1
|
Fuchs J, Hübner J, Schmidt A, Irrgang P, Maier C, Vieira Antão A, Oltmanns F, Thirion C, Lapuente D, Tenbusch M. Evaluation of adenoviral vector Ad19a encoding RSV-F as novel vaccine against respiratory syncytial virus. NPJ Vaccines 2024; 9:205. [PMID: 39472590 PMCID: PMC11522487 DOI: 10.1038/s41541-024-01001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants and toddlers. Since natural infections do not induce persistent immunity, there is the need of vaccines providing long-term protection. Here, we evaluated a new adenoviral vector (rAd) vaccine based on the rare serotype rAd19a and compared the immunogenicity and efficacy to the highly immunogenic rAd5. Given as an intranasal boost in DNA primed mice, both vectors encoding the F protein provided efficient protection against a subsequent RSV infection. However, intramuscular immunization with rAd19a vectors provoked vaccine-enhanced disease after RSV infection compared to non-vaccinated animals. While mucosal IgA antibodies and tissue-resident memory T-cells in intranasally vaccinated mice rapidly control RSV replication, a strong anamnestic systemic T-cell response in absence of local immunity might be the reason for immune-mediated enhanced disease. Our study highlighted the potential benefits of developing effective mucosal against respiratory pathogens.
Collapse
Affiliation(s)
- Jana Fuchs
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Julian Hübner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Anna Schmidt
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Pascal Irrgang
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Clara Maier
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Ana Vieira Antão
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Friederike Oltmanns
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany.
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054, Erlangen, Germany.
| |
Collapse
|
2
|
Rasool AT, Li E, Nazir A. Recent advances in natural products and derivatives with antiviral activity against respiratory syncytial virus (RSV). JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-24. [PMID: 39425923 DOI: 10.1080/10286020.2024.2417211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Respiratory syncytial virus (RSV) is a widespread viral infection that causes millions of high-risk illnesses annually. Medicinal herbs such as ginseng root, echinacea purpurea, and radix astragali have a positive effect on antiviral activity by preventing viral adhesion, syncytial development, inhibiting viral internalization, relieving respiratory inflammation, strengthening the immune system, and stimulating the release of interferons. The potential benefits of natural products in terms of lower costs, better patient outcomes, and fewer adverse effects are discussed. This review examines the current evidence on the prevention and control of RSV with natural ingredients and the challenges and opportunities in clinical practice.
Collapse
Affiliation(s)
- Ameena Tur Rasool
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu210093, China
| | - Erguang Li
- Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu210093, China
| | - Ahsan Nazir
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu210094, China
| |
Collapse
|
3
|
Grosse S, Cooymans L, Embrechts W, McGowan D, Jacoby E, Stoops B, Gupta K, Ackermann M, Alnajjar S, Guillemont J, Jin Z, Kesteleyn B, Matcha K, Sriboonyapirat P, Truong A, Van Den Berg J, Yu X, Herschke F, Roymans D, Raboisson P, Rigaux P, Jonckers THM. Discovery of gem-Dimethyl-hydroxymethylpyridine Derivatives as Potent Non-nucleoside RSV Polymerase Inhibitors. J Med Chem 2024; 67:13723-13736. [PMID: 39105710 DOI: 10.1021/acs.jmedchem.4c00525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Respiratory syncytial virus (RSV) is an RNA virus infecting the upper and lower respiratory tract and is recognized as a major respiratory health threat, particularly to older adults, immunocompromised individuals, and young children. Around 64 million children and adults are infected every year worldwide. Despite two vaccines and a new generation monoclonal antibody recently approved, no effective antiviral treatment is available. In this manuscript, we present the medicinal chemistry efforts resulting in the identification of compound 28 (JNJ-8003), a novel RSV non-nucleoside inhibitor displaying subnanomolar activity in vitro as well as prominent efficacy in mice and a neonatal lamb models.
Collapse
Affiliation(s)
- Sandrine Grosse
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Ludwig Cooymans
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Werner Embrechts
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | | | - Edgar Jacoby
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Bart Stoops
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Kusum Gupta
- Neuron23 Inc. 343 Oyster Point Blvd, South San Francisco, California 94080, United States
| | | | - Sarhad Alnajjar
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, U.K
| | | | - Zhinan Jin
- Janssen Pharmaceutica NV, Brisbane, California 94005, United States
| | - Bart Kesteleyn
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Kiran Matcha
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | | | - Anh Truong
- Neuron23 Inc. 343 Oyster Point Blvd, South San Francisco, California 94080, United States
| | - Joke Van Den Berg
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Xiaodi Yu
- Janssen Pharmaceutica NV, Spring House, Pennsylvania 19477 United States
| | - Florence Herschke
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Dirk Roymans
- DNS Life Sciences Consulting, Brandhoefstraat 63, 2300 Turnhout, Belgium
| | - Pierre Raboisson
- Galapagos, General De Wittelaan L112, A3, 2800 Mechelen, Belgium
| | - Peter Rigaux
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Tim H M Jonckers
- Janssen Research & Development, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| |
Collapse
|
4
|
Drury RE, Camara S, Chelysheva I, Bibi S, Sanders K, Felle S, Emary K, Phillips D, Voysey M, Ferreira DM, Klenerman P, Gilbert SC, Lambe T, Pollard AJ, O'Connor D. Multi-omics analysis reveals COVID-19 vaccine induced attenuation of inflammatory responses during breakthrough disease. Nat Commun 2024; 15:3402. [PMID: 38649734 PMCID: PMC11035709 DOI: 10.1038/s41467-024-47463-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
The immune mechanisms mediating COVID-19 vaccine attenuation of COVID-19 remain undescribed. We conducted comprehensive analyses detailing immune responses to SARS-CoV-2 virus in blood post-vaccination with ChAdOx1 nCoV-19 or a placebo. Samples from randomised placebo-controlled trials (NCT04324606 and NCT04400838) were taken at baseline, onset of COVID-19-like symptoms, and 7 days later, confirming COVID-19 using nucleic amplification test (NAAT test) via real-time PCR (RT-PCR). Serum cytokines were measured with multiplexed immunoassays. The transcriptome was analysed with long, short and small RNA sequencing. We found attenuation of RNA inflammatory signatures in ChAdOx1 nCoV-19 compared with placebo vaccinees and reduced levels of serum proteins associated with COVID-19 severity. KREMEN1, a putative alternative SARS-CoV-2 receptor, was downregulated in placebo compared with ChAdOx1 nCoV-19 vaccinees. Vaccination ameliorates reductions in cell counts across leukocyte populations and platelets noted at COVID-19 onset, without inducing potentially deleterious Th2-skewed immune responses. Multi-omics integration links a global reduction in miRNA expression at COVID-19 onset to increased pro-inflammatory responses at the mRNA level. This study reveals insights into the role of COVID-19 vaccines in mitigating disease severity by abrogating pro-inflammatory responses associated with severe COVID-19, affirming vaccine-mediated benefit in breakthrough infection, and highlighting the importance of clinically relevant endpoints in vaccine evaluation.
Collapse
Affiliation(s)
- Ruth E Drury
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Susana Camara
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Irina Chelysheva
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katherine Sanders
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Salle Felle
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katherine Emary
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Daniel Phillips
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Daniela M Ferreira
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Paul Klenerman
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Peter Medawar Building for Pathogen Research, Nuffield Dept. of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
5
|
Sun BW, Zhang PP, Wang ZH, Yao X, He ML, Bai RT, Che H, Lin J, Xie T, Hui Z, Ye XY, Wang LW. Prevention and Potential Treatment Strategies for Respiratory Syncytial Virus. Molecules 2024; 29:598. [PMID: 38338343 PMCID: PMC10856762 DOI: 10.3390/molecules29030598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a significant viral pathogen that causes respiratory infections in infants, the elderly, and immunocompromised individuals. RSV-related illnesses impose a substantial economic burden worldwide annually. The molecular structure, function, and in vivo interaction mechanisms of RSV have received more comprehensive attention in recent times, and significant progress has been made in developing inhibitors targeting various stages of the RSV replication cycle. These include fusion inhibitors, RSV polymerase inhibitors, and nucleoprotein inhibitors, as well as FDA-approved RSV prophylactic drugs palivizumab and nirsevimab. The research community is hopeful that these developments might provide easier access to knowledge and might spark new ideas for research programs.
Collapse
Affiliation(s)
- Bo-Wen Sun
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Peng-Peng Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zong-Hao Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xia Yao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Meng-Lan He
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Rui-Ting Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Che
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jing Lin
- Drug Discovery, Hangzhou Haolu Pharma Co., Hangzhou 311121, China;
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Li-Wei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (B.-W.S.); (P.-P.Z.); (Z.-H.W.); (X.Y.); (M.-L.H.); (R.-T.B.); (H.C.); (T.X.); (Z.H.)
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
6
|
Thimmiraju SR, Kimata JT, Pollet J. Pseudoviruses, a safer toolbox for vaccine development against enveloped viruses. Expert Rev Vaccines 2024; 23:174-185. [PMID: 38164690 DOI: 10.1080/14760584.2023.2299380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Pseudoviruses are recombinant, replication-incompetent, viral particles designed to mimic the surface characteristics of native enveloped viruses. They are a safer, and cost-effective research alternative to live viruses. With the potential emergence of the next major infectious disease, more vaccine scientists must become familiar with the pseudovirus platform as a vaccine development tool to mitigate future outbreaks. AREAS COVERED This review aims at vaccine developers to provide a basic understanding of pseudoviruses, list their production methods, and discuss their utility to assess vaccine efficacy against enveloped viral pathogens. We further illustrate their usefulness as wet-lab simulators for emerging mutant variants, and new viruses to help prepare for current and future viral outbreaks, minimizing the need for gain-of-function experiments with highly infectious or lethal enveloped viruses. EXPERT OPINION With this platform, researchers can better understand the role of virus-receptor interactions and entry in infections, prepare for dangerous mutations, and develop effective vaccines.
Collapse
Affiliation(s)
- Syamala R Thimmiraju
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
7
|
Proaño A, Flannery DD. EBNEO Commentary: Respiratory syncytial virus vaccine in pregnancy for infant protection. Acta Paediatr 2023; 112:2449-2450. [PMID: 37608492 DOI: 10.1111/apa.16950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Affiliation(s)
- Alvaro Proaño
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
8
|
Pokharel SM, Mohanty I, Mariasoosai C, Miura TA, Maddison LA, Natesan S, Bose S. Human beta defensin-3 mediated activation of β-catenin during human respiratory syncytial virus infection: interaction of HBD3 with LDL receptor-related protein 5. Front Microbiol 2023; 14:1186510. [PMID: 37426017 PMCID: PMC10324619 DOI: 10.3389/fmicb.2023.1186510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is a non-segmented negative-sense RNA virus belonging to the paramyxovirus family. RSV infects the respiratory tract to cause pneumonia and bronchiolitis in infants, elderly, and immunocompromised patients. Effective clinical therapeutic options and vaccines to combat RSV infection are still lacking. Therefore, to develop effective therapeutic interventions, it is imperative to understand virus-host interactions during RSV infection. Cytoplasmic stabilization of β-catenin protein results in activation of canonical Wingless (Wnt)/β-catenin signaling pathway that culminates in transcriptional activation of various genes regulated by T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors. This pathway is involved in various biological and physiological functions. Our study shows RSV infection of human lung epithelial A549 cells triggering β-catenin protein stabilization and induction of β-catenin mediated transcriptional activity. Functionally, the activated β-catenin pathway promoted a pro-inflammatory response during RSV infection of lung epithelial cells. Studies with β-catenin inhibitors and A549 cells lacking optimal β-catenin activity demonstrated a significant loss of pro-inflammatory chemokine interleukin-8 (IL-8) release from RSV-infected cells. Mechanistically, our studies revealed a role of extracellular human beta defensin-3 (HBD3) in interacting with cell surface Wnt receptor LDL receptor-related protein-5 (LRP5) to activate the non-canonical Wnt independent β-catenin pathway during RSV infection. We showed gene expression and release of HBD3 from RSV-infected cells and silencing of HBD3 expression resulted in reduced stabilization of β-catenin protein during RSV infection. Furthermore, we observed the binding of extracellular HBD3 with cell surface localized LRP5 protein, and our in silico and protein-protein interaction studies have highlighted a direct interaction of HBD3 with LRP5. Thus, our studies have identified the β-catenin pathway as a key regulator of pro-inflammatory response during RSV infection of human lung epithelial cells. This pathway was induced during RSV infection via a non-canonical Wnt-independent mechanism involving paracrine/autocrine action of extracellular HBD3 activating cell surface Wnt receptor complex by directly interacting with the LRP5 receptor.
Collapse
Affiliation(s)
- Swechha M. Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Indira Mohanty
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Charles Mariasoosai
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Tanya A. Miura
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Lisette A. Maddison
- Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Senthil Natesan
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| |
Collapse
|
9
|
Moin AT, Ullah MA, Patil RB, Faruqui NA, Araf Y, Das S, Uddin KMK, Hossain MS, Miah MF, Moni MA, Chowdhury DUS, Islam S. A computational approach to design a polyvalent vaccine against human respiratory syncytial virus. Sci Rep 2023; 13:9702. [PMID: 37322049 PMCID: PMC10272159 DOI: 10.1038/s41598-023-35309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Human Respiratory Syncytial Virus (RSV) is one of the leading causes of lower respiratory tract infections (LRTI), responsible for infecting people from all age groups-a majority of which comprises infants and children. Primarily, severe RSV infections are accountable for multitudes of deaths worldwide, predominantly of children, every year. Despite several efforts to develop a vaccine against RSV as a potential countermeasure, there has been no approved or licensed vaccine available yet, to control the RSV infection effectively. Therefore, through the utilization of immunoinformatics tools, a computational approach was taken in this study, to design a multi-epitope polyvalent vaccine against two major antigenic subtypes of RSV, RSV-A and RSV-B. Potential predictions of the T-cell and B-cell epitopes were followed by extensive tests of antigenicity, allergenicity, toxicity, conservancy, homology to human proteome, transmembrane topology, and cytokine-inducing ability. The peptide vaccine was modeled, refined, and validated. Molecular docking analysis with specific Toll-like receptors (TLRs) revealed excellent interactions with suitable global binding energies. Additionally, molecular dynamics (MD) simulation ensured the stability of the docking interactions between the vaccine and TLRs. Mechanistic approaches to imitate and predict the potential immune response generated by the administration of vaccines were determined through immune simulations. Subsequent mass production of the vaccine peptide was evaluated; however, there remains a necessity for further in vitro and in vivo experiments to validate its efficacy against RSV infections.
Collapse
Affiliation(s)
- Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh.
| | - Md Asad Ullah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | - Rajesh B Patil
- Department of Pharmaceutical Chemistry, Sinhgad Technical Education Society's, Sinhgad College of Pharmacy, Pune, Maharashtra, India
| | - Nairita Ahsan Faruqui
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Sowmen Das
- Department of Computer Science and Engineering, School of Physical Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Khaza Md Kapil Uddin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Md Shakhawat Hossain
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh
| | - Md Faruque Miah
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Mohammad Ali Moni
- Bone Biology Division, The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- WHO Collaborating Centre on eHealth, UNSW Digital Health, School of Public Health and Community Medicine, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Artificial Intelligence and Data Science, Faculty of Health and Behavioural Sciences, School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | - Dil Umme Salma Chowdhury
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, Bangladesh.
| | - Saiful Islam
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram Laboratories, Chattogram, Bangladesh.
| |
Collapse
|
10
|
De C, Pickles RJ, Yao W, Liao B, Boone A, Choi M, Battaglia DM, Askin FB, Whitmire JK, Silvestri G, Garcia JV, Wahl A. Human T cells efficiently control RSV infection. JCI Insight 2023; 8:e168110. [PMID: 37159271 PMCID: PMC10393221 DOI: 10.1172/jci.insight.168110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection causes significant morbidity and mortality in infants, immunocompromised individuals, and older individuals. There is an urgent need for effective antivirals and vaccines for high-risk individuals. We used 2 complementary in vivo models to analyze RSV-associated human lung pathology and human immune correlates of protection. RSV infection resulted in widespread human lung epithelial damage, a proinflammatory innate immune response, and elicited a natural adaptive human immune response that conferred protective immunity. We demonstrated a key role for human T cells in controlling RSV infection. Specifically, primed human CD8+ T cells or CD4+ T cells effectively and independently control RSV replication in human lung tissue in the absence of an RSV-specific antibody response. These preclinical data support the development of RSV vaccines, which also elicit effective T cell responses to improve RSV vaccine efficacy.
Collapse
Affiliation(s)
- Chandrav De
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Raymond J. Pickles
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wenbo Yao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Baolin Liao
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
- Department of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Allison Boone
- Department of Microbiology and Immunology, and
- Marsico Lung Institute, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mingyu Choi
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Diana M. Battaglia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | | | - Jason K. Whitmire
- Department of Microbiology and Immunology, and
- Department of Genetics, and
- Lineberger Comprehensive Cancer Center, UNC at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J. Victor Garcia
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| | - Angela Wahl
- International Center for the Advancement of Translational Science
- Division of Infectious Diseases, Department of Medicine
- Center for AIDS Research
| |
Collapse
|
11
|
Anandhan G, Narkhede YB, Mohan M, Paramasivam P. Immunoinformatics aided approach for predicting potent cytotoxic T cell epitopes of respiratory syncytial virus. J Biomol Struct Dyn 2023; 41:12093-12105. [PMID: 36935101 DOI: 10.1080/07391102.2023.2191136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/27/2022] [Indexed: 03/21/2023]
Abstract
Respiratory syncytial virus (RSV) is an infectious viral pathogen that causing serious respiratory infection in adults and neonates. The only approved therapies for RSV are the monoclonal antibodies palivizumab and its derivative motavizumab. Both treatments are expensive and require a hospital setting for administration. A vaccine represents a safe, effective and cheaper alternative for preventing RSV infection. In silico prediction methods have proven to be valuable in speeding up the process of vaccine design. In this study, reverse vaccinology methods were used to predict the cytotoxic T lymphocytes (CTL) epitopes from the entire proteome of RSV strain A. From amongst 3402 predicted binders to 12 high frequency alleles from the Immune Epitope Database (IEDB), 567 had positive processing scores while 327 epitopes were predicted to be immunogenic. A thorough examination of the 327 epitopes for possible antigenicity, allergenicity and toxicity resulted in 95 epitopes with desirable properties. A BLASTp analysis revealed 94 unique and non-homologous epitopes that were subjected to molecular docking across the 12 high frequency alleles. The final dataset of 70 epitopes contained 13 experimentally proven and 57 unique epitopes from a total of 11 RSV proteins. From our findings on selected T-cell-specific RSV antigen epitopes, notably the four epitopes confirmed to exhibit stable binding by molecular dynamics. The prediction pipeline used in this study represents an effective way to screen the immunogenic epitopes from other pathogens.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gayathri Anandhan
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Manikandan Mohan
- College of Pharmacy, University of Georgia, Athens, USA
- Vaxigen International Research Center, Coimbatore, Tamil Nadu, India
| | - Premasudha Paramasivam
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
12
|
Bartsch YC, Cizmeci D, Kang J, Zohar T, Periasamy S, Mehta N, Tolboom J, Van der Fits L, Sadoff J, Comeaux C, Callendret B, Bukreyev A, Lauffenburger DA, Bastian AR, Alter G. Antibody effector functions are associated with protection from respiratory syncytial virus. Cell 2022; 185:4873-4886.e10. [PMID: 36513064 DOI: 10.1016/j.cell.2022.11.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 08/29/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022]
Abstract
Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.
Collapse
Affiliation(s)
- Yannic C Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tomer Zohar
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sivakumar Periasamy
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nickita Mehta
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeroen Tolboom
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | | | - Jerry Sadoff
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | - Christy Comeaux
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | | | - Alexander Bukreyev
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
Soto JA, Galvez NMS, Rivera DB, Díaz FE, Riedel CA, Bueno SM, Kalergis AM. From animal studies into clinical trials: the relevance of animal models to develop vaccines and therapies to reduce disease severity and prevent hRSV infection. Expert Opin Drug Discov 2022; 17:1237-1259. [PMID: 36093605 DOI: 10.1080/17460441.2022.2123468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Human respiratory syncytial virus (hRSV) is an important cause of lower respiratory tract infections in the pediatric and the geriatric population worldwide. There is a substantial economic burden resulting from hRSV disease during winter. Although no vaccines have been approved for human use, prophylactic therapies are available for high-risk populations. Choosing the proper animal models to evaluate different vaccine prototypes or pharmacological treatments is essential for developing efficient therapies against hRSV. AREAS COVERED This article describes the relevance of using different animal models to evaluate the effect of antiviral drugs, pharmacological molecules, vaccine prototypes, and antibodies in the protection against hRSV. The animal models covered are rodents, mustelids, bovines, and nonhuman primates. Animals included were chosen based on the available literature and their role in the development of the drugs discussed in this manuscript. EXPERT OPINION Choosing the correct animal model is critical for exploring and testing treatments that could decrease the impact of hRSV in high-risk populations. Mice will continue to be the most used preclinical model to evaluate this. However, researchers must also explore the use of other models such as nonhuman primates, as they are more similar to humans, prior to escalating into clinical trials.
Collapse
Affiliation(s)
- J A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - N M S Galvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D B Rivera
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - S M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
14
|
Ahuja R, Shelly A, Meena J, Panda AK. Microparticles entrapping pneumococcal protein SP0845 show improved immunogenicity and temperature stability. Int J Biol Macromol 2022; 203:661-670. [PMID: 35120939 DOI: 10.1016/j.ijbiomac.2022.01.175] [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: 09/19/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
Protein based vaccines are the most safe and affordable strategy to combat pneumococcal disease circumventing the limitations of conventional polysaccharide-based vaccines like serotype dependence, high cost and inability to be administered to immunocompromised. SP0845 is a highly conserved vaccine candidate shown to provide protection against heterologous strains of Streptococcus pneumoniae, the primal cause of pneumonia. However, the associated poor immunogenicity warrants the need for adjuvants and multiple doses to mount desired responses. The present study relates to improve the immunogenicity of pneumococcal protein SP0845 by use of poly lactic acid biodegradable polymer microparticles. The immunization studies showed that microparticles elicited higher antibody response compared to alum adjuvanted protein and this immunopotentiation was achieved without the use of any additional adjuvant. They were also capable of eliciting secondary antibody response upon boosting after four months. Further, the particles upon storage at 25 and 37 °C for one month were still capable of mounting an immune response equivalent to those stored in cold chain. Thus, using microparticles entrapping SP0845 for immunization not only improve the immunogenicity but also offer better temperature stability. This can greatly reduce the cost and increase access of protein-based vaccine to resource limited settings.
Collapse
Affiliation(s)
- Rahul Ahuja
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Asha Shelly
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Jairam Meena
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Amulya K Panda
- Product Development Cell, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
15
|
Protein and Peptide Substances in the Treatment of Respiratory Syncytial Infection: Current State. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072263. [PMID: 35408661 PMCID: PMC9000545 DOI: 10.3390/molecules27072263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 12/05/2022]
Abstract
Respiratory syncytial virus infection (RSVI) is an acute medical and social problem in many countries globally. Infection is most dangerous for infants under one year old and the elderly. Despite its epidemiological relevance, only two drugs are registered for clinical use against RSVI: ribavirin (approved in a limited number of countries due to side effects) and palivizumab (Synagis), which is intended only for the prevention, but not the treatment, of infection. Currently, various research groups are searching for new drugs against RSV, with three main areas of research: small molecules, polymeric drugs (proteins and peptides), and plant extracts. This review is devoted to currently developed protein and peptide anti-RSV drugs.
Collapse
|
16
|
Andreano E, Paciello I, Bardelli M, Tavarini S, Sammicheli C, Frigimelica E, Guidotti S, Torricelli G, Biancucci M, D’Oro U, Chandramouli S, Bottomley MJ, Rappuoli R, Finco O, Buricchi F. The respiratory syncytial virus (RSV) prefusion F-protein functional antibody repertoire in adult healthy donors. EMBO Mol Med 2021; 13:e14035. [PMID: 33998144 PMCID: PMC8185550 DOI: 10.15252/emmm.202114035] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/27/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of death from lower respiratory tract infection in infants and children, and is responsible for considerable morbidity and mortality in older adults. Vaccines for pregnant women and elderly which are in phase III clinical studies target people with pre-existing natural immunity against RSV. To investigate the background immunity which will be impacted by vaccination, we single cell-sorted human memory B cells and dissected functional and genetic features of neutralizing antibodies (nAbs) induced by natural infection. Most nAbs recognized both the prefusion and postfusion conformations of the RSV F-protein (cross-binders) while a smaller fraction bound exclusively to the prefusion conformation. Cross-binder nAbs used a wide array of gene rearrangements, while preF-binder nAbs derived mostly from the expansion of B-cell clonotypes from the IGHV1 germline. This latter class of nAbs recognizes an epitope located between Site Ø, Site II, and Site V on the F-protein, identifying an important site of pathogen vulnerability.
Collapse
Affiliation(s)
- Emanuele Andreano
- Department of Life SciencesUniversity of SienaSienaItaly
- GSK VaccinesSienaItaly
- Present address:
Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | - Ida Paciello
- GSK VaccinesSienaItaly
- Present address:
Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | | | | | | | | | | | | | | | | | - Sumana Chandramouli
- GSK VaccinesRockvilleMDUSA
- Present address:
Moderna Therapeutics IncCambridgeMAUSA
| | | | - Rino Rappuoli
- GSK VaccinesSienaItaly
- Faculty of MedicineImperial CollegeLondonUK
- Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | | | | |
Collapse
|
17
|
Díaz FE, Guerra-Maupome M, McDonald PO, Rivera-Pérez D, Kalergis AM, McGill JL. A Recombinant BCG Vaccine Is Safe and Immunogenic in Neonatal Calves and Reduces the Clinical Disease Caused by the Respiratory Syncytial Virus. Front Immunol 2021; 12:664212. [PMID: 33981309 PMCID: PMC8108697 DOI: 10.3389/fimmu.2021.664212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) constitutes a major health burden, causing millions of hospitalizations in children under five years old worldwide due to acute lower respiratory tract infections. Despite decades of research, licensed vaccines to prevent hRSV are not available. Development of vaccines against hRSV targeting young infants requires ruling out potential vaccine-enhanced disease presentations. To achieve this goal, vaccine testing in proper animal models is essential. A recombinant BCG vaccine that expresses the Nucleoprotein of hRSV (rBCG-N-hRSV) protects mice against hRSV infection, eliciting humoral and cellular immune protection. Further, this vaccine was shown to be safe and immunogenic in human adult volunteers. Here, we evaluated the safety, immunogenicity, and protective efficacy of the rBCG-N-hRSV vaccine in a neonatal bovine RSV calf infection model. Newborn, colostrum-replete Holstein calves were either vaccinated with rBCG-N-hRSV, WT-BCG, or left unvaccinated, and then inoculated via aerosol challenge with bRSV strain 375. Vaccination with rBCG-N-hRSV was safe and well-tolerated, with no systemic adverse effects. There was no evidence of vaccine-enhanced disease following bRSV challenge of rBCG-N-hRSV vaccinated animals, suggesting that the vaccine is safe for use in neonates. Vaccination increased virus-specific IgA and virus-neutralization activity in nasal fluid and increased the proliferation of virus- and BCG-specific CD4+ and CD8+ T cells in PBMCs and lymph nodes at 7dpi. Furthermore, rBCG-N-hRSV vaccinated calves developed reduced clinical disease as compared to unvaccinated control calves, although neither pathology nor viral burden were significantly reduced in the lungs. These results suggest that the rBCG-N-hRSV vaccine is safe in neonatal calves and induces protective humoral and cellular immunity against this respiratory virus. These data from a newborn animal model provide further support to the notion that this vaccine approach could be considered as a candidate for infant immunization against RSV.
Collapse
Affiliation(s)
- Fabián E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Paiton O McDonald
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Daniela Rivera-Pérez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Hamza A, Shafat Z, Parray ZA, Hisamuddin M, Khan WH, Ahmed A, Almajhdi FN, Farrag MA, Mohammed AA, Islam A, Parveen S. Structural Characterization and Binding Studies of the Ectodomain G Protein of Respiratory Syncytial Virus Reveal the Crucial Role of pH with Possible Implications in Host-Pathogen Interactions. ACS OMEGA 2021; 6:10403-10414. [PMID: 34056193 PMCID: PMC8153753 DOI: 10.1021/acsomega.1c00800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/25/2021] [Indexed: 06/12/2023]
Abstract
Respiratory syncytial virus (RSV) is a leading viral pathogen causing acute lower respiratory tract infection in children. The G protein of RSV is involved in attachment with the host cell. It is a neutralizing antigen and thus a vaccine candidate. Heparan sulfate is a type of glycosaminoglycan (GAG) present on the host cell membrane that is involved in attachment with the G protein of RSV. We describe a novel approach for efficient expression and purification of the ectodomain G protein in the prokaryotic system and its biophysical characterization. The native ectodomain G protein was purified using a two-step process by Ni-NTA and DEAE weak anion-exchange chromatography through the supernatant obtained after cell lysis. In addition, the denatured form of the protein was also purified from the solubilized inclusion bodies (IBs) by Ni-NTA affinity chromatography with a higher yield. Dynamic light scattering (DLS) was performed to confirm the homogeneity of the purified protein. The effect of pH on the stability and structure of the purified protein was studied by circular dichroism (CD), fluorescence, and absorbance spectroscopy techniques. Isothermal titration calorimetry (ITC) and microscale thermophoresis (MST) were exploited to demonstrate the interaction of heparan sulfate with the ectodomain G protein. The dynamic light scattering results showed that the purified protein was homogenic and had a well-folded native conformation. Biophysical characterization of the protein revealed that it was stable and had intact secondary and tertiary structures at pH 7.5. CD analysis revealed that the protein showed a loss in the secondary structure at pH values 5.5 and 3.5, while absorbance spectroscopy suggested a stable tertiary structure at pH values 7.5 and 5.5 with a probable aggregation pattern at pH 3.5. This loss in the structure of the ectodomain G protein at low pH can be correlated with its physiological activity. A slight change in pH might play a crucial role in host-pathogen interactions. The fluorescence intensity of the protein decreased on moving toward a lower pH with no spectral shift in emission maxima. In addition, isothermal titration calorimetry and microscale thermophoresis results showed strong binding affinity of the ectodomain G protein with heparan sulfate. The binding of heparan sulfate with protein was probably due to the electrostatic interaction of positively charged amino acid residues of the heparin-binding domain of the protein and the negatively charged group of GAGs. Future studies may involve the development of possible therapeutic agents interacting with the G protein and affecting the overall charge and pH that might hinder the host-pathogen interaction.
Collapse
Affiliation(s)
- Abu Hamza
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Zoya Shafat
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Zahoor Ahmad Parray
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Malik Hisamuddin
- Interdisciplinary
Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Wajihul Hasan Khan
- Kusuma
School of Biological Sciences, Indian Institute
of Technology Delhi, New Delhi 110016, India
| | - Anwar Ahmed
- Centre
of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fahad N. Almajhdi
- Department
of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
- Centre
of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed A. Farrag
- Department
of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Arif Ahmed Mohammed
- Centre
of Excellence in Biotechnology Research, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Asimul Islam
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shama Parveen
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
19
|
Goswami R, O’Hagan DT, Adamo R, Baudner BC. Conjugation of Mannans to Enhance the Potency of Liposome Nanoparticles for the Delivery of RNA Vaccines. Pharmaceutics 2021; 13:pharmaceutics13020240. [PMID: 33572332 PMCID: PMC7916126 DOI: 10.3390/pharmaceutics13020240] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022] Open
Abstract
Recent approval of mRNA vaccines to combat COVID-19 have highlighted the potential of this platform. Lipid nanoparticles (LNP) is the delivery vehicle of choice for mRNA as they prevent its enzymatic degradation by encapsulation. We have recently shown that surface exposition of mannose, incorporated in LNPs as stable cholesterol-amine conjugate, enhances the potency of self-amplifying RNA (SAM) replicon vaccines through augmented uptake by antigen presenting cells (APCs). Here, we generated a new set of LNPs whose surface was modified with mannans of different length (from mono to tetrasaccharide), in order to study the effect on antibody response of model SAM replicon encoding for the respiratory syncytial virus fusion F protein. Furthermore, the impact of the mannosylated liposomal delivery through intradermal as well as intramuscular routes was investigated. The vaccine priming response showed to improve consistently with increase in the chain length of mannoses; however, the booster dose response plateaued above the length of disaccharide. An increase in levels of IgG1 and IgG2a was observed for mannnosylated lipid nanoparticles (MLNPs) as compared to LNPs. This work confirms the potential of mannosylated SAM LNPs for both intramuscular and intradermal delivery, and highlights a disaccharide length as sufficient to ensure improved immunogenicity compared to the un-glycosylated delivery system.
Collapse
Affiliation(s)
- Roshan Goswami
- mAbxience, Julia Morros s/n, Armunia, 24009 León, Spain;
- GSK, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Roberto Adamo
- GSK, Via Fiorentina 1, 53100 Siena, Italy
- Correspondence: (R.A.); (B.C.B.)
| | - Barbara C. Baudner
- GSK, Via Fiorentina 1, 53100 Siena, Italy
- Correspondence: (R.A.); (B.C.B.)
| |
Collapse
|
20
|
Mustfa SA, Maurizi E, McGrath J, Chiappini C. Nanomedicine Approaches to Negotiate Local Biobarriers for Topical Drug Delivery. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Salman Ahmad Mustfa
- Centre for Craniofacial and Regenerative Biology King's College London London SE1 9RT UK
| | - Eleonora Maurizi
- Dipartimento di Medicina e Chirurgia Università di Parma Parma 43121 Italy
| | - John McGrath
- St John's Institute of Dermatology King's College London London SE1 9RT UK
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology King's College London London SE1 9RT UK
- London Centre for Nanotechnology King's College London London WC2R 2LS UK
| |
Collapse
|
21
|
Brouwers TJ, Van der Zeijst BA. Vaccine Production, Safety, and Efficacy. ENCYCLOPEDIA OF VIROLOGY 2021. [PMCID: PMC7917445 DOI: 10.1016/b978-0-12-814515-9.00121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Vaccination was a key factor in reducing viral diseases after the transition of a nomadic lifestyle to a sedentary society. Without vaccination, our highly urbanized society would not be sustainable. Vaccine development is an, increasingly, long and expensive process. This poses severe problems for the development of vaccines against new emerging viral diseases. Therefore, interest to speed up development timelines has increased considerably. Clinical evaluation of protection and safety and licensing take up most of the time. But also building scalable production facilities require time and money. This article describes the traditional approach to vaccine R&D and possible improvements, particularly towards being better preparared for emerging viral diseases. It was written just before the covid-19 outbreeak in 2020. We have added a final paragraph on the development of covid-19 vaccines.
Collapse
|
22
|
Harshbarger W, Tian S, Wahome N, Balsaraf A, Bhattacharya D, Jiang D, Pandey R, Tungare K, Friedrich K, Mehzabeen N, Biancucci M, Chinchilla-Olszar D, Mallett CP, Huang Y, Wang Z, Bottomley MJ, Malito E, Chandramouli S. Convergent structural features of respiratory syncytial virus neutralizing antibodies and plasticity of the site V epitope on prefusion F. PLoS Pathog 2020; 16:e1008943. [PMID: 33137810 PMCID: PMC7660905 DOI: 10.1371/journal.ppat.1008943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 11/12/2020] [Accepted: 08/28/2020] [Indexed: 11/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a global public health burden for which no licensed vaccine exists. To aid vaccine development via increased understanding of the protective antibody response to RSV prefusion glycoprotein F (PreF), we performed structural and functional studies using the human neutralizing antibody (nAb) RSB1. The crystal structure of PreF complexed with RSB1 reveals a conformational, pre-fusion specific site V epitope with a unique cross-protomer binding mechanism. We identify shared structural features between nAbs RSB1 and CR9501, elucidating for the first time how diverse germlines obtained from different subjects can develop convergent molecular mechanisms for recognition of the same PreF site of vulnerability. Importantly, RSB1-like nAbs were induced upon immunization with PreF in naturally-primed cattle. Together, this work reveals new details underlying the immunogenicity of site V and further supports PreF-based vaccine development efforts. Respiratory syncytial virus (RSV) is a persistent, contagious seasonal pathogen and a serious public health threat. While infants are the most at-risk population, with infections potentially leading to bronchiolitis, adults, especially the elderly, are also burdened by RSV-induced respiratory infections. The only treatment currently available for RSV is passive immunization for high-risk infants. Thus, there is a critical need to develop a vaccine for the vast majority of the vulnerable population for which there is no preventative treatment. The RSV fusion protein in its prefusion form (PreF) is the target of the majority of naturally-induced neutralizing antibodies, and several clinical trials are currently evaluating PreF as a promising vaccine candidate. In this study, we solved the X-ray structure of PreF bound to the Fab fragment of a human neutralizing antibody. The structure reveals plasticity of the epitope, as well as a unique molecular signature for antibodies elicited towards this region of PreF. We also find that similar antibodies are induced upon immunization of naturally-primed cattle with a PreF vaccine antigen, suggesting that this epitope is highly immunogenic. These results will help us better understand the human immune response to RSV infection and vaccination, and guide future vaccine-design efforts.
Collapse
Affiliation(s)
| | - Sai Tian
- GSK, Rockville, MD, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | - Ying Huang
- GSK, Rockville, MD, United States of America
| | - Zihao Wang
- GSK, Rockville, MD, United States of America
| | | | - Enrico Malito
- GSK, Rockville, MD, United States of America
- * E-mail: (EM); (SC)
| | | |
Collapse
|
23
|
Sadoff J, De Paepe E, Haazen W, Omoruyi E, Bastian AR, Comeaux C, Heijnen E, Strout C, Schuitemaker H, Callendret B. Safety and Immunogenicity of the Ad26.RSV.preF Investigational Vaccine Coadministered With an Influenza Vaccine in Older Adults. J Infect Dis 2020; 223:699-708. [DOI: 10.1093/infdis/jiaa409] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
Abstract
Background
Respiratory syncytial virus (RSV) and influenza cause significant disease burden in older adults. Overlapping RSV and influenza seasonality presents the opportunity to coadminister vaccines for both infections. This study assessed coadministration of the investigational vaccine, Ad26.RSV.preF, an adenovirus serotype 26 (Ad26) vector encoding RSV F protein stabilized in its prefusion conformation (pre-F), with a seasonal influenza vaccine in older adults.
Methods
In this phase 2a, double-blind, placebo-controlled study, 180 adults aged ≥60 years received Ad26.RSV.preF plus Fluarix on day 1 and placebo on day 29, or placebo plus Fluarix on day 1 and Ad26.RSV.preF on day 29 (control).
Results
The coadministration regimen had an acceptable tolerability profile. Reactogenicity was generally higher after Ad26.RSV.preF versus Fluarix, but symptoms were generally transient and mild or moderate. At 28 days after the first vaccination, the upper confidence intervals of the hemagglutination inhibition antibody geometric mean ratio (control/coadministration) for all influenza strains were <2, demonstrating noninferiority. Robust neutralizing and binding antibody responses to RSV A2 were observed in both groups.
Conclusions
Coadministration of Fluarix with Ad26.RSV.preF vaccine had an acceptable safety profile and showed no evidence of interference in immune response. The results are compatible with simultaneous seasonal vaccination with both vaccines.
Clinical Trials Registration
NCT03339713.
Collapse
Affiliation(s)
- Jerald Sadoff
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | | | | | | | | | | | - Cynthia Strout
- Coastal Carolina Research Center, Mount Pleasant, South Carolina
| | | | | |
Collapse
|
24
|
Bedient L, Pokharel SM, Chiok KR, Mohanty I, Beach SS, Miura TA, Bose S. Lytic Cell Death Mechanisms in Human Respiratory Syncytial Virus-Infected Macrophages: Roles of Pyroptosis and Necroptosis. Viruses 2020; 12:v12090932. [PMID: 32854254 PMCID: PMC7552060 DOI: 10.3390/v12090932] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is the most common cause of viral bronchiolitis and pneumonia in infants and children worldwide. Inflammation induced by RSV infection is responsible for its hallmark manifestation of bronchiolitis and pneumonia. The cellular debris created through lytic cell death of infected cells is a potent initiator of this inflammation. Macrophages are known to play a pivotal role in the early innate immune and inflammatory response to viral pathogens. However, the lytic cell death mechanisms associated with RSV infection in macrophages remains unknown. Two distinct mechanisms involved in lytic cell death are pyroptosis and necroptosis. Our studies revealed that RSV induces lytic cell death in macrophages via both of these mechanisms, specifically through the ASC (Apoptosis-associated speck like protein containing a caspase recruitment domain)-NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome activation of both caspase-1 dependent pyroptosis and receptor-interacting serine/threonine-protein kinase 3 (RIPK3), as well as a mixed lineage kinase domain like pseudokinase (MLKL)-dependent necroptosis. In addition, we demonstrated an important role of reactive oxygen species (ROS) during lytic cell death of RSV-infected macrophages.
Collapse
Affiliation(s)
- Lori Bedient
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Swechha Mainali Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Kim R. Chiok
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Indira Mohanty
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
| | - Sierra S. Beach
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA; (S.S.B.); (T.A.M.)
| | - Tanya A. Miura
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA; (S.S.B.); (T.A.M.)
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; (L.B.); (S.M.P.); (K.R.C.); (I.M.)
- Correspondence:
| |
Collapse
|
25
|
Sebina I, Phipps S. The Contribution of Neutrophils to the Pathogenesis of RSV Bronchiolitis. Viruses 2020; 12:E808. [PMID: 32726921 PMCID: PMC7472258 DOI: 10.3390/v12080808] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Acute viral bronchiolitis causes significant mortality in the developing world, is the number one cause of infant hospitalisation in the developed world, and is associated with the later development of chronic lung diseases such as asthma. A vaccine against respiratory syncytial virus (RSV), the leading cause of viral bronchiolitis in infancy, remains elusive, and hence new therapeutic modalities are needed to limit disease severity. However, much remains unknown about the underlying pathogenic mechanisms. Neutrophilic inflammation is the predominant phenotype observed in infants with both mild and severe disease, however, a clear understanding of the beneficial and deleterious effects of neutrophils is lacking. In this review, we describe the multifaceted roles of neutrophils in host defence and antiviral immunity, consider their contribution to bronchiolitis pathogenesis, and discuss whether new approaches that target neutrophil effector functions will be suitable for treating severe RSV bronchiolitis.
Collapse
Affiliation(s)
- Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Australia;
| | | |
Collapse
|
26
|
Bhat R, Farrag MA, Almajhdi FN. Double-edged role of natural killer cells during RSV infection. Int Rev Immunol 2020; 39:233-244. [PMID: 32469615 DOI: 10.1080/08830185.2020.1770748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Natural killer cells play a vital role in the rejection of tumors and pathogen-infected cells. NK cells are indispensable in the early immune response against viral infections by directly targeting infected cells. Furthermore, NK cells influence adaptive immunity by driving virus-specific T-cell responses. Respiratory syncytial virus, a highly contagious virus that causes bronchiolitis, is the main reason for mortality in infants and elderly patients. RSV infection triggers both innate and adaptive immune responses. However, immunity against RSV is ephemeral due to the impaired development of immunological memory. The role of NK cells during RSV infection remains ambiguous. NK cells play a dual role in RSV infection; initially, their role is a protective one as they utilize their intrinsic cytotoxicity, followed by a detrimental one that induces lung injury due to the inhibition of antibody responses and the secretion of pro-inflammatory factors. Noteworthy, IFN-γ released from NK cells play a critical role in promoting a shift to adaptive responses and inhibiting antibody responses in neonates. Indeed, NK cells have a pro-inflammatory and inhibitory role rather than a cytotoxic one that contributes to the severity of the disease. Therapeutic options, including DNA-protein-based vaccines, synthetic peptides, and attenuated strains, are presently under tests. However, there is a need for effective strategies to augment NK cell activity and circumvent the pro-inflammatory activity to benefit the host. In this review, we focused on the role played by NK cells in the immune response and its outcome on the immunopathogenesis of RSV disease.
Collapse
Affiliation(s)
- Rauf Bhat
- Virology Research Group, Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Farrag
- Virology Research Group, Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fahad N Almajhdi
- Virology Research Group, Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Penkert RR, Hankins JS, Young NS, Hurwitz JL. Vaccine Design Informed by Virus-Induced Immunity. Viral Immunol 2020; 33:342-350. [PMID: 32366204 PMCID: PMC7247049 DOI: 10.1089/vim.2019.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
When an individual is exposed to a viral pathogen for the first time, the adaptive immune system is naive and cannot prevent virus replication. The consequence may be severe disease. At the same time, the host may rapidly generate a pathogen-specific immune response that will prevent disease if the virus is encountered again. Parvovirus B19 provides one such example. Children with sickle cell disease can experience life-threatening transient aplastic crisis when first exposed to parvovirus B19, but an effective immune response confers lifelong protection. We briefly examine the induction and benefits of virus-induced immunity. We focus on three human viruses for which there are no licensed vaccines (respiratory syncytial virus, human immunodeficiency virus type 1, and parvovirus B19) and consider how virus-induced immunity may inform successful vaccine design.
Collapse
Affiliation(s)
- Rhiannon R. Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jane S. Hankins
- Pathology Department, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
28
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Subversion of Host Cell Mitochondria by RSV to Favor Virus Production is Dependent on Inhibition of Mitochondrial Complex I and ROS Generation. Cells 2019; 8:cells8111417. [PMID: 31717900 PMCID: PMC6912631 DOI: 10.3390/cells8111417] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/01/2019] [Accepted: 11/06/2019] [Indexed: 12/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a key cause of severe respiratory infection in infants, immunosuppressed adults, and the elderly worldwide, but there is no licensed vaccine or effective, widely-available antiviral therapeutic. We recently reported staged redistribution of host cell mitochondria in RSV infected cells, which results in compromised respiratory activities and increased reactive oxygen species (ROS) generation. Here, bioenergetic measurements, mitochondrial redox-sensitive dye, and high-resolution quantitative imaging were performed, revealing for the first time that mitochondrial complex I is key to this effect on the host cell, whereby mitochondrial complex I subunit knock-out (KO) cells, with markedly decreased mitochondrial respiration, show elevated levels of RSV infectious virus production compared to wild-type cells or KO cells with re-expressed complex I subunits. This effect correlates strongly with elevated ROS generation in the KO cells compared to wild-type cells or retrovirus-rescued KO cells re-expressing complex I subunits. Strikingly, blocking mitochondrial ROS levels using the mitochondrial ROS scavenger, mitoquinone mesylate (MitoQ), inhibits RSV virus production, even in the KO cells. The results highlight RSV's unique ability to usurp host cell mitochondrial ROS to facilitate viral infection and reinforce the idea of MitoQ as a potential therapeutic for RSV.
Collapse
|
30
|
Schneider-Ohrum K, Snell Bennett A, Rajani GM, Hostetler L, Maynard SK, Lazzaro M, Cheng LI, O'Day T, Cayatte C. CD4 + T Cells Drive Lung Disease Enhancement Induced by Immunization with Suboptimal Doses of Respiratory Syncytial Virus Fusion Protein in the Mouse Model. J Virol 2019; 93:e00695-19. [PMID: 31092578 PMCID: PMC6639276 DOI: 10.1128/jvi.00695-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of seronegative children previously immunized with formalin-inactivated (FI) RSV has been associated with serious enhanced respiratory disease (ERD). The phenomenon was reproduced in the cotton rat and the mouse, and both preclinical models have been routinely used to evaluate the safety of new RSV vaccine candidates. More recently, we demonstrated that immunizations with suboptimal doses of the RSV fusion (F) antigen, in its post- or prefusion conformation, and in the presence of a Th1-biasing adjuvant, unexpectedly led to ERD in the cotton rat model. To assess if those observations are specific to the cotton rat and to elucidate the mechanism by which vaccination with low antigen doses can drive ERD post-RSV challenge, we evaluated RSV post-F antigen dose de-escalation in BALB/c mice in the presence of a Th1-biasing adjuvant. While decreasing antigen doses, we observed an increase in lung inflammation associated with an upregulation of proinflammatory cytokines. The amplitude of the lung histopathology was comparable to that of FI-RSV-induced ERD, confirming the observations made in the cotton rat. Importantly, depletion of CD4+ T cells prior to viral challenge completely abrogated ERD, preventing proinflammatory cytokine upregulation and the infiltration of T cells, neutrophils, eosinophils, and macrophages into the lung. Overall, low-antigen-dose-induced ERD resembles FI-RSV-induced ERD, except that the former appears in the absence of detectable levels of viral replication and in the context of a Th1-biased immune response. Taken together, our observations reinforce the recent concept that vaccines developed for RSV-naïve individuals should be systematically tested under suboptimal dosing conditions.IMPORTANCE RSV poses a significant health care burden and is the leading cause of serious lower-respiratory-tract infections in young children. A formalin-inactivated RSV vaccine developed in the 1960s not only showed a complete lack of efficacy against RSV infection but also induced severe lung disease enhancement in vaccinated children. Since then, establishing safety in preclinical models has been one of the major challenges to RSV vaccine development. We recently observed in the cotton rat model that suboptimal immunizations with RSV fusion protein could induce lung disease enhancement. In the present study, we extended suboptimal dosing evaluation to the mouse model. We confirmed the induction of lung disease enhancement by vaccinations with low antigen doses and dissected the associated immune mechanisms. Our results stress the need to evaluate suboptimal dosing for any new RSV vaccine candidate developed for seronegative infants.
Collapse
Affiliation(s)
| | - Angie Snell Bennett
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | - Leigh Hostetler
- Laboratory Animal Resources, MedImmune, Gaithersburg, Maryland, USA
| | - Sean K Maynard
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Michelle Lazzaro
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Lily I Cheng
- Pathology Department, MedImmune, Gaithersburg, Maryland, USA
| | - Terrence O'Day
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, USA
| | - Corinne Cayatte
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| |
Collapse
|
31
|
Hu M, Schulze KE, Ghildyal R, Henstridge DC, Kolanowski JL, New EJ, Hong Y, Hsu AC, Hansbro PM, Wark PA, Bogoyevitch MA, Jans DA. Respiratory syncytial virus co-opts host mitochondrial function to favour infectious virus production. eLife 2019; 8:42448. [PMID: 31246170 PMCID: PMC6598784 DOI: 10.7554/elife.42448] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 06/10/2019] [Indexed: 12/15/2022] Open
Abstract
Although respiratory syncytial virus (RSV) is responsible for more human deaths each year than influenza, its pathogenic mechanisms are poorly understood. Here high-resolution quantitative imaging, bioenergetics measurements and mitochondrial membrane potential- and redox-sensitive dyes are used to define RSV’s impact on host mitochondria for the first time, delineating RSV-induced microtubule/dynein-dependent mitochondrial perinuclear clustering, and translocation towards the microtubule-organizing centre. These changes are concomitant with impaired mitochondrial respiration, loss of mitochondrial membrane potential and increased production of mitochondrial reactive oxygen species (ROS). Strikingly, agents that target microtubule integrity the dynein motor protein, or inhibit mitochondrial ROS production strongly suppresses RSV virus production, including in a mouse model with concomitantly reduced virus-induced lung inflammation. The results establish RSV’s unique ability to co-opt host cell mitochondria to facilitate viral infection, revealing the RSV-mitochondrial interface for the first time as a viable target for therapeutic intervention.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Keith E Schulze
- Monash Micro Imaging, Monash University, Melbourne, Australia
| | - Reena Ghildyal
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | | | | | - Elizabeth J New
- School of Chemistry, The University of Sydney, Sydney, Australia
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Peter Ab Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| |
Collapse
|
32
|
Qiu X, Duvvuri VR, Bahl J. Computational Approaches and Challenges to Developing Universal Influenza Vaccines. Vaccines (Basel) 2019; 7:E45. [PMID: 31141933 PMCID: PMC6631137 DOI: 10.3390/vaccines7020045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/15/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022] Open
Abstract
The traditional design of effective vaccines for rapidly-evolving pathogens, such as influenza A virus, has failed to provide broad spectrum and long-lasting protection. With low cost whole genome sequencing technology and powerful computing capabilities, novel computational approaches have demonstrated the potential to facilitate the design of a universal influenza vaccine. However, few studies have integrated computational optimization in the design and discovery of new vaccines. Understanding the potential of computational vaccine design is necessary before these approaches can be implemented on a broad scale. This review summarizes some promising computational approaches under current development, including computationally optimized broadly reactive antigens with consensus sequences, phylogenetic model-based ancestral sequence reconstruction, and immunomics to compute conserved cross-reactive T-cell epitopes. Interactions between virus-host-environment determine the evolvability of the influenza population. We propose that with the development of novel technologies that allow the integration of data sources such as protein structural modeling, host antibody repertoire analysis and advanced phylodynamic modeling, computational approaches will be crucial for the development of a long-lasting universal influenza vaccine. Taken together, computational approaches are powerful and promising tools for the development of a universal influenza vaccine with durable and broad protection.
Collapse
Affiliation(s)
- Xueting Qiu
- Center for Ecology of Infectious Diseases, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| | - Venkata R Duvvuri
- Center for Ecology of Infectious Diseases, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
| | - Justin Bahl
- Center for Ecology of Infectious Diseases, Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, GA 30606, USA.
- Duke-NUS Graduate Medical School, Singapore 169857, Singapore.
| |
Collapse
|
33
|
Altamirano-Lagos MJ, Díaz FE, Mansilla MA, Rivera-Pérez D, Soto D, McGill JL, Vasquez AE, Kalergis AM. Current Animal Models for Understanding the Pathology Caused by the Respiratory Syncytial Virus. Front Microbiol 2019; 10:873. [PMID: 31130923 PMCID: PMC6510261 DOI: 10.3389/fmicb.2019.00873] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the main etiologic agent of severe lower respiratory tract infections that affect young children throughout the world, associated with significant morbidity and mortality, becoming a serious public health problem globally. Up to date, no licensed vaccines are available to prevent severe hRSV-induced disease, and the generation of safe-effective vaccines has been a challenging task, requiring constant biomedical research aimed to overcome this ailment. Among the difficulties presented by the study of this pathogen, it arises the fact that there is no single animal model that resembles all aspects of the human pathology, which is due to the specificity that this pathogen has for the human host. Thus, for the study of hRSV, different animal models might be employed, depending on the goal of the study. Of all the existing models, the murine model has been the most frequent model of choice for biomedical studies worldwide and has been of great importance at contributing to the development and understanding of vaccines and therapies against hRSV. The most notable use of the murine model is that it is very useful as a first approach in the development of vaccines or therapies such as monoclonal antibodies, suggesting in this way the direction that research could have in other preclinical models that have higher maintenance costs and more complex requirements in its management. However, several additional different models for studying hRSV, such as other rodents, mustelids, ruminants, and non-human primates, have been explored, offering advantages over the murine model. In this review, we discuss the various applications of animal models to the study of hRSV-induced disease and the advantages and disadvantages of each model, highlighting the potential of each model to elucidate different features of the pathology caused by the hRSV infection.
Collapse
Affiliation(s)
- María José Altamirano-Lagos
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián E. Díaz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Andrés Mansilla
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Abel E. Vasquez
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
34
|
Guerra-Maupome M, Palmer MV, McGill JL, Sacco RE. Utility of the Neonatal Calf Model for Testing Vaccines and Intervention Strategies for Use against Human RSV Infection. Vaccines (Basel) 2019; 7:vaccines7010007. [PMID: 30626099 PMCID: PMC6466205 DOI: 10.3390/vaccines7010007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of pediatric respiratory tract infections. It is estimated that two-thirds of infants are infected with RSV during the first year of life and it is one of the leading causes of death in this age group worldwide. Similarly, bovine RSV is a primary viral pathogen in cases of pneumonia in young calves and plays a significant role in bovine respiratory disease complex. Importantly, naturally occurring infection of calves with bovine RSV shares many features in common with human RSV infection. Herein, we update our current understanding of RSV infection in cattle, with particular focus on similarities between the calf and human infection, and the recent reports in which the neonatal calf has been employed for the development and testing of vaccines and therapeutics which may be applied to hRSV infection in humans.
Collapse
Affiliation(s)
- Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Mitchell V Palmer
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| |
Collapse
|
35
|
Khan IU, Huang J, Li X, Xie J, Zhu N. Nasal immunization with RSV F and G protein fragments conjugated to an M cell-targeting ligand induces an enhanced immune response and protection against RSV infection. Antiviral Res 2018; 159:95-103. [PMID: 30290196 DOI: 10.1016/j.antiviral.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 01/24/2023]
Abstract
Human respiratory syncytial virus (RSV) is a major paediatric health concern worldwide. The development of an effective and safe vaccine against RSV is urgently needed. As RSV infects via the mucosal surfaces, developing a nasal vaccine may offer protective benefits over alternative administration routes. In this study, we tested a recombinant protein FG-Gb1 as an intranasal vaccine candidate against RSV. FG-Gb1 consists of the core fragments of the RSV fusion (F) and attachment (G) proteins conjugated to an microfold (M) cell-specific ligand Gb-1. Intranasal immunization with FG-Gb1 induced efficient systemic and mucosal immune responses as measured by the level of antigen-specific antibodies, cytokine-secreting cells and antigen-specific lymphocyte proliferation after exposure to antigen. Moreover, intranasal immunization induced protective immunity against nasal challenge with RSV, which was confirmed by a lack of weight loss and by viral clearance after challenge. Collectively, we confirmed that a ligand capable of targeting the conjugated antigen to nasopharynx-associated lymphoid tissue (NALT) can be used as an effective nasal vaccine adjuvant to induce protective immunity against RSV infection. Moreover, FG-Gb1 may have promise as an RSV vaccine but requires further studies.
Collapse
Affiliation(s)
- Inam Ullah Khan
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | - Jiansheng Huang
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | - Xue Li
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | - Jun Xie
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, PR China.
| | - Naishuo Zhu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, PR China.
| |
Collapse
|
36
|
Turi KN, Romick-Rosendale L, Gebretsadik T, Watanabe M, Brunwasser S, Anderson LJ, Moore ML, Larkin EK, Peebles RS, Hartert TV. Using urine metabolomics to understand the pathogenesis of infant respiratory syncytial virus (RSV) infection and its role in childhood wheezing. Metabolomics 2018; 14:135. [PMID: 30830453 PMCID: PMC6557166 DOI: 10.1007/s11306-018-1431-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/21/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection in infants causes significant morbidity and is the strongest risk factor associated with asthma. Metabolites, which reflect the interactions between host cell and virus, provide an opportunity to identify the pathways that underlie severe infections and asthma development. OBJECTIVE To study metabolic profile differences between infants with RSV infection, and human rhinovirus (HRV) infection, and healthy infants. To compare infant metabolic differences between children who do and do not wheeze. METHODS In a term birth cohort, urine was collected while healthy and during acute viral respiratory infection with RSV and HRV. We used 1H-NMR to identify urinary metabolites. Multivariate and univariate statistics were used to discriminate metabolic profiles of infants with either RSV ARI, or HRV ARI, and healthy infants. Multivariable logistic regression was used to assess the association of urine metabolites with 1st-, 2nd-, and 3rd-year recurrent wheezing. RESULTS Several metabolites in nicotinate and nicotinamide metabolism pathways were down-regulated in infants with RSV infection compared to healthy controls. There were no significant differences in metabolite profiles between infants with RSV infection and infants with HRV Infection. Alanine was strongly associated with reduced risk of 1st-year wheezing (OR 0.18[0.0, 0.46]) and 2nd-year wheezing (OR 0.31[0.13, 0.73]), while 2-hydroxyisobutyric acid was associated with increased 3rd-year wheezing (OR 5.02[1.49, 16.93]) only among the RSV infected subset. CONCLUSION The metabolites associated with infant RSV infection and recurrent-wheezing are indicative of viral takeover of the cellular machinery and resources to enhance virulence, replication, and subversion of the host immune-response, highlighting metabolic pathways important in the pathogenesis of RSV infection and wheeze development.
Collapse
Affiliation(s)
- Kedir N Turi
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2525 West End Avenue, Suite 450, Nashville, TN, 37203, USA
| | - Lindsey Romick-Rosendale
- Department of Pathology, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tebeb Gebretsadik
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Miki Watanabe
- Department of Pathology, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Steven Brunwasser
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2525 West End Avenue, Suite 450, Nashville, TN, 37203, USA
| | | | - Martin L Moore
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Emma K Larkin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2525 West End Avenue, Suite 450, Nashville, TN, 37203, USA
| | - Ray Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2525 West End Avenue, Suite 450, Nashville, TN, 37203, USA
| | - Tina V Hartert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2525 West End Avenue, Suite 450, Nashville, TN, 37203, USA.
| |
Collapse
|
37
|
Vaccination with RSV M 209-223 peptide promotes a protective immune response associated with reduced pulmonary inflammation. Antiviral Res 2018; 157:102-110. [DOI: 10.1016/j.antiviral.2018.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/29/2022]
|
38
|
Anderson AJ, Snelling TL, Moore HC, Blyth CC. Advances in Vaccines to Prevent Viral Respiratory Illnesses in Children. Paediatr Drugs 2017; 19:523-531. [PMID: 28808938 DOI: 10.1007/s40272-017-0257-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Childhood vaccination has played a critical role in the reduction of morbidity and mortality from communicable diseases, including specific respiratory pathogens. Acute lower respiratory infection (ALRI) of both bacterial and viral aetiology continues to impact global child health. Key bacterial pathogens including Streptococcus pneumoniae and Haemophilus influenza type b are specifically targeted with current vaccination programmes, while at present there are less effective strategies for the prevention of viral disease. Influenza vaccines, including both live attenuated intranasal vaccines and inactivated influenza vaccines, are limited by seasonal strain variation and unsustained immunity. Research into the development of a universal influenza vaccine is ongoing; potential targets are the conserved regions of the virus such as the M2e antigen and hemagglutinin stalk. Respiratory syncytial virus (RSV) and parainfluenza virus 3 (PIV3) are the viral pathogens most commonly causing ALRI in children, particularly the infant population. Currently, no vaccine exists for either virus. Over the last decade, promising advances have been made. Protection of neonates via maternal RSV immunisation is being assessed in a phase III clinical trial, with many other candidates for RSV and PIV3 at less advanced stages of development.
Collapse
Affiliation(s)
- Aleisha J Anderson
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia
| | - Tom L Snelling
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Hannah C Moore
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia
| | - Christopher C Blyth
- Department of Infectious Diseases, Princess Margaret Hospital, Roberts Rd, Subiaco, WA, Australia. .,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, Roberts Rd, Subiaco, WA, Australia. .,Discipline of Paediatrics, School of Medicine, Princess Margaret Hospital, University of Western Australia, Roberts Rd, Subiaco, WA, Australia. .,PathWest Laboratory Medicine WA, Department of Microbiology, QEII Medical Centre, Nedlands, WA, Australia.
| |
Collapse
|
39
|
Radke L, Sandig G, Lubitz A, Schließer U, von Horsten HH, Blanchard V, Keil K, Sandig V, Giese C, Hummel M, Hinderlich S, Frohme M. In Vitro Evaluation of Glycoengineered RSV-F in the Human Artificial Lymph Node Reactor. Bioengineering (Basel) 2017; 4:bioengineering4030070. [PMID: 28952549 PMCID: PMC5615316 DOI: 10.3390/bioengineering4030070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 11/16/2022] Open
Abstract
Subunit vaccines often require adjuvants to elicit sustained immune activity. Here, a method is described to evaluate the efficacy of single vaccine candidates in the preclinical stage based on cytokine and gene expression analysis. As a model, the recombinant human respiratory syncytial virus (RSV) fusion protein (RSV-F) was produced in CHO cells. For comparison, wild-type and glycoengineered, afucosylated RSV-F were established. Both glycoprotein vaccines were tested in a commercial Human Artificial Lymph Node in vitro model (HuALN®). The analysis of six key cytokines in cell culture supernatants showed well-balanced immune responses for the afucosylated RSV-F, while immune response of wild-type RSV-F was more Th1 accentuated. In particular, stronger and specific secretion of interleukin-4 after each round of re-stimulation underlined higher potency and efficacy of the afucosylated vaccine candidate. Comprehensive gene expression analysis by nCounter gene expression assay confirmed the stronger onset of the immunologic reaction in stimulation experiments with the afucosylated vaccine in comparison to wild-type RSV-F and particularly revealed prominent activation of Th17 related genes, innate immunity, and comprehensive activation of humoral immunity. We, therefore, show that our method is suited to distinguish the potency of two vaccine candidates with minor structural differences.
Collapse
Affiliation(s)
- Lars Radke
- Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, Wildau 15745, Germany.
- Institute of Pathology, Charitè-University Medicine Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Grit Sandig
- Laboratory of Biochemistry, Department of Life Sciences and Technology, Beuth University of Applied Sciences, Seestraße 64, Berlin 13347, Germany.
| | - Annika Lubitz
- ProBioGen AG, Goethestraße 54, Berlin 13086, Germany.
| | | | - Hans Henning von Horsten
- Department of Life Science Engineering, HTW Berlin University of Applied Sciences, Wilhelminenhofstraße 75a, Berlin 12459, Germany.
| | - Veronique Blanchard
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité Medical University Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Karolin Keil
- Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, Wildau 15745, Germany.
| | - Volker Sandig
- ProBioGen AG, Goethestraße 54, Berlin 13086, Germany.
| | | | - Michael Hummel
- Institute of Pathology, Charitè-University Medicine Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Stephan Hinderlich
- Laboratory of Biochemistry, Department of Life Sciences and Technology, Beuth University of Applied Sciences, Seestraße 64, Berlin 13347, Germany.
| | - Marcus Frohme
- Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, Wildau 15745, Germany.
| |
Collapse
|
40
|
CD4 + T cells support establishment of RSV-specific IgG and IgA antibody secreting cells in the upper and lower murine respiratory tract following RSV infection. Vaccine 2017; 35:2617-2621. [PMID: 28410812 DOI: 10.1016/j.vaccine.2017.03.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 02/28/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022]
Abstract
The RSV vaccine field suffered a major set-back when children were vaccinated with a formalin-inactivated RSV vaccine (FI-RSV). Unexpectedly, the vaccinated children fared worse than unvaccinated children when they were naturally infected with RSV. Mouse models were then developed that implicated the CD4+ T helper cell population as a contributor to adverse events. Today, the T cell is viewed with much caution in the RSV field, and its induction by vaccination is sometimes discouraged. Here we re-emphasize the beneficial role of the CD4+ T cell. Experiments were performed with RSV-infected nude mice that received CD4+ T cells by adoptive transfer. Data demonstrated that CD4+ T cells were necessary for the induction of mucosal and systemic RSV-specific antibodies, for the establishment of RSV-specific IgG and IgA antibody secreting cells in the upper and lower respiratory tract, and for RSV clearance.
Collapse
|
41
|
Immunization with Low Doses of Recombinant Postfusion or Prefusion Respiratory Syncytial Virus F Primes for Vaccine-Enhanced Disease in the Cotton Rat Model Independently of the Presence of a Th1-Biasing (GLA-SE) or Th2-Biasing (Alum) Adjuvant. J Virol 2017; 91:JVI.02180-16. [PMID: 28148790 DOI: 10.1128/jvi.02180-16] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of children previously immunized with a nonlive, formalin-inactivated (FI)-RSV vaccine has been associated with serious enhanced respiratory disease (ERD). Consequently, detailed studies of potential ERD are a critical step in the development of nonlive RSV vaccines targeting RSV-naive children and infants. The fusion glycoprotein (F) of RSV in either its postfusion (post-F) or prefusion (pre-F) conformation is a target for neutralizing antibodies and therefore an attractive antigen candidate for a pediatric RSV subunit vaccine. Here, we report the evaluation of RSV post-F and pre-F in combination with glucopyranosyl lipid A (GLA) integrated into stable emulsion (SE) (GLA-SE) and alum adjuvants in the cotton rat model. Immunization with optimal doses of RSV F antigens in the presence of GLA-SE induced high titers of virus-neutralizing antibodies and conferred complete lung protection from virus challenge, with no ERD signs in the form of alveolitis. To mimic a waning immune response, and to assess priming for ERD under suboptimal conditions, an antigen dose de-escalation study was performed in the presence of either GLA-SE or alum. At low RSV F doses, alveolitis-associated histopathology was unexpectedly observed with either adjuvant at levels comparable to FI-RSV-immunized controls. This occurred despite neutralizing-antibody titers above the minimum levels required for protection and with no/low virus replication in the lungs. These results emphasize the need to investigate a pediatric RSV vaccine candidate carefully for priming of ERD over a wide dose range, even in the presence of strong neutralizing activity, Th1 bias-inducing adjuvant, and protection from virus replication in the lower respiratory tract.IMPORTANCE RSV disease is of great importance worldwide, with the highest burden of serious disease occurring upon primary infection in infants and children. FI-RSV-induced enhanced disease, observed in the 1960s, presented a major and ongoing obstacle for the development of nonlive RSV vaccine candidates. The findings presented here underscore the need to evaluate a nonlive RSV vaccine candidate during preclinical development over a wide dose range in the cotton rat RSV enhanced-disease model, as suboptimal dosing of several RSV F subunit vaccine candidates led to the priming for ERD. These observations are relevant to the validity of the cotton rat model itself and to safe development of nonlive RSV vaccines for seronegative infants and children.
Collapse
|
42
|
Flight W, Jones A. The diagnosis and management of respiratory viral infections in cystic fibrosis. Expert Rev Respir Med 2017; 11:221-227. [PMID: 28132571 DOI: 10.1080/17476348.2017.1288102] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Respiratory viruses, such as those that cause influenza and the common cold, are a regular feature of life for the entire human population. Among people with CF, these viruses are associated with prolonged respiratory illness and show a clear association with pulmonary exacerbations which in turn are associated with lung function decline and risk of death. Human rhinovirus is the most commonly encountered respiratory viral pathogen in CF although adenovirus, bocavirus, coronavirus, influenza, parainfluenza, metapneumovirus and respiratory syncytial virus are all also responsible for infections in this population. Areas covered: This article reviews the epidemiology, clinical impact and therapeutic options for respiratory virus infection in both children and adults with CF. Expert commentary: The management of CF to date has largely focused on airway clearance strategies, nutritional support and aggressive antibacterial therapy. We highlight the significant role that respiratory viruses play in CF lung disease and argue that these pathogens represent an under-exploited target in the battle to control patients' symptoms and disease progression.
Collapse
Affiliation(s)
- William Flight
- a Oxford Adult Cystic Fibrosis Centre, Oxford University Hospitals NHS Foundation Trust , Oxford , UK
| | - Andrew Jones
- b Manchester Adult Cystic Fibrosis Centre, University Hospital of South Manchester NHS Foundation Trust , Manchester , UK.,c Institute of Inflammation & Repair, University of Manchester , Manchester , UK
| |
Collapse
|
43
|
Oliveira-Santos M, Santos JA, Soares J, Dias A, Quaresma M. Influence of meteorological conditions on RSV infection in Portugal. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2016; 60:1807-1817. [PMID: 27059367 DOI: 10.1007/s00484-016-1168-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/29/2016] [Accepted: 04/03/2016] [Indexed: 06/05/2023]
Abstract
Acute viral bronchiolitis is a common cause for infant hospital admissions. Of all etiological agents, respiratory syncytial virus (RSV) is commonly the most frequent. The present study assesses relationships between atmospheric factors and RSV infections in under 3-year-old patients admitted to the Inpatient Paediatric Service of Vila Real (North of Portugal). For this purpose, (1) clinical files of children admitted with a diagnosis of acute bronchiolitis from September 2005 to December 2015 (>10 years) were scrutinised and (2) local daily temperature/precipitation series, as well as six weather types controlling meteorological conditions in Portugal, were used. Fifty-five percent of all 770 admitted children were effectively infected with a given virus, whilst 48 % (367) were RSV+, i.e. 87 % of virus-infected children were RSV+. The bulk of incidence is verified in the first year of age (82 %, 302), slightly higher in males. RSV outbreaks are typically from December to March, but important inter-annual variability is found in both magnitude and shape. Although no clear connections were found between monthly temperatures/precipitation and RSV outbreaks apart from seasonality, a linkage to wintertime cold spells is apparent on a daily basis. Anomalously low minimum temperatures from the day of admittance back to 10 days before are observed. This relationship is supported by anomalously high occurrences of the E and AA weather types over the same period, which usually trigger dry and cold weather. These findings highlight some predictability in the RSV occurrences, revealing potential for modelling and risk assessments.
Collapse
Affiliation(s)
- M Oliveira-Santos
- Department of Psychiatry and Mental Health, Centro Hospitalar de Trás-os-Montes e Alto Douro EPE, CHTMAD, Vila Real, Portugal.
| | - J A Santos
- Centre for the Research and Technology of Agro-environmental and Biological Sciences, CITAB, Universidade de Trás-os-Montes e Alto Douro, UTAD, Vila Real, Portugal
| | - J Soares
- Paediatric Department, Centro Hospitalar de Trás-os-Montes e Alto Douro EPE, CHTMAD, Vila Real, Portugal
| | - A Dias
- Paediatric Department, Centro Hospitalar de Trás-os-Montes e Alto Douro EPE, CHTMAD, Vila Real, Portugal
| | - M Quaresma
- Paediatric Department, Centro Hospitalar de Trás-os-Montes e Alto Douro EPE, CHTMAD, Vila Real, Portugal
| |
Collapse
|
44
|
Martinez EC, Garg R, Shrivastava P, Gomis S, van Drunen Littel-van den Hurk S. Intranasal treatment with a novel immunomodulator mediates innate immune protection against lethal pneumonia virus of mice. Antiviral Res 2016; 135:108-119. [PMID: 27771388 PMCID: PMC7126411 DOI: 10.1016/j.antiviral.2016.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections. P-I-P pre-treatment, consisting of poly(I:C), IDR peptide and PCEP, was tested in the context of PVM infection in mice. P-I-P confers complete protection against lethal PVM infection by reducing clinical signs and immunopathology. P-I-P minimizes viral titers in the lungs reduces the influx of neutrophils and eosinophils into the tissue. P-I-P induces early upregulation of genes involved in host defense without any observable adverse effects. Survivor mice were PVM negative, suggesting that P-I-P mediates the successfully clearance of the virus in vivo.
Collapse
Affiliation(s)
- Elisa C Martinez
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Ravendra Garg
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Pratima Shrivastava
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, Saskatchewan, 52 Campus Drive, S7N 5B4, Canada
| | - Sylvia van Drunen Littel-van den Hurk
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada.
| |
Collapse
|
45
|
Esposito S, Scarselli E, Lelii M, Scala A, Vitelli A, Capone S, Fornili M, Biganzoli E, Orenti A, Nicosia A, Cortese R, Principi N. Antibody response to respiratory syncytial virus infection in children <18 months old. Hum Vaccin Immunother 2016; 12:1700-6. [PMID: 26901128 DOI: 10.1080/21645515.2016.1145847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The development of a safe and effective respiratory syncytial virus (RSV) vaccine might be facilitated by knowledge of the natural immune response to this virus. The aims of this study were to evaluate the neutralizing antibody response of a cohort of healthy children <18 months old to RSV infection. During the RSV season, 89 healthy children <18 months old were enrolled and followed up weekly for 12 weeks. At each visit, a nasopharyngeal swab was obtained for RSV detection by real-time polymerase chain reaction (PCR). During the study period, 2 blood samples were drawn and they were used to determine RSV geometric mean neutralizing antibody titres (GMT) against RSV. A total of 35 (39.3%) children had RSV detected during the study period. Among RSV-positive patients, children ≥7 months showed a significantly higher increase in antibody response (p<0.001). A significantly higher number of patients with a ≥4 -fold increase in GMT were ≥7 months old (p = 0.02) and presented lower respiratory tract infections (LRTIs) during the study period (p = 0.01). Viral shedding was longer among children aged ≥7 months (p = 0.06), those with viral load ≥10(6) copies/mL (p = 0.03), and those with LRTIs during the study period (p = 0.03), but it was not associated with the immune response (p = 0.41). In conclusion, natural RSV infection seems to evoke a low immune response in younger children. To be effective in this infant population, which is at highest risk of developing severe LRTIs, vaccines must be able to induce in the first months of life a stronger immune response than that produced by the natural infection.
Collapse
Affiliation(s)
- Susanna Esposito
- a Paediatric Highly Intensive Care Unit , Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Elisa Scarselli
- b ReiThera Srl (formerly Okairos) , Viale Città d'Europa 679, Rome , Italy
| | - Mara Lelii
- a Paediatric Highly Intensive Care Unit , Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Alessia Scala
- a Paediatric Highly Intensive Care Unit , Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Alessandra Vitelli
- b ReiThera Srl (formerly Okairos) , Viale Città d'Europa 679, Rome , Italy
| | - Stefania Capone
- b ReiThera Srl (formerly Okairos) , Viale Città d'Europa 679, Rome , Italy
| | - Marco Fornili
- c Unit of Medical Statistics, Biometry and Bioinformatics "G.A. Maccacaro"; Università degli Studi di Milano, Fondazione IRCCS Istituto Nazionale Tumori , Milan , Italy
| | - Elia Biganzoli
- c Unit of Medical Statistics, Biometry and Bioinformatics "G.A. Maccacaro"; Università degli Studi di Milano, Fondazione IRCCS Istituto Nazionale Tumori , Milan , Italy
| | - Annalisa Orenti
- c Unit of Medical Statistics, Biometry and Bioinformatics "G.A. Maccacaro"; Università degli Studi di Milano, Fondazione IRCCS Istituto Nazionale Tumori , Milan , Italy
| | - Alfredo Nicosia
- b ReiThera Srl (formerly Okairos) , Viale Città d'Europa 679, Rome , Italy
| | | | - Nicola Principi
- a Paediatric Highly Intensive Care Unit , Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
46
|
Gu H, Li T, Han L, Zhu P, Zhang P, Zhang S, Sun S, Duan Y, Xing L, Zhao Z, Lai C, Wen B, Wang X, Yang P. Protection conferred by virus-like particle vaccines against respiratory syncytial virus infection in mice by intranasal vaccination. Hum Vaccin Immunother 2016; 11:1057-64. [PMID: 25933187 DOI: 10.1080/21645515.2015.1011993] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major pathogen in infants and the elderly, causing pneumonia and bronchiolitis. Despite decades of research, to date there is still no approved RSV vaccine available. In this study, we developed RSV virus-like particle (VLP) vaccines containing an RSV fusion (F) and/or attachment (G) protein with Newcastle disease virus (NDV) as the platform. The VLPs were expressed in a baculovirus system and purified by sucrose gradient centrifugation. BALB/c mice immunized intranasally (i.n.) with rNDV/RSV/F plus rNDV/RSV/G developed robust humoral, mucosal RSV-specific antibodies and cellular immune responses. Furthermore, rNDV/RSV/F plus rNDV/RSV/G provided better protection than did rNDV/RSV/F or rNDV/RSV/G alone, as shown by an obvious decrease in viral replication together with alleviation of histopathological changes in the lungs of the challenged mice. Our data demonstrate that the intranasal vaccination of combined RSV virus-like particle vaccine candidates has great potential for protection against RSV infection.
Collapse
Affiliation(s)
- Hongjing Gu
- a Beijing Institute of Microbiology and Epidemiology; State Key Laboratory of Pathogen and Biosecurity Beijing , China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Russell CJ, Hurwitz JL. Sendai virus as a backbone for vaccines against RSV and other human paramyxoviruses. Expert Rev Vaccines 2015; 15:189-200. [PMID: 26648515 DOI: 10.1586/14760584.2016.1114418] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human paramyxoviruses are the etiological agents for life-threatening respiratory virus infections of infants and young children. These viruses, including respiratory syncytial virus (RSV), the human parainfluenza viruses (hPIV1-4) and human metapneumovirus (hMPV), are responsible for millions of serious lower respiratory tract infections each year worldwide. There are currently no standard treatments and no licensed vaccines for any of these pathogens. Here we review research with which Sendai virus, a mouse parainfluenza virus type 1, is being advanced as a Jennerian vaccine for hPIV1 and as a backbone for RSV, hMPV and other hPIV vaccines for children.
Collapse
Affiliation(s)
- Charles J Russell
- a Department of Infectious Diseases , St. Jude Children's Research Hospital , Memphis , TN , USA.,b Department of Microbiology, Immunology and Biochemistry , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Julia L Hurwitz
- a Department of Infectious Diseases , St. Jude Children's Research Hospital , Memphis , TN , USA.,b Department of Microbiology, Immunology and Biochemistry , University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
48
|
Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35 expressing the respiratory syncytial virus (RSV) fusion protein induce protective immunity against RSV infection in cotton rats. Vaccine 2015; 33:5406-5414. [PMID: 26319741 DOI: 10.1016/j.vaccine.2015.08.056] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/12/2015] [Accepted: 08/13/2015] [Indexed: 11/22/2022]
Abstract
RSV is an important cause of lower respiratory tract infections in children, the elderly and in those with underlying medical conditions. Although the high disease burden indicates an urgent need for a vaccine against RSV, no licensed RSV vaccine is currently available. We developed an RSV vaccine candidate based on the low-seroprevalent human adenovirus serotypes 26 and 35 (Ad26 and Ad35) encoding the RSV fusion (F) gene. Single immunization of mice with either one of these vectors induced high titers of RSV neutralizing antibodies and high levels of F specific interferon-gamma-producing T cells. A Th1-type immune response was indicated by a high IgG2a/IgG1 ratio of RSV-specific antibodies, strong induction of RSV-specific interferon-gamma and tumor necrosis factor-alpha cytokine producing CD8 Tcells, and low RSV-specific CD4 T-cell induction. Both humoral and cellular responses were increased upon a boost with RSV-F expressing heterologous adenovirus vector (Ad35 boost after Ad26 prime or vice versa). Both single immunization and prime-boost immunization of cotton rats induced high and long-lasting RSV neutralizing antibody titers and protective immunity against lung and nasal RSV A2 virus load up to at least 30 weeks after immunization. Cotton rats were also completely protected against challenge with a RSV B strain (B15/97) after heterologous prime-boost immunization. Lungs from vaccinated animals showed minimal damage or inflammatory infiltrates post-challenge, in contrast to animals vaccinated with formalin-inactivated virus. Our results suggest that recombinant human adenoviral Ad26 and Ad35 vectors encoding the RSV F gene have the potential to provide broad and durable protection against RSV in humans, and appear safe to be investigated in infants.
Collapse
|
49
|
Taylor G, Thom M, Capone S, Pierantoni A, Guzman E, Herbert R, Scarselli E, Napolitano F, Giuliani A, Folgori A, Colloca S, Cortese R, Nicosia A, Vitelli A. Efficacy of a virus-vectored vaccine against human and bovine respiratory syncytial virus infections. Sci Transl Med 2015; 7:300ra127. [DOI: 10.1126/scitranslmed.aac5757] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
50
|
Enhanced immunogenicity of a respiratory syncytial virus (RSV) F subunit vaccine formulated with the adjuvant GLA-SE in cynomolgus macaques. Vaccine 2015. [DOI: 10.1016/j.vaccine.2015.07.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|