1
|
Juarez MG, O'Rourke SM, Dzimianski JV, Gagnon D, Penunuri G, Serrão VHB, Corbett-Detig RB, Kauvar LM, DuBois RM. Structures of respiratory syncytial virus G bound to broadly reactive antibodies provide insights into vaccine design. Sci Rep 2025; 15:8666. [PMID: 40082629 PMCID: PMC11906780 DOI: 10.1038/s41598-025-92886-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in infants and older adults. The attachment glycoprotein (RSV G) binds to the chemokine receptor CX3CR1 to promote viral entry and modulate host immunity. Antibodies against RSV G are a known correlate of protection. Previously, several broadly reactive, high-affinity anti-RSV G human monoclonal antibodies were isolated from RSV-exposed individuals and were shown to be protective in vitro and in vivo. Here, we determined the structures of three of these antibodies in complex with RSV G and defined distinct conformational epitopes comprised of highly conserved RSV G residues. Binding competition and structural studies demonstrated that this highly conserved region displays two non-overlapping antigenic sites. Analyses of anti-RSV G antibody sequences reveal that antigenic site flexibility may promote the elicitation of diverse antibody germlines. Together, these findings provide a foundation for next-generation RSV prophylactics, and they expand concepts in vaccine design for the elicitation of germline lineage-diverse, broadly reactive, high-affinity antibodies.
Collapse
Affiliation(s)
- Maria G Juarez
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Sara M O'Rourke
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - John V Dzimianski
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Delia Gagnon
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Gabriel Penunuri
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Vitor H B Serrão
- Department of Chemistry & Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
- Biomolecular Cryo-Electron Microscopy Facility, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Russell B Corbett-Detig
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | | | - Rebecca M DuBois
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
2
|
Li S, Mei C, Chen S, Wang C, Gao Y, Ma J, Zhong L, Luo T, Zhao X, Bu H, Lyu Y, Kuang X, Jia Z, Wang X, Wang Y, Tian D. Protective or limited? Maternal antibodies and RSV-associated lower respiratory tract infection in hospitalized infants aged 28-90 days. Front Immunol 2025; 15:1437616. [PMID: 39845974 PMCID: PMC11753235 DOI: 10.3389/fimmu.2024.1437616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025] Open
Abstract
Background Respiratory syncytial virus (RSV) is a major cause of severe health problems in newborns and young children. The protective role and limitations of serum maternal RSV antibodies in infants under 3 months remain controversial. Methods A two-center prospective study from 2020 to 2023 recruited infants (n=286) admitted to the respiratory departments of two children's hospitals in southwestern and southeastern China during RSV epidemic. These infants were hospitalized with lower respiratory tract infections (LRTI). We evaluated the relationship between serum RSV Prefusion (Pre-F), postfusion (Post-F) IgG levels, subtype neutralizing antibodies, and the incidence of RSV infection, as well as the relationship between these maternal antibodies and severity of disease. Since this prospective study only included data from RSV epidemic, we retrospectively reviewed medical records from the Children's Hospital of Chongqing Medical University for the years 2019 to 2023 (n=3467) to analyze population characteristics during both RSV epidemic and non-epidemic periods, using the same inclusion and exclusion criteria. Result There were no significant differences in RSV Pre-F IgG, Post-F IgG, or RSV A or B neutralizing antibody levels between the RSV infected and non-infected groups during the epidemic. While RSV Pre-F IgG antibody was inversely correlated with disease severity, RSV Post-F IgG, and RSV A and B neutralizing antibodies did not show a similar correlation across the three illness severity categories. Additionally, there were no differences in age, gender, or illness severity distribution among hospitalized patients during epidemic and non-epidemic periods. Conclusion Serum maternal antibody levels offer insufficient protection against RSV-associated LRTI in hospitalized infants aged 28 to 90 days.
Collapse
Affiliation(s)
- Shuanglian Li
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chenghao Mei
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Sainan Chen
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Soochow, China
| | - Chenglin Wang
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yelei Gao
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jinhua Ma
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhong
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tingting Luo
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Zhao
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Huaqin Bu
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Lyu
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohu Kuang
- Department of Respiratory Medicine, Yibin Hospital Affiliated to Children’s Hospital of Chongqing Medical University, Yibin, China
| | - Zhenxing Jia
- Department of Respiratory Medicine, Yibin Hospital Affiliated to Children’s Hospital of Chongqing Medical University, Yibin, China
| | - Xiaoli Wang
- Department of Respiratory Medicine, Yibin Hospital Affiliated to Children’s Hospital of Chongqing Medical University, Yibin, China
| | - Yuqing Wang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Soochow, China
| | - Daiyin Tian
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity. Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Respiratory Medicine, Yibin Hospital Affiliated to Children’s Hospital of Chongqing Medical University, Yibin, China
| |
Collapse
|
3
|
Anderson EJ, Tippett A, Begier E, Gibson T, Ess G, Patel V, Taylor M, Reese O, Salazar L, Jadhao S, Sun HY, Hsiao HM, Gupta S, Li W, Stephens K, Keane A, Ciric C, Hellmeister K, Cheng A, Al-Husein Z, Bristow L, Hubler R, Liu Q, Gessner BD, Jodar L, Swerdlow D, Kalina W, Uppal S, Kamidani S, Rouphael N, Anderson LJ, Rostad CA. Relative Contribution of Diagnostic Testing to the Diagnosis of Respiratory Syncytial Virus in Hospitalized Adults in the United States. J Infect Dis 2024; 230:1342-1351. [PMID: 38995029 PMCID: PMC11646620 DOI: 10.1093/infdis/jiae346] [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: 03/08/2024] [Revised: 06/13/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a leading cause of acute respiratory illness (ARI) in older adults. Optimizing diagnosis could improve understanding of RSV burden. METHODS We enrolled adults ≥50 years of age hospitalized with ARI and adults of any age hospitalized with congestive heart failure or chronic obstructive pulmonary disease exacerbations at 2 hospitals during 2 respiratory seasons (2018-2020). We collected nasopharyngeal (NP) and oropharyngeal (OP) swabs (n = 1558), acute and convalescent sera (n = 568), and expectorated sputum (n = 153) from participants, and recorded standard-of-care (SOC) NP results (n = 805). We measured RSV antibodies by 2 immunoassays and performed BioFire testing on respiratory specimens. RESULTS Of 1558 eligible participants, 92 (5.9%) tested positive for RSV by any diagnostic method. Combined NP/OP polymerase chain reaction (PCR) testing yielded 58 positives, while separate NP and OP testing identified 11 additional positives (18.9% increase). Compared to study NP/OP PCR alone, the addition of paired serology increased RSV detection by 42.9% (28 vs 40) among those with both specimen types, while the addition of SOC swab PCR increased RSV detection by 25.9% (47 vs 59). CONCLUSIONS The addition of paired serology testing, SOC swab results, and separate testing of NP and OP swabs improved RSV diagnostic yield in hospitalized adults.
Collapse
Affiliation(s)
- Evan J Anderson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ashley Tippett
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Elizabeth Begier
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Theda Gibson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Gabby Ess
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Vikash Patel
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Meg Taylor
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Olivia Reese
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Luis Salazar
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Samadhan Jadhao
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - He-Ying Sun
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Hui-Mien Hsiao
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Shadwal Gupta
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Wensheng Li
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Kathleen Stephens
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Amy Keane
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Caroline Ciric
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Kieffer Hellmeister
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Andrew Cheng
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zayna Al-Husein
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Laurel Bristow
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Robin Hubler
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Qing Liu
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Bradford D Gessner
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Luis Jodar
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - David Swerdlow
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Warren Kalina
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Sonal Uppal
- Vaccines Medical Development, Scientific and Clinical Affairs, Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Satoshi Kamidani
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Larry J Anderson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Christina A Rostad
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Riccò M, Abu-Raya B, Icardi G, Spoulou V, Greenberg D, Pecurariu OF, Hung IFN, Osterhaus A, Sambri V, Esposito S. Respiratory Syncytial Virus: A WAidid Consensus Document on New Preventive Options. Vaccines (Basel) 2024; 12:1317. [PMID: 39771979 PMCID: PMC11679680 DOI: 10.3390/vaccines12121317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Respiratory syncytial virus (RSV) is a leading cause of respiratory infections, particularly affecting young infants, older adults, and individuals with comorbidities. Methods: This document, developed as a consensus by an international group of experts affiliated with the World Association of Infectious Diseases and Immunological Disorders (WAidid), focuses on recent advancements in RSV prevention, highlighting the introduction of monoclonal antibodies (mAbs) and vaccines. Results: Historically, RSV treatment options were limited to supportive care and the monoclonal antibody palivizumab, which required multiple doses. Recent innovations have led to the development of long-acting mAbs, such as nirsevimab, which provide season-long protection with a single dose. Nirsevimab has shown high efficacy in preventing severe RSV-related lower respiratory tract infections (LRTIs) in infants, reducing hospitalizations and ICU admissions. Additionally, new vaccines, such as RSVpreF and RSVpreF3, target older adults and have demonstrated significant efficacy in preventing LRTIs in clinical trials. Maternal vaccination strategies also show promise in providing passive immunity to newborns, protecting them during the most vulnerable early months of life. This document further discusses the global burden of RSV, its economic impact, and the challenges of implementing these preventative strategies in different healthcare settings. Conclusions: The evidence supports the integration of both passive (mAbs) and active (vaccines) immunization approaches as effective tools to mitigate the public health impact of RSV. The combined use of these interventions could substantially reduce RSV-related morbidity and mortality across various age groups and populations, emphasizing the importance of widespread immunization efforts.
Collapse
Affiliation(s)
- Matteo Riccò
- Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy;
| | - Bahaa Abu-Raya
- Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre and the Nova Scotia Health Authority, Halifax, NS B3K 6R8, Canada;
- Departments of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Giancarlo Icardi
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece;
| | - David Greenberg
- Pediatric Infectious Diseases Unit, Soroka University Medical Center, Faculty of Health Sciences, Ben Gurion University, Beer Sheva 8410501, Israel;
| | - Oana Falup Pecurariu
- Children’s Clinical Hospital Brasov, 500063 Brasov, Romania;
- Faculty of Medicine Brasov, Transilvania University, 500019 Brasov, Romania
| | - Ivan Fan-Ngai Hung
- Division of Infectious Diseases, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR 999077, China;
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Vittorio Sambri
- Unit of Microbiology, The Greater Romagna Area Hub Laboratory, 47522 Cesena, Italy;
- Department Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
5
|
Bonnin FA, Talarico LB, Ferolla FM, Acosta PL, Phung E, Kumar A, Toledano A, Caratozzolo A, Neira P, Mascardi N, Satragno D, Contrini MM, Graham BS, Ruckwardt TJ, López EL. Antibody levels against respiratory syncytial virus fusion protein conformations and lack of association with life-threatening infection in previously healthy infants. Vaccine 2024; 42:126119. [PMID: 39003106 PMCID: PMC11401757 DOI: 10.1016/j.vaccine.2024.07.020] [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: 10/19/2023] [Revised: 04/13/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Humoral immune response against the pre-fusion (pre-F) conformation of respiratory syncytial virus (RSV) F protein has been proposed to play a protective role against infection. An RSV pre-F maternal vaccine has been recently approved in several countries to protect young infants against RSV. We aimed to assess serum IgG titers against the pre-F and post-F conformations of RSV F protein and their association with life-threatening RSV disease (LTD) in previously healthy infants. METHODS A prospective cohort study including hospitalized infants <12 months with a first RSV infection was conducted during 2017-2019. Patients with LTD required intensive care and mechanical respiratory assistance. RSV pre-F exclusive and post-F antibody responses were determined by post-F competition and non-competition immunoassays, respectively, and neutralizing activity was measured by plaque reduction neutralization test. RESULTS Fifty-eight patients were included; the median age was 3.5 months and 41 % were females. Fifteen patients developed LTD. RSV F-specific antibody titers positively correlated with neutralizing antibody titers in acute and convalescent phases but, importantly, they did not associate with LTD. Acute RSV pre-F exclusive and post-F IgG titers negatively correlated with patient age (P = 0.0007 and P < 0.0001), while a positive correlation was observed between the fold changes in RSV F-specific antibody titers between convalescent and acute phase and patient age (P = 0.0014 and P < 0.0001). Infants ≤2 months exhibited significantly lower fold-changes in RSV F-specific and neutralizing antibody titers between convalescence and acute phase than older infants. Additionally, acute RSV antibody titers showed no correlation with nasal RSV load and, furthermore, nasal viral load was not associated with the development of LTD. CONCLUSIONS This study highlights that protection against life-threatening RSV disease is not necessarily antibody-dependent. Further characterization of the immune response against RSV and its role in protection against severe disease is important for the development of the safest possible preventive strategies.
Collapse
Affiliation(s)
- Florencia A Bonnin
- Laboratory of Infectious Diseases and Molecular Biology, Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires 1428, Argentina
| | - Laura B Talarico
- Laboratory of Infectious Diseases and Molecular Biology, Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires 1425, Argentina.
| | - Fausto M Ferolla
- Department of Medicine, Pediatric Infectious Diseases Program, Hospital de Niños Dr. Ricardo Gutiérrez, Universidad de Buenos Aires, Buenos Aires 1425, Argentina
| | - Patricio L Acosta
- Laboratory of Infectious Diseases and Molecular Biology, Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires 1425, Argentina
| | - Emily Phung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Azad Kumar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Analía Toledano
- Laboratory of Infectious Diseases and Molecular Biology, Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina
| | - Ana Caratozzolo
- Laboratory of Infectious Diseases and Molecular Biology, Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina
| | - Pablo Neira
- Intensive Care Unit, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina
| | - Normando Mascardi
- Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina
| | - Daniela Satragno
- Department of Medicine, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1425, Argentina
| | - María M Contrini
- Department of Medicine, Pediatric Infectious Diseases Program, Hospital de Niños Dr. Ricardo Gutiérrez, Universidad de Buenos Aires, Buenos Aires 1425, Argentina
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eduardo L López
- Department of Medicine, Pediatric Infectious Diseases Program, Hospital de Niños Dr. Ricardo Gutiérrez, Universidad de Buenos Aires, Buenos Aires 1425, Argentina.
| |
Collapse
|
6
|
Wilkins D, Wählby Hamrén U, Chang Y, Clegg LE, Domachowske J, Englund JA, Muller WJ, Leach A, Kelly EJ, Villafana T. RSV Neutralizing Antibodies Following Nirsevimab and Palivizumab Dosing. Pediatrics 2024; 154:e2024067174. [PMID: 39350745 DOI: 10.1542/peds.2024-067174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Data describing respiratory syncytial virus (RSV) neutralizing antibody (nAb) levels for nirsevimab, a recently approved, extended half-life, anti-RSV fusion protein (F protein) monoclonal antibody, relative to the previous standard of care, palivizumab, have not been reported. METHODS MEDLEY was a randomized, palivizumab-controlled, phase 2/3 study of nirsevimab during 2 RSV seasons (season 1 and 2) in infants born preterm (≤35 weeks' gestational age; dosed season 1 only) or with congenital heart disease or chronic lung disease of prematurity (dosed seasons 1 and 2). Participants were randomly assigned to receive a single dose of nirsevimab followed by 4 monthly placebo doses, or 5 once-monthly doses of palivizumab. Anti-RSV F protein serology (ie, levels of prefusion [pre-F]/postfusion [post-F] conformation antibodies), nirsevimab and palivizumab concentrations, and RSV nAbs were measured in participant serum collected at baseline (pre-dose) and days 31, 151, and 361. RESULTS Serologic data were similar in seasons 1 and 2. Nirsevimab predominately conferred pre-F antibodies, whereas palivizumab conferred pre-F and post-F antibodies. Nirsevimab and palivizumab serum concentrations highly correlated with nAb levels in both seasons. In season 1, nAb levels in nirsevimab recipients were highest in day 31 samples and gradually declined but remained 17-fold above baseline at day 361. nAb levels in palivizumab recipients increased incrementally with monthly doses to day 151. nAb levels followed similar patterns in season 2. nAb levels were ∼10-fold higher with nirsevimab compared with palivizumab across both seasons. CONCLUSIONS Nirsevimab prophylaxis confers ∼10-fold higher and more sustained RSV nAb levels relative to palivizumab.
Collapse
Affiliation(s)
- Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Yue Chang
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | | | - Joseph Domachowske
- Department of Pediatrics, State University of New York Upstate Medical University, Syracuse, New York
| | - Janet A Englund
- Department of Pediatrics, Seattle Children's Hospital Research Institute, University of Washington, Seattle, Washington
| | - William J Muller
- Ann & Robert H Lurie Children's Hospital of Chicago and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amanda Leach
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
7
|
Tuffy KM, Ahani B, Domachowske JB, Furuno K, Ji H, Madhi SA, Mankad VS, Hamrén UW, Villafana T, Wang Y, Kelly EJ, Wilkins D. Molecular and phenotypic characteristics of respiratory syncytial virus isolates recovered from medically vulnerable children: An exploratory analysis of a phase 2/3 randomized, double-blind, palivizumab-controlled trial of nirsevimab (MEDLEY). Vaccine 2024; 42:126276. [PMID: 39241352 DOI: 10.1016/j.vaccine.2024.126276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/09/2024] [Accepted: 08/25/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Nirsevimab is an extended half-life monoclonal antibody (mAb) licensed for the prevention of respiratory syncytial virus (RSV)-associated lower respiratory tract disease in neonates, infants and medically vulnerable children. We characterized RSV isolates recovered from participants enrolled in MEDLEY: a randomized, palivizumab-controlled phase 2/3 trial of nirsevimab in infants born preterm and/or with congenital heart disease or chronic lung disease of prematurity. METHODS Participants were assessed in two RSV seasons (Season 1 and 2). Season 1 participants were randomized (2:1) to receive a single dose of nirsevimab (50 mg if weight <5 kg or 100 mg if weight ≥5 kg in Season 1; 200 mg in Season 2) followed by four monthly doses of placebo, or five once-monthly doses of palivizumab (15 mg/kg weight per dose). Season 2 participants continued nirsevimab and placebo (nirsevimab/nirsevimab) or were re-randomized (1:1) to switch to nirsevimab (palivizumab/nirsevimab) or continue palivizumab (palivizumab/palivizumab). Cases of RSV infection were identified by central testing of nasal swabs from participants seeking medical attention for respiratory illnesses. Nirsevimab and palivizumab binding site substitutions were assessed via microneutralization assay. RESULTS Twenty-five cases of confirmed RSV infection were observed during the trial and sequenced: 12 in nirsevimab recipients and 10 in palivizumab recipients during Season 1, and 1 case in each Season 2 group. Molecular sequencing of RSV A (n = 14) isolates detected no nirsevimab binding site substitutions, and 3 palivizumab neutralization-resistant substitutions (Lys272Met, Lys272Thr, Ser275Leu). The nirsevimab binding site Ile206Met:Gln209Arg and Ile206Met:Gln209Arg:Ser211Asn substitutions were the only anti-RSV mAb binding site substitutions detected among RSV B isolates (n = 11). Nirsevimab neutralized all nirsevimab and palivizumab binding site substitutions in RSV A and B isolates recovered from MEDLEY participants. CONCLUSION No binding site substitution detected during MEDLEY affected RSV susceptibility to nirsevimab neutralization.
Collapse
MESH Headings
- Humans
- Palivizumab/therapeutic use
- Palivizumab/administration & dosage
- Respiratory Syncytial Virus Infections/prevention & control
- Infant
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antiviral Agents/therapeutic use
- Antiviral Agents/administration & dosage
- Double-Blind Method
- Male
- Respiratory Syncytial Virus, Human/immunology
- Respiratory Syncytial Virus, Human/drug effects
- Respiratory Syncytial Virus, Human/genetics
- Female
- Infant, Newborn
- Antibodies, Viral/immunology
- Child, Preschool
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
Collapse
Affiliation(s)
- Kevin M Tuffy
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Bahar Ahani
- Bioinformatics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Kenji Furuno
- Department of General Pediatrics and Interdisciplinary Medicine, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Hong Ji
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vaishali S Mankad
- Clinical Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Durham, NC, USA
| | - Ulrika Wählby Hamrén
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Gothenburg, Sweden
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Yingyi Wang
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
8
|
Coindy EL, Efstathiou C, Talwar S, Moureau A, Vernhes C, Openshaw PJM, Thwaites RS. Antibody-mediated protection against respiratory syncytial virus in children. Eur Respir Rev 2024; 33:240106. [PMID: 39384305 PMCID: PMC11462297 DOI: 10.1183/16000617.0106-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/31/2024] [Indexed: 10/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a major global pathogen, causing lower respiratory tract disease in at-risk populations including young children. Antibodies form a crucial layer of protection from RSV disease, particularly in immunologically naïve infants. Such antibodies are derived from the mother via transplacental transfer and breast milk, but may be particularly low in high-risk infants such as those born preterm. Maternally derived antibodies can now be supplemented by the administration of anti-RSV monoclonal antibodies, while a rising wave of maternal and paediatric vaccine strategies are approaching. The implementation of these prophylactics may profoundly decrease the healthcare burden of RSV. In this article, we review the role of antibody-mediated immunity in protecting children from RSV. We focus on maternally derived antibodies as the main source of protection against RSV and study factors that influence the scale of this transfer. The role of passive and active prophylactic approaches in protecting infants against RSV are discussed and knowledge gaps in our understanding of antibody-mediated protection against RSV are identified.
Collapse
Affiliation(s)
- Emma L Coindy
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Shubha Talwar
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
9
|
Mazur NI, Caballero MT, Nunes MC. Severe respiratory syncytial virus infection in children: burden, management, and emerging therapies. Lancet 2024; 404:1143-1156. [PMID: 39265587 DOI: 10.1016/s0140-6736(24)01716-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024]
Abstract
The global burden of respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) in young children is high. The RSV prevention strategies approved in 2023 will be essential to lowering the global disease burden. In this Series paper, we describe clinical presentation, burden of disease, hospital management, emerging therapies, and targeted prevention focusing on developments and groundbreaking publications for RSV. We conducted a systematic search for literature published in the past 15 years and used a non-systematic approach to analyse the results, prioritising important papers and the most recent reviews per subtopic. Annually, 33 million episodes of RSV LRTI occur in children younger than 5 years, resulting in 3·6 million hospitalisations and 118 200 deaths. RSV LRTI is a clinical diagnosis but a clinical case definition and universal clinical tool to predict severe disease are non-existent. The advent of molecular point-of-care testing allows rapid and accurate confirmation of RSV infection and could reduce antibiotic use. There is no evidence-based treatment of RSV, only supportive care. Despite widespread use, evidence for high-flow nasal cannula (HFNC) therapy is insufficient and increased paediatric intensive care admissions and intubation indicate the need to remove HFNC therapy from standard care. RSV is now a vaccine-preventable disease in young children with a market-approved long-acting monoclonal antibody and a maternal vaccine targeting the RSV prefusion protein. To have a high impact on life-threatening RSV infection, infants at high risk, especially in low-income and middle-income countries, should be prioritised as an interim strategy towards universal immunisation. The implementation of RSV preventive strategies will clarify the full burden of RSV infection. Vaccine probe studies can address existing knowledge gaps including the effect of RSV prevention on transmission dynamics, antibiotic misuse, the respiratory microbiome composition, and long-term sequalae.
Collapse
Affiliation(s)
- Natalie I Mazur
- Department of Pediatrics, Wilhelmina Children's Hospital, Utrecht, Netherlands.
| | - Mauricio T Caballero
- Centro INFANT de Medicina Traslacional (CIMeT), Escuela de Bio y Nanotecnología, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Marta C Nunes
- Center of Excellence in Respiratory Pathogens, Hospices Civils de Lyon and Centre International de Recherche en Infectiologie, Équipe Santé Publique, Épidémiologie et Écologie Évolutive des Maladies Infectieuses, Inserm U1111, CNRS UMR5308, ENS de Lyon, Lyon, France; South African Medical Research Council, Vaccines & Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
10
|
Phijffer EW, de Bruin O, Ahmadizar F, Bont LJ, Van der Maas NA, Sturkenboom MC, Wildenbeest JG, Bloemenkamp KW. Respiratory syncytial virus vaccination during pregnancy for improving infant outcomes. Cochrane Database Syst Rev 2024; 5:CD015134. [PMID: 38695784 PMCID: PMC11064886 DOI: 10.1002/14651858.cd015134.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2024]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections (LRTIs) in infants. Maternal RSV vaccination is a preventive strategy of great interest, as it could have a substantial impact on infant RSV disease burden. In recent years, the clinical development of maternal RSV vaccines has advanced rapidly. OBJECTIVES To assess the efficacy and safety of maternal respiratory syncytial virus (RSV) vaccination for preventing RSV disease in infants. SEARCH METHODS We searched Cochrane Pregnancy and Childbirth's Trials Register and two other trials registries on 21 October 2022. We updated the search on 27 July 2023, when we searched MEDLINE, Embase, CENTRAL, CINAHL, and two trials registries. Additionally, we searched the reference lists of retrieved studies and conference proceedings. There were no language restrictions on our searches. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing maternal RSV vaccination with placebo or no intervention in pregnant women of any age. The primary outcomes were hospitalisation with clinically confirmed or laboratory-confirmed RSV disease in infants. The secondary outcomes covered adverse pregnancy outcomes (intrauterine growth restriction, stillbirth, and maternal death) and adverse infant outcomes (preterm birth, congenital abnormalities, and infant death). DATA COLLECTION AND ANALYSIS We used standard Cochrane methods and assessed the certainty of evidence using the GRADE approach. MAIN RESULTS We included six RCTs (25 study reports) involving 17,991 pregnant women. The intervention was an RSV pre-F protein vaccine in four studies, and an RSV F protein nanoparticle vaccine in two studies. In all studies, the comparator was a placebo (saline, formulation buffer, or sterile water). We judged four studies at overall low risk of bias and two studies at overall high risk (mainly due to selection bias). All studies were funded by pharmaceutical companies. Maternal RSV vaccination compared with placebo reduces infant hospitalisation with laboratory-confirmed RSV disease (risk ratio (RR) 0.50, 95% confidence interval (CI) 0.31 to 0.82; 4 RCTs, 12,216 infants; high-certainty evidence). Based on an absolute risk with placebo of 22 hospitalisations per 1000 infants, our results represent 11 fewer hospitalisations per 1000 infants from vaccinated pregnant women (15 fewer to 4 fewer). No studies reported infant hospitalisation with clinically confirmed RSV disease. Maternal RSV vaccination compared with placebo has little or no effect on the risk of congenital abnormalities (RR 0.96, 95% CI 0.88 to 1.04; 140 per 1000 with placebo, 5 fewer per 1000 with RSV vaccination (17 fewer to 6 more); 4 RCTs, 12,304 infants; high-certainty evidence). Maternal RSV vaccination likely has little or no effect on the risk of intrauterine growth restriction (RR 1.32, 95% CI 0.75 to 2.33; 3 per 1000 with placebo, 1 more per 1000 with RSV vaccination (1 fewer to 4 more); 4 RCTs, 12,545 pregnant women; moderate-certainty evidence). Maternal RSV vaccination may have little or no effect on the risk of stillbirth (RR 0.81, 95% CI 0.38 to 1.72; 3 per 1000 with placebo, no difference with RSV vaccination (2 fewer to 3 more); 5 RCTs, 12,652 pregnant women). There may be a safety signal warranting further investigation related to preterm birth. This outcome may be more likely with maternal RSV vaccination, although the 95% CI includes no effect, and the evidence is very uncertain (RR 1.16, 95% CI 0.99 to 1.36; 6 RCTs, 17,560 infants; very low-certainty evidence). Based on an absolute risk of 51 preterm births per 1000 infants from pregnant women who received placebo, there may be 8 more per 1000 infants from pregnant women with RSV vaccination (1 fewer to 18 more). There was one maternal death in the RSV vaccination group and none in the placebo group. Our meta-analysis suggests that RSV vaccination compared with placebo may have little or no effect on the risk of maternal death (RR 3.00, 95% CI 0.12 to 73.50; 3 RCTs, 7977 pregnant women; low-certainty evidence). The effect of maternal RSV vaccination on the risk of infant death is very uncertain (RR 0.81, 95% CI 0.36 to 1.81; 6 RCTs, 17,589 infants; very low-certainty evidence). AUTHORS' CONCLUSIONS The findings of this review suggest that maternal RSV vaccination reduces laboratory-confirmed RSV hospitalisations in infants. There are no safety concerns about intrauterine growth restriction and congenital abnormalities. We must be careful in drawing conclusions about other safety outcomes owing to the low and very low certainty of the evidence. The evidence available to date suggests RSV vaccination may have little or no effect on stillbirth, maternal death, and infant death (although the evidence for infant death is very uncertain). However, there may be a safety signal warranting further investigation related to preterm birth. This is driven by data from one trial, which is not fully published yet. The evidence base would be much improved by more RCTs with substantial sample sizes and well-designed observational studies with long-term follow-up for assessment of safety outcomes. Future studies should aim to use standard outcome measures, collect data on concomitant vaccines, and stratify data by timing of vaccination, gestational age at birth, race, and geographical setting.
Collapse
Affiliation(s)
- Emily Wem Phijffer
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Odette de Bruin
- Department of Obstetrics, Division Woman and Baby, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| | - Fariba Ahmadizar
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| | - Louis J Bont
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Childrens Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nicoline At Van der Maas
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Miriam Cjm Sturkenboom
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| | - Joanne G Wildenbeest
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Childrens Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kitty Wm Bloemenkamp
- Department of Obstetrics, Division Woman and Baby, Wilhelmina Childrens Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
11
|
Anderson LJ, Jadhao SJ, Hussaini L, Ha B, McCracken CE, Gibson T, Yildirim I, Yi J, Stephens K, Korski C, Kao C, Sun H, Lee CY, Jaunarajs A, Rostad CA, Anderson EJ. Development and comparison of immunologic assays to detect primary RSV infections in infants. Front Immunol 2024; 14:1332772. [PMID: 38283339 PMCID: PMC10811012 DOI: 10.3389/fimmu.2023.1332772] [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: 11/03/2023] [Accepted: 12/18/2023] [Indexed: 01/30/2024] Open
Abstract
Effective respiratory syncytial virus (RSV) vaccines have been developed and licensed for elderly adults and pregnant women but not yet for infants and young children. The RSV immune state of the young child, i.e., previously RSV infected or not, is important to the conduct and interpretation of epidemiology studies and vaccine clinical trials. To address the need for sensitive assays to detect immunologic evidence of past infection, we developed, characterized, and evaluated 7 assays including 4 IgG antibody enzyme immunoassays (EIAs), two neutralizing antibody assays, and an IFN-γ EliSpot (EliSpot) assay. The four IgG EIAs used a subgroup A plus subgroup B RSV-infected Hep-2 cell lysate antigen (Lysate), an expressed RSV F protein antigen (F), an expressed subgroup A G protein antigen (Ga), or an expressed subgroup B G protein (Gb) antigen. The two neutralizing antibody assays used either a subgroup A or a subgroup B RSV strain. The EliSpot assay used a sucrose cushion purified combination of subgroup A and subgroup B infected cell lysate. All seven assays had acceptable repeatability, signal against control antigen, lower limit of detection, and, for the antibody assays, effect of red cell lysis, lipemia and anticoagulation of sample on results. In 44 sera collected from children >6 months after an RSV positive illness, the lysate, F, Ga and Gb IgG EIAs, and the subgroup A and B neutralizing antibody assays, and the EliSpot assays were positive in 100%, 100%, 86%, 95%, 43%, and 57%, respectively. The Lysate and F EIAs were most sensitive for detecting RSV antibody in young children with a documented RSV infection. Unexpectedly, the EliSpot assay was positive in 9/15 (60%) of PBMC specimens from infants not exposed to an RSV season, possibly from maternal microchimerism. The Lysate and F EIAs provide good options to reliably detect RSV antibodies in young children for epidemiologic studies and vaccine trials.
Collapse
Affiliation(s)
- Larry J. Anderson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Samadhan J. Jadhao
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Laila Hussaini
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Binh Ha
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Courtney E. McCracken
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Theda Gibson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Inci Yildirim
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Jumi Yi
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Kathy Stephens
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Chelsea Korski
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Carol Kao
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Heying Sun
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Chun Yi Lee
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | | | - Christina A. Rostad
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
12
|
Di Gennaro F, Guido G, Frallonardo L, Segala FV, De Nola R, Damiani GR, De Vita E, Totaro V, Barbagallo M, Nicastri E, Vimercati A, Cicinelli E, Liuzzi G, Veronese N, Saracino A. Efficacy and safety of therapies for COVID-19 in pregnancy: a systematic review and meta-analysis. BMC Infect Dis 2023; 23:776. [PMID: 37946100 PMCID: PMC10634005 DOI: 10.1186/s12879-023-08747-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Clinical evidence suggests that pregnant women are more vulnerable to COVID-19, since they are at increased risk for disease progression and for obstetric complications, such as premature labor, miscarriage, preeclampsia, cesarean delivery, fetal growth restriction and perinatal death. Despite this evidence, pregnant women are often excluded from clinical trials, resulting in limited knowledge on COVID-19 management. The aim of this systematic review and meta-analysis is to provide better evidence on the efficacy and safety of available COVID-19 treatment in pregnant women. METHODS Four authors searched major electronic databases from inception until 1 st November-2022 for controlled trials/observational studies, investigating outcomes after the administration of anti-SARS-CoV-2 treatments in pregnant women affected by COVID-19. The analyses investigated the cumulative incidence of delivery and maternal outcomes in pregnant women, comparing those taking active medication vs standard care. Risk ratios (RRs) with 95% confidence intervals were calculated. Statistical significance was assessed using the random effects model and inverse-variance method. This systematic review and meta-analysis was conducted in accordance with the updated 2020 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The protocol has been registered in Prospero (number registration: CRD42023397445). RESULTS From initially 937 non duplicate records, we assessed the full texts of 40 articles, finally including ten studies. In six studies, including 1627 patients, the use of casirivimab/imdevimab (CAS/IMD), remdesivir, and IFN-alpha 2b significantly decreased the need of cesarean section ((RR = 0.665; 95%CI: 0.491-0.899; p = 0.008; I 2 = 19.5%;) (Table 1, (Fig. 1). Treatments did not decrease the risk of preterm delivery, admission to neonatal ICU, or stillbirth/perinatal loss (p-values > 0.50 for all these outcomes) and did not prevent the progression of disease towards severe degrees (k = 8; 2,374 pregnant women; RR = 0.778; 95%CI: 0.550-1.099; p = 0.15; I 2 = 0%). Moreover, the use of medications during pregnancy did not modify the incidence of maternal death in two studies (Table 2). CONCLUSIONS To our analysis, CAS/IMD, remdesivir, and IFN alpha 2b reduced the number of cesarean sections but demonstrated no effect on disease progression and other obstetric and COVID-19 related outcomes. The inability to evaluate the influence of viral load on illness development in pregnant women was attributed to lack of data. In our systematic review, no major side effects were reported. Though, it is essential for the medical community to focus more on clinical trials and less on episodic case reports and case series, with standardization of fetal and maternal outcomes.
Collapse
Affiliation(s)
- Francesco Di Gennaro
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| | - Giacomo Guido
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| | - Luisa Frallonardo
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy.
| | - Francesco Vladimiro Segala
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| | - Rosalba De Nola
- Clinic of Obstetrics & Gynaecology, University of Bari "Aldo Moro", Bari, Italy
| | | | - Elda De Vita
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| | - Valentina Totaro
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| | - Mario Barbagallo
- Geriatrics Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Emanuele Nicastri
- National Institute for Infectious Diseases 'Lazzaro Spallanzani' (IRCCS), Rome, Italy
| | - Antonella Vimercati
- Clinic of Obstetrics & Gynaecology, University of Bari "Aldo Moro", Bari, Italy
| | - Ettore Cicinelli
- Clinic of Obstetrics & Gynaecology, University of Bari "Aldo Moro", Bari, Italy
| | - Giuseppina Liuzzi
- National Institute for Infectious Diseases 'Lazzaro Spallanzani' (IRCCS), Rome, Italy
| | - Nicola Veronese
- Geriatrics Section, Department of Internal Medicine, University of Palermo, Palermo, Italy
| | - Annalisa Saracino
- Clinic of Infectious Diseases, Department of Precision and Regenerative Medicine and Ionian Area - (DiMePRe-J), University of "Aldo Moro", University of Bari "Aldo Moro", Piazza Giulio Cesare N. 11 Cap 70124, Bari, Italy
| |
Collapse
|
13
|
Nenna R, Stern DA, Carr TF, Spangenberg A, Wright AL, Martinez FD, Halonen M. Prenatal exposure to RSV season influences first-year risk of RSV lower respiratory illness and RSV-specific immune responses assessed at birth. J Virol 2023; 97:e0076723. [PMID: 37671863 PMCID: PMC10537569 DOI: 10.1128/jvi.00767-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/01/2023] [Indexed: 09/07/2023] Open
Abstract
Maternal-to-fetal transmission of respiratory syncytial virus (RSV) has been shown to occur but whether late prenatal exposure to RSV season influences offspring postnatal RSV-lower respiratory illness (LRI) risk in early life or RSV immune status at birth is unclear. In this study, the duration of third trimester RSV season exposure was determined for 1,094 newborns of the Tucson Children's Respiratory Study (TCRS) and found to show an inverse relation to risk for first RSV-LRI in the first year. Cord blood anti-RSV antibody is related to third trimester RSV season exposure but not to first year RSV-LRI risk. In a separate birth cohort (the Infant Immune Study), supernatants from cord blood mononuclear cells stimulated with the recall antigen, UV-inactivated RSV, were assayed for IFN-γ and IL-4. The frequency of detectable IFN-γ (but not IL-4) was increased for those with at least 2 mo of third trimester RSV season exposure, suggestive of a fetal immune response to RSV. IMPORTANCE Our study found that duration of third trimester exposure to RSV season related inversely to subsequent risk of postnatal RSV-LRI in the first year, thus implicating this exposure as an important factor in reducing risk of postnatal RSV-LRIs, a risk reduction that appears to be independent of maternally transferred anti-RSV antibody level. The increase in frequency of detectable IFN-γ and not IL-4 in response to UV-inactivated RSV in cord blood immune cells for infants with greater third trimester exposure to RSV season is suggestive of a Type-1 immune response to RSV occurring in utero.
Collapse
Affiliation(s)
- Raffaella Nenna
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
- Department of Maternal, Infantile, and Urological Services, Sapienza University of Rome, Rome, Italy
| | - Debra A. Stern
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Tara F. Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Amber Spangenberg
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Anne L. Wright
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Fernando D. Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Marilyn Halonen
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
14
|
Respiratory syncytial virus vaccination during pregnancy for improving infant outcomes. Cochrane Database Syst Rev 2022; 2022:CD015134. [PMCID: PMC9585514 DOI: 10.1002/14651858.cd015134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To assess the efficacy and safety of maternal RSV vaccination for preventing respiratory syncytial virus disease in infants.
Collapse
|
15
|
Koch CM, Prigge AD, Setar L, Anekalla KR, Do-Umehara HC, Abdala-Valencia H, Politanska Y, Shukla A, Chavez J, Hahn GR, Coates BM. Cilia-related gene signature in the nasal mucosa correlates with disease severity and outcomes in critical respiratory syncytial virus bronchiolitis. Front Immunol 2022; 13:924792. [PMID: 36211387 PMCID: PMC9540395 DOI: 10.3389/fimmu.2022.924792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) can cause life-threatening respiratory failure in infants. We sought to characterize the local host response to RSV infection in the nasal mucosa of infants with critical bronchiolitis and to identify early admission gene signatures associated with clinical outcomes. Methods Nasal scrape biopsies were obtained from 33 infants admitted to the pediatric intensive care unit (PICU) with critical RSV bronchiolitis requiring non-invasive respiratory support (NIS) or invasive mechanical ventilation (IMV), and RNA sequencing (RNA-seq) was performed. Gene expression in participants who required shortened NIS (</= 3 days), prolonged NIS (> 3 days), and IMV was compared. Findings Increased expression of ciliated cell genes and estimated ciliated cell abundance, but not immune cell abundance, positively correlated with duration of hospitalization in infants with critical bronchiolitis. A ciliated cell signature characterized infants who required NIS for > 3 days while a basal cell signature was present in infants who required NIS for </= 3 days, despite both groups requiring an equal degree of respiratory support at the time of sampling. Infants who required invasive mechanical ventilation had increased expression of genes involved in neutrophil activation and cell death. Interpretation Increased expression of cilia-related genes in clinically indistinguishable infants with critical RSV may differentiate between infants who will require prolonged hospitalization and infants who will recover quickly. Validation of these findings in a larger cohort is needed to determine whether a cilia-related gene signature can predict duration of illness in infants with critical bronchiolitis. The ability to identify which infants with critical RSV bronchiolitis may require prolonged hospitalization using non-invasive nasal samples would provide invaluable prognostic information to parents and medical providers.
Collapse
Affiliation(s)
- Clarissa M. Koch
- Department of Medicine, Northwestern University, Chicago, IL, United States
| | - Andrew D. Prigge
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Leah Setar
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | | | | | | | - Yuliya Politanska
- Department of Medicine, Northwestern University, Chicago, IL, United States
| | - Avani Shukla
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Jairo Chavez
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Grant R. Hahn
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Bria M. Coates
- Department of Pediatrics, Northwestern University, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
- *Correspondence: Bria M. Coates,
| |
Collapse
|
16
|
Bylsma LC, Suh M, Movva N, Fryzek JP, Nelson CB. Mortality Among US Infants and Children Under 5 Years of Age with Respiratory Syncytial Virus and Bronchiolitis: A Systematic Literature Review. J Infect Dis 2022; 226:S267-S281. [PMID: 35968871 PMCID: PMC9377034 DOI: 10.1093/infdis/jiac226] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background A systematic literature review was conducted to summarize the mortality (overall and by disease severity factors) of US infants and children aged <5 years with respiratory syncytial virus (RSV) or all-cause bronchiolitis (ACB). Methods Comprehensive, systematic literature searches were conducted; articles were screened using prespecified eligibility criteria. A standard risk of bias tool was used to evaluate studies. Mortality was extracted as the rate per 100 000 or the case fatality ratio (CFR; proportion of deaths among RSV/ACB cases). Results Among 42 included studies, 36 evaluated inpatient deaths; 10 used nationally representative populations updated through 2013, and only 2 included late-preterm/full-term otherwise healthy infants and children. The RSV/ACB definition varied across studies (multiple International Classification of Diseases [ICD] codes; laboratory confirmation); no study reported systematic testing for RSV. No studies reported RSV mortality rates, while 3 studies provided ACB mortality rates (0.57–9.4 per 100 000). CFRs ranged from 0% to 1.7% for RSV (n = 15) and from 0% to 0.17% for ACB (n = 6); higher CFRs were reported among premature, intensive care unit-admitted, and publicly insured infants and children. Conclusions RSV mortality reported among US infants and children is variable. Current, nationally representative estimates are needed for otherwise healthy, late-preterm to full-term infants and children.
Collapse
Affiliation(s)
- Lauren C Bylsma
- EpidStrategies, A Division of ToxStrategies, Inc, Rockville, Maryland, USA
| | - Mina Suh
- EpidStrategies, A Division of ToxStrategies, Inc, Rockville, Maryland, USA
| | - Naimisha Movva
- EpidStrategies, A Division of ToxStrategies, Inc, Rockville, Maryland, USA
| | - Jon P Fryzek
- EpidStrategies, A Division of ToxStrategies, Inc, Rockville, Maryland, USA
| | | |
Collapse
|
17
|
Baraldi E, Checcucci Lisi G, Costantino C, Heinrichs JH, Manzoni P, Riccò M, Roberts M, Vassilouthis N. RSV disease in infants and young children: Can we see a brighter future? Hum Vaccin Immunother 2022; 18:2079322. [PMID: 35724340 DOI: 10.1080/21645515.2022.2079322] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a highly contagious seasonal virus and the leading cause of Lower Respiratory Tract Infections (LRTI), including pneumonia and bronchiolitis in children. RSV-related LRTI cause approximately 3 million hospitalizations and 120,000 deaths annually among children <5 years of age. The majority of the burden of RSV occurs in previously healthy infants. Only a monoclonal antibody (mAb) has been approved against RSV infections in a restricted group, leaving an urgent unmet need for a large number of children potentially benefiting from preventive measures. Approaches under development include maternal vaccines to protect newborns, extended half-life monoclonal antibodies to provide rapid long-lasting protection, and pediatric vaccines. RSV has been identified as a major global priority but a solution to tackle this unmet need for all children has yet to be implemented. New technologies represent the avenue for effectively addressing the leading-cause of hospitalization in children <1 years old.
Collapse
Affiliation(s)
- Eugenio Baraldi
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | | | - Claudio Costantino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | | | - Paolo Manzoni
- Department of Pediatrics and Neonatology, University Hospital Degli Infermi, Biella, Italy
| | - Matteo Riccò
- Dipartimento di Sanità Pubblica, Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | | |
Collapse
|
18
|
Respiratory Syncytial Virus in Pregnant Women: Systematic Review and Meta-Analysis. WOMEN 2022. [DOI: 10.3390/women2020016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Human Respiratory Syncytial Virus (RSV) is a highly contagious viral pathogen. In infants, it is usually listed among the main causes of medical referrals and hospitalizations, particularly among newborns. While waiting for the results of early randomized controlled trials on maternal vaccination against RSV, the present systematic review and meta-analysis aimed to collect available evidence on maternal RSV infections. According to the PRISMA statement, Pubmed, Embase, and pre-print archive medRxiv.og were searched for eligible studies published up to 1 April 2022. Raw data included the incidence of RSV infection among sampled pregnant women, and the occurrence of complications. Data were then pooled in a random-effects model. Heterogeneity was assessed using the I2 measure, while reporting bias was assessed by means of funnel plots and regression analysis. A total of 5 studies for 282,918 pregnancies were retrieved, with a pooled prevalence of 0.2 per 100 pregnancies and 2.5 per 100 pregnancies with respiratory tract infections. Neither maternal deaths nor miscarriages were reported. Even though detailed data were available only for 6309 pregnancies and 33 RSV cases, infant outcomes such as low birth weight and preterm delivery were rare (in both cases 0.04%), but up to 9.1% in cases where RSV diagnosis was confirmed. No substantially increased risk for preterm delivery (RR 1.395; 95%CI 0.566 to 3.434) and giving birth to a low-birth-weight infant (RR 0.509; 95%CI 0.134 to 1.924) was eventually identified. Conclusions. Although RSV is uncommonly detected among pregnant women, incident cases were associated with a relatively high share of complications. However, heterogeneous design and the quality of retrieved reports stress the need for specifically designed studies.
Collapse
|
19
|
McCall MN, Chu CY, Wang L, Benoodt L, Thakar J, Corbett A, Holden-Wiltse J, Slaunwhite C, Grier A, Gill SR, Falsey AR, Topham DJ, Caserta MT, Walsh EE, Qiu X, Mariani TJ. A systems genomics approach uncovers molecular associates of RSV severity. PLoS Comput Biol 2021; 17:e1009617. [PMID: 34962914 PMCID: PMC8746750 DOI: 10.1371/journal.pcbi.1009617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 01/10/2022] [Accepted: 11/05/2021] [Indexed: 01/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection results in millions of hospitalizations and thousands of deaths each year. Variations in the adaptive and innate immune response appear to be associated with RSV severity. To investigate the host response to RSV infection in infants, we performed a systems-level study of RSV pathophysiology, incorporating high-throughput measurements of the peripheral innate and adaptive immune systems and the airway epithelium and microbiota. We implemented a novel multi-omic data integration method based on multilayered principal component analysis, penalized regression, and feature weight back-propagation, which enabled us to identify cellular pathways associated with RSV severity. In both airway and immune cells, we found an association between RSV severity and activation of pathways controlling Th17 and acute phase response signaling, as well as inhibition of B cell receptor signaling. Dysregulation of both the humoral and mucosal response to RSV may play a critical role in determining illness severity.
Collapse
Affiliation(s)
- Matthew N. McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Chin-Yi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Lauren Benoodt
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Juilee Thakar
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Anthony Corbett
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester New York, United States of America
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester New York, United States of America
| | - Christopher Slaunwhite
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Ann R. Falsey
- Department of Medicine, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Medicine, Rochester General Hospital, Rochester New York, United States of America
| | - David J. Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Mary T. Caserta
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| | - Edward E. Walsh
- Department of Medicine, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Medicine, Rochester General Hospital, Rochester New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Thomas J. Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester New York, United States of America
- Department of Pediatrics, University of Rochester Medical Center, Rochester New York, United States of America
| |
Collapse
|
20
|
Citron MP, McAnulty J, Callahan C, Knapp W, Fontenot J, Morales P, Flynn JA, Douglas CM, Espeseth AS. Transplacental Antibody Transfer of Respiratory Syncytial Virus Specific IgG in Non-Human Primate Mother-Infant Pairs. Pathogens 2021; 10:pathogens10111441. [PMID: 34832599 PMCID: PMC8624788 DOI: 10.3390/pathogens10111441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
One approach to protect new-borns against respiratory syncytial virus (RSV) is to vaccinate pregnant women in the last trimester of pregnancy. The boosting of circulating antibodies which can be transferred to the foetus would offer immune protection against the virus and ultimately the disease. Since non-human primates (NHPs) have similar reproductive anatomy, physiology, and antibody architecture and kinetics to humans, we utilized this preclinical species to evaluate maternal immunization (MI) using an RSV F subunit vaccine. Three species of NHPs known for their ability to be infected with human RSV in experimental challenge studies were tested for RSV-specific antibodies. African green monkeys had the highest overall antibody levels of the old-world monkeys evaluated and they gave birth to offspring with anti-RSV titers that were proportional to their mother. These higher overall antibody levels are associated with greater durability found in their offspring. Immunization of RSV seropositive AGMs during late pregnancy boosts RSV titers, which consequentially results in significantly higher titers in the vaccinated new-borns compared to the new-borns of unvaccinated mothers. These findings, accomplished in small treatment group sizes, demonstrate a model that provides an efficient, resource sparing and translatable preclinical in vivo system for evaluating vaccine candidates for maternal immunization.
Collapse
Affiliation(s)
- Michael P. Citron
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
- Correspondence:
| | - Jessica McAnulty
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Cheryl Callahan
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Walter Knapp
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., Kenilworth, NJ 07033, USA;
| | - Jane Fontenot
- The New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA;
| | - Pablo Morales
- The Mannheimer Foundation, Homestead, FL 33034, USA;
| | - Jessica A. Flynn
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Cameron M. Douglas
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Amy S. Espeseth
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| |
Collapse
|
21
|
Khan S, Dobrovolny HM. A study of the effects of age on the dynamics of RSV in animal models. Virus Res 2021; 304:198524. [PMID: 34329697 DOI: 10.1016/j.virusres.2021.198524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/24/2021] [Accepted: 07/17/2021] [Indexed: 01/18/2023]
Abstract
Respiratory syncytial virus can cause severe illness and even death, particularly in infants. The increased severity of disease in young children is thought to be due to a lack of previous exposure to the virus as well as the limited immune response in infants. While studies have examined the clinical differences in disease between infants and adults, there has been limited examination of how the viral dynamics differ as infants develop. In this study, we apply a mathematical model to data from cotton rats and ferrets of different ages to assess how viral kinetics parameters change as the animals age. We find no clear trend in the viral decay rate, infecting time, and basic reproduction number as the animals age. We discuss possible reasons for the null result including the limited data, lack of detail of the mathematical model, and the limitations of animal models.
Collapse
Affiliation(s)
- Shaheer Khan
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX USA
| | - Hana M Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX USA.
| |
Collapse
|
22
|
Anderson LJ, Jadhao SJ, Paden CR, Tong S. Functional Features of the Respiratory Syncytial Virus G Protein. Viruses 2021; 13:1214. [PMID: 34372490 PMCID: PMC8310105 DOI: 10.3390/v13071214] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/28/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of serious lower respiratory tract infections in children <5 years of age worldwide and repeated infections throughout life leading to serious disease in the elderly and persons with compromised immune, cardiac, and pulmonary systems. The disease burden has made it a high priority for vaccine and antiviral drug development but without success except for immune prophylaxis for certain young infants. Two RSV proteins are associated with protection, F and G, and F is most often pursued for vaccine and antiviral drug development. Several features of the G protein suggest it could also be an important to vaccine or antiviral drug target design. We review features of G that effect biology of infection, the host immune response, and disease associated with infection. Though it is not clear how to fit these together into an integrated picture, it is clear that G mediates cell surface binding and facilitates cellular infection, modulates host responses that affect both immunity and disease, and its CX3C aa motif contributes to many of these effects. These features of G and the ability to block the effects with antibody, suggest G has substantial potential in vaccine and antiviral drug design.
Collapse
Affiliation(s)
- Larry J. Anderson
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
| | - Samadhan J. Jadhao
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
| | - Clinton R. Paden
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30322, USA; (C.R.P.); (S.T.)
| | - Suxiang Tong
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30322, USA; (C.R.P.); (S.T.)
| |
Collapse
|
23
|
Koivisto K, Nieminen T, Mejias A, Capella C, Ye F, Mertz S, Peeples M, Ramilo O, Saxén H. RSV Specific Antibodies in Pregnant Women and Subsequent Risk of RSV Hospitalization in Young Infants. J Infect Dis 2021; 225:1189-1196. [PMID: 34129040 PMCID: PMC8974854 DOI: 10.1093/infdis/jiab315] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background The fusion (F) glycoprotein of respiratory syncytial virus (RSV) represents the major neutralizing antigen, and antibodies against the pre-F conformation have the most potent neutralizing activity. This study aimed to assess the correlation between maternal antibody titers against the pre-F, post-F, and G glycoproteins and the child’s risk of developing severe RSV bronchiolitis early in infancy. Methods We identified previously healthy term infants <3 months of age hospitalized with RSV bronchiolitis from December 2015 to March 2016. We measured IgG antibody titers to pre-F, post-F, and G proteins in maternal sera obtained at 9–12 weeks of pregnancy of these hospitalized infants’ mothers (n = 94) and compared them with serum antibody titers of control pregnant mothers (n = 130) whose children were not hospitalized. Results All maternal samples (n = 224) had detectable pre-F antibodies. Pre-F antibody titers were significantly lower in mothers whose infants were hospitalized with RSV bronchiolitis compared with those mothers whose infants were not hospitalized (23.9 [range (or antibody titer range), 1.4–273.7] µg/L vs 30.6 [XXX, 3.4–220.0] µg/L; P = .0026). There were no significant differences in maternal post-F and G antibody titers between hospitalized and nonhospitalized infants. Conclusions Our findings indicate that maternal pre-F antibodies are fundamental for providing immune protection to the infant.
Collapse
Affiliation(s)
- K Koivisto
- Helsinki University Hospital and University of Helsinki, Children's Hospital, Helsinki, Finland
| | - T Nieminen
- Helsinki University Hospital and University of Helsinki, Children's Hospital, Helsinki, Finland
| | - A Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - C Capella
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - F Ye
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - S Mertz
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - M Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - O Ramilo
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - H Saxén
- Helsinki University Hospital and University of Helsinki, Children's Hospital, Helsinki, Finland
| |
Collapse
|
24
|
Jadhao SJ, Ha B, McCracken C, Gebretsadik T, Rosas-Salazar C, Chappell J, Das S, Hartert T, Anderson LJ. Performance evaluation of antibody tests for detecting infant respiratory syncytial virus infection. J Med Virol 2021; 93:3439-3445. [PMID: 33325064 PMCID: PMC8046717 DOI: 10.1002/jmv.26736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Accepted: 12/12/2020] [Indexed: 11/12/2022]
Abstract
Respiratory syncytial virus (RSV) infection is a major cause of respiratory tract disease in young children and throughout life. Infant infection is also associated with later respiratory morbidity including asthma. With a prospective birth cohort study of RSV and asthma, we evaluated the performance of an RSV antibody enzyme-linked immunoassay (EIA) for detecting prior infant RSV infection. Infant RSV infection was determined by biweekly respiratory illness surveillance plus RSV polymerase chain reaction (PCR) testing in their first RSV season and serum RSV antibodies after the season at approximately 1 year of age. RSV antibodies were detected by RSV A and B lysate EIA. Antibody and PCR results on 1707 children included 327 RSV PCR positive (PCR+) and 1380 not RSV+. Of 327 PCR+ children, 314 (96%) were lysate EIA positive and 583 out of 1380 (42%) children not PCR+ were positive. We compared the lysate EIA to RSV F, group A G (Ga), and group B G (Gb) protein antibody EIAs in a subset of 226 sera, 118 PCR+ children (97 group A and 21 group B) and 108 not PCR+. In this subset, 117 out of 118 (99%) RSV PCR+ children were positive by both the F and lysate EIAs and 103 out of 118 (87%) were positive by the Ga and/or Gb EIAs. Comparison of the two G EIAs indicated the infecting group correctly in 100 out of 118 (86%) and incorrectly in 1 out of 118 (1%). The lysate and F EIAs are sensitive for detecting infant infection and the two G EIAs can indicate the group of an earlier primary infection.
Collapse
Affiliation(s)
- Samadhan J. Jadhao
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Binh Ha
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Courtney McCracken
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Tebeb Gebretsadik
- Department of Medicine, Biostatistics and Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Christian Rosas-Salazar
- Department of Medicine, Biostatistics and Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - James Chappell
- Department of Medicine, Biostatistics and Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Suman Das
- Department of Medicine, Biostatistics and Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tina Hartert
- Department of Medicine, Biostatistics and Pathology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Larry J. Anderson
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
25
|
Chu CY, Qiu X, McCall MN, Wang L, Corbett A, Holden-Wiltse J, Slaunwhite C, Grier A, Gill SR, Pryhuber GS, Falsey AR, Topham DJ, Caserta MT, Walsh EE, Mariani TJ. Airway Gene Expression Correlates of Respiratory Syncytial Virus Disease Severity and Microbiome Composition in Infants. J Infect Dis 2021; 223:1639-1649. [PMID: 32926149 PMCID: PMC8136980 DOI: 10.1093/infdis/jiaa576] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants. The causes and correlates of severe illness in the majority of infants are poorly defined. METHODS We recruited a cohort of RSV-infected infants and simultaneously assayed the molecular status of their airways and the presence of airway microbiota. We used rigorous statistical approaches to identify gene expression patterns associated with disease severity and microbiota composition, separately and in combination. RESULTS We measured comprehensive airway gene expression patterns in 106 infants with primary RSV infection. We identified an airway gene expression signature of severe illness dominated by excessive chemokine expression. We also found an association between Haemophilus influenzae, disease severity, and airway lymphocyte accumulation. Exploring the time of onset of clinical symptoms revealed acute activation of interferon signaling following RSV infection in infants with mild or moderate illness, which was absent in subjects with severe illness. CONCLUSIONS Our data reveal that airway gene expression patterns distinguish mild/moderate from severe illness. Furthermore, our data identify biomarkers that may be therapeutic targets or useful for measuring efficacy of intervention responses.
Collapse
Affiliation(s)
- Chin-Yi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York, USA
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew N McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, USA
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, USA
| | - Anthony Corbett
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, USA
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, USA
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Christopher Slaunwhite
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York, USA
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Gloria S Pryhuber
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Ann R Falsey
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Medicine, Rochester General Hospital, Rochester, New York, USA
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Mary T Caserta
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward E Walsh
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Medicine, Rochester General Hospital, Rochester, New York, USA
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York, USA
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
26
|
Wang L, Chu CY, McCall MN, Slaunwhite C, Holden-Wiltse J, Corbett A, Falsey AR, Topham DJ, Caserta MT, Mariani TJ, Walsh EE, Qiu X. Airway gene-expression classifiers for respiratory syncytial virus (RSV) disease severity in infants. BMC Med Genomics 2021; 14:57. [PMID: 33632195 PMCID: PMC7908785 DOI: 10.1186/s12920-021-00913-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A substantial number of infants infected with RSV develop severe symptoms requiring hospitalization. We currently lack accurate biomarkers that are associated with severe illness. METHOD We defined airway gene expression profiles based on RNA sequencing from nasal brush samples from 106 full-tem previously healthy RSV infected subjects during acute infection (day 1-10 of illness) and convalescence stage (day 28 of illness). All subjects were assigned a clinical illness severity score (GRSS). Using AIC-based model selection, we built a sparse linear correlate of GRSS based on 41 genes (NGSS1). We also built an alternate model based upon 13 genes associated with severe infection acutely but displaying stable expression over time (NGSS2). RESULTS NGSS1 is strongly correlated with the disease severity, demonstrating a naïve correlation (ρ) of ρ = 0.935 and cross-validated correlation of 0.813. As a binary classifier (mild versus severe), NGSS1 correctly classifies disease severity in 89.6% of the subjects following cross-validation. NGSS2 has slightly less, but comparable, accuracy with a cross-validated correlation of 0.741 and classification accuracy of 84.0%. CONCLUSION Airway gene expression patterns, obtained following a minimally-invasive procedure, have potential utility for development of clinically useful biomarkers that correlate with disease severity in primary RSV infection.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester School Medicine, Rochester, NY, USA
| | - Chin-Yi Chu
- Department of Pediatrics, University of Rochester School Medicine, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biostatistics and Computational Biology, University of Rochester School Medicine, Rochester, NY, USA
| | | | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester School Medicine, Rochester, NY, USA
| | - Anthony Corbett
- Department of Biostatistics and Computational Biology, University of Rochester School Medicine, Rochester, NY, USA
| | - Ann R Falsey
- Department of Medicine, University of Rochester School Medicine, Rochester, NY, USA
- Department of Medicine, Rochester General Hospital, Rochester, NY, USA
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester School Medicine, Rochester, NY, USA
| | - Mary T Caserta
- Department of Pediatrics, University of Rochester School Medicine, Rochester, NY, USA
| | - Thomas J Mariani
- Department of Pediatrics, University of Rochester School Medicine, Rochester, NY, USA.
| | - Edward E Walsh
- Department of Medicine, University of Rochester School Medicine, Rochester, NY, USA.
- Department of Medicine, Rochester General Hospital, Rochester, NY, USA.
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School Medicine, Rochester, NY, USA.
| |
Collapse
|
27
|
Eichinger KM, Kosanovich JL, Lipp M, Empey KM, Petrovsky N. Strategies for active and passive pediatric RSV immunization. Ther Adv Vaccines Immunother 2021; 9:2515135520981516. [PMID: 33623860 PMCID: PMC7879001 DOI: 10.1177/2515135520981516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide, with the most severe disease occurring in very young infants. Despite half a century of research there still are no licensed RSV vaccines. Difficulties in RSV vaccine development stem from a number of factors, including: (a) a very short time frame between birth and first RSV exposure; (b) interfering effects of maternal antibodies; and (c) differentially regulated immune responses in infants causing a marked T helper 2 (Th2) immune bias. This review seeks to provide an age-specific understanding of RSV immunity critical to the development of a successful pediatric RSV vaccine. Historical and future approaches to the prevention of infant RSV are reviewed, including passive protection using monoclonal antibodies or maternal immunization strategies versus active infant immunization using pre-fusion forms of RSV F protein antigens formulated with novel adjuvants such as Advax that avoid excess Th2 immune polarization.
Collapse
Affiliation(s)
- Katherine M. Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, and Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L. Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kerry M. Empey
- Department of Pharmacy and Therapeutics, Department of Pharmaceutical Sciences, School of Medicine and Clinical and Translational Science Institute, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia and Vaxine Pty Ltd, Warradale, SA 5046, Australia
| |
Collapse
|
28
|
Siefker DT, Vu L, You D, McBride A, Taylor R, Jones TL, DeVincenzo J, Cormier SA. Respiratory Syncytial Virus Disease Severity Is Associated with Distinct CD8 + T-Cell Profiles. Am J Respir Crit Care Med 2020; 201:325-334. [PMID: 31644878 PMCID: PMC6999109 DOI: 10.1164/rccm.201903-0588oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 10/21/2019] [Indexed: 01/03/2023] Open
Abstract
Rationale: Respiratory syncytial virus (RSV) causes significant morbidity and mortality in infants worldwide. Although T-helper type 2 (Th2) cell pathology is implicated in severe disease, the mechanisms underlying the development of immunopathology are incompletely understood.Objectives: We aimed to identify local immune responses associated with severe RSV in infants. Our hypothesis was that disease severity would correlate with enhanced Th2 cellular responses.Methods: Nasal aspirates were collected from infants hospitalized with severe (admitted to the pediatric ICU) or moderate (maintained in the general ward) RSV disease at 5 to 9 days after enrollment. The immune response was investigated by evaluating T-lymphocyte cellularity, cytokine concentration, and viral load.Measurements and Main Results: Patients with severe disease had increased proportions of CD8 (cluster of differentiation 8)-positive T cells expressing IL-4 (Tc2) and reduced proportions of CD8+ T cells expressing IFNγ (Tc1). Nasal aspirates from patients with severe disease had reduced concentrations of IL-17. Patients with greater frequencies of Tc1, CD8+ T cells expressing IL-17 (Tc17), and CD4+ T cells expressing IL-17 (Th17) had shorter durations of hospitalization.Conclusions: Severe RSV disease was associated with distinct T-cell profiles. Tc1, Tc17, and Th17 were associated with shorter hospital stay and may play a protective role, whereas Tc2 cells may play a previously underappreciated role in pathology.
Collapse
Affiliation(s)
- David T. Siefker
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Luan Vu
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Dahui You
- Department of Pediatrics, and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | | | - Tamekia L. Jones
- Department of Pediatrics, and
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | - John DeVincenzo
- Department of Pediatrics, and
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | - Stephania A. Cormier
- Department of Biological Sciences, College of Science, and Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
29
|
Muňoz FM, Swamy GK, Hickman SP, Agrawal S, Piedra PA, Glenn GM, Patel N, August AM, Cho I, Fries L. Safety and Immunogenicity of a Respiratory Syncytial Virus Fusion (F) Protein Nanoparticle Vaccine in Healthy Third-Trimester Pregnant Women and Their Infants. J Infect Dis 2019; 220:1802-1815. [PMID: 31402384 DOI: 10.1093/infdis/jiz390] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/31/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the leading cause of infant lower respiratory tract disease and hospitalization worldwide. METHODS Safety and immunogenicity of RSV fusion (F) protein nanoparticle vaccine or placebo were evaluated in 50 healthy third-trimester pregnant women. Assessments included vaccine tolerability and safety in women and infants, and RSV-specific antibody measures in women before and after vaccination, at delivery and post partum. RESULTS The vaccine was well tolerated; no meaningful differences in pregnancy or infant outcomes were observed between study groups. RSV-specific antibody levels increased significantly among vaccine recipients, including responses competitive with well-described monoclonal antibodies specific for multiple RSV neutralizing epitopes. No significant antibody increase was seen among placebo recipients, although a shallow upward trend across the RSV season was noted. Transplacental antibody transfer was 90%-120% across assays for infants of vaccinated women. Women with an interval of ≥30 days between vaccination and delivery demonstrated higher placental antibody transfer rates than women with an interval <30 days. Half-lives of RSV-specific antibodies in infants approximated 40 days. There was no evidence of severe RSV disease in infants of vaccinated mothers. CONCLUSIONS Data from this phase 2 study support a maternal immunization strategy to protect infants from RSV disease. CLINICAL TRIALS REGISTRATION NCT02247726.
Collapse
Affiliation(s)
| | - Geeta K Swamy
- Duke University School of Medicine, Durham, North Carolina
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kwon YM, Lee Y, Kim KH, Jung YJ, Li Z, Jeeva S, Lee S, Moore ML, Kang SM. Antigenicity and immunogenicity of unique prefusion-mimic F proteins presented on enveloped virus-like particles. Vaccine 2019; 37:6656-6664. [PMID: 31542260 DOI: 10.1016/j.vaccine.2019.09.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 11/27/2022]
Abstract
Pre-fusion stabilizing mutations (DS-Cav1) in soluble fusion (F) proteins of human respiratory syncytial virus (RSV) were previously reported. Here we investigated the antigenic and immunogenic properties of pre-fusion like RSV F proteins on enveloped virus-like particles (VLP). Additional mutations were introduced to DS-Cav1 (F-dcmTM VLP); fusion peptide deletion and cleavage mutation site 1 (F1d-dcmTM VLP) or both sites (F12d-dcmTM VLP). F1d-dcmTM VLP and F12d-dcmTM VLP displayed higher reactivity against pre-fusion specific site Ø and antigenic site I and II specific monoclonal antibodies, compared to F-dcmTM VLP with DS-Cav1 only. Mice immunized with F1d-dcmTM VLP and F12d-dcmTM VLP induced higher levels of DS-Cav1 pre-fusion specific IgG antibodies, RSV neutralizing activity titers, and effective lung viral clearance after challenge. These results suggest that cleavage site mutations and fusion peptide deletion in addition to DS-Cav1 mutations have contributed to structural stabilization of pre-fusion like F conformation on enveloped VLP, capable of inducing high levels of pre-fusion F specific and RSV neutralizing antibodies.
Collapse
Affiliation(s)
- Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Ki Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yu Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Zhuo Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sujin Lee
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
31
|
Sonawane AR, Tian L, Chu CY, Qiu X, Wang L, Holden-Wiltse J, Grier A, Gill SR, Caserta MT, Falsey AR, Topham DJ, Walsh EE, Mariani TJ, Weiss ST, Silverman EK, Glass K, Liu YY. Microbiome-Transcriptome Interactions Related to Severity of Respiratory Syncytial Virus Infection. Sci Rep 2019; 9:13824. [PMID: 31554845 PMCID: PMC6761288 DOI: 10.1038/s41598-019-50217-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/09/2019] [Indexed: 01/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections and hospital visits during infancy and childhood. Although risk factors for RSV infection have been identified, the role of microbial species in the respiratory tract is only partially known. We aimed to understand the impact of interactions between the nasal microbiome and host transcriptome on the severity and clinical outcomes of RSV infection. We used 16 S rRNA sequencing to characterize the nasal microbiome of infants with RSV infection. We used RNA sequencing to interrogate the transcriptome of CD4+ T cells obtained from the same set of infants. After dimension reduction through principal component (PC) analysis, we performed an integrative analysis to identify significant co-variation between microbial clade and gene expression PCs. We then employed LIONESS (Linear Interpolation to Obtain Network Estimates for Single Samples) to estimate the clade-gene association patterns for each infant. Our network-based integrative analysis identified several clade-gene associations significantly related to the severity of RSV infection. The microbial taxa with the highest loadings in the implicated clade PCs included Moraxella, Corynebacterium, Streptococcus, Haemophilus influenzae, and Staphylococcus. Interestingly, many of the genes with the highest loadings in the implicated gene PCs are encoded in mitochondrial DNA, while others are involved in the host immune response. This study on microbiome-transcriptome interactions provides insights into how the host immune system mounts a response against RSV and specific infectious agents in nasal microbiota.
Collapse
Affiliation(s)
- Abhijeet R Sonawane
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Liang Tian
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Chin-Yi Chu
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Mary T Caserta
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ann R Falsey
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Edward E Walsh
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Thomas J Mariani
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
- Center for Cancer Systems Biology, Dana Farber Cancer Institute, Boston, MA, 02115, USA.
| |
Collapse
|
32
|
Olagoke O, Quigley BL, Eiden MV, Timms P. Antibody response against koala retrovirus (KoRV) in koalas harboring KoRV-A in the presence or absence of KoRV-B. Sci Rep 2019; 9:12416. [PMID: 31455828 PMCID: PMC6711960 DOI: 10.1038/s41598-019-48880-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/13/2019] [Indexed: 01/13/2023] Open
Abstract
Koala retrovirus (KoRV) is in the process of endogenization into the koala (Phascolarctos cinereus) genome and is currently spreading through the Australian koala population. Understanding how the koala's immune system responds to KoRV infection is critical for developing an efficacious vaccine to protect koalas. To this end, we analyzed the antibody response of 235 wild koalas, sampled longitudinally over a four-year period, that harbored KoRV-A, and with or without KoRV-B. We found that the majority of the sampled koalas were able to make anti-KoRV antibodies, and that there was a linear increase in anti-KoRV IgG levels in koalas up to approximately seven years of age and then a gradual decrease thereafter. Koalas infected with both KoRV-A and KoRV-B were found to have slightly higher anti-KoRV IgG titers than koalas with KoRV-A alone and there was an inverse relationship between anti-KoRV IgG levels and circulating KoRV viral load. Finally, we identified distinct epitopes on the KoRV envelope protein that were recognized by antibodies. Together, these findings provide insight into the koala's immune response to KoRV and may be useful in the development of a therapeutic KoRV vaccine.
Collapse
Affiliation(s)
- O Olagoke
- Genecology Research Center, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, 4556, Queensland, Australia
| | - B L Quigley
- Genecology Research Center, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, 4556, Queensland, Australia
| | - M V Eiden
- Section on Directed Gene Transfer, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - P Timms
- Genecology Research Center, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, 4556, Queensland, Australia.
| |
Collapse
|
33
|
Respiratory syncytial virus prefusogenic fusion (F) protein nanoparticle vaccine: Structure, antigenic profile, immunogenicity, and protection. Vaccine 2019; 37:6112-6124. [PMID: 31416644 DOI: 10.1016/j.vaccine.2019.07.089] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 07/06/2019] [Accepted: 07/26/2019] [Indexed: 12/15/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in the very young, elderly, and immunocompromised for which there is no vaccine. The surface exposed RSV fusion (F) glycoprotein is required for membrane fusion and infection and is a desirable vaccine candidate. RSV F glycoprotein structure is dynamic and undergoes significant rearrangements during virus assembly, fusion, and infection. We have previously described an RSV fusion-inactive prefusogenic F with a mutation of one of two furin cleavage sites resulting in the p27 region on the N-terminus of F1 with a truncated fusion peptide covalently linked to F2. A processing intermediate RSV prefusogenic F has been reported in infected cells, purified F, budded virus, and elicited a strong immune response against p27 in RSV infected young children. In this report, we demonstrate that prefusogenic F, when expressed on the cell surface of Sf9 insect and human 293T cells, binds monoclonal antibodies (mAbs) that target prefusion-specific antigenic sites Ø and VIII, and mAbs targeting epitopes common to pre- and postfusion F sites II and IV. Purified prefusogenic F bound prefusion F specific mAbs to antigenic sites Ø and VIII and mAbs targeting pre- and postfusion sites II, IV, and p27. Mice immunized with prefusogenic F antigen produced significantly higher levels of anti-F IgG and RSV neutralizing antibodies than prefusion or postfusion F antigens and induced antibodies competitive with mAbs to sites Ø, VIII, II, and IV. RSV prefusogenic F neutralization antibody responses were enhanced with aluminum phosphate adjuvant and significantly higher than prefusion F. Prefusogenic F vaccine protected cotton rats against upper and lower respiratory tract infection by RSV/A. For the first time, we present the structure, antigenic profile, immunogenicity, and protective efficacy of RSV prefusogenic F nanoparticle vaccine.
Collapse
|
34
|
Boyoglu-Barnum S, Chirkova T, Anderson LJ. Biology of Infection and Disease Pathogenesis to Guide RSV Vaccine Development. Front Immunol 2019; 10:1675. [PMID: 31402910 PMCID: PMC6677153 DOI: 10.3389/fimmu.2019.01675] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in young children and a substantial contributor to respiratory tract disease throughout life and as such a high priority for vaccine development. However, after nearly 60 years of research no vaccine is yet available. The challenges to developing an RSV vaccine include the young age, 2-4 months of age, for the peak of disease, the enhanced RSV disease associated with the first RSV vaccine, formalin-inactivated RSV with an alum adjuvant (FI-RSV), and difficulty achieving protection as illustrated by repeat infections with disease that occur throughout life. Understanding the biology of infection and disease pathogenesis has and will continue to guide vaccine development. In this paper, we review the roles that RSV proteins play in the biology of infection and disease pathogenesis and the corresponding contribution to live attenuated and subunit RSV vaccines. Each of RSV's 11 proteins are in the design of one or more vaccines. The G protein's contribution to disease pathogenesis through altering host immune responses as well as its role in the biology of infection suggest it can make a unique contribution to an RSV vaccine, both live attenuated and subunit vaccines. One of G's potential unique contributions to a vaccine is the potential for anti-G immunity to have an anti-inflammatory effect independent of virus replication. Though an anti-viral effect is essential to an effective RSV vaccine, it is important to remember that the goal of a vaccine is to prevent disease. Thus, other effects of the infection, such as G's alteration of the host immune response may provide opportunities to induce responses that block this effect and improve an RSV vaccine. Keeping in mind the goal of a vaccine is to prevent disease and not virus replication may help identify new strategies for other vaccine challenges, such as improving influenza vaccines and developing HIV vaccines.
Collapse
Affiliation(s)
| | - Tatiana Chirkova
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Larry J. Anderson
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
35
|
Walsh EE, Mariani TJ, Chu C, Grier A, Gill SR, Qiu X, Wang L, Holden-Wiltse J, Corbett A, Thakar J, Benoodt L, McCall MN, Topham DJ, Falsey AR, Caserta MT. Aims, Study Design, and Enrollment Results From the Assessing Predictors of Infant Respiratory Syncytial Virus Effects and Severity Study. JMIR Res Protoc 2019; 8:e12907. [PMID: 31199303 PMCID: PMC6595944 DOI: 10.2196/12907] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The majority of infants hospitalized with primary respiratory syncytial virus (RSV) infection have no obvious risk factors for severe disease. OBJECTIVE The aim of this study (Assessing Predictors of Infant RSV Effects and Severity, AsPIRES) was to identify factors associated with severe disease in full-term healthy infants younger than 10 months with primary RSV infection. METHODS RSV infected infants were enrolled from 3 cohorts during consecutive winters from August 2012 to April 2016 in Rochester, New York. A birth cohort was prospectively enrolled and followed through their first winter for development of RSV infection. An outpatient supplemental cohort was enrolled in the emergency department or pediatric offices, and a hospital cohort was enrolled on admission with RSV infection. RSV was diagnosed by reverse transcriptase-polymerase chain reaction. Demographic and clinical data were recorded and samples collected for assays: buccal swab (cytomegalovirus polymerase chain reaction, PCR), nasal swab (RSV qualitative PCR, complete viral gene sequence, 16S ribosomal ribonucleic acid [RNA] amplicon microbiota analysis), nasal wash (chemokine and cytokine assays), nasal brush (nasal respiratory epithelial cell gene expression using RNA sequencing [RNAseq]), and 2 to 3 ml of heparinized blood (flow cytometry, RNAseq analysis of purified cluster of differentiation [CD]4+, CD8+, B cells and natural killer cells, and RSV-specific antibody). Cord blood (RSV-specific antibody) was also collected for the birth cohort. Univariate and multivariate logistic regression will be used for analysis of data using a continuous Global Respiratory Severity Score (GRSS) as the outcome variable. Novel statistical methods will be developed for integration of the large complex datasets. RESULTS A total of 453 infants were enrolled into the 3 cohorts; 226 in the birth cohort, 60 in the supplemental cohort, and 78 in the hospital cohort. A total of 126 birth cohort infants remained in the study and were evaluated for 150 respiratory illnesses. Of the 60 RSV positive infants in the supplemental cohort, 42 completed the study, whereas all 78 of the RSV positive hospital cohort infants completed the study. A GRSS was calculated for each RSV-infected infant and is being used to analyze each of the complex datasets by correlation with disease severity in univariate and multivariate methods. CONCLUSIONS The AsPIRES study will provide insights into the complex pathogenesis of RSV infection in healthy full-term infants with primary RSV infection. The analysis will allow assessment of multiple factors potentially influencing the severity of RSV infection including the level of RSV specific antibodies, the innate immune response of nasal epithelial cells, the adaptive response by various lymphocyte subsets, the resident airway microbiota, and viral factors. Results of this study will inform disease interventions such as vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Edward E Walsh
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Thomas J Mariani
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - ChinYi Chu
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Alex Grier
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Steven R Gill
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Xing Qiu
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Lu Wang
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Jeanne Holden-Wiltse
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Anthony Corbett
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Juilee Thakar
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Lauren Benoodt
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Matthew N McCall
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - David J Topham
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Ann R Falsey
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Mary T Caserta
- University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
36
|
Hijano DR, Vu LD, Kauvar LM, Tripp RA, Polack FP, Cormier SA. Role of Type I Interferon (IFN) in the Respiratory Syncytial Virus (RSV) Immune Response and Disease Severity. Front Immunol 2019; 10:566. [PMID: 30972063 PMCID: PMC6443902 DOI: 10.3389/fimmu.2019.00566] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract disease in children <2 years of age. Increased morbidity and mortality have been reported in high-risk patients, such as premature infants, patients with cardiac disease, and severely immune compromised patients. Severe disease is associated with the virulence of the virus as well as host factors specifically including the innate immune response. The role of type I interferons (IFNs) in the response to RSV infection is important in regulating the rate of virus clearance and in directing the character of the immune response, which is normally associated with protection and less severe disease. Two RSV non-structural proteins, NS1 and NS2, as well as the envelope G glycoprotein are known to suppress type I IFN production and a robust type I IFN response to RSV does not occur in human infants or neonatal mouse models of RSV infection. Additionally, presence of type I IFNs are associated with mild symptoms in infants and administration of IFN-α prior to infection of neonatal mice with RSV reduces immunopathology. This evidence has driven RSV prophylaxis and therapeutic efforts to consider strategies for enhancing type I IFN production.
Collapse
Affiliation(s)
- Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Luan D Vu
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | | - Ralph A Tripp
- Department of Infectious Disease, University of Georgia, Athens, GA, United States
| | | | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
37
|
Epitope-Specific Serological Assays for RSV: Conformation Matters. Vaccines (Basel) 2019; 7:vaccines7010023. [PMID: 30813394 PMCID: PMC6466065 DOI: 10.3390/vaccines7010023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes substantial morbidity and mortality in children and older adults. An effective vaccine must elicit neutralizing antibodies targeting the RSV fusion (F) protein, which exists in two major conformations, pre-fusion (pre-F) and post-fusion (post-F). Although 50% of the surface is shared, pre-F contains highly neutralization-sensitive antigenic sites not present on post-F. Recent advancement of several subunit F-based vaccine trials has spurred interest in quantifying and understanding the protective potential of antibodies directed to individual antigenic sites. Monoclonal antibody competition ELISAs are being used to measure these endpoints, but the impact of F conformation and competition from antibodies binding to adjacent antigenic sites has not been thoroughly investigated. Since this information is critical for interpreting clinical trial outcomes and defining serological correlates of protection, we optimized assays to evaluate D25-competing antibodies (DCA) to antigenic site Ø on pre-F, and compared readouts of palivizumab-competing antibodies (PCA) to site II on both pre-F and post-F. We show that antibodies to adjacent antigenic sites can contribute to DCA and PCA readouts, and that cross-competition from non-targeted sites is especially confounding when PCA is measured using a post-F substrate. While measuring DCA and PCA levels may be useful to delineate the role of antibodies targeting the apex and side of the F protein, respectively, the assay limitations and caveats should be considered when conducting immune monitoring during vaccine trials and defining correlates of protection.
Collapse
|
38
|
Hijano DR, Maron G, Hayden RT. Respiratory Viral Infections in Patients With Cancer or Undergoing Hematopoietic Cell Transplant. Front Microbiol 2018; 9:3097. [PMID: 30619176 PMCID: PMC6299032 DOI: 10.3389/fmicb.2018.03097] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/29/2018] [Indexed: 12/25/2022] Open
Abstract
Survival rates for pediatric cancer have steadily improved over time but it remains a significant cause of morbidity and mortality among children. Infections are a major complication of cancer and its treatment. Community acquired respiratory viral infections (CRV) in these patients increase morbidity, mortality and can lead to delay in chemotherapy. These are the result of infections with a heterogeneous group of viruses including RNA viruses, such as respiratory syncytial virus (RSV), influenza virus (IV), parainfluenza virus (PIV), metapneumovirus (HMPV), rhinovirus (RhV), and coronavirus (CoV). These infections maintain a similar seasonal pattern to those of immunocompetent patients. Clinical manifestations vary significantly depending on the type of virus and the type and degree of immunosuppression, ranging from asymptomatic or mild disease to rapidly progressive fatal pneumonia Infections in this population are characterized by a high rate of progression from upper to lower respiratory tract infection and prolonged viral shedding. Use of corticosteroids and immunosuppressive therapy are risk factors for severe disease. The clinical course is often difficult to predict, and clinical signs are unreliable. Accurate prognostic viral and immune markers, which have become part of the standard of care for systemic viral infections, are currently lacking; and management of CRV infections remains controversial. Defining effective prophylactic and therapeutic strategies is challenging, especially considering, the spectrum of immunocompromised patients, the variety of respiratory viruses, and the presence of other opportunistic infections and medical problems. Prevention remains one of the most important strategies against these viruses. Early diagnosis, supportive care and antivirals at an early stage, when available and indicated, have proven beneficial. However, with the exception of neuraminidase inhibitors for influenza infection, there are no accepted treatments. In high-risk patients, pre-emptive treatment with antivirals for upper respiratory tract infection (URTI) to decrease progression to LRTI is a common strategy. In the future, viral load and immune markers may prove beneficial in predicting severe disease, supporting decision making and monitor treatment in this population.
Collapse
Affiliation(s)
- Diego R. Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Randall T. Hayden
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
39
|
Abstract
Respiratory syncytial virus (RSV) is the most common cause of infant hospitalization and causes a high burden of disease in the elderly, too. This enveloped negative-stranded RNA virus has been recently reclassified in the Pneumoviridae family. Infections of the respiratory cells happens when the two major surface glycoproteins, G and F, take contact with the cell receptor CX3CR1 and mediate entry by fusion, respectively. Viral mRNA transcription, genomic RNA synthesis and nucleocapsid formation occur in large cytoplasmic inclusion bodies to avoid recognition by the host innate immune response. Most progeny virions remain associated to the infected cell surface; fusion of infected with adjacent cells results in the formation of large multinucleated syncytia that eventually undergo apoptosis. Desquamated epithelial cells form the plugs that with mucus and fibrin may cause lower airway obstructions. Pathogenetic mechanism of severe RSV disease likely involve both the extent of viral replication and the host immune response. Regarding the latter, single nucleotide polymorphism analysis and genome-wide association studies showed that genetic susceptibility to severe RSV infection is likely a complex trait, in which many different host genetic variants contribute. Recent studies pointed to the fact that bronchiolitis severity depends more on the specific infecting RSV genotypes than on the amount of viral loads. A population-based surveillance system to better define RSV burden of disease would be of valuable help for implementing future vaccination programs.
Collapse
Affiliation(s)
- Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy -
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|