1
|
Peña-López Y, Sabater-Riera J, Raj P. Severe respiratory syncytial virus disease. JOURNAL OF INTENSIVE MEDICINE 2024; 4:405-416. [PMID: 39310066 PMCID: PMC11411437 DOI: 10.1016/j.jointm.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 09/25/2024]
Abstract
The burden of respiratory syncytial virus (RSV) disease is widely recognized. Main risk factors for severe disease, such as extreme ages, chronic cardiopulmonary conditions, and immunosuppression, typically coincide with poorer outcomes. While the majority of RSV hospitalizations involve healthy children, a higher proportion of hospitalized adults with underlying conditions need intensive care. Presently, treatment primarily consists of supportive measures. RSV-induced wheezing should be distinguished from respiratory tract thickening, without response to bronchodilators. Obstructive RSV disease frequently overlaps with viral pneumonia. Non-invasive mechanical ventilation and high-flow oxygen therapy represented significant advancements in the management of severe RSV disease in children and may also hold considerable importance in specific phenotypes of RSV disease in adults. Most severe infections manifest with refractory hypoxemia necessitating more advanced ventilatory support and/or extracorporeal membrane oxygenation therapy. Although bacterial co-infection rates are low, they have been associated with worse outcomes. Antibiotic prescription rates are high. Accurately diagnosing bacterial co-infections remains a challenge. Current evidence and antibiotic stewardship policies advise against indiscriminate antibiotic usage, even in severe cases. The role of currently developing antiviral therapies in severe RSV disease will be elucidated in the coming years, contingent upon the success of new vaccines and immune passive strategies involving nirsevimab.
Collapse
Affiliation(s)
- Yolanda Peña-López
- Microbiome Research Laboratory (MRL), Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Pediatric Critical Care Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Global Health eCore, Vall d'Hebron Institute of Research, Barcelona, Spain
| | - Joan Sabater-Riera
- Intensive Care Department, Servei de Medicina Intensiva, IDIBELL-Hospital Universitari de Bellvitge, L´Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Prithvi Raj
- Microbiome Research Laboratory (MRL), Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Peng R, Chen C, Chen Q, Zhang Y, Huang R, Zhang Y, Li J. Global progress in clinical research on human respiratory syncytial virus vaccines. Front Microbiol 2024; 15:1457703. [PMID: 39286350 PMCID: PMC11402711 DOI: 10.3389/fmicb.2024.1457703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Human respiratory syncytial virus (hRSV) not only affects newborns but also older adults, contributing to a substantial worldwide burden of disease. However, only three approved hRSV vaccines remain commercially available to date. The development of a safe, practical and broad-spectrum vaccine suitable for all age groups remains extremely challenging. Using five different approaches-live-attenuated, recombinant-vector, subunit, particle-based, and mRNA-nearly 30 hRSV vaccine candidates are currently conducting clinical trials worldwide; moreover, > 30 vaccines are under preclinical evaluation. This review presents a comprehensive overview of these hRSV vaccines along with prospects for the development of infectious disease vaccines in the post-COVID-19 pandemic era.
Collapse
Affiliation(s)
- Ruofan Peng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qian Chen
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Yuwen Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Renjin Huang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
3
|
Luganini A, Sibille G, Pavan M, Mello Grand M, Sainas S, Boschi D, Lolli ML, Chiorino G, Gribaudo G. Mechanisms of antiviral activity of the new hDHODH inhibitor MEDS433 against respiratory syncytial virus replication. Antiviral Res 2023; 219:105734. [PMID: 37852322 DOI: 10.1016/j.antiviral.2023.105734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Human respiratory syncytial virus (RSV) is an important cause of acute lower respiratory infections, for which no effective drugs are currently available. The development of new effective anti-RSV agents is therefore an urgent priority, and Host-Targeting Antivirals (HTAs) can be considered to target RSV infections. As a contribution to this antiviral avenue, we have characterized the molecular mechanisms of the anti-RSV activity of MEDS433, a new inhibitor of human dihydroorotate dehydrogenase (hDHODH), a key cellular enzyme of de novo pyrimidine biosynthesis. MEDS433 was found to exert a potent antiviral activity against RSV-A and RSV-B in the one-digit nanomolar range. Analysis of the RSV replication cycle in MEDS433-treated cells, revealed that the hDHODH inhibitor suppressed the synthesis of viral genome, consistently with its ability to specifically target hDHODH enzymatic activity. Then, the capability of MEDS433 to induce the expression of antiviral proteins encoded by Interferon-Stimulated Genes (ISGs) was identified as a second mechanism of its antiviral activity against RSV. Indeed, MEDS433 stimulated secretion of IFN-β and IFN-λ1 that, in turn, induced the expression of some ISG antiviral proteins, such as IFI6, IFITM1 and IRF7. Singly expression of these ISG proteins reduced RSV-A replication, thus likely contributing to the overall anti-RSV activity of MEDS433. Lastly, MEDS433 proved to be effective against RSV-A replication even in a primary human small airway epithelial cell model. Taken as a whole, these observations provide new insights for further development of MEDS433, as a promising candidate to develop new strategies for treatment of RSV infections.
Collapse
Affiliation(s)
- Anna Luganini
- Department of Life Sciences and Systems Biology, University of Torino, 10123, Torino, Italy
| | - Giulia Sibille
- Department of Life Sciences and Systems Biology, University of Torino, 10123, Torino, Italy
| | - Marta Pavan
- Department of Life Sciences and Systems Biology, University of Torino, 10123, Torino, Italy
| | | | - Stefano Sainas
- Department of Drug Sciences and Technology, University of Torino, 10125, Torino, Italy
| | - Donatella Boschi
- Department of Drug Sciences and Technology, University of Torino, 10125, Torino, Italy
| | - Marco L Lolli
- Department of Drug Sciences and Technology, University of Torino, 10125, Torino, Italy
| | | | - Giorgio Gribaudo
- Department of Life Sciences and Systems Biology, University of Torino, 10123, Torino, Italy.
| |
Collapse
|
4
|
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
|
5
|
Saunders JL, Daniels IA, Edwards TL, Relich RF, Zhao Y, Smith LA, Gaston BM, Davis MD. Effects of pH alteration on respiratory syncytial virus in human airway epithelial cells. ERJ Open Res 2023; 9:00404-2022. [PMID: 37465558 PMCID: PMC10351676 DOI: 10.1183/23120541.00404-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/05/2023] [Indexed: 07/20/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a leading cause of respiratory distress and hospitalisation in the paediatric population. Low airway surface pH impairs antimicrobial host defence and worsens airway inflammation. Inhaled Optate safely raises airway surface pH in humans and raises intracellular pH in primary human airway epithelial cells (HAECs) in vitro. We aimed to determine whether raising intracellular pH with Optate would decrease infection and replication of RSV in primary HAECs. Methods We cultured HAECs from healthy subjects in both air-liquid interface and submerged conditions. We infected HAECs with green fluorescent protein-labelled RSV (GFP-RSV; multiplicity of infection=1) and treated them with Optate or PBS control. We collected supernatant after a 4-h incubation and then every 24 h. We used fluorescence intensity, fluorescent particle counts, plaque assays, Western blots and ELISA to quantitate infection. Results In submerged culture, fluorescence intensity decreased in Optate-treated cells (48 h p=0.0174, 72 h p≤0.001). Similarly, Optate treatment resulted in decreased fluorescent particle count (48 h p=0.0178, 72 h p=0.0019) and plaque-forming units (48 h p=0.0011, 72 h p=0.0148) from cell culture supernatant. In differentiated HAECs cultured at ALI, Optate treatment decreased fluorescence intensity (p≤0.01), GFP via Western blot and ELISA (p<0.0001), and RSV-fusion protein via ELISA (p=0.001). Additionally, RSV infection decreased as Optate concentration increased in a dose-dependent manner (p<0.001). Conclusions Optate inhibits RSV infection in primary HAECs in a dose-dependent manner. These findings suggest that Optate may have potential as an inhaled therapeutic for patients with RSV.
Collapse
Affiliation(s)
- Jessica L. Saunders
- Division of Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ivana A. Daniels
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Taiya L. Edwards
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ryan F. Relich
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Zhao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura A. Smith
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benjamin M. Gaston
- Division of Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael D. Davis
- Division of Pulmonology, Allergy and Sleep Medicine, Riley Hospital for Children, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
6
|
Abstract
Human respiratory syncytial virus (RSV) is a negative sense single-stranded RNA virus that can result in epidemics of seasonal respiratory infections. Generally, one of the two genotypes (A and B) predominates in a single season and alternate annually with regional variation. RSV is a known cause of disease and death at both extremes of ages in the pediatric and elderly, as well as immunocompromised populations. The clinical impact of RSV on the hospitalized adults has been recently clarified with the expanded use of multiplex molecular assays. Among adults, RSV can produce a wide range of clinical symptoms due to upper respiratory tract infections potentially leading to severe lower respiratory tract infections, as well as exacerbations of underlying cardiac and lung diseases. While supportive care is the mainstay of therapy, there are currently multiple therapeutic and preventative options under development.
Collapse
Affiliation(s)
- Hannah H Nam
- Department of Infectious Diseases, University of California, Irvine, Orange, California
| | - Michael G Ison
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
7
|
Rahman F, Libre C, Oleinikov A, Tcherniuk S. Chloroquine and pyrimethamine inhibit the replication of human respiratory syncytial virus A. J Gen Virol 2021; 102. [PMID: 34342560 DOI: 10.1099/jgv.0.001627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human respiratory syncytial virus (hRSV) is a major cause of respiratory illness in young children and can cause severe infections in the elderly or in immunocompromised adults. To date, there is no vaccine to prevent hRSV infections, and disease management is limited to preventive care by palivizumab in infants and supportive care for adults. Intervention with small-molecule antivirals specific for hRSV represents a good alternative, but no such compounds are currently approved. The investigation of existing drugs for new therapeutic purposes (drug repositioning) can be a faster approach to address this issue. In this study, we show that chloroquine and pyrimethamine inhibit the replication of human respiratory syncytial virus A (long strain) and synergistically increase the anti-replicative effect of ribavirin in cellulo. Moreover, chloroquine, but not pyrimethamine, inhibits hRSV replication in the mouse model. Our results show that chloroquine can potentially be an interesting compound for treatment of hRSV infection in monotherapy or in combination with other antivirals.
Collapse
Affiliation(s)
- Fryad Rahman
- Department of Biology, College of Science, University of Sulaimani, Kurdistan Region, Iraq.,Department of Molecular Biology, High Quality Laboratory, Anwar Sheikha Medical City, Sulaymaniyah, Iraq
| | - Camille Libre
- Cancer Research Center of Lyon, INSERM U1052 UMR CNRS 5286, Equipe labellisée Ligue Contre le Cancer, Université de Lyon, 69008 Lyon, France
| | - Andrew Oleinikov
- Charles E. Schmidt College of Medicine, Department of Biomedical Science, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, USA
| | - Sergey Tcherniuk
- Unité de Virologie et Immunologie Moléculaires, INRA, Université Paris SaclayJouy-en-Josas, France.,Department of Biological Sciences, Youth Academy of Sciences, Kiev, Ukraine
| |
Collapse
|
8
|
Global Molecular Epidemiology of Respiratory Syncytial Virus from the 2017-2018 INFORM-RSV Study. J Clin Microbiol 2020; 59:JCM.01828-20. [PMID: 33087438 PMCID: PMC7771447 DOI: 10.1128/jcm.01828-20] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children, resulting in annual epidemics worldwide. INFORM-RSV is a multiyear clinical study designed to describe the global molecular epidemiology of RSV in children under 5 years of age by monitoring temporal and geographical evolution of current circulating RSV strains, F protein antigenic sites, and their relationships with clinical features of RSV disease. During the pilot season (2017–2018), 410 RSV G-F gene sequences were obtained from 476 RSV-positive nasal samples collected from 8 countries (United Kingdom, Spain, The Netherlands, Finland, Japan, Brazil, South Africa, and Australia). Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children, resulting in annual epidemics worldwide. INFORM-RSV is a multiyear clinical study designed to describe the global molecular epidemiology of RSV in children under 5 years of age by monitoring temporal and geographical evolution of current circulating RSV strains, F protein antigenic sites, and their relationships with clinical features of RSV disease. During the pilot season (2017–2018), 410 RSV G-F gene sequences were obtained from 476 RSV-positive nasal samples collected from 8 countries (United Kingdom, Spain, The Netherlands, Finland, Japan, Brazil, South Africa, and Australia). RSV B (all BA9 genotype) predominated over RSV A (all ON1 genotype) globally (69.0% versus 31.0%) and in all countries except South Africa. Geographic clustering patterns highlighted wide transmission and continued evolution with viral spread. Most RSV strains were from infants of <1 year of age (81.2%), males (56.3%), and patients hospitalized for >24 h (70.5%), with no differences in subtype distribution. Compared to 2013 reference sequences, variations at F protein antigenic sites were observed for both RSV A and B strains, with high-frequency polymorphisms at antigenic site Ø (I206M/Q209R) and site V (L172Q/S173L/K191R) in RSV B strains. The INFORM-RSV 2017–2018 pilot season establishes an important molecular baseline of RSV strain distribution and sequence variability with which to track the emergence of new strains and provide an early warning system of neutralization escape variants that may impact transmission or the effectiveness of vaccines and MAbs under development.
Collapse
|
9
|
Elawar F, Oraby AK, Kieser Q, Jensen LD, Culp T, West FG, Marchant DJ. Pharmacological targets and emerging treatments for respiratory syncytial virus bronchiolitis. Pharmacol Ther 2020; 220:107712. [PMID: 33121940 DOI: 10.1016/j.pharmthera.2020.107712] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022]
Abstract
RSV infection of the lower respiratory tract in infants is the leading cause of pediatric hospitalizations and second to malaria in causing infant deaths worldwide. RSV also causes substantial morbidity in immunocompromised and elderly populations. The only available therapeutic is a prophylactic drug called Palivizumab that is a humanized monoclonal antibody, given to high-risk infants. However, this intervention is expensive and has a limited impact on annual hospitalization rates caused by RSV. No vaccine is available, nor are efficacious antivirals to treat an active infection, and there is still no consensus on how infants with bronchiolitis should be treated during hospital admission. In this comprehensive review, we briefly outline the function of the RSV proteins and their suitability as therapeutic targets. We then discuss the most promising drug candidates, their inhibitory mechanisms, and whether they are in the process of clinical trials. We also briefly discuss the reasons for some of the failures in RSV therapeutics and vaccines. In summary, we provide insight into current antiviral development and the considerations toward producing licensed antivirals and therapeutics.
Collapse
Affiliation(s)
- Farah Elawar
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Ahmed K Oraby
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Misr University for Science &Technology, Al-Motamayez District, 6th of October City, P.O. Box 77, Egypt
| | - Quinten Kieser
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Lionel D Jensen
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Tyce Culp
- Li Ka Shing Institute of Virology, 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
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
10
|
Liu H, Lu B, Tabor DE, Tovchigrechko A, Wilkins D, Jin H, Madhi SA, Soofie N, Esser MT, Nunes MC. Characterization of human respiratory syncytial virus (RSV) isolated from HIV-exposed-uninfected and HIV-unexposed infants in South Africa during 2015-2017. Influenza Other Respir Viruses 2020; 14:403-411. [PMID: 32126161 PMCID: PMC7298309 DOI: 10.1111/irv.12727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND RSV is a leading cause of lower respiratory tract infection in infants. Monitoring RSV glycoprotein sequences is critical for understanding RSV epidemiology and viral antigenicity in the effort to develop anti-RSV prophylactics and therapeutics. OBJECTIVES The objective is to characterize the circulating RSV strains collected from infants in South Africa during 2015-2017. METHODS A subset of 150 RSV-positive samples obtained in South Africa from HIV-unexposed and HIV-exposed-uninfected infants from 2015 to 2017, were selected for high-throughput next-generation sequencing of the RSV F and G glycoprotein genes. The RSV G and F sequences were analyzed by a bioinformatic pipeline and compared to the USA samples from the same three-year period. RESULTS Both RSV A and RSV B co-circulated in South Africa during 2015-2017, with a shift from RSV A (58%-61% in 2015-2016) to RSV B (69%) in 2017. RSV A ON1 and RSV B BA9 genotypes emerged as the most prevalent genotypes in 2017. Variations at the F protein antigenic sites were observed for both RSV A and B strains, with dominant changes (L172Q/S173L) at antigenic site V observed in RSV B strains. RSV A and B F protein sequences from South Africa were very similar to the USA isolates except for a higher rate of RSV A NA1 and RSV B BA10 genotypes in South Africa. CONCLUSION RSV G and F genes continue to evolve and exhibit both local and global circulation patterns in South Africa, supporting the need for continued national surveillance.
Collapse
Affiliation(s)
- Hui Liu
- AstraZeneca, South San Francisco, CA, USA
| | - Bin Lu
- AstraZeneca, South San Francisco, CA, USA
| | | | | | | | - Hong Jin
- AstraZeneca, South San Francisco, CA, USA
| | - 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
| | - Nasiha Soofie
- 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
| | | | - Marta C Nunes
- 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
| |
Collapse
|
11
|
Comparative Therapeutic Potential of ALX-0171 and Palivizumab against Respiratory Syncytial Virus Clinical Isolate Infection of Well-Differentiated Primary Pediatric Bronchial Epithelial Cell Cultures. Antimicrob Agents Chemother 2020; 64:AAC.02034-19. [PMID: 31767728 DOI: 10.1128/aac.02034-19] [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: 10/09/2019] [Accepted: 11/20/2019] [Indexed: 01/09/2023] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract infections in young infants. There are no RSV-specific treatments available. Ablynx has been developing an anti-RSV F-specific nanobody, ALX-0171. To characterize the therapeutic potential of ALX-0171, we exploited our well-differentiated primary pediatric bronchial epithelial cell (WD-PBEC)/RSV infection model, which replicates several hallmarks of RSV disease in vivo Using 2 clinical isolates (BT2a and Memphis 37), we compared the therapeutic potential of ALX-0171 with that of palivizumab, which is currently prescribed for RSV prophylaxis in high-risk infants. ALX-0171 treatment (900 nM) at 24 h postinfection reduced apically released RSV titers to near or below the limit of detection within 24 h for both strains. Progressively lower doses resulted in concomitantly diminished RSV neutralization. ALX-0171 was approximately 3-fold more potent in this therapeutic RSV/WD-PBEC model than palivizumab (mean 50% inhibitory concentration [IC50] = 346.9 to 363.6 nM and 1,048 to 1,090 nM for ALX-0171 and palivizumab, respectively), irrespective of the clinical isolate. The number of viral genomic copies (GC) was determined by quantitative reverse transcription-PCR (RT-qPCR), and the therapeutic effect of ALX-0171 treatment at 300 and 900 nM was found to be considerably lower and the number of GCs reduced only moderately (0.62 to 1.28 log10 copies/ml). Similar findings were evident for palivizumab. Therefore, ALX-0171 was very potent at neutralizing RSV released from apical surfaces but had only a limited impact on virus replication. The data indicate a clear disparity between viable virus neutralization and GC viral load, the latter of which does not discriminate between viable and neutralized RSV. This report validates the RSV/WD-PBEC model for the preclinical evaluation of RSV antivirals.
Collapse
|
12
|
Quan FS, Basak S, Chu KB, Kim SS, Kang SM. Progress in the development of virus-like particle vaccines against respiratory viruses. Expert Rev Vaccines 2020; 19:11-24. [PMID: 31903811 PMCID: PMC7103727 DOI: 10.1080/14760584.2020.1711053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Influenza virus, human respiratory syncytial virus (RSV), and human metapneumovirus (HMPV) are important human respiratory pathogens. Recombinant virus-like particle (VLP) vaccines are suggested to be potential promising platforms to protect against these respiratory viruses. This review updates important progress in the development of VLP vaccines against respiratory viruses.Areas Covered: This review summarizes progress in developing VLP and nanoparticle-based vaccines against influenza virus, RSV, and HMPV. The PubMed was mainly used to search for important research articles published since 2010 although earlier key articles were also referenced. The research area covered includes VLP and nanoparticle platform vaccines against seasonal, pandemic, and avian influenza viruses as well as RSV and HMPV respiratory viruses. The production methods, immunogenic properties, and vaccine efficacy of respiratory VLP vaccines in preclinical animal models and clinical studies were reviewed in this article.Expert opinion: Previous and current preclinical and clinical studies suggest that recombinant VLP and nanoparticle vaccines are expected to be developed as promising alternative platforms against respiratory viruses in future. Therefore, continued research efforts are warranted.
Collapse
Affiliation(s)
- Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea
| | - Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
13
|
Abstract
Respiratory syncytial virus (RSV) is worldwide a very important virus leading to infection of the respiratory system. In particular preterm babies, infants and elderly adults are prone to developing severe diseases such as bronchiolitis or pneumonia, which require intensive care and cause increased mortality. Although RSV is rapidly detected, preventive and therapeutic measures are limited. New antivirals are already in clinical trials.
Collapse
Affiliation(s)
- Jürgen Seidenberg
- Universitätsklinik für Kinder- und Jugendmedizin, Klinik für Pädiatrische Pneumologie und Allergologie, Klinikum Oldenburg AöR, Rahel-Straus-Straße 10, 26133, Oldenburg, Deutschland.
| |
Collapse
|
14
|
Goldstein E, Finelli L, O’Halloran A, Liu P, Karaca Z, Steiner CA, Viboud C, Lipsitch M. Hospitalizations Associated with Respiratory Syncytial Virus and Influenza in Children, Including Children Diagnosed with Asthma. Epidemiology 2019; 30:918-926. [PMID: 31469696 PMCID: PMC6768705 DOI: 10.1097/ede.0000000000001092] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is uncertainty about the burden of hospitalization associated with respiratory syncytial virus (RSV) and influenza in children, including those with underlying medical conditions. METHODS We applied previously developed methodology to Health Care Cost and Utilization Project hospitalization data and additional data related to asthma diagnosis/previous history in hospitalized children to estimate RSV and influenza-associated hospitalization rates in different subpopulations of US children between 2003 and 2010. RESULTS The estimated average annual rates (per 100,000 children) of RSV-associated hospitalization with a respiratory cause (ICD-9 codes 460-519) present anywhere in the discharge diagnosis were 2,381 (95% CI(2252,2515)) in children <1 year of age; 710.6 (609.1, 809.2) (1 y old); 395 (327.7, 462.4) (2 y old); 211.3 (154.6, 266.8) (3 y old); 111.1 (62.4, 160.1) (4 y old); 72.3 (29.3, 116.4) (5-6 y of age); 35.6 (9.9,62.2) (7-11 y of age); and 39 (17.5, 60.6) (12-17 y of age). The corresponding rates of influenza-associated hospitalization were lower, ranging from 181 (142.5, 220.3) in <1 year old to 17.9 (11.7, 24.2) in 12-17 years of age. The relative risks for RSV-related hospitalization associated with a prior diagnosis of asthma in age groups <5 y ranged between 3.1 (2.1, 4.7) (<1 y old) and 6.7 (4.2, 11.8) (2 y old; the corresponding risks for influenza-related hospitalization ranged from 2.8 (2.1, 4) (<1y old) to 4.9 (3.8, 6.4) (3 y old). CONCLUSION RSV-associated hospitalization rates in young children are high and decline rapidly with age. There are additional risks for both RSV and influenza hospitalization associated with a prior diagnosis of asthma, with the rates of RSV-related hospitalization in the youngest children diagnosed with asthma being particularly high.
Collapse
Affiliation(s)
- Edward Goldstein
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115 USA
| | | | - Alissa O’Halloran
- Influenza Division, National Center for Immunization and Respiratory Diseases, US CDC, Atlanta, GA 30329 USA
| | - Patrick Liu
- Yale School of Medicine, New Haven, CT 06510 USA
| | - Zeynal Karaca
- Agency for HealthCare Research and Quality, U.S. Department of Health & Human Services, Rockville, MD 20850 USA
| | - Claudia A. Steiner
- Agency for HealthCare Research and Quality, U.S. Department of Health & Human Services, Rockville, MD 20850 USA (work performed in that capacity; currently works at Institute for Health Research, Kaiser Permanente Colorado, Denver, CO 80231 USA)
| | - Cecile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, MD 20892 USA
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115 USA
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA 02115 USA
| |
Collapse
|
15
|
Abstract
Human respiratory syncytial virus (RSV) belongs to the recently defined Pneumoviridae family, Orthopneumovirus genus. It is a negative sense, single stranded RNA virus that results in epidemics of respiratory infections that typically peak in the winter in temperate climates and during the rainy season in tropical climates. Generally, one of the two genotypes (A and B) predominates in a single season, alternating annually, although regional variation occurs. RSV is a cause of disease and death in children, older people, and immunocompromised patients, and its clinical effect on adults admitted to hospital is clarified with expanded use of multiplex molecular assays. Among adults, RSV produces a wide range of clinical symptoms including upper respiratory tract infections, severe lower respiratory tract infections, and exacerbations of underlying disease. Here we discuss the latest evidence on the burden of RSV related disease in adults, especially in those with immunocompromise or other comorbidities. We review current therapeutic and prevention options, as well as those in development.
Collapse
Affiliation(s)
- Hannah H Nam
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael G Ison
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
16
|
Original Antigenic Sin and Respiratory Syncytial Virus Vaccines. Vaccines (Basel) 2019; 7:vaccines7030107. [PMID: 31500131 PMCID: PMC6789633 DOI: 10.3390/vaccines7030107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 02/08/2023] Open
Abstract
The original antigenic sin (OAS) theory considers the outcome of the first encounter with an antigen. It favors a memory response to the original antigen upon exposure to a similar or related antigen, and includes both positive and negative impacts of past exposure on the memory response to challenge, and, in particular, on vaccine efficacy. This phenomenon is closely linked with imprinting and the hierarchical nature of immune responses to previously encountered antigens. The focus of this commentary centers on the potential role of OAS or immunological imprinting on respiratory syncytial virus memory responses.
Collapse
|
17
|
de Souza RP, Ribeiro ALR, de Menezes SAF, Machado LFA. Incidence of respiratory syncytial virus infection in children with congenital heart disease undergoing immunoprophylaxis with palivizumab in Pará state, north region of Brazil. BMC Pediatr 2019; 19:299. [PMID: 31462289 PMCID: PMC6714430 DOI: 10.1186/s12887-019-1681-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/21/2019] [Indexed: 12/03/2022] Open
Abstract
Background Palivizumab prophylaxis for the human respiratory syncytial virus (HRSV) has been reported to reduce the risk of hospital admissions related to HRSV in children with congenital heart disease (CHD). These children are at high risk of developing severe lower respiratory tract infection (LRTI) due to HRSV infection. Our goal was to evaluate the incidence of HRSV infection in children with CHD after being submitted to immunoprophylaxis with palivizumab in Pará state, North region of Brazil. Methods A prospective and observational cohort study was performed in children ≤2 years of age with CHD who received palivizumab immunoprophylaxis between January 1 and June 31, 2016. A questionnaire about basic non-medical care measures was applied to parents/legal representatives. Data on patients’ demographic characteristics, household environment, and respiratory infections were evaluated. HRSV infection was determined by qPCR. Results There were 104 children enrolled in this investigation and the results showed a mean age of 10.6 months, an average weight of 7.3 kg and 3.5 doses of palivizumab per children during seasonality of HRSV. Respiratory infection was observed in 27.9% of cases, of which 9.6% were LRTI. No case of children who received palivizumab immunoprophylaxis and developed influenza-like symptoms tested positive for HRSV. Conclusion Although the lack of a control group doesn’t allow to affirm the effectiveness of HRSV passive immunization, the immunoprophylaxis with palivizumab appeared to be totally efficient in preventing respiratory infection by HRSV in children up to two years of age with CHD.
Collapse
Affiliation(s)
- Roseane Porfírio de Souza
- Biology of Infectious and Parasitic Agents Post-Graduate Program, Federal University of Pará, Belém, Pará, Brazil.,Gaspar Vianna Clinic Hospital Foundation, Belém, Pará, Brazil
| | - Andre Luis Ribeiro Ribeiro
- Postdoctoral fellowship, Cell Culture Laboratory, School of Dentistry, Federal University of Para - UFPA, Belém, Pará, Brazil
| | | | - Luiz Fernando Almeida Machado
- Biology of Infectious and Parasitic Agents Post-Graduate Program, Federal University of Pará, Belém, Pará, Brazil. .,Virology Laboratory, Institute of Biological Sciences, Federal University of Pará, Cidade Universitária Prof. José da Silveira Netto, Rua Augusto Correa 1, Guamá, 66.075-110, Belém, Pará, Brazil.
| |
Collapse
|
18
|
Sanders SL, Agwan S, Hassan M, van Driel ML, Del Mar CB. 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: 3.0] [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
|
19
|
Goldstein E, Nguyen HH, Liu P, Viboud C, Steiner CA, Worby CJ, Lipsitch M. On the Relative Role of Different Age Groups During Epidemics Associated With Respiratory Syncytial Virus. J Infect Dis 2019; 217:238-244. [PMID: 29112722 DOI: 10.1093/infdis/jix575] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 10/31/2017] [Indexed: 11/14/2022] Open
Abstract
Background While circulation of respiratory syncytial virus (RSV) results in high rates of hospitalization, particularly among young children and elderly individuals, little is known about the role of different age groups in propagating annual RSV epidemics. Methods We evaluate the roles played by individuals in different age groups during RSV epidemics in the United States between 2001 and 2012, using the previously defined relative risk (RR) statistic estimated from the hospitalization data from the Healthcare Cost and Utilization Project. Transmission modeling was used to examine the robustness of our inference method. Results Children aged 3-4 years and 5-6 years each had the highest RR estimate for 5 of 11 seasons included in this study, with RSV hospitalization rates in infants being generally higher during seasons when children aged 5-6 years had the highest RR estimate. Children aged 2 years had the highest RR estimate during one season. RR estimates in infants and individuals aged ≥11 years were mostly lower than in children aged 1-10 years. Highest RR values aligned with groups for which vaccination had the largest impact on epidemic dynamics in most model simulations. Conclusions Our estimates suggest the prominent relative roles of children aged ≤10 years (particularly among those aged 3-6 years) in propagating RSV epidemics. These results, combined with further modeling work, should help inform RSV vaccination policies.
Collapse
Affiliation(s)
- Edward Goldstein
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Hieu H Nguyen
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Patrick Liu
- Yale School of Medicine, New Haven, Connecticut
| | - Cecile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda
| | - Claudia A Steiner
- Agency for HealthCare Research and Quality, Department of Health and Human Services, Rockville, Maryland.,Institute for Health Research, Kaiser Permanente Colorado, Denver
| | - Colin J Worby
- Department of Ecology and Evolutionary Biology, Princeton University, New Jersey
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
20
|
Bin Lu, Liu H, Tabor DE, Tovchigrechko A, Qi Y, Ruzin A, Esser MT, Jin H. Emergence of new antigenic epitopes in the glycoproteins of human respiratory syncytial virus collected from a US surveillance study, 2015-17. Sci Rep 2019; 9:3898. [PMID: 30846850 PMCID: PMC6405860 DOI: 10.1038/s41598-019-40387-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of lower respiratory tract infection in infants and elderly. To understand the evolution of neutralizing epitopes on the RSV glycoprotein (G) and fusion (F) proteins, we conducted a multi-year surveillance program (OUTSMART-RSV) in the US. Analysis of 1,146 RSV samples from 2015-2017 revealed a slight shift in prevalence from RSV A (58.7%) to B (53.7%) between the two seasons. RSV B was more prevalent in elderly (52.9% and 73.4%). Approximately 1% of the samples contained both RSV A and B viruses. All RSV A isolates were ON1 and almost all the B isolates were BA9 genotypes. Compared with the 2013 reference sequences, changes at the F antigenic sites of RSV B were greater than RSV A, which mainly occurred at antigenic sites V (L172Q/S173L at 99.6%), Ø (I206M/Q209K at 18.6%) and IV (E463D at 7%) of RSV B F. Sequence diversities in the G protein second hypervariable region were observed in the duplicated regions for RSV A and B, and at the G stop codon resulting in extension of 7 amino acids (22.1%) for RSV B in 2016-17. Thus, RSV surface glycoproteins are continuously evolving, and continued surveillance is important for the clinical evaluation of immunoprophylactic products.
Collapse
Affiliation(s)
- Bin Lu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | - Hui Liu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Yanping Qi
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Hong Jin
- MedImmune/AstraZeneca, South San Francisco, CA, USA.
| |
Collapse
|
21
|
|
22
|
Abstract
INTRODUCTION Nanoparticles are under discussion in drug delivery for more than 20 years now, but examples for nanoparticulate formulations in the treatment of respiratory diseases are rare and mostly limited to the administration of sub-micron drug particles (ultrafine particles). However, nanoparticles may also carry specific benefits for respiratory treatment. Are nanoparticles the next-generation drug carrier system to facilitate systemic delivery, sustained release and cancer treatment in the lungs? AREAS COVERED This review will look into the promises and opportunities of the use of nanoparticles in the treatment of respiratory diseases. Important aspects to discuss are the fate of nanoparticles in the lung and mechanisms for reproducible delivery of nanoparticulate formulations to the lungs. Examples are given where nanoparticles may be advantageous over for traditional formulations and further aspects to explore are mentioned. EXPERT OPINION The benefit of nanoparticulate systems for respiratory delivery adds to the portfolio of possible formulation strategies, depends on the intended functionality and needs more exploration. Advantages of such systems are only seen in special cases.
Collapse
Affiliation(s)
- Regina Scherließ
- a Department of Pharmaceutics and Biopharmaceutics , Kiel University , Kiel , Germany
| |
Collapse
|
23
|
Simões EAF, Bont L, Manzoni P, Fauroux B, Paes B, Figueras-Aloy J, Checchia PA, Carbonell-Estrany X. Past, Present and Future Approaches to the Prevention and Treatment of Respiratory Syncytial Virus Infection in Children. Infect Dis Ther 2018; 7:87-120. [PMID: 29470837 PMCID: PMC5840107 DOI: 10.1007/s40121-018-0188-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION The REGAL (RSV Evidence - A Geographical Archive of the Literature) series has provided a comprehensive review of the published evidence in the field of respiratory syncytial virus (RSV) in Western countries over the last 20 years. This seventh and final publication covers the past, present and future approaches to the prevention and treatment of RSV infection among infants and children. METHODS A systematic review was undertaken of publications between January 1, 1995 and December 31, 2017 across PubMed, Embase and The Cochrane Library. Studies reporting data on the effectiveness and tolerability of prophylactic and therapeutic agents for RSV infection were included. Study quality and strength of evidence (SOE) were graded using recognized criteria. A further nonsystematic search of the published literature and Clinicaltrials.gov on antiviral therapies and RSV vaccines currently in development was also undertaken. RESULTS The systematic review identified 1441 studies of which 161 were included. Management of RSV remains centered around prophylaxis with the monoclonal antibody palivizumab, which has proven effective in reducing RSV hospitalization (RSVH) in preterm infants < 36 weeks' gestational age (72% reduction), children with bronchopulmonary dysplasia (65% reduction), and infants with hemodynamically significant congenital heart disease (53% reduction) (high SOE). Palivizumab has also shown to be effective in reducing recurrent wheezing following RSVH (high SOE). Treatment of RSV with ribavirin has conflicting success (moderate SOE). Antibodies with increased potency and extended half-life are currently entering phase 3 trials. There are approximately 15 RSV vaccines in clinical development targeting the infant directly or indirectly via the mother. CONCLUSION Palivizumab remains the only product licensed for RSV prophylaxis, and only available for high-risk infants. For the general population, there are several promising vaccines and monoclonal antibodies in various stages of clinical development, with the aim to significantly reduce the global healthcare impact of this common viral infection. FUNDING AbbVie.
Collapse
Affiliation(s)
- Eric A F Simões
- Center for Global Health, Colorado School of Public Health, University of Colorado School of Medicine, Aurora, CO, USA
| | - Louis Bont
- University Medical Center Utrecht, Utrecht, The Netherlands
- ReSViNET (Respiratory Syncytial Virus Network), Utrecht, The Netherlands
| | - Paolo Manzoni
- ReSViNET (Respiratory Syncytial Virus Network), Utrecht, The Netherlands
- Neonatology and NICU, Sant'Anna Hospital, Turin, Italy
| | - Brigitte Fauroux
- Necker University Hospital and Paris 5 University, Paris, France
| | - Bosco Paes
- Department of Paediatrics (Neonatal Division), McMaster University, Hamilton, Canada
| | - Josep Figueras-Aloy
- Hospital Clínic, Catedràtic de Pediatria, Universitat de Barcelona, Barcelona, Spain
| | - Paul A Checchia
- Baylor College of Medicine, Texas Children's Hospital Houston, Houston, TX, USA
| | - Xavier Carbonell-Estrany
- Hospital Clinic, Institut d'Investigacions Biomediques August Pi Suñer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
24
|
Rodrigues CMC, Groves H. Community-Acquired Pneumonia in Children: the Challenges of Microbiological Diagnosis. J Clin Microbiol 2018; 56:e01318-17. [PMID: 29237789 PMCID: PMC5824044 DOI: 10.1128/jcm.01318-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Community-acquired pneumonia (CAP) is the leading cause of mortality in children under 5 years of age globally. To improve the management of CAP, we must distinguish CAP from other common pediatric conditions and develop better diagnostic methods to detect the causative organism, so as to best direct appropriate resources in both industrialized and developing countries. Here, we review the diagnostic modalities available for identifying viruses and bacteria in the upper and lower respiratory tract of children, with a discussion of their utility and limitations in diagnosing CAP in children.
Collapse
Affiliation(s)
- C M C Rodrigues
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Department of Paediatric Infectious Diseases and Immunology, Great North Children's Hospital, Newcastle Upon Tyne, United Kingdom
| | - H Groves
- Centre for Experimental Medicine, Queens University, Belfast, United Kingdom
- Department of Paediatrics, Royal Belfast Hospital for Sick Children, Belfast, United Kingdom
| |
Collapse
|
25
|
Xin Y, Weng W, Murray BP, Eisenberg EJ, Chien JW, Ling J, Silverman JA. The Drug-Drug Interaction Profile of Presatovir. J Clin Pharmacol 2018; 58:771-780. [PMID: 29412463 DOI: 10.1002/jcph.1073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/04/2017] [Indexed: 11/10/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in young children. Presatovir (previously GS-5806) is a novel, orally administered RSV fusion inhibitor with a favorable safety profile and proven antiviral efficacy in preclinical and clinical studies. In vitro, presatovir is a substrate of the efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) and hepatic uptake transporters organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 and is slowly metabolized by cytochrome P450 (CYP) 3A4 and CYP3A5. This study enrolled 64 healthy subjects to evaluate the effect of cyclosporine, a P-gp, BCRP, and OATP1B1/1B3 inhibitor; rifampin, a strong CYP3A4 and P-gp inducer; efavirenz, a moderate CYP3A4 inducer; and cobicistat, a potent CYP3A inhibitor, on presatovir pharmacokinetics. Presatovir plasma exposures (maximum observed plasma concentration [Cmax ] and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf ]) were not affected by coadministration of cyclosporine, suggesting presatovir is not a sensitive substrate of P-gp, BCRP, or OATP1B1/1B3. As expected, based on the role of CYP3A in presatovir metabolism, presatovir exposure was increased by cobicistat (122% in AUCinf ), and decreased by rifampin (40.3% in Cmax and 82.5% in AUCinf ) and efavirenz (55.7% in AUCinf ). These data support coadministration of presatovir with inhibitors of P-gp, BCRP, OATP1B1/1B3, or CYP3A, but not with moderate or strong CYP3A4 inducers. Presatovir was well-tolerated with the most common drug-related adverse events of dizziness (n = 12) and somnolence (n = 4) reported during efavirenz treatment.
Collapse
Affiliation(s)
- Yan Xin
- Gilead Sciences, Inc., Foster City, CA, USA
| | | | | | | | | | - John Ling
- Gilead Sciences, Inc., Foster City, CA, USA
| | | |
Collapse
|
26
|
Respiratory Syncytial Virus: Targeting the G Protein Provides a New Approach for an Old Problem. J Virol 2018; 92:JVI.01302-17. [PMID: 29118126 DOI: 10.1128/jvi.01302-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection (LRTI) annually affecting >2 million children in the United States <5 years old. In the elderly (>65 years old), RSV results in ∼175,000 hospitalizations annually in the United States with a worldwide incidence of ∼34 million. There is no approved RSV vaccine, and treatments are limited. Recently, a phase 3 trial in the elderly using a recombinant RSV F protein vaccine failed to meet its efficacy objectives, namely, prevention of moderate-to-severe RSV-associated LRTI and reduced incidence of acute respiratory disease. Moreover, a recent phase 3 trial evaluating suptavumab (REGN2222), an antibody to RSV F protein, did not meet its primary endpoint of preventing medically attended RSV infections in preterm infants. Despite these setbacks, numerous efforts targeting the RSV F protein with vaccines, antibodies, and small molecules continue based on the commercial success of a monoclonal antibody (MAb) against the RSV F protein (palivizumab). As the understanding of RSV biology has improved, the other major coat protein, the RSV G protein, has reemerged as an alternative target reflecting progress in understanding its roles in infecting bronchial epithelial cells and in altering the host immune response. In mouse models, a high-affinity, strain-independent human MAb to the RSV G protein has shown potent direct antiviral activity combined with the alleviation of virus-induced immune system effects that contribute to disease pathology. This MAb, being prepared for clinical trials, provides a qualitatively new approach to managing RSV for populations not eligible for prophylaxis with palivizumab.
Collapse
|
27
|
Russell CJ, Simões EAF, Hurwitz JL. Vaccines for the Paramyxoviruses and Pneumoviruses: Successes, Candidates, and Hurdles. Viral Immunol 2018; 31:133-141. [PMID: 29323621 DOI: 10.1089/vim.2017.0137] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human parainfluenza viruses (family Paramyxoviridae), human metapneumovirus, and respiratory syncytial virus (family Pneumoviridae) infect most infants and children within the first few years of life and are the etiologic agents for many serious acute respiratory illnesses. These virus infections are also associated with long-term diseases that impact quality of life, including asthma. Despite over a half-century of vaccine research, development, and clinical trials, no vaccine has been licensed to date for the paramyxoviruses or pneumoviruses for the youngest infants. In this study, we describe the recent reclassification of paramyxoviruses and pneumoviruses into distinct families by the International Committee on the Taxonomy of Viruses. We also discuss some past unsuccessful vaccine trials and some currently preferred vaccine strategies. Finally, we discuss hurdles that must be overcome to support successful respiratory virus vaccine development for the youngest children.
Collapse
Affiliation(s)
- Charles J Russell
- 1 Department of Infectious Diseases, St. Jude Children's Research Hospital , Memphis, Tennessee.,2 Department of Molecular Biology, Immunology, and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Eric A F Simões
- 3 Department of Pediatrics, University of Colorado School of Medicine, Department of Epidemiology, Colorado School of Public Health, Section of Infectious Diseases, Children's Hospital Colorado, Aurora, Colorado
| | - Julia L Hurwitz
- 1 Department of Infectious Diseases, St. Jude Children's Research Hospital , Memphis, Tennessee.,2 Department of Molecular Biology, Immunology, and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
28
|
Jordan PC, Stevens SK, Deval J. Nucleosides for the treatment of respiratory RNA virus infections. Antivir Chem Chemother 2018; 26:2040206618764483. [PMID: 29562753 PMCID: PMC5890544 DOI: 10.1177/2040206618764483] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/07/2018] [Indexed: 12/20/2022] Open
Abstract
Influenza virus, respiratory syncytial virus, human metapneumovirus, parainfluenza virus, coronaviruses, and rhinoviruses are among the most common viruses causing mild seasonal colds. These RNA viruses can also cause lower respiratory tract infections leading to bronchiolitis and pneumonia. Young children, the elderly, and patients with compromised cardiac, pulmonary, or immune systems are at greatest risk for serious disease associated with these RNA virus respiratory infections. In addition, swine and avian influenza viruses, together with severe acute respiratory syndrome-associated and Middle Eastern respiratory syndrome coronaviruses, represent significant pandemic threats to the general population. In this review, we describe the current medical need resulting from respiratory infections caused by RNA viruses, which justifies drug discovery efforts to identify new therapeutic agents. The RNA polymerase of respiratory viruses represents an attractive target for nucleoside and nucleotide analogs acting as inhibitors of RNA chain synthesis. Here, we present the molecular, biochemical, and structural fundamentals of the polymerase of the four major families of RNA respiratory viruses: Orthomyxoviridae, Pneumoviridae/Paramyxoviridae, Coronaviridae, and Picornaviridae. We summarize past and current efforts to develop nucleoside and nucleotide analogs as antiviral agents against respiratory virus infections. This includes molecules with very broad antiviral spectrum such as ribavirin and T-705 (favipiravir), and others targeting more specifically one or a few virus families. Recent advances in our understanding of the structure(s) and function(s) of respiratory virus polymerases will likely support the discovery and development of novel nucleoside analogs.
Collapse
Affiliation(s)
- Paul C Jordan
- Alios BioPharma, Inc., a Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, USA
| | - Sarah K Stevens
- Alios BioPharma, Inc., a Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, USA
| | - Jerome Deval
- Alios BioPharma, Inc., a Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, USA
| |
Collapse
|
29
|
Salimi V, Mirzaei H, Ramezani A, Tahamtan A, Jamali A, Shahabi S, Golaram M, Minaei B, Gharagozlou MJ, Mahmoodi M, Bont L, Shokri F, Mokhtari-Azad T. Blocking of opioid receptors in experimental formaline-inactivated respiratory syncytial virus (FI-RSV) immunopathogenesis: from beneficial to harmful impacts. Med Microbiol Immunol 2017; 207:105-115. [DOI: 10.1007/s00430-017-0531-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/09/2017] [Indexed: 11/29/2022]
|
30
|
Anderson AJ, Snelling TL, Moore HC, Blyth CC. Advances in Vaccines to Prevent Viral Respiratory Illnesses in Children. Paediatr Drugs 2017; 19:523-531. [PMID: 28808938 DOI: 10.1007/s40272-017-0257-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Childhood vaccination has played a critical role in the reduction of morbidity and mortality from communicable diseases, including specific respiratory pathogens. Acute lower respiratory infection (ALRI) of both bacterial and viral aetiology continues to impact global child health. Key bacterial pathogens including Streptococcus pneumoniae and Haemophilus influenza type b are specifically targeted with current vaccination programmes, while at present there are less effective strategies for the prevention of viral disease. Influenza vaccines, including both live attenuated intranasal vaccines and inactivated influenza vaccines, are limited by seasonal strain variation and unsustained immunity. Research into the development of a universal influenza vaccine is ongoing; potential targets are the conserved regions of the virus such as the M2e antigen and hemagglutinin stalk. Respiratory syncytial virus (RSV) and parainfluenza virus 3 (PIV3) are the viral pathogens most commonly causing ALRI in children, particularly the infant population. Currently, no vaccine exists for either virus. Over the last decade, promising advances have been made. Protection of neonates via maternal RSV immunisation is being assessed in a phase III clinical trial, with many other candidates for RSV and PIV3 at less advanced stages of development.
Collapse
Affiliation(s)
- Aleisha J Anderson
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia
| | - Tom L Snelling
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Hannah C Moore
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Christopher C Blyth
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia. .,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia. .,Discipline of Paediatrics, School of Medicine, Princess Margaret Hospital, University of Western Australia, Roberts Rd, Subiaco, WA, Australia. .,PathWest Laboratory Medicine WA, Department of Microbiology, QEII Medical Centre, Nedlands, WA, Australia.
| |
Collapse
|
31
|
Li H, Callahan C, Citron M, Wen Z, Touch S, Monslow MA, Cox KS, DiStefano DJ, Vora KA, Bett A, Espeseth A. Respiratory syncytial virus elicits enriched CD8+ T lymphocyte responses in lung compared with blood in African green monkeys. PLoS One 2017; 12:e0187642. [PMID: 29121080 PMCID: PMC5679537 DOI: 10.1371/journal.pone.0187642] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/23/2017] [Indexed: 11/18/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of serious lower respiratory tract disease in young children and older adults throughout the world. Prevention of severe RSV disease through active immunization is optimal but no RSV vaccine has been licensed so far. Immune mechanisms of protection against RSV infection in humans have not been fully established, thus a comprehensive characterization of virus-specific immune responses in a relevant animal model will be beneficial in defining correlates of protection. In this study, we infected juvenile naive AGMs with RSV A2 strain and longitudinally assessed virus-specific humoral and cellular immune responses in both peripheral blood and the respiratory tract. RSV viral loads at nasopharyngeal surfaces and in the lung peaked at around day 5 following infection, and then largely resolved by day 10. Low levels of neutralizing antibody titers were detected in serum, with similar kinetics as RSV fusion (F) protein-binding IgG antibodies. RSV infection induced CD8+, but very little CD4+, T lymphocyte responses in peripheral blood. Virus-specific CD8+ T cell frequencies were ~10 fold higher in bronchoaveolar lavage (BAL) compared to peripheral blood and exhibited effector memory (CD95+CD28-) / tissue resident memory (CD69+CD103+) T (TRM) cell phenotypes. The kinetics of virus-specific CD8+ T cells emerging in peripheral blood and BAL correlated with declining viral titers, suggesting that virus-specific cellular responses contribute to the clearance of RSV infection. RSV-experienced AGMs were protected from subsequent exposure to RSV infection. Additional studies are underway to understand protective correlates in these seropositive monkeys.
Collapse
Affiliation(s)
- Hualin Li
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Cheryl Callahan
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Michael Citron
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Zhiyun Wen
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Sinoeun Touch
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Morgan A. Monslow
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Kara S. Cox
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Daniel J. DiStefano
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Kalpit A. Vora
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Andrew Bett
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| | - Amy Espeseth
- Department of Infectious Diseases and Vaccines, MRL, Merck & Co., Inc., West Point, PA, United States of America
| |
Collapse
|
32
|
Antibody therapies for the prevention and treatment of viral infections. NPJ Vaccines 2017; 2:19. [PMID: 29263875 PMCID: PMC5627241 DOI: 10.1038/s41541-017-0019-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Antibodies are an important component in host immune responses to viral pathogens. Because of their unique maturation process, antibodies can evolve to be highly specific to viral antigens. Physicians and researchers have been relying on such high specificity in their quest to understand host–viral interaction and viral pathogenesis mechanisms and to find potential cures for viral infection and disease. With more than 60 recombinant monoclonal antibodies developed for human use in the last 20 years, monoclonal antibodies are now considered a viable therapeutic modality for infectious disease targets, including newly emerging viral pathogens such as Ebola representing heightened public health concerns, as well as pathogens that have long been known, such as human cytomegalovirus. Here, we summarize some recent advances in identification and characterization of monoclonal antibodies suitable as drug candidates for clinical evaluation, and review some promising candidates in the development pipeline.
Collapse
|
33
|
Lee BH, Chathuranga K, Uddin MB, Weeratunga P, Kim MS, Cho WK, Kim HI, Ma JY, Lee JS. Coptidis Rhizoma extract inhibits replication of respiratory syncytial virus in vitro and in vivo by inducing antiviral state. J Microbiol 2017; 55:488-498. [DOI: 10.1007/s12275-017-7088-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023]
|
34
|
Rezaee F, Linfield DT, Harford TJ, Piedimonte G. Ongoing developments in RSV prophylaxis: a clinician's analysis. Curr Opin Virol 2017; 24:70-78. [PMID: 28500974 DOI: 10.1016/j.coviro.2017.03.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/10/2017] [Accepted: 03/30/2017] [Indexed: 01/10/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common respiratory pathogen in infants and young children worldwide. Lower respiratory tract infection due to RSV is one of the most common causes of hospitalization for infants, especially those born premature or with chronic lung or heart disease. Furthermore, RSV infection is an important cause of morbidity in adults, particularly in the elderly and immunocompromised individuals. The acute phase of this infection is often followed by episodes of wheezing that recur for months or years and usually lead to a physician diagnosis of asthma. RSV was discovered more than 50 years ago, and despite extensive research to identify pharmacological therapies, the most effective management of this infection remains supportive care. The trial of a formalin-inactivated RSV vaccine in the 1960s resulted in priming the severe illness upon natural infection. Currently, Palivizumab is the only available option for RSV prophylaxis, and because of restricted clinical benefits and high costs, it has been limited to a group of high-risk infants. There are several ongoing trials in preclinical, Phase-I, Phase-II, or Phase-III clinical stages for RSV vaccine development based on various strategies. Here we review the existing available prophylactic options, the current stages of RSV vaccine clinical trials, different strategies, and major hurdles in the development of an effective RSV vaccine.
Collapse
Affiliation(s)
- Fariba Rezaee
- Pediatric Research Center and Pediatric Institute, Cleveland Clinic Children's, United States; Pathobiology Department, Lerner Research Institute, United States.
| | - Debra T Linfield
- Pediatric Research Center and Pediatric Institute, Cleveland Clinic Children's, United States; Pathobiology Department, Lerner Research Institute, United States
| | - Terri J Harford
- Pediatric Research Center and Pediatric Institute, Cleveland Clinic Children's, United States; Pathobiology Department, Lerner Research Institute, United States
| | - Giovanni Piedimonte
- Pediatric Research Center and Pediatric Institute, Cleveland Clinic Children's, United States; Pathobiology Department, Lerner Research Institute, United States
| |
Collapse
|
35
|
Immunization with Low Doses of Recombinant Postfusion or Prefusion Respiratory Syncytial Virus F Primes for Vaccine-Enhanced Disease in the Cotton Rat Model Independently of the Presence of a Th1-Biasing (GLA-SE) or Th2-Biasing (Alum) Adjuvant. J Virol 2017; 91:JVI.02180-16. [PMID: 28148790 DOI: 10.1128/jvi.02180-16] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of children previously immunized with a nonlive, formalin-inactivated (FI)-RSV vaccine has been associated with serious enhanced respiratory disease (ERD). Consequently, detailed studies of potential ERD are a critical step in the development of nonlive RSV vaccines targeting RSV-naive children and infants. The fusion glycoprotein (F) of RSV in either its postfusion (post-F) or prefusion (pre-F) conformation is a target for neutralizing antibodies and therefore an attractive antigen candidate for a pediatric RSV subunit vaccine. Here, we report the evaluation of RSV post-F and pre-F in combination with glucopyranosyl lipid A (GLA) integrated into stable emulsion (SE) (GLA-SE) and alum adjuvants in the cotton rat model. Immunization with optimal doses of RSV F antigens in the presence of GLA-SE induced high titers of virus-neutralizing antibodies and conferred complete lung protection from virus challenge, with no ERD signs in the form of alveolitis. To mimic a waning immune response, and to assess priming for ERD under suboptimal conditions, an antigen dose de-escalation study was performed in the presence of either GLA-SE or alum. At low RSV F doses, alveolitis-associated histopathology was unexpectedly observed with either adjuvant at levels comparable to FI-RSV-immunized controls. This occurred despite neutralizing-antibody titers above the minimum levels required for protection and with no/low virus replication in the lungs. These results emphasize the need to investigate a pediatric RSV vaccine candidate carefully for priming of ERD over a wide dose range, even in the presence of strong neutralizing activity, Th1 bias-inducing adjuvant, and protection from virus replication in the lower respiratory tract.IMPORTANCE RSV disease is of great importance worldwide, with the highest burden of serious disease occurring upon primary infection in infants and children. FI-RSV-induced enhanced disease, observed in the 1960s, presented a major and ongoing obstacle for the development of nonlive RSV vaccine candidates. The findings presented here underscore the need to evaluate a nonlive RSV vaccine candidate during preclinical development over a wide dose range in the cotton rat RSV enhanced-disease model, as suboptimal dosing of several RSV F subunit vaccine candidates led to the priming for ERD. These observations are relevant to the validity of the cotton rat model itself and to safe development of nonlive RSV vaccines for seronegative infants and children.
Collapse
|
36
|
Bont L, Checchia PA, Fauroux B, Figueras-Aloy J, Manzoni P, Paes B, Simões EAF, Carbonell-Estrany X. Defining the Epidemiology and Burden of Severe Respiratory Syncytial Virus Infection Among Infants and Children in Western Countries. Infect Dis Ther 2016; 5:271-98. [PMID: 27480325 PMCID: PMC5019979 DOI: 10.1007/s40121-016-0123-0] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION The REGAL (RSV [respiratory syncytial virus] Evidence-a Geographical Archive of the Literature) series provides a comprehensive review of the published evidence in the field of RSV in Western countries over the last 20 years. This first of seven publications covers the epidemiology and burden of RSV infection. METHODS A systematic review was undertaken for articles published between Jan 1, 1995 and Dec 31, 2015 across PubMed, Embase, The Cochrane Library, and Clinicaltrials.gov. Studies reporting data for hospital visits/admissions for RSV infection among children (≤18 years of age), as well as studies reporting RSV-associated morbidity, mortality, and risk factors were included. Study quality and strength of evidence (SOE) were graded using recognized criteria. RESULT 2315 studies were identified of which 98 were included. RSV was associated with 12-63% of all acute respiratory infections (ARIs) and 19-81% of all viral ARIs causing hospitalizations in children (high SOE). Annual RSV hospitalization (RSVH) rates increased with decreasing age and varied by a factor of 2-3 across seasons (high SOE). Studies were conflicting on whether the incidence of RSVH has increased, decreased, or remained stable over the last 20 years (moderate SOE). Length of hospital stay ranged from 2 to 11 days, with 2-12% of cases requiring intensive care unit admission (moderate SOE). Case-fatality rates were <0.5% (moderate SOE). Risk factors associated with RSVH included: male sex; age <6 months; birth during the first half of the RSV season; crowding/siblings; and day-care exposure (high SOE). CONCLUSION RSV infection remains a major burden on Western healthcare systems and has been associated with significant morbidity. Further studies focusing on the epidemiology of RSV infection (particularly in the outpatient setting), the impact of co-infection, better estimates of case-fatality rates and associated risk factors (all currently moderate/low SOE) are needed to determine the true burden of disease. FUNDING Abbvie.
Collapse
Affiliation(s)
- Louis Bont
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul A Checchia
- Baylor College of Medicine, Texas Children's Hospital Houston, Texas, USA
| | - Brigitte Fauroux
- Necker University Hospital and Paris 5 University, Paris, France
| | - Josep Figueras-Aloy
- Hospital Clínic, Catedràtic de Pediatria, Universitat de Barcelona, Barcelona, Spain
| | - Paolo Manzoni
- Neonatology and NICU, Sant'Anna Hospital, Turin, Italy
| | - Bosco Paes
- Department of Pediatrics (Neonatal Division), McMaster University, Hamilton, ON, Canada
| | - Eric A F Simões
- Colorado School of Public Health, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xavier Carbonell-Estrany
- Hospital Clinic, Institut d'Investigacions Biomediques August Pi Suñer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
37
|
Abstract
Since the first antiviral drug, idoxuridine, was approved in 1963, 90 antiviral drugs categorized into 13 functional groups have been formally approved for the treatment of the following 9 human infectious diseases: (i) HIV infections (protease inhibitors, integrase inhibitors, entry inhibitors, nucleoside reverse transcriptase inhibitors, nonnucleoside reverse transcriptase inhibitors, and acyclic nucleoside phosphonate analogues), (ii) hepatitis B virus (HBV) infections (lamivudine, interferons, nucleoside analogues, and acyclic nucleoside phosphonate analogues), (iii) hepatitis C virus (HCV) infections (ribavirin, interferons, NS3/4A protease inhibitors, NS5A inhibitors, and NS5B polymerase inhibitors), (iv) herpesvirus infections (5-substituted 2'-deoxyuridine analogues, entry inhibitors, nucleoside analogues, pyrophosphate analogues, and acyclic guanosine analogues), (v) influenza virus infections (ribavirin, matrix 2 protein inhibitors, RNA polymerase inhibitors, and neuraminidase inhibitors), (vi) human cytomegalovirus infections (acyclic guanosine analogues, acyclic nucleoside phosphonate analogues, pyrophosphate analogues, and oligonucleotides), (vii) varicella-zoster virus infections (acyclic guanosine analogues, nucleoside analogues, 5-substituted 2'-deoxyuridine analogues, and antibodies), (viii) respiratory syncytial virus infections (ribavirin and antibodies), and (ix) external anogenital warts caused by human papillomavirus infections (imiquimod, sinecatechins, and podofilox). Here, we present for the first time a comprehensive overview of antiviral drugs approved over the past 50 years, shedding light on the development of effective antiviral treatments against current and emerging infectious diseases worldwide.
Collapse
Affiliation(s)
- Erik De Clercq
- KU Leuven-University of Leuven, Rega Institute for Medical Research, Department of Microbiology and Immunology, Leuven, Belgium
| | - Guangdi Li
- KU Leuven-University of Leuven, Rega Institute for Medical Research, Department of Microbiology and Immunology, Leuven, Belgium Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Hogan AB, Anderssen RS, Davis S, Moore HC, Lim FJ, Fathima P, Glass K. Time series analysis of RSV and bronchiolitis seasonality in temperate and tropical Western Australia. Epidemics 2016; 16:49-55. [PMID: 27294794 DOI: 10.1016/j.epidem.2016.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/08/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes respiratory illness in young children and is most commonly associated with bronchiolitis. RSV typically occurs as annual or biennial winter epidemics in temperate regions, with less pronounced seasonality in the tropics. We sought to characterise and compare the seasonality of RSV and bronchiolitis in temperate and tropical Western Australia. We examined over 13 years of RSV laboratory identifications and bronchiolitis hospitalisations in children, using an extensive linked dataset from Western Australia. We applied mathematical time series analyses to identify the dominant seasonal cycle, and changes in epidemic size and timing over this period. Both the RSV and bronchiolitis data showed clear winter epidemic peaks in July or August in the southern Western Australia regions, but less identifiable seasonality in the northern regions. Use of complex demodulation proved very effective at comparing disease epidemics. The timing of RSV and bronchiolitis epidemics coincided well, but the size of the epidemics differed, with more consistent peak sizes for bronchiolitis than for RSV. Our results show that bronchiolitis hospitalisations are a reasonable proxy for the timing of RSV detections, but may not fully capture the magnitude of RSV epidemics.
Collapse
Affiliation(s)
- Alexandra B Hogan
- National Centre for Epidemiology and Population Health, Research School of Population Health, The Australian National University, Australia.
| | - Robert S Anderssen
- CSIRO Data61; Mathematical Sciences Institute, The Australian National University; Mathematics and Statistics, La Trobe University, Australia
| | - Stephanie Davis
- National Centre for Epidemiology and Population Health, Research School of Population Health, The Australian National University, Australia
| | - Hannah C Moore
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, The University of Western Australia, Australia
| | - Faye J Lim
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, The University of Western Australia, Australia
| | - Parveen Fathima
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, The University of Western Australia, Australia
| | - Kathryn Glass
- National Centre for Epidemiology and Population Health, Research School of Population Health, The Australian National University, Australia
| |
Collapse
|
39
|
Korsten K, Blanken MO, Nibbelke EE, Moons KGM, Bont L. Prediction model of RSV-hospitalization in late preterm infants: An update and validation study. Early Hum Dev 2016; 95:35-40. [PMID: 26930376 DOI: 10.1016/j.earlhumdev.2016.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND New vaccines and RSV therapeutics have been developed in the past decade. With approval of these new pharmaceuticals on the horizon, new challenges lie ahead in selecting the appropriate target population. We aimed to improve a previously published prediction model for prediction of RSV-hospitalization within the first year of life. METHODS Two consecutive prospective multicenter birth cohort studies were performed from June 2008 until February 2015. The first cohort (RISK-I, n=2524, 2008-2011) was used to update the existing model. The updated model was subsequently validated in the RISK-II cohort (n=1564, 2011-2015). We used the TRIPOD criteria for transparent reporting. RESULTS 181 infants (n=127 in RISK-I, n=54 in RISK-II) were hospitalized for RSV within their first year of life. The updated model included the following predictors; day care attendance and/or siblings (OR: 5.3; 95% CI 2.8-10.1), birth between Aug. 14th and Dec. 1st (OR: 2.4; 1.8-3.2), neonatal respiratory support (OR 2.2; 1.6-3.0), breastfeeding ≤4 months (OR 1.6; 1.2-2.2) and maternal atopic constitution (OR 1.5; 1.1-2.1). The updated models' discrimination was superior to the original model in the RISK-II cohort (AUROC 0.72 95% CI 0.65-0.78 versus AUROC 0.66, 95% CI 0.60-0.73, respectively). The updated model was translated into a simple nomogram to be able to distinguish infants with high versus low risk of RSV-hospitalization. CONCLUSION We developed and validated a clinical prediction model to be able to predict RSV-hospitalization in preterm infants born within 32-35 weeks gestational age. A simple nomogram was developed to target RSV therapeutics to those children who will benefit the most.
Collapse
Affiliation(s)
- Koos Korsten
- Division of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten O Blanken
- Division of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elisabeth E Nibbelke
- Division of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karel G M Moons
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Louis Bont
- Division of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
40
|
French CE, McKenzie BC, Coope C, Rajanaidu S, Paranthaman K, Pebody R, Nguyen-Van-Tam JS, Higgins JPT, Beck CR. Risk of nosocomial respiratory syncytial virus infection and effectiveness of control measures to prevent transmission events: a systematic review. Influenza Other Respir Viruses 2016; 10:268-90. [PMID: 26901358 PMCID: PMC4910170 DOI: 10.1111/irv.12379] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 01/14/2023] Open
Abstract
Respiratory syncytial virus (RSV) causes a significant public health burden, and outbreaks among vulnerable patients in hospital settings are of particular concern. We reviewed published and unpublished literature from hospital settings to assess: (i) nosocomial RSV transmission risk (attack rate) during outbreaks, (ii) effectiveness of infection control measures. We searched the following databases: MEDLINE, EMBASE, CINAHL, Cochrane Library, together with key websites, journals and grey literature, to end of 2012. Risk of bias was assessed using the Cochrane risk of bias tool or Newcastle–Ottawa scale. A narrative synthesis was conducted. Forty studies were included (19 addressing research question one, 21 addressing question two). RSV transmission risk varied by hospital setting; 6–56% (median: 28·5%) in neonatal/paediatric settings (n = 14), 6–12% (median: 7%) in adult haematology and transplant units (n = 3), and 30–32% in other adult settings (n = 2). For question two, most studies (n = 13) employed multi‐component interventions (e.g. cohort nursing, personal protective equipment (PPE), isolation), and these were largely reported to be effective in reducing nosocomial transmission. Four studies examined staff PPE; eye protection appeared more effective than gowns and masks. One study reported on RSV prophylaxis for patients (RSV‐Ig/palivizumab); there was no statistical evidence of effectiveness although the sample size was small. Overall, risk of bias for included studies tended to be high. We conclude that RSV transmission risk varies widely during hospital outbreaks. Although multi‐component control strategies appear broadly successful, further research is required to disaggregate the effectiveness of individual components including the potential role of palivizumab prophylaxis.
Collapse
Affiliation(s)
- Clare E French
- School of Social and Community Medicine, University of Bristol, Bristol, UK.,NIHR Health Protection Research Unit in Evaluation of Interventions at University of Bristol, Bristol, UK
| | | | - Caroline Coope
- School of Social and Community Medicine, University of Bristol, Bristol, UK.,NIHR Health Protection Research Unit in Evaluation of Interventions at University of Bristol, Bristol, UK.,Public Health England, London, UK
| | | | | | | | | | | | - Julian P T Higgins
- School of Social and Community Medicine, University of Bristol, Bristol, UK.,NIHR Health Protection Research Unit in Evaluation of Interventions at University of Bristol, Bristol, UK
| | - Charles R Beck
- School of Social and Community Medicine, University of Bristol, Bristol, UK.,NIHR Health Protection Research Unit in Evaluation of Interventions at University of Bristol, Bristol, UK.,Public Health England, London, UK
| |
Collapse
|
41
|
Zhou J, Zhang X, Liu S, Wang Z, Chen Q, Wu Y, He Z, Huang Z. Genetic association of TLR4 Asp299Gly, TLR4 Thr399Ile, and CD14 C-159T polymorphisms with the risk of severe RSV infection: a meta-analysis. Influenza Other Respir Viruses 2016; 10:224-33. [PMID: 26901241 PMCID: PMC4814857 DOI: 10.1111/irv.12378] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 01/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the most frequent cause of hospitalization in infants worldwide. It is recognized by Toll‐like receptor 4 (TLR 4) and cluster of differentiation 14 (CD14) in the innate immune response. Previous case–control studies reported the influence of TLR4 Asp299Gly, TLR4 Thr399Ile, and CD14 C‐159T polymorphisms on the risk of severe RSV infection. However, a decisive conclusion has not been achieved. Therefore, we performed this meta‐analysis to examine the association between these three polymorphisms and the development of RSV bronchiolitis. A systematic literature search was performed using the PubMed, EMbase, Google Scholar Search, China National Knowledge Infrastructure, China Biological Medicine, and Wanfang Databases. The data were extracted and pooled odds ratios with 95% confidence intervals were calculated under six genetic models. A total of six studies with 1009 cases and 1348 controls, three studies with 473 cases and 481 controls, or four studies with 325 cases and 650 controls relating to each of the three polymorphisms were included in this meta‐analysis. The analyzed data indicated that all of these polymorphisms were not associated with the risk of severe RSV infection. This is the first meta‐analysis to investigate the relationship of TLR4 Asp299Gly, TLR4 Thr399Ile, and CD14 C‐159T polymorphisms with the risk of severe RSV infection. Although the results of this retrospective analysis indicated a lack of the association, more extensive multicentric studies with large sample sizes are necessary to provide a more reliable estimation of the association between these three polymorphisms and RSV bronchiolitis susceptibility.
Collapse
Affiliation(s)
- Jiahui Zhou
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Xiangning Zhang
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Shuming Liu
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Ziyou Wang
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Qicong Chen
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yongfu Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhiwei He
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Zunnan Huang
- China-America Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, Guangdong, China.,Department of Pathophysiology, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
42
|
Rivera CA, Gómez RS, Díaz RA, Céspedes PF, Espinoza JA, González PA, Riedel CA, Bueno SM, Kalergis AM. Novel therapies and vaccines against the human respiratory syncytial virus. Expert Opin Investig Drugs 2015; 24:1613-30. [DOI: 10.1517/13543784.2015.1099626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Sanchez JL, Cooper MJ, Myers CA, Cummings JF, Vest KG, Russell KL, Sanchez JL, Hiser MJ, Gaydos CA. Respiratory Infections in the U.S. Military: Recent Experience and Control. Clin Microbiol Rev 2015; 28:743-800. [PMID: 26085551 PMCID: PMC4475643 DOI: 10.1128/cmr.00039-14] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This comprehensive review outlines the impact of military-relevant respiratory infections, with special attention to recruit training environments, influenza pandemics in 1918 to 1919 and 2009 to 2010, and peacetime operations and conflicts in the past 25 years. Outbreaks and epidemiologic investigations of viral and bacterial infections among high-risk groups are presented, including (i) experience by recruits at training centers, (ii) impact on advanced trainees in special settings, (iii) morbidity sustained by shipboard personnel at sea, and (iv) experience of deployed personnel. Utilizing a pathogen-by-pathogen approach, we examine (i) epidemiology, (ii) impact in terms of morbidity and operational readiness, (iii) clinical presentation and outbreak potential, (iv) diagnostic modalities, (v) treatment approaches, and (vi) vaccine and other control measures. We also outline military-specific initiatives in (i) surveillance, (ii) vaccine development and policy, (iii) novel influenza and coronavirus diagnostic test development and surveillance methods, (iv) influenza virus transmission and severity prediction modeling efforts, and (v) evaluation and implementation of nonvaccine, nonpharmacologic interventions.
Collapse
Affiliation(s)
- Jose L Sanchez
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA
| | - Michael J Cooper
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA
| | | | - James F Cummings
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA
| | - Kelly G Vest
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA
| | - Kevin L Russell
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA
| | - Joyce L Sanchez
- Mayo Clinic, Division of General Internal Medicine, Rochester, Minnesota, USA
| | - Michelle J Hiser
- Armed Forces Health Surveillance Center, Silver Spring, Maryland, USA Oak Ridge Institute for Science and Education, Postgraduate Research Participation Program, U.S. Army Public Health Command, Aberdeen Proving Ground, Aberdeen, Maryland, USA
| | - Charlotte A Gaydos
- International STD, Respiratory, and Biothreat Research Laboratory, Division of Infectious Diseases, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|