1
|
Perčinić A, Vuletić T, Lizzul N, Vukić Dugac A, Gverić Grginić A, Tabain I, Jurić D, Budimir A. Epidemiological and Clinical Characteristics of Adult RSV Infections: A Retrospective Analysis at University Hospital Center Zagreb (2022-2024). Pathogens 2025; 14:284. [PMID: 40137769 PMCID: PMC11946814 DOI: 10.3390/pathogens14030284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of respiratory infections in adults, particularly among older adults and individuals with chronic diseases. While traditionally linked to pediatric populations, RSV's impact on adults, especially the elderly, is increasingly recognized but remains understudied in many regions. This retrospective study, conducted at the University Hospital Center Zagreb from October 2022 to April 2024, is the first to analyze RSV-positive adults in Croatia. Using RT-PCR testing, we evaluated clinical and epidemiological characteristics in both hospitalized and outpatient populations, focusing on those aged > 65 years. Among 2631 tested individuals, the RSV prevalence was 5.25%, with older adults experiencing the most severe outcomes, including pneumonia, COPD exacerbation, and intensive care admissions. Seasonal analysis confirmed a winter peak in RSV cases, while chronic conditions such as cardiovascular and respiratory diseases were strongly associated with higher complication rates. These findings demonstrate that older adults with comorbidities bear the greatest burden of RSV infection, highlighting the need for the early identification of high-risk patients. By providing detailed insights into RSV-related outcomes in this population, this study supports the development of targeted prevention and management strategies to reduce the burden of RSV in vulnerable groups.
Collapse
Affiliation(s)
- Antonio Perčinić
- Department of Clinical Microbiology, Prevention, and Infection Control, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (N.L.); (A.B.)
| | - Tara Vuletić
- Department of Respiratory Infections, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (T.V.); (A.V.D.)
| | - Nina Lizzul
- Department of Clinical Microbiology, Prevention, and Infection Control, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (N.L.); (A.B.)
| | - Andrea Vukić Dugac
- Department of Respiratory Infections, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (T.V.); (A.V.D.)
| | - Ana Gverić Grginić
- Microbiological Service, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (A.G.G.); (I.T.); (D.J.)
| | - Irena Tabain
- Microbiological Service, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (A.G.G.); (I.T.); (D.J.)
| | - Dragan Jurić
- Microbiological Service, Croatian Institute of Public Health, 10000 Zagreb, Croatia; (A.G.G.); (I.T.); (D.J.)
| | - Ana Budimir
- Department of Clinical Microbiology, Prevention, and Infection Control, University Hospital Center Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia; (N.L.); (A.B.)
| |
Collapse
|
2
|
Parsons EL, Kim JS, Malloy AMW. Development of innate and adaptive immunity to RSV in young children. Cell Immunol 2024; 399-400:104824. [PMID: 38615612 DOI: 10.1016/j.cellimm.2024.104824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Infection of the respiratory tract with respiratory syncytial virus (RSV) is common and occurs repeatedly throughout life with most severe disease occurring at the extremes of age: in young infants and the elderly. Effective anti-viral therapeutics are not available and therefore prevention has been the primary strategy for reducing the disease burden. Our current understanding of respiratory mucosal cell biology and the immune response within the respiratory tract is inadequate to prevent infection caused by a pathogen like RSV that does not disseminate outside of this environment. Gaps in our understanding of the activation of innate and adaptive immunity in response to RSV and the role of age upon infection also limit improvements in the design of therapeutics and vaccines for young infants. However, advancements in structural biology have improved our ability to characterize antibodies against viral proteins and in 2023 the first vaccines for those over 60 years and pregnant women became available, potentially reducing the burden of disease. This review will examine our current understanding of the critical facets of anti-RSV immune responses in infants and young children as well as highlight areas where more research is needed.
Collapse
Affiliation(s)
| | - Jisung S Kim
- Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation, Bethesda, MD, USA
| | | |
Collapse
|
3
|
Verwey C, Dangor Z, Madhi SA. Approaches to the Prevention and Treatment of Respiratory Syncytial Virus Infection in Children: Rationale and Progress to Date. Paediatr Drugs 2024; 26:101-112. [PMID: 38032456 PMCID: PMC10891269 DOI: 10.1007/s40272-023-00606-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 12/01/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection (LRTI) in children, and is associated with long-term pulmonary sequelae for up to 30 years after infection. The mainstay of RSV management is supportive therapy such as supplemental oxygen. Palivizumab (Synagis™-AstraZeneca), a monoclonal antibody targeting the RSV F protein site II, has been licensed for the prevention of RSV in high-risk groups since 1998. There has been recent promising progress in preventative strategies that include vaccines and long-acting, high-potency monoclonal antibodies. Nirsevimab (Beyfortus™-AstraZeneca/Sanofi), a monoclonal antibody with an extended half-life, has recently been registered in the European Union and granted licensure by the US Food and Drug Administration. Furthermore, a pre-fusion sub-unit protein vaccine has been granted licensure for pregnant women, aimed at protecting their young infants, following established safety and efficacy in clinical trials (Abrysvo™-Pfizer). Also, multiple novel antiviral therapeutic options are in early phase clinical trials. The next few years have the potential to change the landscape of LRTI through improvements in the prevention and management of RSV LRTI. Here, we discuss these new approaches, current research, and clinical trials in novel therapeutics, monoclonal antibodies, and vaccines against RSV infection in infants and children.
Collapse
Affiliation(s)
- Charl Verwey
- Department of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Ziyaad Dangor
- Department of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
4
|
Raphael A, Schlesinger Y, Nir A. Sinus arrest during respiratory syncytial virus bronchiolitis: A report of two cases and literature review. Pediatr Pulmonol 2023; 58:3330-3332. [PMID: 37417816 DOI: 10.1002/ppul.26589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/04/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Affiliation(s)
- Allon Raphael
- Department of Pediatrics, Wilf Children Hospital, Sha'are Zedek Medical Center, Affiliated to the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| | - Yechiel Schlesinger
- Department of Pediatrics, Wilf Children Hospital, Sha'are Zedek Medical Center, Affiliated to the Hadassah-Hebrew University Medical School, Jerusalem, Israel
- Pediatric Infectious Diseases Unit, Wilf Children Hospital, Shaare Zedek Medical Center, Affiliated to the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| | - Amiram Nir
- Department of Pediatrics, Wilf Children Hospital, Sha'are Zedek Medical Center, Affiliated to the Hadassah-Hebrew University Medical School, Jerusalem, Israel
- Pediatric Cardiology and Adult Congenital Heart Disease Unit, Wilf Children Hospital, Sha'are Zedek Medical Center, Affiliated to the Hadassah-Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
5
|
Singh S, Maheshwari A, Namazova I, Benjamin JT, Wang Y. Respiratory Syncytial Virus Infections in Neonates: A Persisting Problem. NEWBORN (CLARKSVILLE, MD.) 2023; 2:222-234. [PMID: 38348152 PMCID: PMC10860331 DOI: 10.5005/jp-journals-11002-0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infections in young infants. It is an enveloped, single-stranded, nonsegmented, negative-strand RNA virus, a member of the family Pneumoviridae. Globally, RSV is responsible for 2.3% of deaths among neonates 0-27 days of age. Respiratory syncytial virus infection is most common in children aged below 24 months. Neonates present with cough and fever. Respiratory syncytial virus-associated wheezing is seen in 20% infants during the first year of life of which 2-3% require hospitalization. Reverse transcriptase polymerase chain reaction (RT-PCR) gives fast results and has higher sensitivity compared with culture and rapid antigen tests and are not affected by passively administered antibody to RSV. Therapy for RSV infection of the LRT is mainly supportive, and preventive measures like good hygiene and isolation are the mainstay of management. Standard precautions, hand hygiene, breastfeeding and contact isolation should be followed for RSV-infected newborns. Recent AAP guidelines do not recommend pavilizumab prophylaxis for preterm infants born at 29-35 weeks without chronic lung disease, hemodynamically significant congenital heart disease and coexisting conditions. RSV can lead to long-term sequelae such as wheezing and asthma, associated with increased healthcare costs and reduced quality of life.
Collapse
Affiliation(s)
- Srijan Singh
- Neonatologist, Kailash Hospital, Noida, Uttar Pradesh, India
- Global Newborn Society (https://www.globalnewbornsociety.org/)
| | - Akhil Maheshwari
- Global Newborn Society (https://www.globalnewbornsociety.org/)
- Department of Pediatrics, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Ilhama Namazova
- Global Newborn Society (https://www.globalnewbornsociety.org/)
- Department of Pediatrics, Azerbaijan Tibb Universiteti, Baku, Azerbaijan
| | - John T Benjamin
- Global Newborn Society (https://www.globalnewbornsociety.org/)
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Yuping Wang
- Department of Obstetrics and Gynaecology, Louisiana State University, Shreveport, Louisiana, United States of America
| |
Collapse
|
6
|
Verwey C, Madhi SA. Review and Update of Active and Passive Immunization Against Respiratory Syncytial Virus. BioDrugs 2023; 37:295-309. [PMID: 37097594 PMCID: PMC10127166 DOI: 10.1007/s40259-023-00596-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/26/2023]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection (LRTI) in children, causing approximately 3.6 million hospitalizations per year, and has been associated with long-term pulmonary sequelae for up to 30 years after infection, yet preventative strategies and active treatment options remain elusive. The associated morbidity and healthcare related costs could be decreased substantially with the development of these much-needed medications. After an initial false start in the development of an RSV vaccine, gradual progress is now being made with the development of multiple vaccine candidates using numerous different mechanisms of action. Furthermore, nirsevimab, a new monoclonal antibody for the prevention of RSV, has recently been registered in the European Union. New novel treatments for RSV infection are also in the pipeline, which would provide the clinician with much needed ammunition in the management of the acute disease. The next few years have the potential to change the landscape of LRTI forever through the prevention and management of RSV LRTI and thereby decrease the mortality and morbidity associated with it. In this review, we discuss these new approaches, current research, and clinical trials in monoclonal antibody and vaccine development against RSV.
Collapse
Affiliation(s)
- Charl Verwey
- Department of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise (ALIVE), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
7
|
Huang L, Liu MQ, Wan CQ, Cheng NN, Su YB, Zheng YP, Peng XL, Yu JM, Fu YH, He JS. The protective immunity induced by intranasally inoculated serotype 63 chimpanzee adenovirus vector expressing human respiratory syncytial virus prefusion fusion glycoprotein in BALB/c mice. Front Microbiol 2022; 13:1041338. [PMID: 36466668 PMCID: PMC9716990 DOI: 10.3389/fmicb.2022.1041338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/21/2022] [Indexed: 12/23/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is a ubiquitous pediatric pathogen causing serious lower respiratory tract disease worldwide. No licensed vaccine is currently available. In this work, the coding gene for mDS-Dav1, the full-length and prefusion conformation RSV fusion glycoprotein (F), was designed by introducing the stabilized prefusion F (preF) mutations from DS-Cav1 into the encoding gene of wild-type RSV (wtRSV) F protein. The recombinant adenovirus encoding mDS-Cav1, rChAd63-mDS-Cav1, was constructed based on serotype 63 chimpanzee adenovirus vector and characterized in vitro. After immunizing mice via intranasal route, the rChAd63-mDS-Cav1 induced enhanced neutralizing antibody and F-specific CD8+ T cell responses as well as good immune protection against RSV challenge with the absence of enhanced RSV disease (ERD) in BALB/c mice. The results indicate that rChAd63-mDS-Cav1 is a promising mucosal vaccine candidate against RSV infection and warrants further development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuan-Hui Fu
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Jin-Sheng He
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
8
|
Šantak M, Matić Z. The Role of Nucleoprotein in Immunity to Human Negative-Stranded RNA Viruses—Not Just Another Brick in the Viral Nucleocapsid. Viruses 2022; 14:v14030521. [PMID: 35336928 PMCID: PMC8955406 DOI: 10.3390/v14030521] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Negative-stranded RNA viruses (NSVs) are important human pathogens, including emerging and reemerging viruses that cause respiratory, hemorrhagic and other severe illnesses. Vaccine design traditionally relies on the viral surface glycoproteins. However, surface glycoproteins rarely elicit effective long-term immunity due to high variability. Therefore, an alternative approach is to include conserved structural proteins such as nucleoprotein (NP). NP is engaged in myriad processes in the viral life cycle: coating and protection of viral RNA, regulation of transcription/replication processes and induction of immunosuppression of the host. A broad heterosubtypic T-cellular protection was ascribed very early to this protein. In contrast, the understanding of the humoral immunity to NP is very limited in spite of the high titer of non-neutralizing NP-specific antibodies raised upon natural infection or immunization. In this review, the data with important implications for the understanding of the role of NP in the immune response to human NSVs are revisited. Major implications of the elicited T-cell immune responses to NP are evaluated, and the possible multiple mechanisms of the neglected humoral response to NP are discussed. The intention of this review is to remind that NP is a very promising target for the development of future vaccines.
Collapse
|
9
|
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
|
10
|
McGinley J, Thwaites R, Brebner W, Greenan-Barrett L, Aerssens J, Öner D, Bont L, Wildenbeest J, Martinón-Torres F, Nair H, Pollard AJ, Openshaw P, Drysdale S. A Systematic Review and Meta-analysis of Animal Studies Investigating the Relationship Between Serum Antibody, T Lymphocytes, and Respiratory Syncytial Virus Disease. J Infect Dis 2021; 226:S117-S129. [PMID: 34522970 DOI: 10.1093/infdis/jiab370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infections occur in human populations around the globe, causing disease of variable severity, disproportionately affecting infants and older adults (>65 years of age). Immune responses can be protective but also contribute to disease. Experimental studies in animals enable detailed investigation of immune responses, provide insights into clinical questions, and accelerate the development of passive and active vaccination. We aimed to review the role of antibody and T-cell responses in relation to RSV disease severity in animals. METHODS Systematic review and meta-analysis of animal studies examining the association between T-cell responses/phenotype or antibody titers and severity of RSV disease. The PubMed, Zoological Record, and Embase databases were screened from January 1980 to May 2018 to identify animal studies of RSV infection that assessed serum antibody titer or T lymphocytes with disease severity as an outcome. Sixty-three studies were included in the final review. RESULTS RSV-specific antibody appears to protect from disease in mice, but such an effect was less evident in bovine RSV. Strong T-cell, Th1, Th2, Th17, CD4/CD8 responses, and weak Treg responses accompany severe disease in mice. CONCLUSIONS Murine studies suggest that measures of T-lymphocyte activity (particularly CD4 and CD8 T cells) may be predictive biomarkers of severity. Further inquiry is merited to validate these results and assess relevance as biomarkers for human disease.
Collapse
Affiliation(s)
- Joseph McGinley
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Will Brebner
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Jeroen Aerssens
- Biomarkers Infectious Diseases, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Deniz Öner
- Biomarkers Infectious Diseases, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Louis Bont
- Department of Paediatric Infectious Diseases and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Joanne Wildenbeest
- Department of Paediatric Infectious Diseases and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Harish Nair
- University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Simon Drysdale
- Oxford Vaccine Group, Paediatrics, University of Oxford, Oxford, United Kingdom.,Paediatric Infectious Diseases Unit, St George's University Hospitals NHS Foundation Trust, London, United Kingdom.,Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | | |
Collapse
|
11
|
Pathogenesis of a novel porcine parainfluenza virus type 1 isolate in conventional and colostrum deprived/caesarean derived pigs. Virology 2021; 563:88-97. [PMID: 34500147 DOI: 10.1016/j.virol.2021.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 11/19/2022]
Abstract
Two experimental challenge studies were conducted to evaluate the pathogenesis of a porcine parainfluenza virus type 1 (PPIV-1) isolate. Four-week-old conventional (CON) pigs were challenged in Study 1 and six-week-old caesarean derived/colostrum deprived (CDCD) pigs were challenged in Study 2. Results indicate that PPIV-1 shedding and replication occur in the upper and lower respiratory tracts of CON and CDCD pigs as detected by RT-qPCR and immunohistochemistry. Mild macroscopic lung lesions were observed in CON pigs but not in CDCD pigs. Microscopic lung lesions were mild and consisted of peribronchiolar lymphocytic cuffing and epithelial proliferation in CON and CDCD pigs. Serum neutralizing antibodies were detected in the CON and CDCD pigs by 14 and 7 days post inoculation, respectively. This study provides evidence that in spite of PPIV-1 infection and replication in challenged swine, significant clinical respiratory disease was not observed.
Collapse
|
12
|
The specific features of the developing T cell compartment of the neonatal lung are a determinant of respiratory syncytial virus immunopathogenesis. PLoS Pathog 2021; 17:e1009529. [PMID: 33909707 PMCID: PMC8109812 DOI: 10.1371/journal.ppat.1009529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/10/2021] [Accepted: 04/05/2021] [Indexed: 11/19/2022] Open
Abstract
The human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants, possibly due to the properties of the immature neonatal pulmonary immune system. Using the newborn lamb, a classical model of human lung development and a translational model of RSV infection, we aimed to explore the role of cell-mediated immunity in RSV disease during early life. Remarkably, in healthy conditions, the developing T cell compartment of the neonatal lung showed major differences to that seen in the mature adult lung. The most striking observation being a high baseline frequency of bronchoalveolar IL-4-producing CD4+ and CD8+ T cells, which declined progressively over developmental age. RSV infection exacerbated this pro-type 2 environment in the bronchoalveolar space, rather than inducing a type 2 response per se. Moreover, regulatory T cell suppressive functions occurred very early to dampen this pro-type 2 environment, rather than shutting them down afterwards, while γδ T cells dropped and failed to produce IL-17. Importantly, RSV disease severity was related to the magnitude of those unconventional bronchoalveolar T cell responses. These findings provide novel insights in the mechanisms of RSV immunopathogenesis in early life, and constitute a major step for the understanding of RSV disease severity.
Collapse
|
13
|
Asha K, Khanna M, Kumar B. Current Insights into the Host Immune Response to Respiratory Viral Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:59-83. [PMID: 34661891 DOI: 10.1007/978-3-030-67452-6_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Respiratory viral infections often lead to severe illnesses varying from mild or asymptomatic upper respiratory tract infections to severe bronchiolitis and pneumonia or/and chronic obstructive pulmonary disease. Common viral infections, including but not limited to influenza virus, respiratory syncytial virus, rhinovirus and coronavirus, are often the leading cause of morbidity and mortality. Since the lungs are continuously exposed to foreign particles, including respiratory pathogens, it is also well equipped for recognition and antiviral defense utilizing the complex network of innate and adaptive immune cells. Immediately upon infection, a range of proinflammatory cytokines, chemokines and an interferon response is generated, thereby making the immune response a two edged sword, on one hand it is required to eliminate viral pathogens while on other hand it's prolonged response can lead to chronic infection and significant pulmonary damage. Since vaccines to all respiratory viruses are not available, a better understanding of the virus-host interactions, leading to the development of immune response, is critically needed to design effective therapies to limit the severity of inflammatory damage, enhance viral clearance and to compliment the current strategies targeting the virus. In this chapter, we discuss the host responses to common respiratory viral infections, the key players of adaptive and innate immunity and the fine balance that exists between the viral clearance and immune-mediated damage.
Collapse
Affiliation(s)
- Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Madhu Khanna
- Department of Virology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Binod Kumar
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
14
|
Samy N, Reichhardt D, Schmidt D, Chen LM, Silbernagl G, Vidojkovic S, Meyer TP, Jordan E, Adams T, Weidenthaler H, Stroukova D, De Carli S, Chaplin P. Safety and immunogenicity of novel modified vaccinia Ankara-vectored RSV vaccine: A randomized phase I clinical trial. Vaccine 2020; 38:2608-2619. [PMID: 32057576 DOI: 10.1016/j.vaccine.2020.01.055] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/18/2019] [Accepted: 01/18/2020] [Indexed: 10/25/2022]
Abstract
Respiratory disease caused by RSV infection is recognized as a severe public health issue in infants, young children and elderly with no specific treatment option. Vaccination may be the most effective strategy to combat this highly infectious virus although no vaccine has been approved. The novel vaccine candidate MVA-BN-RSV encodes RSV surface proteins F and G (subtypes A, B) as well as internal proteins N and M2 in the MVA-BN viral vector backbone to provide broad protection against RSV. This was a first in human study to investigate safety, reactogenicity and immunogenicity of MVA-BN-RSV. Sixty-three participants were allocated to 3 groups: adult (18-49 years) low (1 × 107 TCID50) or high (1 × 108 TCID50) dose and older adult (50-65 years) high dose. Participants in each group were randomized in a 6:1 ratio to receive 2 doses of MVA-BN-RSV or placebo 4 weeks apart and were monitored for 30 weeks. All participants completed the study, receiving both doses. No serious AEs or AEs of special interest were reported. The most common AEs were injection site pain (56% in the combined high dose groups, 17% in the low dose group). MVA-BN-RSV induced robust T cell responses covering all 5 inserts with fold increases ranging from 1.8 to 3.8. Higher and broader responses were observed in the high dose groups (83% responders to at least 3 peptide pools in the combined high dose groups compared to 63% in the low dose group). Moderate but consistent humoral responses were observed against A and B RSV subtypes (up to approximately 2-fold increases in the high dose groups). No differences were observed between the adult and the older adult groups in safety, reactogenicity or immunogenicity. The study demonstrated that the well tolerated MVA-BN-RSV vaccine candidate induces broad cellular and humoral immune responses, warranting further development.
Collapse
Affiliation(s)
- Nathaly Samy
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | | | - Darja Schmidt
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Liddy M Chen
- Bavarian Nordic Inc, 3025 Carrington Mill Boulevard, Morrisville, NC 27560, United States
| | - Günter Silbernagl
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Sanja Vidojkovic
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Thomas Ph Meyer
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Elke Jordan
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Tatiana Adams
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | | | - Daria Stroukova
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Sonja De Carli
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, 82152 Martinsried, Germany
| | - Paul Chaplin
- Bavarian Nordic A/S, Hejreskovvej 10A, DK-3490 Kvistgård, Denmark
| |
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
|
Ivey KS, Edwards KM, Talbot HK. Respiratory Syncytial Virus and Associations With Cardiovascular Disease in Adults. J Am Coll Cardiol 2019; 71:1574-1583. [PMID: 29622165 DOI: 10.1016/j.jacc.2018.02.013] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/02/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022]
Abstract
Respiratory syncytial virus (RSV) is historically known for causing respiratory illness in young children, but the appreciation of its impact on older adults is growing. Studies have shown that hospitalization for respiratory illness due to RSV is complicated by cardiovascular events in 14% to 22% of adult patients, including worsening congestive heart failure, acute coronary syndrome, and arrhythmias. Additionally, underlying cardiovascular disease is associated with hospitalization in 45% to 63% of adults with confirmed RSV. In summary, patients with cardiopulmonary disease have higher rates of health care utilization for RSV-related illness and worse outcomes. Patients with cardiovascular disease likely represent an important target population for the rapidly developing field of RSV vaccines.
Collapse
Affiliation(s)
- Kelsey S Ivey
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kathryn M Edwards
- Department of Pediatrics, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| | - H Keipp Talbot
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Health Policy, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
17
|
López D, Barriga A, Lorente E, Mir C. Immunoproteomic Lessons for Human Respiratory Syncytial Virus Vaccine Design. J Clin Med 2019; 8:E486. [PMID: 30974886 PMCID: PMC6518116 DOI: 10.3390/jcm8040486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 11/25/2022] Open
Abstract
Accurate antiviral humoral and cellular immune responses require prior recognition of antigenic peptides presented by human leukocyte antigen (HLA) class I and II molecules on the surface of antigen-presenting cells. Both the helper and the cytotoxic immune responses are critical for the control and the clearance of human respiratory syncytial virus (HRSV) infection, which is a significant cause of morbidity and mortality in infected pediatric, immunocompromised and elderly populations. In this article we review the immunoproteomics studies which have defined the general antigen processing and presentation rules that determine both the immunoprevalence and the immunodominance of the cellular immune response to HRSV. Mass spectrometry and functional analyses have shown that the HLA class I and II cellular immune responses against HRSV are mainly focused on three viral proteins: fusion, matrix, and nucleoprotein. Thus, these studies have important implications for vaccine development against this virus, since a vaccine construct including these three relevant HRSV proteins could efficiently stimulate the major components of the adaptive immune system: humoral, helper, and cytotoxic effector immune responses.
Collapse
Affiliation(s)
- Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain.
| | - Alejandro Barriga
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain.
| | - Elena Lorente
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain.
| | - Carmen Mir
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda (Madrid), Spain.
| |
Collapse
|
18
|
Ye X, Iwuchukwu OP, Avadhanula V, Aideyan LO, McBride TJ, Ferlic-Stark LL, Patel KD, Piedra FA, Shah DP, Chemaly RF, Piedra PA. Antigenic Site-Specific Competitive Antibody Responses to the Fusion Protein of Respiratory Syncytial Virus Were Associated With Viral Clearance in Hematopoietic Cell Transplantation Adults. Front Immunol 2019; 10:706. [PMID: 30984206 PMCID: PMC6449644 DOI: 10.3389/fimmu.2019.00706] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 03/14/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Recent studies of human sera showed that the majority of the respiratory syncytial virus (RSV) neutralizing antibodies are directed against pre-fusion conformation of the fusion (F) protein of RSV and revealed the importance of pre-fusion antigenic site Ø specific antibodies. However, detailed analysis of multiple antigenic site-specific competitive antibody responses to RSV F protein and their contribution to virus clearance in humans are lacking. Methods: We prospectively enrolled a cohort of RSV infected hematopoietic cell transplantation (HCT) adults (n = 40). Serum samples were collected at enrollment (acute, n = 40) and 14 to 60 days post-enrollment (convalescent, n = 40). Antigenic site-specific F protein antibodies were measured against pre-fusion site Ø, post-fusion site I, and sites II and IV present in both the pre-fusion and post-fusion F protein conformations utilizing four different competitive antibody assays developed with biotinylated monoclonal antibodies (mAb) D25, 131-2A, palivizumab, and 101F, respectively. The lower limit of detection were 7.8 and 1.0 μg/mL for the competitive antibody assays that measured site Ø specific response, as well as sites I, II, and IV specific responses, respectively. Neutralizing antibody titers to RSV A and B subgroups was determined by microneutralization assays. Results: The overall findings in RSV infected HCT adults revealed: (1) a significant increase in antigenic site-specific competitive antibodies in convalescent sera except for site Ø competitive antibody (p < 0.01); (2) comparable concentrations in the acute and convalescent serum samples of antigenic site-specific competitive antibodies between RSV/A and RSV/B infected HCT adults (p > 0.05); (3) significantly increased concentrations of the antigenic site-specific competitive antibodies in HCT adults who had genomic RSV detected in the upper respiratory tract for <14 days compared to those for ≥14 days (p < 0.01); and (4) statistically significant correlation between the antigenic site-specific competitive antibody concentrations and neutralizing antibody titers against RSV/A and RSV/B (r ranged from 0.33 to 0.83 for acute sera, and 0.50-0.88 for convalescent sera; p < 0.05). Conclusions: In RSV infected HCT adults, antigenic site-specific antibody responses were induced against multiple antigenic sites found in both the pre-fusion and post-fusion F conformations, and were associated with a more rapid viral clearance and neutralizing antibody activity. However, the association is not necessarily the cause and the consequence.
Collapse
Affiliation(s)
- Xunyan Ye
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Obinna P Iwuchukwu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Letisha O Aideyan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Trevor J McBride
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Laura L Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Kirtida D Patel
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Dimpy P Shah
- Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Roy F Chemaly
- Departments of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
19
|
Science M, Akseer N, Asner S, Allen U. Risk stratification of immunocompromised children, including pediatric transplant recipients at risk of severe respiratory syncytial virus disease. Pediatr Transplant 2019; 23:e13336. [PMID: 30604582 DOI: 10.1111/petr.13336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/18/2018] [Accepted: 11/02/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is associated with increased morbidity and mortality in immunocompromised patients. Our goal was to develop a framework for risk stratifying immunocompromised patients, including transplant patients, for RSV prophylaxis. METHODS Risk factors for severe RSV disease in immunocompromised patients were identified in the literature and by an expert panel via survey. Experts assigned a probability of developing severe disease (0 to 100 scale) to the risk factors for each immunocompromised population. The results were validated using a clinical dataset. Linear mixed models adjusted for within-expert clustering of ranks were used to estimate average scores, and differences were tested using paired t tests. Logistic regression was utilized to identify important determinants of severe RSV disease. RESULTS The survey was emailed to twenty-seven experts and thirteen responded (48%). Across all transplant groups, age <2 years (mean 77.1, 95% CI 71.7, 82.5) and day care attendance (mean 72.8, 95% CI 67.3, 78.3) were assigned the highest risk of severe disease. The highest risk groups were lung transplant recipients (mean 73.2, 95% CI 67.6, 78.8), combined lung and heart transplant recipients (mean 75.2, 95% CI 69.6, 80.7), allogeneic stem cell transplant (mean 76.0, 95% CI 70.4, 81.6), and severe combined immunodeficiency (mean 74.7, 95% CI 69.1, 80.3). CONCLUSION The results provide a logical validity to current practice and provide guidance for prioritizing patients to receive prophylactic agents to prevent severe RSV disease. The results will facilitate the development of a risk stratification tool for RSV prophylaxis for immunocompromised patients.
Collapse
Affiliation(s)
- Michelle Science
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nadia Akseer
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,University of Toronto, Toronto, Ontario, Canada
| | - Sandra Asner
- Pediatric Infectious Diseases Unit, Department of Pediatrics, University Hospital Lausanne, Lausanne, Switzerland
| | - Upton Allen
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.,University of Toronto, Toronto, Ontario, Canada.,Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Schmidt ME, Varga SM. Identification of Novel Respiratory Syncytial Virus CD4 + and CD8 + T Cell Epitopes in C57BL/6 Mice. Immunohorizons 2019; 3:1-12. [PMID: 31356172 DOI: 10.4049/immunohorizons.1800056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/15/2018] [Indexed: 11/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection and hospitalization in infants. It is well established that both CD4+ and CD8+ T cells are critical for mediating viral clearance but also contribute to the induction of immunopathology following RSV infection. C57BL/6 mice are often used to study T cell responses following RSV infection given the wide variety of genetically modified animals available. To date, few RSV-derived CD4+ and CD8+ T cell epitopes have been identified in C57BL/6 mice. Using an overlapping peptide library spanning the entire RSV proteome, intracellular cytokine staining for IFN-γ was performed to identify novel CD4+ and CD8+ T cell epitopes in C57BL/6 mice. We identified two novel CD4+ T cell epitopes and three novel CD8+ T cell epitopes located within multiple RSV proteins. Additionally, we characterized the newly described T cell epitopes by determining their TCR Vβ expression profiles and MHC restriction. Overall, the novel RSV-derived CD4+ and CD8+ T cell epitopes identified in C57BL/6 mice will aid in future studies of RSV-specific T cell responses.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242; .,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242; and.,Department of Pathology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
21
|
Schmidt ME, Varga SM. Cytokines and CD8 T cell immunity during respiratory syncytial virus infection. Cytokine 2018; 133:154481. [PMID: 30031680 PMCID: PMC6551303 DOI: 10.1016/j.cyto.2018.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 01/10/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization in infants. In spite of the enormous clinical burden caused by RSV infections, there remains no efficacious RSV vaccine. CD8 T cells mediate viral clearance as well as provide protection against a secondary RSV infection. However, RSV-specific CD8 T cells may also induce immunopathology leading to exacerbated morbidity and mortality. Many of the crucial functions performed by CD8 T cells are mediated by the cytokines they produce. IFN-γ and TNF are produced by CD8 T cells following RSV infection and contribute to both the acceleration of viral clearance and the induction of immunopathology. To prevent immunopathology, regulatory mechanisms are in place within the immune system to inhibit CD8 T cell effector functions after the infection has been cleared. The actions of a variety of cytokines, including IL-10 and IL-4, play a critical role in the regulation of CD8 T cell effector activity. Herein, we review the current literature on CD8 T cell responses and the functions of the cytokines they produce following RSV infection. Additionally, we discuss the regulation of CD8 T cell activation and effector functions through the actions of various cytokines.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Department of Pathology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
22
|
Schmidt ME, Varga SM. The CD8 T Cell Response to Respiratory Virus Infections. Front Immunol 2018; 9:678. [PMID: 29686673 PMCID: PMC5900024 DOI: 10.3389/fimmu.2018.00678] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Humans are highly susceptible to infection with respiratory viruses including respiratory syncytial virus (RSV), influenza virus, human metapneumovirus, rhinovirus, coronavirus, and parainfluenza virus. While some viruses simply cause symptoms of the common cold, many respiratory viruses induce severe bronchiolitis, pneumonia, and even death following infection. Despite the immense clinical burden, the majority of the most common pulmonary viruses lack long-lasting efficacious vaccines. Nearly all current vaccination strategies are designed to elicit broadly neutralizing antibodies, which prevent severe disease following a subsequent infection. However, the mucosal antibody response to many respiratory viruses is not long-lasting and declines with age. CD8 T cells are critical for mediating clearance following many acute viral infections in the lung. In addition, memory CD8 T cells are capable of providing protection against secondary infections. Therefore, the combined induction of virus-specific CD8 T cells and antibodies may provide optimal protective immunity. Herein, we review the current literature on CD8 T cell responses induced by respiratory virus infections. Additionally, we explore how this knowledge could be utilized in the development of future vaccines against respiratory viruses, with a special emphasis on RSV vaccination.
Collapse
Affiliation(s)
- Megan E Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
23
|
Shafique M, Rasool MH, Khurshid M. Respiratory syncytial virus: an overview of infection biology and vaccination strategies. Future Virol 2017. [DOI: 10.2217/fvl-2017-0120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Respiratory syncytial virus (RSV) is the foremost cause of lower respiratory tract infections, especially in infants and young children. To date, there is no licensed vaccine available for RSV. Only option to restrain RSV is a prophylactic treatment in the form of monoclonal antibody (palivizumab). However, it is quite expensive and used in few patients with co-morbidities. In ongoing research, virologists contemplate about various vaccine candidates to control RSV infection. This review will help in understating the RSV pathobiology and encompass the advancement on various vaccine candidates that would lead to reduce the incidence, mortality and morbidity. Furthermore, it will lighten up the different avenues which might be useful for the development of novel vaccination approaches.
Collapse
Affiliation(s)
- Muhammad Shafique
- Department of Microbiology, Government College University Faisalabad, Pakistan
| | | | - Mohsin Khurshid
- Department of Microbiology, Government College University Faisalabad, Pakistan
- College of Allied Health Professionals, Directorate of Medical Sciences, Government College University Faisalabad, Pakistan
| |
Collapse
|
24
|
Shafique M, Rasool MH, Khurshid M. Respiratory Syncytial Virus: An Overview of Infection Biology and Vaccination Strategies. Future Virol 2017; 12:297-313. [DOI: doi.org/10.2217/fvl-2017-0120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/27/2017] [Indexed: 04/10/2025]
Affiliation(s)
- Muhammad Shafique
- Department of Microbiology Government College University Faisalabad
Pakistan
| | | | - Mohsin Khurshid
- Department of Microbiology Government College University Faisalabad
Pakistan
- College of Allied Health Professionals Directorate of Medical Sciences Government College University Faisalabad
Pakistan
| |
Collapse
|
25
|
Development of an intradermal DNA vaccine delivery strategy to achieve single-dose immunity against respiratory syncytial virus. Vaccine 2017; 35:2840-2847. [PMID: 28413132 DOI: 10.1016/j.vaccine.2017.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 12/22/2022]
Abstract
Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.
Collapse
|
26
|
CD4 + T cells support establishment of RSV-specific IgG and IgA antibody secreting cells in the upper and lower murine respiratory tract following RSV infection. Vaccine 2017; 35:2617-2621. [PMID: 28410812 DOI: 10.1016/j.vaccine.2017.03.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 02/28/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022]
Abstract
The RSV vaccine field suffered a major set-back when children were vaccinated with a formalin-inactivated RSV vaccine (FI-RSV). Unexpectedly, the vaccinated children fared worse than unvaccinated children when they were naturally infected with RSV. Mouse models were then developed that implicated the CD4+ T helper cell population as a contributor to adverse events. Today, the T cell is viewed with much caution in the RSV field, and its induction by vaccination is sometimes discouraged. Here we re-emphasize the beneficial role of the CD4+ T cell. Experiments were performed with RSV-infected nude mice that received CD4+ T cells by adoptive transfer. Data demonstrated that CD4+ T cells were necessary for the induction of mucosal and systemic RSV-specific antibodies, for the establishment of RSV-specific IgG and IgA antibody secreting cells in the upper and lower respiratory tract, and for RSV clearance.
Collapse
|
27
|
Human parainfluenza virus-3 can be targeted by rapidly ex vivo expanded T lymphocytes. Cytotherapy 2016; 18:1515-1524. [PMID: 27692559 DOI: 10.1016/j.jcyt.2016.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/05/2016] [Accepted: 08/30/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS Human parainfluenza virus-3 (HPIV) is a common cause of respiratory infection in immunocompromised patients and currently has no effective therapies. Virus-specific T-cell therapy has been successful for the treatment or prevention of viral infections in immunocompromised patients but requires determination of T-cell antigens on targeted viruses. METHODS HPIV3-specific T cells were expanded from peripheral blood of healthy donors using a rapid generation protocol targeting four HPIV3 proteins. Immunophenotyping was performed by flow cytometry. Viral specificity was determined by interferon (IFN)-γ ELISpot, intracellular cytokine staining and cytokine measurements from culture supernatants by Luminex assay. Cytotoxic activity was tested by 51Cr release and CD107a mobilization assays. Virus-specific T cells targeting six viruses were then produced by rapid protocol, and the phenotype of HPIV3-specific T cells was determined by immunomagnetic sorting for IFN-γ-producing cells. RESULTS HPIV3-specific T cells were expanded from 13 healthy donors. HPIV3-specific T cells showed a CD4+ predominance (mean CD4:CD8 ratio 2.89) and demonstrated specificity for multiple HPIV3 antigens. The expanded T cells were polyfunctional based on cytokine production but only had a minor cytotoxic component. T cells targeting six viruses in a single product similarly showed HPIV3 specificity, with a predominant effector memory phenotype (CD3+/CD45RA-/CCR7-) in responder cells. DISCUSSION HPIV3-specific T cells can be produced using a rapid ex vivo protocol from healthy donors and are predominantly CD4+ T cells with Th1 activity. HPIV3 epitopes can also be successfully targeted alongside multiple other viral epitopes in production of six-virus T cells, without loss of HPIV3 specificity. These products may be clinically beneficial to combat HPIV3 infections by adoptive T-cell therapy in immune-compromised patients.
Collapse
|
28
|
Bohmwald K, Espinoza JA, Rey-Jurado E, Gómez RS, González PA, Bueno SM, Riedel CA, Kalergis AM. Human Respiratory Syncytial Virus: Infection and Pathology. Semin Respir Crit Care Med 2016; 37:522-37. [PMID: 27486734 PMCID: PMC7171722 DOI: 10.1055/s-0036-1584799] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human respiratory syncytial virus (hRSV) is by far the major cause of acute lower respiratory tract infections (ALRTIs) worldwide in infants and children younger than 2 years. The overwhelming number of hospitalizations due to hRSV-induced ALRTI each year is due, at least in part, to the lack of licensed vaccines against this virus. Thus, hRSV infection is considered a major public health problem and economic burden in most countries. The lung pathology developed in hRSV-infected individuals is characterized by an exacerbated proinflammatory and unbalanced Th2-type immune response. In addition to the adverse effects in airway tissues, hRSV infection can also cause neurologic manifestations in the host, such as seizures and encephalopathy. Although the origins of these extrapulmonary symptoms remain unclear, studies with patients suffering from neurological alterations suggest an involvement of the inflammatory response against hRSV. Furthermore, hRSV has evolved numerous mechanisms to modulate and evade the immune response in the host. Several studies have focused on elucidating the interactions between hRSV virulence factors and the host immune system, to rationally design new vaccines and therapies against this virus. Here, we discuss about the infection, pathology, and immune response triggered by hRSV in the host.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janyra A Espinoza
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Emma Rey-Jurado
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto S Gómez
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas y Facultad de Medicina, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
29
|
Ljungman P, Snydman D, Boeckh M. Respiratory Syncytial Virus and Human Metapneumovirus Infection in Transplant Recipients. TRANSPLANT INFECTIONS 2016. [PMCID: PMC7123147 DOI: 10.1007/978-3-319-28797-3_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Respiratory viral infections due to respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause infections in immunocompromised transplant patients ranging from mild upper respiratory infections to severe lower respiratory tract disease with respiratory failure. These viruses are more readily diagnosed due to improvements in sensitive molecular diagnostic methods. The epidemiology of RSV and hMPV is similarly becoming more readily appreciated in hematopoietic stem cell transplant (HSCT) patients of all ages as well as solid organ transplant (SOT) patients, with lung transplant recipients having evidence of more frequent and severe complications related to these viruses. RSV and hMPV infection typically but not always present with upper respiratory signs and symptoms that progress to lower respiratory tract disease. Treatment options for RSV are limited, with aerosolized, intravenous, and oral ribavirin all studied in HSCT and lung transplant patients. No antiviral therapy for the treatment of hMPV is available, although ribavirin has shown some effectiveness in vitro. New antiviral agents including RSV fusion inhibitors and nucleoside analogs are being developed, with some under clinical evaluation.
Collapse
Affiliation(s)
- Per Ljungman
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Snydman
- Tufts University School of Medicine Tufts Medical Center, Boston, Massachusetts USA
| | - Michael Boeckh
- University of Washington Fred Hutchinson Cancer Research Center, Seattle, Washington USA
| |
Collapse
|
30
|
Abstract
ABSTRACT Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection and hospitalization among infants. Despite the significant healthcare burden, there is no licensed RSV vaccine currently available. This problem is further exacerbated as a natural RSV infection fails to elicit the development of long-lived immunity. It is well established that RSV-specific antibodies play a critical role in mediating protection from severe disease. The CD8 T-cell response is critical for mediating virus clearance following an acute RSV infection. However, the relative contribution of memory CD8 T cells in providing protection against secondary RSV infections remains unclear. In addition, data from animal models indicate that memory CD8 T-cell responses can be pathogenic under certain conditions. Herein, we provide an overview of the CD8 T-cell response elicited by RSV infection and how our current knowledge may impact future studies and vaccine development.
Collapse
Affiliation(s)
- Cory J Knudson
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
31
|
|
32
|
Pierantoni A, Esposito ML, Ammendola V, Napolitano F, Grazioli F, Abbate A, del Sorbo M, Siani L, D’Alise AM, Taglioni A, Perretta G, Siccardi A, Soprana E, Panigada M, Thom M, Scarselli E, Folgori A, Colloca S, Taylor G, Cortese R, Nicosia A, Capone S, Vitelli A. Mucosal delivery of a vectored RSV vaccine is safe and elicits protective immunity in rodents and nonhuman primates. Mol Ther Methods Clin Dev 2015; 2:15018. [PMID: 26015988 PMCID: PMC4441047 DOI: 10.1038/mtm.2015.18] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/20/2015] [Indexed: 01/27/2023]
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of severe respiratory disease in infants and the elderly. No vaccine is presently available to address this major unmet medical need. We generated a new genetic vaccine based on chimpanzee Adenovirus (PanAd3-RSV) and Modified Vaccinia Ankara RSV (MVA-RSV) encoding the F, N, and M2-1 proteins of RSV, for the induction of neutralizing antibodies and broad cellular immunity. Because RSV infection is restricted to the respiratory tract, we compared intranasal (IN) and intramuscular (M) administration for safety, immunogenicity, and efficacy in different species. A single IN or IM vaccination completely protected BALB/c mice and cotton rats against RSV replication in the lungs. However, only IN administration could prevent infection in the upper respiratory tract. IM vaccination with MVA-RSV also protected cotton rats from lower respiratory tract infection in the absence of detectable neutralizing antibodies. Heterologous prime boost with PanAd3-RSV and MVA-RSV elicited high neutralizing antibody titers and broad T-cell responses in nonhuman primates. In addition, animals primed in the nose developed mucosal IgA against the F protein. In conclusion, we have shown that our vectored RSV vaccine induces potent cellular and humoral responses in a primate model, providing strong support for clinical testing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alessandra Taglioni
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | - Gemma Perretta
- Cellular Biology and Neurobiology Institute (IBCN) National Research Council of Italy, Rome, Italy
| | | | | | | | | | | | | | | | | | - Riccardo Cortese
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- Keires AG, Basel, Switzerland
| | - Alfredo Nicosia
- ReiThera Srl, Rome, Italy (former Okairos Srl)
- CEINGE, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University Federico II, Naples, Italy
| | | | | |
Collapse
|
33
|
Chiu C, Openshaw PJ. Antiviral B cell and T cell immunity in the lungs. Nat Immunol 2015; 16:18-26. [PMID: 25521681 PMCID: PMC7097128 DOI: 10.1038/ni.3056] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
Abstract
Respiratory viruses are frequent causes of repeated common colds, bronchitis and pneumonia, which often occur unpredictably as epidemics and pandemics. Despite those decimating effects on health and decades of intensive research, treatments remain largely supportive. The only commonly available vaccines are against influenza virus, and even these need improvement. The lung shares some features with other mucosal sites, but preservation of its especially delicate anatomical structures necessitates a fine balance of pro- and anti-inflammatory responses; well-timed, appropriately placed and tightly regulated T cell and B cell responses are essential for protection from infection and limitation of symptoms, whereas poorly regulated inflammation contributes to tissue damage and disease. Recent advances in understanding adaptive immunity should facilitate vaccine development and reduce the global effect of respiratory viruses.
Collapse
Affiliation(s)
- Christopher Chiu
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter J Openshaw
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
34
|
Guzman E, Taylor G. Immunology of bovine respiratory syncytial virus in calves. Mol Immunol 2014; 66:48-56. [PMID: 25553595 DOI: 10.1016/j.molimm.2014.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/28/2014] [Accepted: 12/07/2014] [Indexed: 12/31/2022]
Abstract
Bovine respiratory syncytial virus (BRSV) is an important cause of respiratory disease in young calves. The virus is genetically and antigenically closely related to human (H)RSV, which is a major cause of respiratory disease in young infants. As a natural pathogen of calves, BRSV infection recapitulates the pathogenesis of respiratory disease in man more faithfully than semi-permissive, animal models of HRSV infection. With the increasing availability of immunological reagents, the calf can be used to dissect the pathogenesis of and mechanisms of immunity to RSV infection, to analyse the ways in which the virus proteins interact with components of the innate response, and to evaluate RSV vaccine strategies. Passively transferred, neutralising bovine monoclonal antibodies, which recognise the same epitopes in the HRSV and BRSV fusion (F) protein, can protect calves against BRSV infection, and depletion of different T cells subsets in calves has highlighted the importance of CD8(+) T cells in viral clearance. Calves can be used to model maternal-antibody mediated suppression of RSV vaccine efficacy, and to increase understanding of the mechanisms responsible for RSV vaccine-enhanced respiratory disease.
Collapse
Affiliation(s)
- Efrain Guzman
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| |
Collapse
|
35
|
Kurzweil V, Tang R, Galinski M, Wang K, Zuo F, Cherukuri A, Gasser RA, Malkin E, Sifakis F, Mendel DB, Esser MT. Translational sciences approach to RSV vaccine development. Expert Rev Vaccines 2014; 12:1047-60. [PMID: 24053398 DOI: 10.1586/14760584.2013.824706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in infants and the elderly. Despite its relatively low degree of antigenic variation, it causes frequent reinfection throughout life. Clinical manifestations of RSV disease and the immune response to infection differ in infants and the elderly, suggesting that vaccines designed to protect these two populations may require different attributes. Here, the authors describe the translational approach of utilizing data from epidemiology studies performed in these populations, the use of RSV diagnostics in clinical practice, lessons learned from previous vaccine clinical trials and the success of palivizumab in prevention of RSV disease in premature and high-risk infants to aid the development of safe and effective RSV vaccines.
Collapse
Affiliation(s)
- Vanessa Kurzweil
- Cell and Molecular Biology Group, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Cytotoxic T cells (CTL) play a critical role in the clearance of respiratory viral infections, but they also contribute to disease manifestations. In this study, we infected mice with a genetically modified pneumonia virus of mice (PVM) that allowed visualization of virus-specific CTL and infected cells in situ. The first virus-specific T cells entered the lung via blood vessels in the scattered foci of PVM-infected cells, which densely clustered around the bronchi at day 7 after infection. At this time, overall pulmonary virus load was maximal, but the mice showed no overt signs of disease. On days 8 to 9, T cells gained access to the infected bronchial epithelium and to the lung interstitium, which was associated with a reduction in the number of virus-infected cells within the initial clusters but could not prevent further virus spread throughout the lung tissue. Interestingly, recruitment of virus-specific CTL throughout the parenchyma was still ongoing on day 10, when the virus infection was already largely controlled. This also represented the peak of clinical disease. Thus, disease was associated with an exuberant T cell infiltration late in the course of the infection, which may be required to completely eliminate virus at residual foci of infection. PVM-induced immunopathology may thus result from the need to generate widespread T cell infiltrates to complete the elimination of virus-infected cells in a large organ like the lung. This experimental model provides the first insights into the spatiotemporal evolution of pulmonary antiviral T cell immunity in vivo.
Collapse
|
37
|
Yoo JK, Kim TS, Hufford MM, Braciale TJ. Viral infection of the lung: host response and sequelae. J Allergy Clin Immunol 2013; 132:1263-76; quiz 1277. [PMID: 23915713 PMCID: PMC3844062 DOI: 10.1016/j.jaci.2013.06.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/21/2013] [Accepted: 06/05/2013] [Indexed: 02/06/2023]
Abstract
Because of its essential role in gas exchange and oxygen delivery, the lung has evolved a variety of strategies to control inflammation and maintain homeostasis. Invasion of the lung by pathogens (and in some instances exposure to certain noninfectious particulates) disrupts this equilibrium and triggers a cascade of events aimed at preventing or limiting colonization (and more importantly infection) by pathogenic microorganisms. In this review we focus on viral infection of the lung and summarize recent advances in our understanding of the triggering of innate and adaptive immune responses to viral respiratory tract infection, mechanisms of viral clearance, and the well-recognized consequences of acute viral infection complicating underlying lung diseases, such as asthma.
Collapse
Affiliation(s)
- Jae-Kwang Yoo
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
| | - Taeg S. Kim
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Pathology and Molecular Medicine, University of Virginia, Charlottesville, Va
| | - Matthew M. Hufford
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Va
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Va
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Va
- Department of Pathology and Molecular Medicine, University of Virginia, Charlottesville, Va
- Corresponding author: Thomas J. Braciale, MD, PhD, Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908.
| |
Collapse
|
38
|
Eyles JE, Johnson JE, Megati S, Roopchand V, Cockle PJ, Weeratna R, Makinen S, Brown TP, Lang S, Witko SE, Kotash CS, Li J, West K, Maldonado O, Falconer DJ, Lees C, Smith GJ, White P, Wright P, Loudon PT, Merson JR, Jansen KU, Sidhu MK. Nonreplicating vaccines can protect african green monkeys from the memphis 37 strain of respiratory syncytial virus. J Infect Dis 2013; 208:319-29. [PMID: 23596321 DOI: 10.1093/infdis/jit169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We evaluated the immunological responses of African green monkeys immunized with multiple F and G protein-based vaccines and assessed protection against the Memphis 37 strain of respiratory syncytial virus (RSV). METHODS Monkeys were immunized with F and G proteins adjuvanted with immunostimulatory (CpG) oligodeoxyribonucleotides admixed with either Alhydrogel or ISCOMATRIX adjuvant. Delivery of F and G proteins via replication incompetent recombinant vesicular stomatitis viruses (VSVs) and human adenoviruses was also evaluated. Mucosally or parenterally administered recombinant adenoviruses were used in prime-boost regimens with adjuvanted proteins or recombinant DNA. RESULTS Animals primed by intranasal delivery of recombinant adenoviruses, and boosted by intramuscular injection of adjuvanted F and G proteins, developed neutralizing antibodies and F/G protein-specific T cells and were protected from RSV infection. Intramuscular injections of Alhydrogel (plus CpG) adjuvanted F and G proteins reduced peak viral loads in the lungs of challenged monkeys. Granulocyte numbers were not significantly elevated, relative to controls, in postchallenge bronchoalveolar lavage samples from vaccinated animals. CONCLUSIONS This study has validated the use of RSV (Memphis 37) in an African green monkey model of intranasal infection and identified nonreplicating vaccines capable of eliciting protection in this higher species challenge model.
Collapse
Affiliation(s)
- Jim E Eyles
- Pfizer Vaccine Research, La Jolla, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kaaijk P, Luytjes W, Rots NY. Vaccination against RSV: is maternal vaccination a good alternative to other approaches? Hum Vaccin Immunother 2013; 9:1263-7. [PMID: 23442726 PMCID: PMC3901815 DOI: 10.4161/hv.24096] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The respiratory syncytial virus (RSV) is the major cause of lower respiratory tract illness (LRI) in infants worldwide. Also persons with heart/lung disease or an immunodeficiency disorder, and the elderly are at increased risk for severe LRI upon RSV infection. Although there is at present no licensed RSV vaccine available, it is a priority target for several vaccine developers. For the implementation of a future RSV vaccination within national immunization schemes, various strategies can be considered even without the availability of extended clinical data on RSV vaccines. For this purpose, the extensive knowledge on RSV with respect to disease pathology, epidemiology and immunology can be used. This article discusses different aspects that should be considered to enable a successful implementation of a new RSV vaccine in national immunization programs. In addition, gaps in knowledge that needs further attention are identified. The maternal immunization strategy is highlighted, but also vaccination in the youngest infants and specific risk group immunization strategies are evaluated in this paper. Key factors such as the seasonality of RSV disease, interference of maternal antibodies and the immaturity of the infants’ immune system are addressed.
Collapse
Affiliation(s)
- Patricia Kaaijk
- Centre for infectious Diseases Control; National Institute for Public Health and the Environment (RIVM); Bilthoven, the Netherlands
| | | | | |
Collapse
|
40
|
Rudraraju R, Jones BG, Sealy R, Surman SL, Hurwitz JL. Respiratory syncytial virus: current progress in vaccine development. Viruses 2013; 5:577-94. [PMID: 23385470 PMCID: PMC3640515 DOI: 10.3390/v5020577] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 12/18/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the etiological agent for a serious lower respiratory tract disease responsible for close to 200,000 annual deaths worldwide. The first infection is generally most severe, while re-infections usually associate with a milder disease. This observation and the finding that re-infection risks are inversely associated with neutralizing antibody titers suggest that immune responses generated toward a first RSV exposure can significantly reduce morbidity and mortality throughout life. For more than half a century, researchers have endeavored to design a vaccine for RSV that can mimic or improve upon natural protective immunity without adverse events. The virus is herein described together with the hurdles that must be overcome to develop a vaccine and some current vaccine development approaches.
Collapse
Affiliation(s)
- Rajeev Rudraraju
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; E-Mails: (R.R.); (B.J.); (R.S.); (S.S.)
| | - Bart G. Jones
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; E-Mails: (R.R.); (B.J.); (R.S.); (S.S.)
| | - Robert Sealy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; E-Mails: (R.R.); (B.J.); (R.S.); (S.S.)
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; E-Mails: (R.R.); (B.J.); (R.S.); (S.S.)
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; E-Mails: (R.R.); (B.J.); (R.S.); (S.S.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, 858 Madison Avenue, Memphis, TN 38163, USA
| |
Collapse
|
41
|
|
42
|
Varga SM, Braciale TJ. The adaptive immune response to respiratory syncytial virus. Curr Top Microbiol Immunol 2013; 372:155-71. [PMID: 24362689 DOI: 10.1007/978-3-642-38919-1_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Respiratory syncytial virus (RSV) causes severe respiratory disease in children, the elderly and immunocompromised individuals. The combined actions of CD4 and CD8 T cells play a critical role in terminating an acute RSV infection whereas antibodies can provide protection from re-infection. Despite eliciting an immune response that mediates clearance of the virus, immunity to the virus appears to wane over time and individuals remain susceptible to reinfection with RSV throughout their lifetime. The ineffectiveness of the natural infection to induce long-term immunity has hampered vaccine efforts and there is currently no licensed RSV vaccine. In this review, we summarize our current understanding of the adaptive immune response to RSV and its contribution to disease.
Collapse
Affiliation(s)
- Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, 52242, USA,
| | | |
Collapse
|
43
|
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract viral disease in infants and young children. Presently, there are no explicit recommendations for RSV treatment apart from supportive care. The virus is therefore responsible for an estimated 160,000 deaths per year worldwide. Despite half a century of dedicated research, there remains no licensed vaccine product. Herein are described past and current efforts to harness innate and adaptive immune potentials to combat RSV. A plethora of candidate vaccine products and strategies are reviewed. The development of a successful RSV vaccine may ultimately stem from attention to historical lessons, in concert with an integral partnering of immunology and virology research fields.
Collapse
Affiliation(s)
- Julia L Hurwitz
- Department of Infectious Diseases, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
44
|
Collins PL, Melero JA. Progress in understanding and controlling respiratory syncytial virus: still crazy after all these years. Virus Res 2011; 162:80-99. [PMID: 21963675 PMCID: PMC3221877 DOI: 10.1016/j.virusres.2011.09.020] [Citation(s) in RCA: 338] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Revised: 09/13/2011] [Accepted: 09/14/2011] [Indexed: 01/25/2023]
Abstract
Human respiratory syncytial virus (RSV) is a ubiquitous pathogen that infects everyone worldwide early in life and is a leading cause of severe lower respiratory tract disease in the pediatric population as well as in the elderly and in profoundly immunosuppressed individuals. RSV is an enveloped, nonsegmented negative-sense RNA virus that is classified in Family Paramyxoviridae and is one of its more complex members. Although the replicative cycle of RSV follows the general pattern of the Paramyxoviridae, it encodes additional proteins. Two of these (NS1 and NS2) inhibit the host type I and type III interferon (IFN) responses, among other functions, and another gene encodes two novel RNA synthesis factors (M2-1 and M2-2). The attachment (G) glycoprotein also exhibits unusual features, such as high sequence variability, extensive glycosylation, cytokine mimicry, and a shed form that helps the virus evade neutralizing antibodies. RSV is notable for being able to efficiently infect early in life, with the peak of hospitalization at 2-3 months of age. It also is notable for the ability to reinfect symptomatically throughout life without need for significant antigenic change, although immunity from prior infection reduces disease. It is widely thought that re-infection is due to an ability of RSV to inhibit or subvert the host immune response. Mechanisms of viral pathogenesis remain controversial. RSV is notable for a historic, tragic pediatric vaccine failure involving a formalin-inactivated virus preparation that was evaluated in the 1960s and that was poorly protective and paradoxically primed for enhanced RSV disease. RSV also is notable for the development of a successful strategy for passive immunoprophylaxis of high-risk infants using RSV-neutralizing antibodies. Vaccines and new antiviral drugs are in pre-clinical and clinical development, but controlling RSV remains a formidable challenge.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antiviral Agents/administration & dosage
- Child
- Communicable Disease Control/organization & administration
- Cytokines/immunology
- Humans
- Immunity, Innate
- Infant
- RNA, Viral/genetics
- RNA, Viral/immunology
- Respiratory Syncytial Virus Infections/drug therapy
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Infections/virology
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Respiratory Syncytial Virus, Human/genetics
- Respiratory Syncytial Virus, Human/immunology
- Vaccination
- Vaccines, Attenuated/administration & dosage
- Viral Proteins/chemistry
- Viral Proteins/genetics
- Viral Proteins/immunology
- Virus Replication/genetics
- Virus Replication/immunology
Collapse
Affiliation(s)
- Peter L. Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - José A. Melero
- Centro Nacional de Microbiología and CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| |
Collapse
|
45
|
Halfhide CP, Flanagan BF, Brearey SP, Hunt JA, Fonceca AM, McNamara PS, Howarth D, Edwards S, Smyth RL. Respiratory syncytial virus binds and undergoes transcription in neutrophils from the blood and airways of infants with severe bronchiolitis. J Infect Dis 2011; 204:451-8. [PMID: 21742845 PMCID: PMC3132143 DOI: 10.1093/infdis/jir280] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 03/21/2011] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neutrophils are the predominant cell in the lung inflammatory infiltrate of infants with respiratory syncytial virus (RSV) bronchiolitis. Although it has previously been shown that neutrophils from both blood and bronchoalveolar lavage (BAL) are activated, little is understood about their role in response to RSV infection. This study investigated whether RSV proteins and mRNA are present in neutrophils from blood and BAL of infected infants. METHODS We obtained blood and BAL samples from 20 infants with severe RSV bronchiolitis and 8 healthy control infants. Neutrophil RSV F, G, and N proteins, RSV N genomic RNA, and messenger RNA (mRNA) were quantified. RESULTS RSV proteins were found in BAL and blood neutrophils in infants with RSV disease but not in neutrophils from healthy infants. BAL and blood neutrophils from infants with RSV disease, but not those from healthy infants, expressed RSV N genomic RNA, indicating uptake of whole virus; 17 of 20 BAL and 8 of 9 blood neutrophils from patients expressed RSV N mRNA. CONCLUSIONS This work shows, for the first time, the presence of RSV proteins and mRNA transcripts within BAL and blood neutrophils from infants with severe RSV bronchiolitis.
Collapse
Affiliation(s)
- Clare P. Halfhide
- Division of Child Health, Department of Women’s and Children’s Health, Royal Liverpool, Alder Hey Children’s Hospital
| | | | - Stephen P. Brearey
- Division of Child Health, Department of Women’s and Children’s Health, Royal Liverpool, Alder Hey Children’s Hospital
| | | | - Angela M. Fonceca
- Division of Child Health, Department of Women’s and Children’s Health, Royal Liverpool, Alder Hey Children’s Hospital
| | - Paul S. McNamara
- Division of Child Health, Department of Women’s and Children’s Health, Royal Liverpool, Alder Hey Children’s Hospital
| | - Deborah Howarth
- Department of Clinical Infection, Microbiology and Immunology
| | - Steven Edwards
- Department of Biochemistry and Cell Biology, University of Liverpool, UK
| | - Rosalind L. Smyth
- Division of Child Health, Department of Women’s and Children’s Health, Royal Liverpool, Alder Hey Children’s Hospital
| |
Collapse
|
46
|
Moraes TJ, Lin GH, Wen T, Watts TH. Incorporation of 4-1BB ligand into an adenovirus vaccine vector increases the number of functional antigen-specific CD8 T cells and enhances the duration of protection against influenza-induced respiratory disease. Vaccine 2011; 29:6301-12. [DOI: 10.1016/j.vaccine.2011.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 04/07/2011] [Accepted: 06/04/2011] [Indexed: 11/30/2022]
|
47
|
Major histocompatibility complex-dependent cytotoxic T lymphocyte repertoire and functional avidity contribute to strain-specific disease susceptibility after murine respiratory syncytial virus infection. J Virol 2011; 85:10135-43. [PMID: 21795345 DOI: 10.1128/jvi.00816-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Susceptibility to respiratory syncytial virus (RSV) infection in mice is genetically determined. While RSV causes little pathology in C57BL/6 mice, pulmonary inflammation and weight loss occur in BALB/c mice. Using major histocompatibility complex (MHC)-congenic mice, we observed that the H-2(d) allele can partially transfer disease susceptibility to C57BL/6 mice. This was not explained by altered viral elimination or differences in the magnitude of the overall virus-specific cytotoxic T lymphocyte (CTL) response. However, H-2(d) mice showed a more focused response, with 70% of virus-specific CTL representing Vβ8.2(+) CTL directed against the immunodominant epitope M2-1 82, while in H-2(b) mice only 20% of antiviral CTL were Vβ9(+) CTL specific for the immunodominant epitope M187. The immunodominant H-2(d)-restricted CTL lysed target cells less efficiently than the immunodominant H-2(b) CTL, probably contributing to prolonged CTL stimulation and cytokine-mediated immunopathology. Accordingly, reduction of dominance of the M2-1 82-specific CTL population by introduction of an M187 response in the F1 generation of a C57BL/6N × C57BL/6-H-2(d) mating (C57BL/6-H-2(dxb) mice) attenuated disease. Moreover, disease in H-2(d) mice was less pronounced after infection with an RSV mutant failing to activate M2-1 82-specific CTL or after depletion of Vβ8.2(+) cells. These data illustrate how the MHC-determined diversity and functional avidity of CTL responses contribute to disease susceptibility after viral infection.
Collapse
|
48
|
Munir S, Hillyer P, Le Nouën C, Buchholz UJ, Rabin RL, Collins PL, Bukreyev A. Respiratory syncytial virus interferon antagonist NS1 protein suppresses and skews the human T lymphocyte response. PLoS Pathog 2011; 7:e1001336. [PMID: 21533073 PMCID: PMC3080852 DOI: 10.1371/journal.ppat.1001336] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 03/23/2011] [Indexed: 12/17/2022] Open
Abstract
We recently demonstrated that the respiratory syncytial virus (RSV) NS1 protein, an antagonist of host type I interferon (IFN-I) production and signaling, has a suppressive effect on the maturation of human dendritic cells (DC) that was only partly dependent on released IFN-I. Here we investigated whether NS1 affects the ability of DC to activate CD8+ and CD4+ T cells. Human DC were infected with RSV deletion mutants lacking the NS1 and/or NS2 genes and assayed for the ability to activate autologous T cells in vitro, which were analyzed by multi-color flow cytometry. Deletion of the NS1, but not NS2, protein resulted in three major effects: (i) an increased activation and proliferation of CD8+ T cells that express CD103, a tissue homing integrin that directs CD8+ T cells to mucosal epithelial cells of the respiratory tract and triggers cytolytic activity; (ii) an increased activation and proliferation of Th17 cells, which have recently been shown to have anti-viral effects and also indirectly attract neutrophils; and (iii) decreased activation of IL-4-producing CD4+ T cells--which are associated with enhanced RSV disease--and reduced proliferation of total CD4+ T cells. Except for total CD4+ T cell proliferation, none of the T cell effects appeared to be due to increased IFN-I signaling. In the infected DC, deletion of the NS1 and NS2 genes strongly up-regulated the expression of cytokines and other molecules involved in DC maturation. This was partly IFN-I-independent, and thus might account for the T cell effects. Taken together, these data demonstrate that the NS1 protein suppresses proliferation and activation of two of the protective cell populations (CD103+ CD8+ T cells and Th17 cells), and promotes proliferation and activation of Th2 cells that can enhance RSV disease.
Collapse
Affiliation(s)
- Shirin Munir
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Philippa Hillyer
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Cyril Le Nouën
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ronald L. Rabin
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Peter L. Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexander Bukreyev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
49
|
|
50
|
Esposito S, Salice P, Bosis S, Ghiglia S, Tremolati E, Tagliabue C, Gualtieri L, Barbier P, Galeone C, Marchisio P, Principi N. Altered cardiac rhythm in infants with bronchiolitis and respiratory syncytial virus infection. BMC Infect Dis 2010; 10:305. [PMID: 20969802 PMCID: PMC2987303 DOI: 10.1186/1471-2334-10-305] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/24/2010] [Indexed: 11/17/2022] Open
Abstract
Background Although the most frequent extra-pulmonary manifestations of respiratory syncytial virus (RSV) infection involve the cardiovascular system, no data regarding heart function in infants with bronchiolitis associated with RSV infection have yet been systematically collected. The aim of this study was to verify the real frequency of heart involvement in patients with bronchiolitis associated with RSV infection, and whether infants with mild or moderate disease also risk heart malfunction. Methods A total of 69 otherwise healthy infants aged 1-12 months with bronchiolitis hospitalised in standard wards were enrolled. Pernasal flocked swabs were performed to collect specimens for the detection of RSV by real-time polymerase chain reaction, and a blood sample was drawn to assess troponin I concentrations. On the day of admission, all of the infants underwent 24-hour Holter ECG monitoring and a complete heart evaluation with echocardiography. Patients were re-evaluated by investigators blinded to the etiological and cardiac findings four weeks after enrolment. Results Regardless of their clinical presentation, sinoatrial blocks were identified in 26/34 RSV-positive patients (76.5%) and 1/35 RSV-negative patients (2.9%) (p < 0.0001). The blocks recurred more than three times over 24 hours in 25/26 RSV-positive patients (96.2%) and none of the RSV-negative infants. Mean and maximum heart rates were significantly higher in the RSV-positive infants (p < 0.05), as was low-frequency power and the low and high-frequency power ratio (p < 0.05). The blocks were significantly more frequent in the children with an RSV load of ≥100,000 copies/mL than in those with a lower viral load (p < 0.0001). Holter ECG after 28 ± 3 days showed the complete regression of the heart abnormalities. Conclusions RSV seems associated with sinoatrial blocks and transient rhythm alterations even when the related respiratory problems are mild or moderate. Further studies are needed to clarify the mechanisms of these rhythm problems and whether they remain asymptomatic and transient even in presence of severe respiratory involvement or chronic underlying disease.
Collapse
Affiliation(s)
- Susanna Esposito
- Department of Maternal and Pediatric Sciences, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|