis 2024; 24:418. [PMID: 38641577 PMCID: PMC11031882 DOI: 10.1186/s12879-024-09300-5] [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: 06/18/2023] [Accepted: 04/07/2024] [Indexed: 04/21/2024] Open
Abstract
AIM Palivizumab has proven effective in reducing hospitalizations, preventing severe illness, improving health outcomes, and reducing healthcare costs for infants at risk of respiratory syncytial virus (RSV) infection. We aim to assess the value of palivizumab in preventing RSV infection in high-risk infants in Colombia, where RSV poses a significant threat, causing severe respiratory illness and hospitalizations. METHODS We conducted a decision tree analysis to compare five doses of palivizumab with no palivizumab. The study considered three population groups: preterm neonates (≤ 35 weeks gestational age), infants with bronchopulmonary dysplasia (BPD), and infants with hemodynamically significant congenital heart disease (CHD). We obtained clinical efficacy data from IMpact-RSV and Cardiac Synagis trials, while we derived neonatal hospitalization risks from the SENTINEL-1 study. We based hospitalization and recurrent wheezing management costs on Colombian analyses and validated them by experts. We estimated incremental cost-effectiveness ratios and performed 1,000 Monte Carlo simulations for probabilistic sensitivity analyses. RESULTS Palivizumab is a dominant strategy for preventing RSV infection in preterm neonates and infants with BPD and CHD. Its high efficacy (78% in preventing RSV in preterm infants), the substantial risk of illness and hospitalization, and the high costs associated with hospitalization, particularly in neonatal intensive care settings, support this finding. The scatter plots and willingness-to-pay curves align with these results. CONCLUSION Palivizumab is a cost-saving strategy in Colombia, effectively preventing RSV infection in preterm neonates and infants with BPD and CHD by reducing hospitalizations and lowering healthcare costs.
Collapse
|
10
|
Sanders SL, Agwan S, Hassan M, Bont LJ, Venekamp RP. Immunoglobulin treatment for hospitalised infants and young children with respiratory syncytial virus infection. Cochrane Database Syst Rev 2023; 10:CD009417. [PMID: 37870128 PMCID: PMC10591280 DOI: 10.1002/14651858.cd009417.pub3] [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] [Indexed: 10/24/2023]
Abstract
BACKGROUND Millions of children are hospitalised due to respiratory syncytial virus (RSV) infection every year. Treatment is supportive, and current therapies (e.g. inhaled bronchodilators, epinephrine, nebulised hypertonic saline, and corticosteroids) are ineffective or have limited effect. Respiratory syncytial virus immunoglobulin may be used prophylactically to prevent hospital admission from RSV-related illness. It may be considered for the treatment of established severe RSV infection or for treatment in an immunocompromised host, although it is not licensed for this purpose. It is unclear whether immunoglobulins improve outcomes when used as a treatment for established RSV infection in infants and young children admitted to hospital. This is an update of a review first published in 2019. OBJECTIVES To assess the effects of immunoglobulins for the treatment of RSV-proven lower respiratory tract infections (LRTIs) in children aged up to three years, admitted to hospital. SEARCH METHODS For this 2022 update, we searched the Cochrane Central Register of Controlled Trials (CENTRAL), which contains the Cochrane Acute Respiratory Infections Specialised Register, Ovid MEDLINE, Embase, CINAHL, and Web of Science (from inception to 2 December 2022) with no restrictions. We searched two trial registries for ongoing trials (to 2 December 2022) and checked the reference lists of reviews and included articles for additional studies. SELECTION CRITERIA Randomised controlled trials comparing immunoglobulins with placebo in hospitalised infants and children aged up to three years with laboratory-diagnosed RSV lower respiratory tract infection. DATA COLLECTION AND ANALYSIS Two review authors independently selected trials, assessed risk of bias, and extracted data. We assessed evidence certainty using GRADE. MAIN RESULTS In total, we included eight trials involving 906 infants and children aged up to three years. We included one new trial in this update. The immunoglobulin preparations used in these trials included anti-RSV immunoglobulin and the monoclonal antibody preparations palivizumab and motavizumab. Five trials were conducted at single or multiple sites within a single high-income country (four in the USA, one in Qatar). Three trials included study sites in different countries. All three of these trials included study sites in one or more high-income countries (USA, Chile, New Zealand, Australia, Qatar), with two trials also including a study site in a middle-income country (Panama). Five of the eight trials were "supported" or "sponsored" by the trial drug manufacturers. The evidence is very uncertain about the effect of immunoglobulins on mortality (risk ratio (RR) 0.87, 95% confidence interval (CI) 0.14 to 5.27; 4 studies, 309 participants). There were four deaths - two amongst 98 children receiving immunoglobulins, and two amongst 98 children receiving placebo. One additional death occurred in a fourth trial, however the study group of the child was not known and the data were not included in the analysis (very low-certainty evidence). The use of immunoglobulins in infants and children admitted to hospital with RSV proven LRTI probably results in little to no difference in the length of hospitalisation (mean difference (MD) -0.13 days, 95% CI -0.37 to 0.12; 6 studies, 737 participants; moderate-certainty evidence). Immunoglobulins may result in little to no difference in the number of children who experience one or more adverse events of any severity or seriousness compared to placebo (RR 1.18, 95% CI 0.78 to 1.78; 5 studies, 340 participants; low-certainty evidence) or the number of children who experience one or more adverse events judged by study investigators to be serious in nature, compared to placebo (RR 1.08, 95% CI 0.65 to 1.79; 4 studies, 238 participants; low-certainty evidence). Certainty of evidence for secondary outcomes was low. This evidence suggests that use of immunoglobulins results in little to no difference in the need for, or duration of, mechanical ventilation and the need for, or duration of, supplemental oxygen. The use of immunoglobulins does not reduce the need for admission to the intensive care unit (ICU) and when children are admitted to the ICU results in little to no difference in the duration of ICU stay. AUTHORS' CONCLUSIONS We are very uncertain about the effect of immunoglobulins on mortality. We are moderately certain that use of immunoglobulins in hospitalised infants and children may result in little to no difference in the length of hospitalisation. Immunoglobulins may result in little to no difference in adverse events, the need for or duration of mechanical ventilation, supplemental oxygen, or admission to the intensive care unit, though we are less certain about this evidence and the true effect of immunoglobulins on these outcomes may differ markedly from the estimated effect observed in this review. All trials were conducted in high-income countries, and data from populations in which the rate of death from RSV infection is higher are lacking.
Collapse
Affiliation(s)
- Sharon L Sanders
- Institute for Evidence-Based Healthcare, Bond University, Gold Coast, Australia
| | - Sushil Agwan
- Gold Coast University Hospital, Gold Coast, Australia
| | | | - Louis J Bont
- Department of Pediatrics, Wilhelmina Childrens Hospital, Utrecht, Netherlands
| | - Roderick P Venekamp
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
11
|
Umemoto S, Nakahashi-Ouchida R, Yuki Y, Kurokawa S, Machita T, Uchida Y, Mori H, Yamanoue T, Shibata T, Sawada SI, Ishige K, Hirano T, Fujihashi K, Akiyoshi K, Kurashima Y, Tokuhara D, Ernst PB, Suzuki M, Kiyono H. Cationic-nanogel nasal vaccine containing the ectodomain of RSV-small hydrophobic protein induces protective immunity in rodents. NPJ Vaccines 2023; 8:106. [PMID: 37488116 PMCID: PMC10366164 DOI: 10.1038/s41541-023-00700-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of upper and lower respiratory tract infection, especially in children and the elderly. Various vaccines containing the major transmembrane surface proteins of RSV (proteins F and G) have been tested; however, they have either afforded inadequate protection or are associated with the risk of vaccine-enhanced disease (VED). Recently, F protein-based maternal immunization and vaccines for elderly patients have shown promising results in phase III clinical trials, however, these vaccines have been administered by injection. Here, we examined the potential of using the ectodomain of small hydrophobic protein (SHe), also an RSV transmembrane surface protein, as a nasal vaccine antigen. A vaccine was formulated using our previously developed cationic cholesteryl-group-bearing pullulan nanogel as the delivery system, and SHe was linked in triplicate to pneumococcal surface protein A as a carrier protein. Nasal immunization of mice and cotton rats induced both SHe-specific serum IgG and mucosal IgA antibodies, preventing viral invasion in both the upper and lower respiratory tracts without inducing VED. Moreover, nasal immunization induced greater protective immunity against RSV in the upper respiratory tract than did systemic immunization, suggesting a critical role for mucosal RSV-specific IgA responses in viral elimination at the airway epithelium. Thus, our nasal vaccine induced effective protection against RSV infection in the airway mucosa and is therefore a promising vaccine candidate for further development.
Collapse
Affiliation(s)
- Shingo Umemoto
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
| | - Rika Nakahashi-Ouchida
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Yoshikazu Yuki
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- HanaVax Inc, Tokyo, Japan
| | - Shiho Kurokawa
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Tomonori Machita
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Yohei Uchida
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Hiromi Mori
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Tomoyuki Yamanoue
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Takehiko Shibata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kazuya Ishige
- Biochemicals Division, Yamasa Corporation, Chiba, Japan
| | - Takashi Hirano
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yosuke Kurashima
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Daisuke Tokuhara
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Peter B Ernst
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, CA, USA
- Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan
| | - Masashi Suzuki
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA.
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan.
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan.
- HanaVax Inc, Tokyo, Japan.
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan.
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan.
| |
Collapse
|
12
|
Ebersjö C, Berggren Broström E, Kull I, Lindholm Olinder A. Home Immunization with Palivizumab-A Randomized Pilot Study Describing Safety Aspects and Parents' Preferences. CHILDREN (BASEL, SWITZERLAND) 2023; 10:198. [PMID: 36832327 PMCID: PMC9955059 DOI: 10.3390/children10020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Among prematurely born infants and newborns with chronic conditions, a respiratory syncytial virus (RSV) infection may cause (re-)admission and later respiratory complications. Therapeutic protection is possible with monthly injections of a specific monoclonal antibody, palivizumab, during RSV season. Standard care is giving up to five injections in clinic-based settings. Immunization at home could be an alternative to standard care for vulnerable infants to reduce the number of revisits and associated risk of RSV infection. The aim of this randomized pilot trial was to evaluate safety aspects and explore parents' preferences of home versus hospital immunization with palivizumab during one RSV season. Immediate adverse events (AEs) were observed and registered by a pediatric specialist nurse. Late-onset AEs were reported by parents. Parents' perceptions were collected through a questionnaire and analyzed using content analysis. The study population consisted of 43 infants in 38 families. No immediate AEs occurred. Three late-onset AEs were reported in two infants in the intervention group. Three categories emerged in the content analysis: (1) protect and watch over the infant, (2) optimal health and well-being for the whole family, and (3) avoid suffering for the infant. The study results show that home immunization with palivizumab is feasible if safety aspects are considered and that parental involvement in the choice of place for immunization after a neonatal intensive care experience can be important.
Collapse
Affiliation(s)
- Christina Ebersjö
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institute, 171 77 Stockholm, Sweden
- Sach’s Children and Youth Hospital, Södersjukhuset AB, 118 83 Stockholm, Sweden
| | - Eva Berggren Broström
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Inger Kull
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institute, 171 77 Stockholm, Sweden
- Sach’s Children and Youth Hospital, Södersjukhuset AB, 118 83 Stockholm, Sweden
| | - Anna Lindholm Olinder
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institute, 171 77 Stockholm, Sweden
- Sach’s Children and Youth Hospital, Södersjukhuset AB, 118 83 Stockholm, Sweden
| |
Collapse
|
13
|
Stuart ASV, Virta M, Williams K, Seppa I, Hartvickson R, Greenland M, Omoruyi E, Bastian AR, Haazen W, Salisch N, Gymnopoulou E, Callendret B, Faust SN, Snape MD, Heijnen E. Phase 1/2a Safety and Immunogenicity of an Adenovirus 26 Vector Respiratory Syncytial Virus (RSV) Vaccine Encoding Prefusion F in Adults 18-50 Years and RSV-Seropositive Children 12-24 Months. J Infect Dis 2022; 227:71-82. [PMID: 36259542 PMCID: PMC9796164 DOI: 10.1093/infdis/jiac407] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/28/2022] [Accepted: 10/17/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) remains a leading cause of pediatric morbidity, with no approved vaccine. We assessed the safety and immunogenicity of the Ad26.RSV.preF vaccine candidate in adults and children. METHODS In this randomized, double-blind, phase 1/2a, placebo-controlled study, 12 adults (18-50 years) and 36 RSV-seropositive children (12-24 months) were randomized 2:1 to Ad26.RSV.preF (1 × 1011 viral particles [vp] for adults, 5 × 1010 vp for children) or placebo, at day 1 and 29, with 6-month immunogenicity and 1-year safety follow-up. Respiratory syncytial virus infection was an exploratory outcome in children. RESULTS In adults, solicited adverse events (AEs) were generally mild to moderate, with no serious AEs. In children, no vaccination-related serious AEs were reported; fever was reported in 14 (58.3%) Ad26.RSV.preF recipients. Baseline pediatric geometric mean titers for RSV A2 neutralization increased from 121 (95% confidence interval [CI], 76-191) to 1608 (95% CI, 730-3544) at day 29, and 2235 (95% CI, 1586-3150) at day 57, remaining elevated over 7 months. Respiratory syncytial virus infection was confirmed in fewer children receiving Ad26.RSV.preF (1, 4.2%) than placebo (5, 41.7%). CONCLUSIONS Ad26.RSV.preF demonstrated immunogenicity in healthy adults and toddlers, with no safety concerns raised. Evaluations in RSV-seronegative children are underway.
Collapse
Affiliation(s)
- Arabella S V Stuart
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Melanie Greenland
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | | | - Wouter Haazen
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Nadine Salisch
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | | | - Saul N Faust
- NIHR Southampton Clinical Research Facility and NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom,Oxford NIHR – Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Esther Heijnen
- Correspondence: Esther Heijnen, MD, Janssen Vaccines & Prevention BV, Leiden, 2333 CN, The Netherlands ()
| |
Collapse
|
14
|
Abu-Raya B, Reicherz F, Lavoie PM. Correlates of Protection Against Respiratory Syncytial Virus Infection in Infancy. Clin Rev Allergy Immunol 2022; 63:371-380. [PMID: 35689745 DOI: 10.1007/s12016-022-08948-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/28/2022]
Abstract
The highest morbidity and mortality from respiratory syncytial virus (RSV) infection occurs in young infants. Immunization of expectant mothers during pregnancy has the potential to substantially reduce the burden of RSV disease in a majority of infants. Correlates of protection (COP) are important in guiding the development of maternal RSV vaccines and the design of maternal RSV vaccine trials, as immune response to candidate vaccines should mirror protective RSV immunity at birth. Here, we review the literature reporting correlations between RSV immune measures at birth and clinical RSV outcomes during infancy. Less than a dozen studies have investigated immunological COP with RSV disease or related hospitalization, yielding inconsistent findings overall. The differences in findings between studies could be due to differences in inclusion/exclusion criteria (e.g., the inclusion of older infants who may benefit less from maternal antibodies or infants followed during inter-seasonal periods where RSV is absent), differences in semi-quantitative RSV antibody neutralization assays, or differences in RSV outcome measures such as the sensititivity/specificity of diagnostic tests. Future research in this field should seek to standardize RSV immunological measures and outcomes, expand the breadth of functional RSV measures beyond antibody neutralization, and consider infants' age and seasonality of RSV infection.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- BC Children's Hospital Research Institute, Vancouver, Canada. .,Department of Pediatrics, University of British Columbia, 950 West 28th Avenue, Vancouver, Canada.
| | - Frederic Reicherz
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Pediatrics, University of British Columbia, 950 West 28th Avenue, Vancouver, Canada
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver, Canada.,Department of Pediatrics, University of British Columbia, 950 West 28th Avenue, Vancouver, Canada
| |
Collapse
|
15
|
Hwang J, Jung Y, Moon S, Yu S, Oh H, Kim S, Kim KW, Yoon JH, Chun J, Kim SJ, Chung WJ, Kweon DH. Nanodisc-Mediated Conversion of Virustatic Antiviral Antibody to Disrupt Virus Envelope in Infected Cells. SMALL METHODS 2022; 6:e2101516. [PMID: 35107214 DOI: 10.1002/smtd.202101516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Many antibody-based antivirals, including broadly neutralizing antibodies (bnAbs) against various influenza virus strains, suffer from limited potency. A booster of the antiviral activity of an antibody is expected to facilitate development of antiviral therapeutics. In this study, a nanodisc (ND), a discoidal lipid bilayer encircled by membrane scaffold proteins, is engineered to provide virucidal properties to antibodies, thereby augmenting their antiviral activity. NDs carrying the Fc-binding peptide sequence form an antibody-ND complex (ANC), which can co-endocytose into cells infected with influenza virus. ANC efficiently inhibits endosome escape of viral RNA by dual complimentary mode of action. While the antibody moiety in an ANC inhibits hemagglutinin-mediated membrane fusion, its ND moiety destroys the viral envelope using free hemagglutinins that are not captured by antibodies. Providing virus-infected host cells with the ability to self-eliminate by the synergistic effect of ANC components dramatically amplifies the antiviral efficacy of a bnAb against influenza virus. When the efficacy of ANC is assessed in mouse models, administration of ANCs dramatically reduces morbidity and mortality compared to bnAb alone. This study is the first to demonstrate the novel nanoparticle ANC and its role in combating viral infections, suggesting that ANC is a versatile platform applicable to various viruses.
Collapse
Affiliation(s)
- Jaehyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Younghun Jung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seokoh Moon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seokhyeon Yu
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyunseok Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soomin Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jeong Hyeon Yoon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihwan Chun
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sang Jick Kim
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Korea
| | - Woo-Jae Chung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
16
|
Saito M, Tsukagoshi H, Sada M, Sunagawa S, Shirai T, Okayama K, Sugai T, Tsugawa T, Hayashi Y, Ryo A, Takeda M, Kawashima H, Saruki N, Kimura H. Detailed Evolutionary Analyses of the F Gene in the Respiratory Syncytial Virus Subgroup A. Viruses 2021; 13:v13122525. [PMID: 34960794 PMCID: PMC8706373 DOI: 10.3390/v13122525] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 11/20/2022] Open
Abstract
We performed evolution, phylodynamics, and reinfection-related antigenicity analyses of respiratory syncytial virus subgroup A (RSV-A) fusion (F) gene in globally collected strains (1465 strains) using authentic bioinformatics methods. The time-scaled evolutionary tree using the Bayesian Markov chain Monte Carlo method estimated that a common ancestor of the RSV-A, RSV-B, and bovine-RSV diverged at around 450 years ago, and RSV-A and RSV-B diverged around 250 years ago. Finally, the RSV-A F gene formed eight genotypes (GA1-GA7 and NA1) over the last 80 years. Phylodynamics of RSV-A F gene, including all genotype strains, increased twice in the 1990s and 2010s, while patterns of each RSV-A genotype were different. Phylogenetic distance analysis suggested that the genetic distances of the strains were relatively short (less than 0.05). No positive selection sites were estimated, while many negative selection sites were found. Moreover, the F protein 3D structure mapping and conformational epitope analysis implied that the conformational epitopes did not correspond to the neutralizing antibody binding sites of the F protein. These results suggested that the RSV-A F gene is relatively conserved, and mismatches between conformational epitopes and neutralizing antibody binding sites of the F protein are responsible for the virus reinfection.
Collapse
Affiliation(s)
- Mariko Saito
- Gunma Prefectural Institute of Public Health and Environmental Sciences, Maebashi-shi 371-0052, Japan; (M.S.); (H.T.); (N.S.)
| | - Hiroyuki Tsukagoshi
- Gunma Prefectural Institute of Public Health and Environmental Sciences, Maebashi-shi 371-0052, Japan; (M.S.); (H.T.); (N.S.)
| | - Mitsuru Sada
- Department of Health Science, Gunma Paz University Graduate School, Takasaki-shi 370-0006, Japan; (M.S.); (S.S.); (K.O.); (Y.H.)
| | - Soyoka Sunagawa
- Department of Health Science, Gunma Paz University Graduate School, Takasaki-shi 370-0006, Japan; (M.S.); (S.S.); (K.O.); (Y.H.)
| | - Tatsuya Shirai
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka-shi 181-8611, Japan;
| | - Kaori Okayama
- Department of Health Science, Gunma Paz University Graduate School, Takasaki-shi 370-0006, Japan; (M.S.); (S.S.); (K.O.); (Y.H.)
| | - Toshiyuki Sugai
- Division of Nursing Science, Hiroshima University, Hiroshima-shi 734-8551, Japan;
| | - Takeshi Tsugawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo-shi 060-8543, Japan;
| | - Yuriko Hayashi
- Department of Health Science, Gunma Paz University Graduate School, Takasaki-shi 370-0006, Japan; (M.S.); (S.S.); (K.O.); (Y.H.)
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama-shi 236-0004, Japan;
| | - Makoto Takeda
- Department of Virology, National Institute of Infectious Diseases, Musashimurayama-shi 208-0011, Japan;
| | - Hisashi Kawashima
- Department of Pediatrics, Tokyo Medical University, Shinjuku-ku 160-0023, Japan;
| | - Nobuhiro Saruki
- Gunma Prefectural Institute of Public Health and Environmental Sciences, Maebashi-shi 371-0052, Japan; (M.S.); (H.T.); (N.S.)
| | - Hirokazu Kimura
- Department of Health Science, Gunma Paz University Graduate School, Takasaki-shi 370-0006, Japan; (M.S.); (S.S.); (K.O.); (Y.H.)
- Correspondence: ; Tel.: +81-27-388-0336
| |
Collapse
|
17
|
Garegnani L, Styrmisdóttir L, Roson Rodriguez P, Escobar Liquitay CM, Esteban I, Franco JV. Palivizumab for preventing severe respiratory syncytial virus (RSV) infection in children. Cochrane Database Syst Rev 2021; 11:CD013757. [PMID: 34783356 PMCID: PMC8594174 DOI: 10.1002/14651858.cd013757.pub2] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Respiratory viruses are the leading cause of lower respiratory tract infection (LRTI) and hospitalisation in infants and young children. Respiratory syncytial virus (RSV) is the main infectious agent in this population. Palivizumab is administered intramuscularly every month during five months in the first RSV season to prevent serious RSV LRTI in children. Given its high cost, it is essential to know if palivizumab continues to be effective in preventing severe RSV disease in children. OBJECTIVES To assess the effects of palivizumab for preventing severe RSV infection in children. SEARCH METHODS We searched CENTRAL, MEDLINE, three other databases and two trials registers to 14 October 2021, together with reference checking, citation searching and contact with study authors to identify additional studies. We searched Embase to October 2020, as we did not have access to this database for 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs), including cluster-RCTs, comparing palivizumab given at a dose of 15 mg/kg once a month (maximum five doses) with placebo, no intervention or standard care in children 0 to 24 months of age from both genders, regardless of RSV infection history. DATA COLLECTION AND ANALYSIS: We used Cochrane's Screen4Me workflow to help assess the search results. Two review authors screened studies for selection, assessed risk of bias and extracted data. We used standard Cochrane methods. We used GRADE to assess the certainty of the evidence. The primary outcomes were hospitalisation due to RSV infection, all-cause mortality and adverse events. Secondary outcomes were hospitalisation due to respiratory-related illness, length of hospital stay, RSV infection, number of wheezing days, days of supplemental oxygen, intensive care unit length of stay and mechanical ventilation days. MAIN RESULTS We included five studies with a total of 3343 participants. All studies were parallel RCTs, assessing the effects of 15 mg/kg of palivizumab every month up to five months compared to placebo or no intervention in an outpatient setting, although one study also included hospitalised infants. Most of the included studies were conducted in children with a high risk of RSV infection due to comorbidities like bronchopulmonary dysplasia and congenital heart disease. The risk of bias of outcomes across all studies was similar and predominately low. Palivizumab reduces hospitalisation due to RSV infection at two years' follow-up (risk ratio (RR) 0.44, 95% confidence interval (CI) 0.30 to 0.64; 5 studies, 3343 participants; high certainty evidence). Based on 98 hospitalisations per 1000 participants in the placebo group, this corresponds to 43 (29 to 62) per 1000 participants in the palivizumab group. Palivizumab probably results in little to no difference in mortality at two years' follow-up (RR 0.69, 95% CI 0.42 to 1.15; 5 studies, 3343 participants; moderate certainty evidence). Based on 23 deaths per 1000 participants in the placebo group, this corresponds to 16 (10 to 27) per 1000 participants in the palivizumab group. Palivizumab probably results in little to no difference in adverse events at 150 days' follow-up (RR 1.09, 95% CI 0.85 to 1.39; 3 studies, 2831 participants; moderate certainty evidence). Based on 84 cases per 1000 participants in the placebo group, this corresponds to 91 (71 to 117) per 1000 participants in the palivizumab group. Palivizumab probably results in a slight reduction in hospitalisation due to respiratory-related illness at two years' follow-up (RR 0.78, 95% CI 0.62 to 0.97; 5 studies, 3343 participants; moderate certainty evidence). Palivizumab may result in a large reduction in RSV infection at two years' follow-up (RR 0.33, 95% CI 0.20 to 0.55; 3 studies, 554 participants; low certainty evidence). Based on 195 cases of RSV infection per 1000 participants in the placebo group, this corresponds to 64 (39 to 107) per 1000 participants in the palivizumab group. Palivizumab also reduces the number of wheezing days at one year's follow-up (RR 0.39, 95% CI 0.35 to 0.44; 1 study, 429 participants; high certainty evidence). AUTHORS' CONCLUSIONS The available evidence suggests that prophylaxis with palivizumab reduces hospitalisation due to RSV infection and results in little to no difference in mortality or adverse events. Moreover, palivizumab results in a slight reduction in hospitalisation due to respiratory-related illness and may result in a large reduction in RSV infections. Palivizumab also reduces the number of wheezing days. These results may be applicable to children with a high risk of RSV infection due to comorbidities. Further research is needed to establish the effect of palivizumab on children with other comorbidities known as risk factors for severe RSV disease (e.g. immune deficiencies) and other social determinants of the disease, including children living in low- and middle-income countries, tropical regions, children lacking breastfeeding, living in poverty, or members of families in overcrowded situations.
Collapse
Affiliation(s)
- Luis Garegnani
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | - Pablo Roson Rodriguez
- Research Department, Instituto Universitario Hospital Italiano, Buenos Aires, Argentina
| | | | - Ignacio Esteban
- Fundación INFANT, Buenos Aires, Argentina
- Pediatric Stepdown Unit, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Juan Va Franco
- Associate Cochrane Centre, Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
18
|
Nourbakhsh S, Shoukat A, Zhang K, Poliquin G, Halperin D, Sheffield H, Halperin SA, Langley JM, Moghadas SM. Effectiveness and cost-effectiveness of RSV infant and maternal immunization programs: A case study of Nunavik, Canada. EClinicalMedicine 2021; 41:101141. [PMID: 34622186 PMCID: PMC8479643 DOI: 10.1016/j.eclinm.2021.101141] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Despite passive immunization with palivizumab to select high-risk children under two years of age, the health and economic burden of respiratory syncytial virus (RSV) remains substantial. We evaluated the effectiveness and cost-effectiveness of immunization programs with new generations of RSV prophylactics, including long-acting monoclonal antibodies (LAMA) and maternal vaccines, in terms of reducing hospitalizations in Nunavik, a Canadian Arctic region. METHODS We developed an agent-based model of RSV transmission and parameterized it with the demographics and burden of RSV in Nunavik, Québec. We compared various immunization strategies, taking into account the costs associated with program delivery and calculating the incremental cost-effectiveness ratio (ICER) using quality-adjusted life-years (QALYs) gained as a measure of effectiveness. Scenario analyses included immunization with palivizumab and LAMA for infants under one year of age, and maternal vaccination in mild, moderate, and severe RSV seasons. Data were analysed from November 1, 2019 to May 1, 2021. FINDINGS We found that a Nunavik pilot program with palivizumab which included healthy full-term infants aged 0-2 months in addition to those considered high-risk for complicated RSV disease is not cost-effective, compared to offering palivizumab only to preterm/chronically ill infants under 1 year of age. Using LAMA as prophylaxis produces ICER values of CAD $39,414/QALY (95% Credible Interval [CrI]: $39,314-$40,017) in a mild season (moderately cost-effective) and CAD $5,255/QALY (95% CrI: $5,222-$5,307) in a moderate season (highly cost-effective). LAMA was a dominant (cost-saving with negative incremental costs and positive incremental effects) strategy in a severe RSV season. Maternal vaccination combined with immunization of preterm/chronically ill infants 3-11 months was also a dominant (cost-saving) strategy in all seasons. INTERPRETATION The switch from palivizumab in RSV immunization programs to new prophylactics would lead to significant savings, with LAMA being an effective strategy without compromising benefits in terms of reducing hospitalizations.
Collapse
Affiliation(s)
- Shokoofeh Nourbakhsh
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario M3J 1P3, Canada
| | - Affan Shoukat
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario M3J 1P3, Canada
| | - Kevin Zhang
- Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Guillaume Poliquin
- Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
- Office of the Scientific Director, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Donna Halperin
- School of Nursing, St. Francis Xavier University, Antigonish, Nova Scotia B2G 2W5, Canada
| | - Holden Sheffield
- Department of Paediatrics, Qikiqtani General Hospital, Iqaluit, Nunavut X0A 0H0, Canada
| | - Scott A. Halperin
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, Nova Scotia B3K 6R8, Canada
| | - Joanne M. Langley
- Canadian Center for Vaccinology, IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, Nova Scotia B3K 6R8, Canada
| | - Seyed M. Moghadas
- Agent-Based Modelling Laboratory, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
19
|
Abstract
Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory tract infection and is responsible for a large proportion of infant morbidity and mortality worldwide. Most RSV-related deaths occur in children under six months, and the majority of these occur in low-income settings. To date, there is no known efficacious treatment for RSV infection; hence, prevention remains an important strategy to reduce the global burden of disease. Monoclonal antibodies and vaccinations are currently the two main approaches for prevention of RSV disease. Maternal RSV vaccination is of particular interest as a strategy to protect infants during their most vulnerable period as this approach has proven highly efficacious in other vaccine-preventable conditions such as pertussis and influenza. As results from ongoing phase III clinical trials become available, important decisions will need to be made about the priority and potential implementation of RSV vaccines alongside other public health measures.
Collapse
Affiliation(s)
- Ahinsa Gunatilaka
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Michelle L Giles
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| |
Collapse
|
20
|
Hervé PL, Dhelft V, Zuniga A, Ghasparian A, Rassek O, Yim KC, Donne N, Lambert PH, Benhamou PH, Sampson HA, Mondoulet L. Epicutaneous immunization using synthetic virus-like particles efficiently boosts protective immunity to respiratory syncytial virus. Vaccine 2021; 39:4555-4563. [PMID: 34154864 DOI: 10.1016/j.vaccine.2021.03.081] [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] [Received: 10/06/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023]
Abstract
Despite the substantial health and economic burden caused by RSV-associated illness, no vaccine is available. The sole licensed treatment (palivizumab), composed of a monoclonal neutralizing antibody, blocks the fusion of the virus to the host cell but does not prevent infection. The development of a safe and efficacious RSV vaccine is therefore a priority, but also a considerable challenge, and new innovative strategies are warranted. Most of the adult population encounter regular RSV infections and can elicit a robust neutralizing antibody response, but unfortunately it wanes over time and reinfections during subsequent seasons are common. One approach to protect the mother and young infant from RSV infection is to administer a vaccine capable of boosting preexisting RSV immunity during pregnancy, which would provide protection to the fetus through passive transfer of maternal antibodies, thus preventing primary RSV infection in newborns during their first months of life. Here, we describe the preclinical evaluation of an epicutaneous RSV vaccine booster that combines epicutaneous patches as a delivery platform and a Synthetic Virus-Like Particles (SVLP)-based vaccine displaying multiple RSV F-protein site II (FsII, palivizumab epitope) mimetic as antigen (V-306). We demonstrated in mice that epicutaneous immunization with V-306 efficiently boosts preexisting immunity induced by the homologous V-306 administered subcutaneously. This boosting was characterized by a significant increase in F- and FsII-specific antibodies capable of competing with palivizumab for its target antigen and neutralize RSV. More importantly, epicutaneous booster immunization with V-306 significantly decreased lung viral replication in experimental mice after intranasal RSV challenge, without inducing enhanced RSV disease. In conclusion, an epicutaneous booster vaccine based on V-306 is safe and efficacious in enhancing RSV preexisting immunity in mice. This needle-free vaccine candidate would be potentially suited as a booster vaccine for vulnerable populations such as young infants via pregnant women, and the elderly.
Collapse
Affiliation(s)
- Pierre-Louis Hervé
- DBV Technologies, 177-181 Avenue Pierre Brossolette, 92120 Montrouge, France.
| | - Véronique Dhelft
- DBV Technologies, 177-181 Avenue Pierre Brossolette, 92120 Montrouge, France
| | - Armando Zuniga
- Virometix AG, Wagisstrasse 14, CH-8952 Schlieren, Switzerland
| | - Arin Ghasparian
- Virometix AG, Wagisstrasse 14, CH-8952 Schlieren, Switzerland
| | - Oliver Rassek
- Virometix AG, Wagisstrasse 14, CH-8952 Schlieren, Switzerland
| | - Kevin C Yim
- Sigmovir Biosystems, Inc., 9610 Medical Center Drive, Suite #100, Rockville, MD 20850, USA
| | - Nathalie Donne
- DBV Technologies, 177-181 Avenue Pierre Brossolette, 92120 Montrouge, France
| | - Paul-Henri Lambert
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | | | - Hugh A Sampson
- DBV Technologies, 12 East 49th Street Tower 49, Suite 4001, New York, NY 10017, USA
| | - Lucie Mondoulet
- DBV Technologies, 177-181 Avenue Pierre Brossolette, 92120 Montrouge, France
| |
Collapse
|
21
|
Thornhill EM, Salpor J, Verhoeven D. Respiratory syntycial virus: Current treatment strategies and vaccine approaches. Antivir Chem Chemother 2021; 28:2040206620947303. [PMID: 32741202 PMCID: PMC7412623 DOI: 10.1177/2040206620947303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Respiratory Syncytial Virus is a yearly respiratory virus that causes significant frequencies of morbidities, particularly in the young and elderly populations. However, preventive vaccines and/or treatment therapies are generally lacking, although much attention is now being placed on this virus. Moreover, there are now multiple strategies currently being explored in a race to the first licensed vaccine. While vaccines are being developed, multiple treatment strategies are being explored to attenuate the severity of infection and thus reduce hospitalization rates in vulnerable populations. This review outlines current strategies to prevent or treat this virus in the hopes of reducing significant human morbidity and mortality that occurs yearly with this seasonal virus.
Collapse
Affiliation(s)
- Elena Margret Thornhill
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, USA
| | - Jessica Salpor
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, USA
| | - David Verhoeven
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, USA
| |
Collapse
|
22
|
Cruz-Teran C, Tiruthani K, McSweeney M, Ma A, Pickles R, Lai SK. Challenges and opportunities for antiviral monoclonal antibodies as COVID-19 therapy. Adv Drug Deliv Rev 2021; 169:100-117. [PMID: 33309815 PMCID: PMC7833882 DOI: 10.1016/j.addr.2020.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 01/08/2023]
Abstract
To address the COVID-19 pandemic, there has been an unprecedented global effort to advance potent neutralizing mAbs against SARS-CoV-2 as therapeutics. However, historical efforts to advance antiviral monoclonal antibodies (mAbs) for the treatment of other respiratory infections have been met with categorical failures in the clinic. By investigating the mechanism by which SARS-CoV-2 and similar viruses spread within the lung, along with available biodistribution data for systemically injected mAb, we highlight the challenges faced by current antiviral mAbs for COVID-19. We summarize some of the leading mAbs currently in development, and present the evidence supporting inhaled delivery of antiviral mAb as an early intervention against COVID-19 that could prevent important pulmonary morbidities associated with the infection.
Collapse
Affiliation(s)
- Carlos Cruz-Teran
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karthik Tiruthani
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Alice Ma
- UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Raymond Pickles
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Inhalon Biopharma, Durham, NC 27709, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
23
|
Rostad CA, Maillis AN, Lai K, Bakshi N, Jerris RC, Lane PA, Yee ME, Yildirim I. The burden of respiratory syncytial virus infections among children with sickle cell disease. Pediatr Blood Cancer 2021; 68:e28759. [PMID: 33034160 PMCID: PMC8246443 DOI: 10.1002/pbc.28759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although respiratory syncytial virus (RSV) is the leading cause of pediatric lower respiratory tract infections, the burden of RSV in children with sickle cell disease (SCD) is unknown. METHODS We conducted a retrospective, nested, case-control study of children with SCD <18 years who had respiratory viral panels (RVPs) performed at Children's Healthcare of Atlanta from 2012 to 2019. We abstracted the medical records to describe the demographics, clinical features, and outcomes of children who tested positive for RSV (cases) versus children who tested negative (controls). We calculated the annual incidence of RSV and related hospitalization rates with 95% confidence intervals (CIs) and used multivariate logistic regression to evaluate associations. RESULTS We identified 3676 RVP tests performed on 2636 patients over seven respiratory seasons resulting in 219/3676 (6.0%) RSV-positive tests among 160/2636 (6.1%) patients. The average annual incidence of laboratory-confirmed RSV infection among children with SCD was 34.3 (95% CI 18.7-49.8) and 3.8 (95% CI 0.5-7.0) cases per 1000 person-years for those <5 years and 5-18 years, respectively. The RSV-related hospitalization rate for children <5 years was 20.7 (95% CI 8.5-32.8) per 1000 person-years. RSV-positive cases were significantly younger than RSV-negative patients (3.8 years vs 7.6 years, P < .001). Of RSV-positive cases, 22 (13.8%) developed acute chest syndrome and nine (5.6%) required intensive care, which was not significantly different from RSV-negative children with SCD. CONCLUSION RSV infections are common in children with SCD with higher burden in younger patients. RSV is associated with considerable morbidity, including higher rates of hospitalization compared to the general population.
Collapse
Affiliation(s)
- Christina A. Rostad
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA,Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA,Corresponding Author: Christina A. Rostad, MD. Current Address: Emory Children’s Center, 2015 Uppergate Dr NE, Atlanta, Georgia, 30322, USA.
| | - Alexander N. Maillis
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Kristina Lai
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Nitya Bakshi
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA,Department of Pediatrics, Division of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Robert C. Jerris
- Department of Pathology and Laboratory Medicine, Children’s Healthcare of Atlanta, Atlanta, GA,Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Peter A. Lane
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA,Department of Pediatrics, Division of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marianne E. Yee
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA,Department of Pediatrics, Division of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Inci Yildirim
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA,Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
24
|
Abu-Raya B, Maertens K. Protection of the Newborn Through Vaccination in Pregnancy. Neoreviews 2021; 22:e25-e39. [PMID: 33386312 DOI: 10.1542/neo.22-1-e25] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Newborns and infants are at risk for severe infections with some pathogens (eg, Bordetella pertussis, influenza, respiratory syncytial virus, group B Streptococcus) during early life. To decrease this window of high susceptibility to some infections during early life and protect young infants, vaccination in pregnancy against some vaccine-preventable diseases (eg, influenza, pertussis, tetanus) has been recommended in an increasing number of countries with notable success. In addition, recent advances have been made in developing vaccines for pregnant women with the aim of reducing the respiratory syncytial virus and group B Streptococcus burden in infancy. In this article, we review the vaccines currently recommended during pregnancy and their benefits to newborns and infants. We also discuss progress made in the development of other vaccines that are expected to be evaluated in pregnant women in the near future.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Vaccine Evaluation Center, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirsten Maertens
- Centre for the Evaluation of Vaccination, Vaccine & Infectious Diseases Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
25
|
Efstathiou C, Abidi SH, Harker J, Stevenson NJ. Revisiting respiratory syncytial virus's interaction with host immunity, towards novel therapeutics. Cell Mol Life Sci 2020; 77:5045-5058. [PMID: 32556372 PMCID: PMC7298439 DOI: 10.1007/s00018-020-03557-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022]
Abstract
Every year there are > 33 million cases of Respiratory Syncytial Virus (RSV)-related respiratory infection in children under the age of five, making RSV the leading cause of lower respiratory tract infection (LRTI) in infants. RSV is a global infection, but 99% of related mortality is in low/middle-income countries. Unbelievably, 62 years after its identification, there remains no effective treatment nor vaccine for this deadly virus, leaving infants, elderly and immunocompromised patients at high risk. The success of all pathogens depends on their ability to evade and modulate the host immune response. RSV has a complex and intricate relationship with our immune systems, but a clearer understanding of these interactions is essential in the development of effective medicines. Therefore, in a bid to update and focus our research community's understanding of RSV's interaction with immune defences, this review aims to discuss how our current knowledgebase could be used to combat this global viral threat.
Collapse
Affiliation(s)
- C Efstathiou
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - S H Abidi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - J Harker
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College London, South Kensington, London, UK
| | - N J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
26
|
Learning from past failures: Challenges with monoclonal antibody therapies for COVID-19. J Control Release 2020; 329:87-95. [PMID: 33276017 PMCID: PMC7836766 DOI: 10.1016/j.jconrel.2020.11.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 01/08/2023]
Abstract
COVID-19, the disease caused by infection with SARS-CoV-2, requires urgent development of therapeutic interventions. Due to their safety, specificity, and potential for rapid advancement into the clinic, monoclonal antibodies (mAbs) represent a highly promising class of antiviral or anti-inflammatory agents. Herein, by analyzing prior efforts to advance antiviral mAbs for other acute respiratory infections (ARIs), we highlight the challenges faced by mAb-based immunotherapies for COVID-19. We present evidence supporting early intervention immediately following a positive diagnosis via inhaled delivery of mAbs with vibrating mesh nebulizers as a promising approach for the treatment of COVID-19.
Collapse
|
27
|
Factors Contributing to Symptom Duration and Viral Reduction in Outpatient Children With Respiratory Syncytial Virus Infection. Pediatr Infect Dis J 2020; 39:678-683. [PMID: 32101910 PMCID: PMC7360094 DOI: 10.1097/inf.0000000000002626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND We investigated the association between age, duration of clinical symptoms and viral shedding in outpatient children infected with respiratory syncytial virus (RSV) in Japan. METHODS Outpatients younger than 2 years of age, with suspected RSV infection between 2014 and 2018, were enrolled in the study. Following informed consent, nasal samples were collected at first and second clinic visits (with 0-9 days gap). RSV-A or -B infection and viral load were determined by real-time polymerase chain reaction. Clinical symptoms were recorded at first clinic visit, and fever and symptoms were recorded at home for up to 8 days. Association between clinical symptoms and patient characteristics, such as age, sex and birth weight, were analyzed using ordered logistic regression analysis. The association between viral reduction and estimated shedding period was examined using linear regression analysis. RESULTS Among the 205 cases enrolled in the study, no difference was found in patient characteristics between RSV-A and -B infection. Duration of fever was prolonged with increased age. Duration of rhinorrhea and cough was shorter in females than in males and in groups with birth weight ≥3 kg than in those with <2.5 kg. Daily viral reduction increased and estimated viral elimination period decreased with age. CONCLUSIONS Fever duration was found to increase while viral shedding decreased with patient age.
Collapse
|
28
|
Abu-Raya B, Maertens K, Edwards KM, Omer SB, Englund JA, Flanagan KL, Snape MD, Amirthalingam G, Leuridan E, Damme PV, Papaevangelou V, Launay O, Dagan R, Campins M, Cavaliere AF, Frusca T, Guidi S, O'Ryan M, Heininger U, Tan T, Alsuwaidi AR, Safadi MA, Vilca LM, Wanlapakorn N, Madhi SA, Giles ML, Prymula R, Ladhani S, Martinón-Torres F, Tan L, Michelin L, Scambia G, Principi N, Esposito S. Global Perspectives on Immunization During Pregnancy and Priorities for Future Research and Development: An International Consensus Statement. Front Immunol 2020; 11:1282. [PMID: 32670282 PMCID: PMC7326941 DOI: 10.3389/fimmu.2020.01282] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022] Open
Abstract
Immunization during pregnancy has been recommended in an increasing number of countries. The aim of this strategy is to protect pregnant women and infants from severe infectious disease, morbidity and mortality and is currently limited to tetanus, inactivated influenza, and pertussis-containing vaccines. There have been recent advancements in the development of vaccines designed primarily for use in pregnant women (respiratory syncytial virus and group B Streptococcus vaccines). Although there is increasing evidence to support vaccination in pregnancy, important gaps in knowledge still exist and need to be addressed by future studies. This collaborative consensus paper provides a review of the current literature on immunization during pregnancy and highlights the gaps in knowledge and a consensus of priorities for future research initiatives, in order to optimize protection for both the mother and the infant.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kirsten Maertens
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Kathryn M. Edwards
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Saad B. Omer
- Department of Internal Medicine (Infectious Diseases), Department of Epidemiology of Microbial Diseases, Yale School of Medicine, Yale School of Public Health, New Haven, CT, United States
| | - Janet A. Englund
- Department of Pediatrics, Seattle Children's Research Institute, University of Washington, Seattle, WA, United States
| | - Katie L. Flanagan
- Faculty of Health Sciences, School of Medicine, University of Tasmania, Launceston, TAS, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Matthew D. Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Gayatri Amirthalingam
- Immunisation and Countermeasures Division, National Infection Service, Public Health England, London, United Kingdom
| | - Elke Leuridan
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Pierre Van Damme
- Faculty of Medicine and Health Sciences, Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Vana Papaevangelou
- Third Department of Pediatrics, University Hospital ATTIKON, National and Kapodistrian University of Athens, Athens, Greece
| | - Odile Launay
- Université de Paris, Inserm, CIC 1417, F-CRIN I REIVAC, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ron Dagan
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Magda Campins
- Preventive Medicine and Epidemiology Department, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Anna Franca Cavaliere
- Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tiziana Frusca
- Department of Medicine and Surgery, Obstetrics and Gynaecology Unit, University of Parma, Parma, Italy
| | - Sofia Guidi
- Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | - Miguel O'Ryan
- Microbiology and Mycology Program, Faculty of Medicine, Institute of Biomedical Sciences and Associate Researcher, Millennium Institute of Immunology and Immunotherapy, University of Chile, Santiago, Chile
| | - Ulrich Heininger
- Pediatric Infectious Diseases, University of Basel Children's Hospital, Basel, Switzerland
| | - Tina Tan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, United States
| | - Ahmed R. Alsuwaidi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Marco. A. Safadi
- Department of Pediatrics, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Luz M. Vilca
- Unit of Obstetrics and Gynecology, Buzzi Hospital - ASST Fatebenefratelli Sacco, University of Milan, Milan, Italy
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Shabir A. Madhi
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michelle L. Giles
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Roman Prymula
- School of Medicine Hradec Kralove, Institute of Social Medicine, Charles University Prague, Prague, Czechia
| | - Shamez Ladhani
- Immunisation and Countermeasures Division, National Infection Service, Public Health England, London, United Kingdom
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago de Compostela, University of Santiago, Santiago de Compostela, Spain
| | - Litjen Tan
- Immunization Action Coalition, St. Paul, MN, United States
| | - Lessandra Michelin
- Infectious Diseases and Vaccinology Division, Health Sciences Post Graduation Program, University of Caxias Do Sul, Caxias Do Sul, Brazil
| | - Giovanni Scambia
- Dipartimento Scienze della Salute della Donna e del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS-Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| |
Collapse
|
29
|
Kimura T, Takeuchi M, Kawakami K. Utilization and efficacy of palivizumab for children with Down syndrome. Pediatr Int 2020; 62:677-682. [PMID: 31961027 DOI: 10.1111/ped.14157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 11/25/2019] [Accepted: 01/15/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Palivizumab is a prophylactic drug used for severe respiratory syncytial virus (RSV) infection in young children with high-risk factors, such as prematurity and congenital heart diseases. Although Japan expanded the use of palivizumab for Down syndrome (DS) in 2013 regardless of additional high-risk factors, there is not enough evidence to support its use. We analyzed health insurance claim data from the JMDC (Tokyo, Japan) to investigate the prescription pattern and efficacy of palivizumab in children with DS. METHODS We enrolled children of ≤24 months of age with DS at the start of the RSV epidemic season (2007-2015). We compared the risk of RSV-related admission with and without palivizumab prophylaxis using mixed effects logistic regression models accounting for patient-level clustering. RESULTS Of 632 children, 30% (81/268) and 83% (303/364) received palivizumab before and after the expanded program (2007-2012 and 2013-2015), respectively. Among children with DS but without additional high-risk factors (n = 135), palivizumab use surged from 0% (0/62) to 73% (53/73). In the whole study population, RSV-related hospitalization occurred in 4.2% (16/384) patients with prophylaxis and 6.0% (15/248) patients without prophylaxis. The multivariate analysis revealed that palivizumab was associated with reduced RSV-related hospitalization (odds ratio: 0.41, 95% confidence interval: 0.18-0.92, P = 0.03). CONCLUSIONS Virtually all children with DS in Japan have now received palivizumab, the use of which was associated with a reduction in RSV-related hospitalization in DS. Further evidence is required to clarify whether palivizumab prophylaxis should be risk-tailored or universal for DS.
Collapse
Affiliation(s)
- Takeshi Kimura
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Masato Takeuchi
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| |
Collapse
|
30
|
Norlander AE, Peebles RS. Innate Type 2 Responses to Respiratory Syncytial Virus Infection. Viruses 2020; 12:E521. [PMID: 32397226 PMCID: PMC7290766 DOI: 10.3390/v12050521] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common and contagious virus that results in acute respiratory tract infections in infants. In many cases, the symptoms of RSV remain mild, however, a subset of individuals develop severe RSV-associated bronchiolitis. As such, RSV is the chief cause of infant hospitalization within the United States. Typically, the immune response to RSV is a type 1 response that involves both the innate and adaptive immune systems. However, type 2 cytokines may also be produced as a result of infection of RSV and there is increasing evidence that children who develop severe RSV-associated bronchiolitis are at a greater risk of developing asthma later in life. This review summarizes the contribution of a newly described cell type, group 2 innate lymphoid cells (ILC2), and epithelial-derived alarmin proteins that activate ILC2, including IL-33, IL-25, thymic stromal lymphopoietin (TSLP), and high mobility group box 1 (HMGB1). ILC2 activation leads to the production of type 2 cytokines and the induction of a type 2 response during RSV infection. Intervening in this innate type 2 inflammatory pathway may have therapeutic implications for severe RSV-induced disease.
Collapse
Affiliation(s)
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA;
| |
Collapse
|
31
|
Jiang XH, Li CQ, Feng GY, Luo MJ, Sun QX. One-Week Nebulization of Mycobacterium vaccae Can Protect Against Pulmonary Respiratory Syncytial Virus Infection in Balb/c Mice. J Aerosol Med Pulm Drug Deliv 2020; 33:249-257. [PMID: 32301643 DOI: 10.1089/jamp.2019.1573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Respiratory syncytial virus (RSV) infection is the most common cause of acute lower respiratory tract infection in children, leading to their death. Currently, no effective prevention and treatment methods for RSV infection are available. RSV and many other unknown viruses pose a serious threat to human health. Our previous study demonstrated that Mycobacterium vaccae nebulization can protect against allergic asthma. As RSV infection and asthma are closely related, we hypothesized that M. vaccae could protect against pulmonary RSV infection. Therefore, we evaluated the effect of M. vaccae on RSV infection in Balb/c mice. Methods: The mice were randomized into three groups: normal, RSV, and M. vaccae. One week before the RSV infection model was established, the mice in the M. vaccae group were nebulized with M. vaccae. On the fourth day after RSV infection, airway responsiveness, airway inflammation, pulmonary RSV infection, mRNA levels of pulmonary toll-like receptor (TLR) 7 and TLR8, and pulmonary NF09, acetylcholine, and epidermal growth factor regulator (EGFR) expression levels in all mice were measured. Results: The airway inflammation in the M. vaccae group was alleviated compared with that in the RSV group. In the M. vaccae group, the pulmonary mRNA level of RSV and the pulmonary expression levels of NF09, acetylcholine, and EGFR were decreased considerably, whereas the mRNA levels of TLR7 and TLR8 were increased significantly. Conclusions: One-week nebulization of M. vaccae can protect against RSV infection in Balb/c mice. The mechanism involves the regulation of neurotransmitters and expression of TLR7, TLR8, and EGFR.
Collapse
Affiliation(s)
- Xiao-Hong Jiang
- Department of Geriatric Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao-Qian Li
- Department of Geriatric Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guang-Yi Feng
- Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Ming-Jie Luo
- Department of Respiratory Medicine, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Qi-Xiang Sun
- The Graduate School of Guangxi Medical University, Nanning, China
| |
Collapse
|
32
|
Kozhikhova KV, Shilovskiy IP, Shatilov AA, Timofeeva AV, Turetskiy EA, Vishniakova LI, Nikolskii AA, Barvinskaya ED, Karthikeyan S, Smirnov VV, Kudlay DA, Andreev SM, Khaitov MR. Linear and dendrimeric antiviral peptides: design, chemical synthesis and activity against human respiratory syncytial virus. J Mater Chem B 2020; 8:2607-2617. [DOI: 10.1039/c9tb02485a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Novel artificial peptides possess anti-RSV activity through a combination of two mechanisms: direct nonspecific destabilization of the viral envelope and competitive interaction with the RSV cellular receptor.
Collapse
Affiliation(s)
| | | | - Artem A. Shatilov
- NRC Institute Immunology FMBA
- Moscow
- Russian Federation
- Sechenov First Moscow State Medical University
- Moscow
| | - Anastasiia V. Timofeeva
- NRC Institute Immunology FMBA
- Moscow
- Russian Federation
- Sechenov First Moscow State Medical University
- Moscow
| | - Evgeny A. Turetskiy
- NRC Institute Immunology FMBA
- Moscow
- Russian Federation
- Sechenov First Moscow State Medical University
- Moscow
| | | | | | | | | | - Valeriy V. Smirnov
- NRC Institute Immunology FMBA
- Moscow
- Russian Federation
- Sechenov First Moscow State Medical University
- Moscow
| | | | | | | |
Collapse
|
33
|
Karampatsas K, Kong J, Cohen J. Bronchiolitis: an update on management and prophylaxis. Br J Hosp Med (Lond) 2019; 80:278-284. [PMID: 31059347 DOI: 10.12968/hmed.2019.80.5.278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bronchiolitis is an acute respiratory illness that is the leading cause of hospitalization in young children less than 2 years of age in the UK. Respiratory syncytial virus is the most common virus associated with bronchiolitis and has the highest disease severity, mortality and cost. Bronchiolitis is generally a self-limiting condition, but can have serious consequences in infants who are very young, premature, or have underlying comorbidities. Management of bronchiolitis in the UK is guided by the National Institute for Health and Care Excellence (2015) guidance. The mainstays of management are largely supportive, consisting of fluid management and respiratory support. Pharmacological interventions including nebulized bronchodilators, steroids and antibiotics generally have limited or no evidence of efficacy and are not advised by National Institute of Health and Care Excellence. Antiviral therapeutics remain in development. As treatments are limited, there have been extensive efforts to develop vaccines, mainly targeting respiratory syncytial virus. At present, the only licensed product is a monoclonal antibody for passive immunisation. Its cost restricts its use to those at highest risk. Vaccines for active immunisation of pregnant women and young infants are also being developed.
Collapse
Affiliation(s)
- Konstantinos Karampatsas
- Specialist Registrar in Paediatrics, Paediatric and Adolescent Division, University College Hospitals NHS Foundation Trust, London NW1 2BU
| | - Jonathan Kong
- Specialty Trainee in Paediatrics, Paediatric and Adolescent Division, University College Hospitals NHS Foundation Trust, London
| | - Jonathan Cohen
- Consultant in General Paediatrics and Paediatric Infectious Diseases, Paediatric and Adolescent Division, University College Hospitals NHS Foundation Trust, London
| |
Collapse
|
34
|
Meeting report: WHO consultation on Respiratory Syncytial Virus (RSV) vaccine development, Geneva, 25–26 April 2016. Vaccine 2019; 37:7355-7362. [DOI: 10.1016/j.vaccine.2017.02.068] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 11/23/2022]
|
35
|
Sanders SL, Agwan S, Hassan M, van Driel ML, Del Mar CB, Cochrane Acute Respiratory Infections Group. Immunoglobulin treatment for hospitalised infants and young children with respiratory syncytial virus infection. Cochrane Database Syst Rev 2019; 8:CD009417. [PMID: 31446622 PMCID: PMC6708604 DOI: 10.1002/14651858.cd009417.pub2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Millions of children are hospitalised due to respiratory syncytial virus (RSV) infection every year. Treatment is supportive, and current therapies (e.g. inhaled bronchodilators, epinephrine, nebulised hypertonic saline, and corticosteroids) are ineffective or have limited effect. Respiratory syncytial virus immunoglobulin is sometimes used prophylactically to prevent hospital admission from RSV-related illness. It may be considered for the treatment of established severe RSV infection or for treatment in an immunocompromised host, although it is not licenced for this purpose. It is unclear whether immunoglobulins improve outcomes when used as a treatment for established RSV infection in infants and young children admitted to hospital. OBJECTIVES: To assess the effects of immunoglobulins for the treatment of RSV-proven lower respiratory tract infections in children aged up to three years, admitted to hospital. SEARCH METHODS: We searched the Cochrane Central Register of Controlled Trials (CENTRAL), which contains the Cochrane Acute Respiratory Infections Group's Specialised Register, Ovid MEDLINE, Embase, CINAHL, and Web of Science (from inception to 6 November 2018) with no restrictions. We searched two trial registries for ongoing trials (to 30 March 2018) and checked the reference lists of reviews and included articles for additional studies. SELECTION CRITERIA Randomised controlled trials comparing immunoglobulins with placebo in hospitalised infants and children aged up to three years with laboratory-diagnosed RSV lower respiratory tract infection. DATA COLLECTION AND ANALYSIS Two review authors independently selected trials, assessed risk of bias, and extracted data. We assessed evidence quality using GRADE. MAIN RESULTS We included seven trials involving 486 infants and children aged up to three years. The immunoglobulin preparations used in these trials included anti-RSV immunoglobulin and the monoclonal antibody preparations palivizumab and motavizumab. We assessed the primary outcomes of mortality, length of hospital stay, and adverse events as providing low- or very low-certainty evidence due to risk of bias and imprecision. All trials were conducted at sites in high-income countries (USA, Chile, New Zealand, Australia), with two studies including a site in a middle-income country (Panama). Five of the seven studies were "supported" or "sponsored" by the trial drug manufacturers. We found no evidence of a difference between immunoglobulins and placebo for mortality (risk ratio (RR) 0.87, 95% confidence interval (CI) 0.14 to 5.27; 3 trials; 196 children; 4 deaths; 2 deaths amongst 98 children receiving immunoglobulins, and 2 deaths amongst 98 children receiving placebo. One additional death occurred in a fourth trial, however, the study group of the child was not known and the data were not included in the analysis; very low-certainty evidence), and length of hospitalisation (mean difference -0.70, 95% CI -1.83 to 0.42; 5 trials; 324 children; low-certainty evidence). There was no evidence of a difference between immunoglobulins and placebo in adverse events of any severity or seriousness (reported in five trials) or serious adverse events (four trials) (RR for any severity 1.18, 95% CI 0.78 to 1.78; 340 children; low-certainty evidence, and for serious adverse events 1.08, 95% CI 0.65 to 1.79; 238 children; low-certainty evidence).We found no evidence of a significant difference between immunoglobulins and placebo for any of our secondary outcomes. We identified one ongoing trial. AUTHORS' CONCLUSIONS We found insufficient evidence of a difference between immunoglobulins and placebo for any review outcomes. We assessed the evidence for the effects of immunoglobulins when used as a treatment for RSV lower respiratory tract infection in hospitalised infants and young children as of low or very low certainty due to risk of bias and imprecision. We are uncertain of the effects of immunoglobulins on these outcomes, and the true effect may be substantially different from the effects reported in this review. All trials were conducted in high-income countries, and data from populations in which the rate of death from RSV infection is higher are lacking.
Collapse
Affiliation(s)
- Sharon L Sanders
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)University DriveRobinaGold CoastQueenslandAustralia4229
| | - Sushil Agwan
- Gold Coast University Hospital1 Hospital BoulevardSouthportGold CoastQueenslandAustralia4215
| | - Mohamed Hassan
- Gold Coast University Hospital1 Hospital BoulevardSouthportGold CoastQueenslandAustralia4215
| | - Mieke L van Driel
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)University DriveRobinaGold CoastQueenslandAustralia4229
- The University of QueenslandPrimary Care Clinical Unit, Faculty of MedicineBrisbaneQueenslandAustralia4029
- Ghent UniversityDepartment of Family Medicine and Primary Health CareCampus UZ 6K3, Corneel Heymanslaan 10GhentBelgium9000
| | - Chris B Del Mar
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)University DriveRobinaGold CoastQueenslandAustralia4229
| | | |
Collapse
|
36
|
Randomized, Double-Blind, Placebo-Controlled, Single-Ascending-Dose Study of the Penetration of a Monoclonal Antibody Combination (ASN100) Targeting Staphylococcus aureus Cytotoxins in the Lung Epithelial Lining Fluid of Healthy Volunteers. Antimicrob Agents Chemother 2019; 63:AAC.00350-19. [PMID: 31138568 PMCID: PMC6658777 DOI: 10.1128/aac.00350-19] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
ASN100 is a novel antibody combination of two fully human IgG1(κ) monoclonal antibodies (MAbs), ASN-1 and ASN-2, which neutralize six Staphylococcus aureus cytotoxins, alpha-hemolysin (Hla) and five bicomponent leukocidins. We assessed the safety, tolerability, and serum and lung pharmacokinetics of ASN100 in a randomized, double-blind, placebo-controlled single-dose-escalation first-in-human study. ASN100 is a novel antibody combination of two fully human IgG1(κ) monoclonal antibodies (MAbs), ASN-1 and ASN-2, which neutralize six Staphylococcus aureus cytotoxins, alpha-hemolysin (Hla) and five bicomponent leukocidins. We assessed the safety, tolerability, and serum and lung pharmacokinetics of ASN100 in a randomized, double-blind, placebo-controlled single-dose-escalation first-in-human study. Fifty-two healthy volunteers were enrolled and randomized to receive either ASN-1, ASN-2, a combination of both MAbs (ASN100), or a corresponding placebo. Thirty-two subjects in the double-blind dose escalation portion of the study received ASN-1 or ASN-2 at a 200-, 600-, 1,800-, or 4,000-mg dose, or placebo. Eight subjects received both MAbs simultaneously in a 1:1 ratio (ASN100) at 3,600 or 8,000 mg, or they received placebos. Twelve additional subjects received open-label ASN100 at 3,600 or 8,000 mg to assess the pharmacokinetics of ASN-1 and ASN-2 in epithelial lining fluid (ELF) by bronchoalveolar lavage fluid sampling. Subjects were monitored for 98 days (double-blind cohorts) or 30 days (open-label cohorts) for safety assessment. No dose-limiting toxicities were observed, and all adverse events were mild and transient, with only two adverse events considered possibly related to the investigational product. ASN100 exhibited linear serum pharmacokinetics with a half-life of approximately 3 weeks and showed detectable penetration into the ELF. No treatment-emergent anti-drug antibody responses were detected. The toxin neutralizing potency of ASN100 in human serum was confirmed up to 58 days postdosing. The favorable safety profile, ELF penetration, and maintained functional activity in serum supported the further clinical development of ASN100.
Collapse
|
37
|
Jun S, Sebastianski M, Featherstone R, Robinson J. Palivizumab and prevention of childhood respiratory syncytial viral infection: protocol for a systematic review and meta-analysis of breakthrough infections. BMJ Open 2019; 9:e029832. [PMID: 31340973 PMCID: PMC6661690 DOI: 10.1136/bmjopen-2019-029832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Childhood respiratory syncytial virus (RSV) infection is a global phenomenon that can lead to fatal respiratory illness. Palivizumab is a drug that is routinely used in affluent countries as a prophylaxis against RSV infection; nevertheless, breakthrough infections are often reported. In light of new findings on potential RSV resistance to palivizumab, an up-to-date synthesis of evidence on effectiveness is needed. Furthering existing reviews, a broadened scope to better reflect effectiveness in a 'real world' clinical context is also important. This systematic review and meta-analysis will enhance our understanding of the effectiveness of palivizumab in varying populations of children. Findings from this review will inform recommendations for best practices regarding palivizumab use for childhood RSV infection as well as research priorities in RSV vaccine development. METHODS AND ANALYSIS We will conduct a systematic review of primary population-based studies that examine the incidence of palivizumab breakthrough infections in children, published between 1997 to present. In collaboration with a research librarian, four electronic databases (MEDLINE, Embase, Cochrane Library, Web of Science) and additional sources will be searched. Study screening and quality assessment will be performed in duplicate. Data will be extracted by one reviewer, with partial and random verification by a second reviewer. The primary outcomes to assess breakthrough RSV infection will be hospitalisation, length of stay and the need for intensive care unit admission and mechanical ventilation in children receiving palivizumab. The secondary outcome will be RSV-associated mortality. We will conduct a meta-analysis using pooled effectiveness data, and include subgroup analyses by patient comorbidities and drug compliance. Sensitivity analyses for risk of bias and study design will also be performed. ETHICS AND DISSEMINATION This systematic review will only include data from previously published literature and is therefore exempt from ethics approval. Final results will be disseminated through peer-reviewed publication and presented at academic conferences and scientific meetings engaging paediatric researchers and healthcare providers. Should findings from this review necessitate updates to current clinical practice guidelines, we intend to establish a working group to engage relevant health administrators and decision makers. PROSPERO REGISTRATION NUMBER CRD42019122120.
Collapse
Affiliation(s)
- Shelly Jun
- Alberta Strategy for Patient-Oriented Research (SPOR) Knowledge Translation Platform, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Meghan Sebastianski
- Alberta Strategy for Patient-Oriented Research (SPOR) Knowledge Translation Platform, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Robin Featherstone
- Alberta Strategy for Patient-Oriented Research (SPOR) Knowledge Translation Platform, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Alberta Research Centre for Health Evidence (ARCHE), Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Joan Robinson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
38
|
Tenenbaum T, Drechsel-Bäuerle U, Oberle D. Cardiorespiratory Events After Monoclonal Antibody Prophylaxis With Palivizumab. Pediatr Infect Dis J 2018; 37:e281-e282. [PMID: 30308606 DOI: 10.1097/inf.0000000000002017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Tobias Tenenbaum
- Pediatric Infectious Diseases and Pulmonology, University Children's Hospital Mannheim, Heidelberg University, Mannheim, Germany Department Safety of Medicinal Products and Medical Devices, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | | | | |
Collapse
|
39
|
Butt ML, Elliott L, Paes BA. Respiratory syncytial virus hospitalization and incurred morbidities the season after prophylaxis. Paediatr Child Health 2018; 23:441-446. [PMID: 30374219 PMCID: PMC6199632 DOI: 10.1093/pch/pxy046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES The primary objective of this study was to determine the incidence and incurred morbidities of Respiratory syncytial virus (RSV)-related hospitalization (RSVH), the season following completion of prophylaxis. METHODS A retrospective study was conducted of all infants enrolled in a prophylaxis clinic in one institution during the 2009 to 2014 RSV seasons. RSV infection was identified by Diseases codes and confirmed by RSV-positivity. Data were classified into five groups based on indications for prophylaxis. The incidence of RSVH was calculated. For each subgroup, differences in characteristics between children with and without RSVH were analyzed by independent t test or chi-square test. RESULTS During five RSV seasons, 827 infants were enrolled. RSVH incidence the season following prophylaxis was 2.1% (n=17/827). Children with chronic lung disease (CLD) had the highest RSVH incidence (7.7%; n=4/52) followed by preterms 33 to 35 weeks gestation (2.5%; n=4/162), those with complex medical disorders (2.2%; n=3/135), those with congenital heart disease (1.5%; n=1/66) and preterms less than or equal to 32 weeks gestation (1.2%; n=5/412). There was no statistically significant association between indications for prophylaxis and RSVH (Fisher exact test, P=0.060). The odds of RSVH were 4.9 times greater (odds ratio [OR]=4.9; 95% CI: 1.53, 15.55; P=0.007) in CLD compared to those without CLD. The median length of RSVH stay was 4 days; 58.8% (n=10/17) required oxygen (median 1 day); 29.4% (n=5/17) required intensive care. CONCLUSIONS Infants with CLD are at highest risk for RSVH in the season postprophylaxis and may merit palivizumab for more than two seasons dependent on disease severity. However, larger prospective studies are necessary to confirm the findings before embarking on a strategy of providing prophylaxis for a third RSV season.
Collapse
Affiliation(s)
- Michelle L Butt
- School of Nursing, McMaster University, Hamilton, Ontario
- Department of Paediatrics, McMaster University, Hamilton, Ontario
| | - LouAnn Elliott
- Paediatrics, McMaster Children’s Hospital, Hamilton, Ontario
| | - Bosco A Paes
- Department of Paediatrics, McMaster University, Hamilton, Ontario
| |
Collapse
|
40
|
Pichler K, Assadian O, Berger A. Viral Respiratory Infections in the Neonatal Intensive Care Unit-A Review. Front Microbiol 2018; 9:2484. [PMID: 30405557 PMCID: PMC6202802 DOI: 10.3389/fmicb.2018.02484] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 09/28/2018] [Indexed: 01/24/2023] Open
Abstract
Although infrequent, respiratory viral infections (RVIs) during birth hospitalization have a significant impact on short- and long-term morbidity in term and preterm neonates. RVI have been associated with increased length of hospital stay, severe disease course, unnecessary antimicrobial exposure and nosocomial outbreaks in the neonatal intensive care unit (NICU). Virus transmission has been described to occur via health care professionals, parents and other visitors. Most at risk are infants born prematurely, due to their immature immune system and the fact that they stay in the NICU for a considerable length of time. A prevalence of RVIs in the NICU in symptomatic infants of 6–30% has been described, although RVIs are most probably underdiagnosed, since testing for viral pathogens is not performed routinely in symptomatic patients in many NICUs. Additional challenges are the wide range of clinical presentation of RVIs, their similarity to bacterial infections and the unreliable detection methods prior to the era of molecular biology based technologies. In this review, current knowledge of early-life RVI in the NICU is discussed. Reviewed viral pathogens include human rhinovirus, respiratory syncytial virus and influenza virus, and discussed literature is restricted to reports based on modern molecular biology techniques. The review highlights therapeutic approaches and possible preventive strategies. Furthermore, short- and long-term consequences of RVIs in infants hospitalized in the NICU are discussed.
Collapse
Affiliation(s)
- Karin Pichler
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ojan Assadian
- Department for Hospital Epidemiology and Infection Control, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Mirra V, Ullmann N, Cherchi C, Onofri A, Paglietti MG, Cutrera R. Respiratory syncytial virus prophylaxis and the "special population". Minerva Pediatr 2018; 70:589-599. [PMID: 30334623 DOI: 10.23736/s0026-4946.18.05316-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bronchiolitis is the most frequent airway infection in the first 2 years of life, and the respiratory syncytial virus (RSV) is the most frequently responsible virus. In selected high-risk groups, RSV may cause severe respiratory disease leading to hospitalization, need for mechanical ventilation, and even death. These high-risk groups include children with congenital heart disease, infants with neuromuscular impairment, cystic fibrosis, Down Syndrome, immunodeficiency syndromes and others specific conditions. In these high-risk populations defined in literature as "special population", a 3- to 10-fold increase in the rate of RSV hospitalization has been observed, justifying RSV specific prophylaxis with palivizumab, a monoclonal antibody that binds a viral glycoprotein epitope and blocks the link between RSV and target cell. Evidence of safety and efficacy of RSV prophylaxis in these populations is lacking. Given the low incidence of these conditions, randomized clinical trials are not feasible. The purpose of this paper is to give an update from the literature of various conditions at higher risk to develop severe RSV infection, and to offer an overview of the efficacy of palivizumab in preventing RSV infection in these specific populations.
Collapse
Affiliation(s)
- Virginia Mirra
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy.,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| | - Nicola Ullmann
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy - .,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| | - Claudio Cherchi
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy.,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| | - Alessandro Onofri
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy.,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| | - Maria G Paglietti
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy.,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| | - Renato Cutrera
- Unit of Paediatric Pulmonology and Respiratory Intermediate Care, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy.,Unit of Sleep and Long-term Ventilation, Academic Department of Paediatrics, Paediatric Hospital "Bambino Gesù" Research Institute, Rome, Italy
| |
Collapse
|
42
|
Alharbi AS, Alqwaiee M, Al-Hindi MY, Mosalli R, Al-Shamrani A, Alharbi S, Yousef A, Al Aidaroos A, Alahmadi T, Alshammary A, Miqdad A, Said Y, Alnemri A. Bronchiolitis in children: The Saudi initiative of bronchiolitis diagnosis, management, and prevention (SIBRO). Ann Thorac Med 2018; 13:127-143. [PMID: 30123331 PMCID: PMC6073791 DOI: 10.4103/atm.atm_60_18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022] Open
Abstract
Bronchiolitis is the leading cause of admissions in children less than two years of age. It has been recognized as highly debated for many decades. Despite the abundance of literature and the well-recognized importance of palivizumab in the high risk groups, and despite the existence of numerous, high-quality, recent guidelines on bronchiolitis, the number of admissions continues to increase. Only supportive therapy and few therapeutic interventions are evidence based and proved to be effective. Since Respiratory Syncytial Virus (RSV) is the major cause of bronchiolitis, we will focus on this virus mostly in high risk groups like the premature babies and children with chronic lung disease and cardiac abnormalities. Further, the prevention of RSV with palivizumab in the high risk groups is effective and well known since 1998; we will discuss the updated criteria for allocating infants to this treatment, as this medication is expensive and should be utilized in the best condition. Usually, diagnosis of bronchiolitis is not challenging, however there has been historically no universally accepted and validated scoring system to assess the severity of the condition. Severe RSV, especially in high risk children, is unique because it can cause serious respiratory sequelae. Currently there is no effective curative treatment for bronchiolitis. The utility of different therapeutic interventions is worth a discussion.
Collapse
Affiliation(s)
- Adel S. Alharbi
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Mansour Alqwaiee
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Mohammed Y Al-Hindi
- King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Department of Pediatric, Ministry of National Guard, Jeddah, Saudi Arabia
| | - Rafat Mosalli
- Department of Pediatrics, Umm Al Qura university, Makkah, Saudi Arabia
| | - Abdullah Al-Shamrani
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Saleh Alharbi
- Department of Pediatrics, Umm Al Qura university, Makkah, Saudi Arabia
| | - Abdullah Yousef
- Department of Pediatrics, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Amal Al Aidaroos
- Department of Pediatrics, Prince Sultan Military City, Ministry of Defense, Riyadh, Saudi Arabia
| | - Turki Alahmadi
- King Abdulaziz University, College of Medicine, Department of Pediatrics, Jeddah, Saudi Arabia
| | | | - Abeer Miqdad
- Department of Pediatrics, Security forces hospital, Riyadh, Saudi Arabia
| | - Yazan Said
- King Fahad Specialist Hospital, Ministry of Health, Dammam, Saudi Arabia
| | - Abdulrahman Alnemri
- College of Medicine, Peadiatric Department, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
German P, Xin Y, Chien JW, Weng W, Mackman R, Lewis SA, Meng A, Ling J, Mathias A. Phase 1 First-in-Human, Single- and Multiple-Ascending Dose, and Food Effect Studies to Assess the Safety, Tolerability, and Pharmacokinetics of Presatovir for the Treatment of Respiratory Syncytial Virus Infection. J Clin Pharmacol 2018; 58:1025-1034. [PMID: 29663420 DOI: 10.1002/jcph.1112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022]
Abstract
Respiratory syncytial virus (RSV)-associated respiratory tract infection is a leading cause of hospitalizations in infants for which no effective treatment exists. RSV infection is also an important cause of respiratory disease in adults and immunocompromised patients. Presatovir (GS-5806) is an orally bioavailable antiviral agent that inhibits fusion of RSV with host cell membranes. Here, results from 2 phase 1 studies that evaluated safety, tolerability, and pharmacokinetics of presatovir in healthy adults following administration of single and multiple (7 days) once- or twice-daily ascending doses (first-in-human study) and in the presence or absence of food (food effect study) are described. Presatovir exhibited favorable safety and pharmacokinetic profiles that supported once-daily dosing. Presatovir exposure increased in an approximately dose-proportional manner across the evaluated dose range (single doses 25-300 mg; multiple doses 10-75 mg once daily for 7 days). Administration of presatovir with a high-fat meal did not alter exposure, supporting administration without regard to a meal in further clinical studies. These data were subsequently used to inform presatovir dosing regimens in a phase 2a challenge study of adults experimentally infected with RSV. Collectively, results from phase 1 evaluations and a phase 2a challenge study support further clinical investigation of presatovir for the treatment of RSV infection.
Collapse
Affiliation(s)
| | - Yan Xin
- Gilead Sciences, Inc, Foster City, CA, USA
| | | | | | | | | | - Amy Meng
- Gilead Sciences, Inc, Foster City, CA, USA
| | - John Ling
- Gilead Sciences, Inc, Foster City, CA, USA
| | | |
Collapse
|
44
|
Abstract
Preterm infants are at an increased risk of morbidity and mortality from vaccine-preventable diseases. Despite this, delays in routine immunization of preterm infants are common. Available guidelines clearly state that they should be immunized according to chronological age, irrespective of gestational age and birth weight or current weight. In this article, we try to assuage parental and provider doubts by reviewing data about immunogenicity, safety, and responses to routine immunizations in preterm infants with and without comorbidities. We also look at evidence for other strategies to help protect this fragile population. [Pediatr Ann. 2018;47(4):e147-e153.].
Collapse
|
45
|
Efficacy of mucosal polyanhydride nanovaccine against respiratory syncytial virus infection in the neonatal calf. Sci Rep 2018; 8:3021. [PMID: 29445124 PMCID: PMC5813012 DOI: 10.1038/s41598-018-21292-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is a leading cause of severe acute lower respiratory tract infection in infants and children worldwide. Bovine RSV (BRSV) is closely related to HRSV and a significant cause of morbidity in young cattle. BRSV infection in calves displays many similarities to RSV infection in humans, including similar age dependency and disease pathogenesis. Polyanhydride nanoparticle-based vaccines (i.e., nanovaccines) have shown promise as adjuvants and vaccine delivery vehicles due to their ability to promote enhanced immunogenicity through the route of administration, provide sustained antigen exposure, and induce both antibody- and cell-mediated immunity. Here, we developed a novel, mucosal nanovaccine that encapsulates the post-fusion F and G glycoproteins from BRSV into polyanhydride nanoparticles and determined the efficacy of the vaccine against RSV infection using a neonatal calf model. Calves receiving the BRSV-F/G nanovaccine exhibited reduced pathology in the lungs, reduced viral burden, and decreased virus shedding compared to unvaccinated control calves, which correlated with BRSV-specific immune responses in the respiratory tract and peripheral blood. Our results indicate that the BRSV-F/G nanovaccine is highly immunogenic and, with optimization, has the potential to significantly reduce the disease burden associated with RSV infection in both humans and animals.
Collapse
|
46
|
Eichinger KM, Kosanovich JL, Empey KM. Localization of the T-cell response to RSV infection is altered in infant mice. Pediatr Pulmonol 2018; 53:145-153. [PMID: 29115050 PMCID: PMC5775046 DOI: 10.1002/ppul.23911] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/22/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections worldwide, causing disproportionate morbidity and mortality in infants and children. Infants with stronger Th1 responses have less severe disease, yet little is known about the infant T-cell response within the air space. Thus, we tested the hypothesis that RSV infected infant mice would have quantitative and qualitative deficiencies in CD4+ and CD8+ T-cell populations isolated from the bronchoalveolar lavage when compared to adults and that local delivery of IFN-γ would increase airway CD4+ Tbet+ and CD8+ Tbet+ T-cell responses. METHODS We compared the localization of T-cell responses in RSV-infected infant and adult mice and investigated the effects of local IFN-γ administration on infant cellular immunity. RESULTS Adult CD8+ CD44HI and CD4+ CD44HI Tbet+ T-cells accumulated in the alveolar space whereas CD4+ CD44HI Tbet+ T-cells were evenly distributed between the infant lung tissue and airway and infant lungs contained higher frequencies of CD8+ T-cells. Delivery of IFN-γ to the infant airway failed to increase the accumulation of T-cells in the airspace and unexpectedly reduced CD4+ CD44HI Tbet+ T-cells. However, intranasal IFN-γ increased RSV F protein-specific CD8+ T-cells in the alveolar space. CONCLUSION Together, these data suggest that quantitative and qualitative defects exist in the infant T-cell response to RSV but early, local IFN-γ exposure can increase the CD8+ RSV-specific T-cell response.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica L Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Janet S, Broad J, Snape MD. Respiratory syncytial virus seasonality and its implications on prevention strategies. Hum Vaccin Immunother 2018; 14:234-244. [PMID: 29194014 PMCID: PMC5791579 DOI: 10.1080/21645515.2017.1403707] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/06/2017] [Indexed: 01/04/2023] Open
Abstract
With maternal and infant vaccines against respiratory syncytial virus (RSV) in development, it is timely to consider how the deployment of these vaccines might vary according to local RSV disease seasonality. In temperate regions RSV infection is predictably limited to a period of 3 to 5 months, while in tropical regions disease seasonality is often both more variable and more prolonged. Accordingly, in tropical regions a year-round immunisation schedule for both maternal and infant immunisation might be appropriate. In contrast, in temperate regions the benefit of year-round maternal immunisation would be heavily dependent on the duration of protection this provided, potentially necessitating a strategy directed at children due to be born in the months immediately prior to the RSV season. This review will consider the impact of seasonality on maternal and infant immunisation strategies against RSV, and the potential of an alternative approach of passive immunisation for all infants immediately prior to the RSV season.
Collapse
Affiliation(s)
- Sophie Janet
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jonathan Broad
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Matthew D. Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
48
|
Hashimoto K, Hosoya M. Neutralizing epitopes of RSV and palivizumab resistance in Japan. Fukushima J Med Sci 2017; 63:127-134. [PMID: 28867684 PMCID: PMC5792496 DOI: 10.5387/fms.2017-09] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/20/2017] [Indexed: 01/26/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is one of the most important viral pathogen related to acute lower respiratory infection in young children. The virus surface envelope contains the G, F, and SH proteins as spike proteins. The F protein is considered to be a major antigenic target for the neutralizing (NT) epitope as only the F protein is essential for cell infection among the three viral envelope proteins, and it is more highly conserved than the G protein. Recently, four antigenic targets related to NT activity have been reported;site I, site II, site IV, and site zero (0). Site II is the target for palivizumab used throughout the world to suppress severe RSV infection as passive immunity in high-risk children since 1998. Under the recent conditions in which indications for palivizumab administered subjects are being expanded, palivizumab-resistant mutations have been confirmed overseas in children with RSV infection, although they remain infrequent. Therefore, continuous genetic analysis of the palivizumab-binding region of the F protein is necessary. In addition, as vaccine development progresses, RSV infection control is expected to improve greatly over the next decade.
Collapse
Affiliation(s)
- Koichi Hashimoto
- Department of Pediatrics, School of Medicine, Fukushima Medical University
| | - Mitsuaki Hosoya
- Department of Pediatrics, School of Medicine, Fukushima Medical University
| |
Collapse
|
49
|
Wong SK, Li A, Lanctôt KL, Paes B. Adherence and outcomes: a systematic review of palivizumab utilization. Expert Rev Respir Med 2017; 12:27-42. [DOI: 10.1080/17476348.2018.1401926] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sophie K. Wong
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Abby Li
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Krista L. Lanctôt
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Bosco Paes
- Division of Neonatology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
50
|
One‐year observational study of palivizumab prophylaxis on infants at risk for respiratory syncytial virus infection in Latin America. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|