101
|
Elawar F, Oraby AK, Kieser Q, Jensen LD, Culp T, West FG, Marchant DJ. Pharmacological targets and emerging treatments for respiratory syncytial virus bronchiolitis. Pharmacol Ther 2020; 220:107712. [PMID: 33121940 DOI: 10.1016/j.pharmthera.2020.107712] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022]
Abstract
RSV infection of the lower respiratory tract in infants is the leading cause of pediatric hospitalizations and second to malaria in causing infant deaths worldwide. RSV also causes substantial morbidity in immunocompromised and elderly populations. The only available therapeutic is a prophylactic drug called Palivizumab that is a humanized monoclonal antibody, given to high-risk infants. However, this intervention is expensive and has a limited impact on annual hospitalization rates caused by RSV. No vaccine is available, nor are efficacious antivirals to treat an active infection, and there is still no consensus on how infants with bronchiolitis should be treated during hospital admission. In this comprehensive review, we briefly outline the function of the RSV proteins and their suitability as therapeutic targets. We then discuss the most promising drug candidates, their inhibitory mechanisms, and whether they are in the process of clinical trials. We also briefly discuss the reasons for some of the failures in RSV therapeutics and vaccines. In summary, we provide insight into current antiviral development and the considerations toward producing licensed antivirals and therapeutics.
Collapse
Affiliation(s)
- Farah Elawar
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Ahmed K Oraby
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Misr University for Science &Technology, Al-Motamayez District, 6th of October City, P.O. Box 77, Egypt
| | - Quinten Kieser
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Lionel D Jensen
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Tyce Culp
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Frederick G West
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - David J Marchant
- Li Ka Shing Institute of Virology, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
102
|
Respiratory Syncytial Virus and Human Metapneumovirus Infections in Three-Dimensional Human Airway Tissues Expose an Interesting Dichotomy in Viral Replication, Spread, and Inhibition by Neutralizing Antibodies. J Virol 2020; 94:JVI.01068-20. [PMID: 32759319 DOI: 10.1128/jvi.01068-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV) are two of the leading causes of respiratory infections in children and elderly and immunocompromised patients worldwide. There is no approved treatment for HMPV and only one prophylactic treatment against RSV, palivizumab, for high-risk infants. Better understanding of the viral lifecycles in a more relevant model system may help identify novel therapeutic targets. By utilizing three-dimensional (3-D) human airway tissues to examine viral infection in a physiologically relevant model system, we showed that RSV infects and spreads more efficiently than HMPV, with the latter requiring higher multiplicities of infection (MOIs) to yield similar levels of infection. Apical ciliated cells were the target for both viruses, but RSV apical release was significantly more efficient than HMPV. In RSV- or HMPV-infected cells, cytosolic inclusion bodies containing the nucleoprotein, phosphoprotein, and respective viral genomic RNA were clearly observed in human airway epithelial (HAE) culture. In HMPV-infected cells, actin-based filamentous extensions were more common (35.8%) than those found in RSV-infected cells (4.4%). Interestingly, neither RSV nor HMPV formed syncytia in HAE tissues. Palivizumab and nirsevimab effectively inhibited entry and spread of RSV in HAE tissues, with nirsevimab displaying significantly higher potency than palivizumab. In contrast, 54G10 completely inhibited HMPV entry but only modestly reduced viral spread, suggesting HMPV may use alternative mechanisms for spread. These results represent the first comparative analysis of infection by the two pneumoviruses in a physiologically relevant model, demonstrating an interesting dichotomy in the mechanisms of infection, spread, and consequent inhibition of the viral lifecycles by neutralizing monoclonal antibodies.IMPORTANCE Respiratory syncytial virus and human metapneumovirus are leading causes of respiratory illness worldwide, but limited treatment options are available. To better target these viruses, we examined key aspects of the viral life cycle in three-dimensional (3-D) human airway tissues. Both viruses establish efficient infection through the apical surface, but efficient spread and apical release were seen for respiratory syncytial virus (RSV) but not human metapneumovirus (HMPV). Both viruses form inclusion bodies, minimally composed of nucleoprotein (N), phosphoprotein (P), and viral RNA (vRNA), indicating that these structures are critical for replication in this more physiological model. HMPV formed significantly more long, filamentous actin-based extensions in human airway epithelial (HAE) tissues than RSV, suggesting HMPV may promote cell-to-cell spread via these extensions. Lastly, RSV entry and spread were fully inhibited by neutralizing antibodies palivizumab and the novel nirsevimab. In contrast, while HMPV entry was fully inhibited by 54G10, a neutralizing antibody, spread was only modestly reduced, further supporting a cell-to-cell spread mechanism.
Collapse
|
103
|
McMorrow ML, Tempia S, Walaza S, Treurnicht FK, Moyes J, Cohen AL, Pretorius M, Hellferscee O, Wolter N, von Gottberg A, Nguweneza A, McAnerney JM, Naby F, Mekgoe O, Venter M, Madhi SA, Cohen C. The Impact of Human Immunodeficiency Virus Exposure on Respiratory Syncytial Virus-associated Severe Respiratory Illness in South African Infants, 2011-2016. Clin Infect Dis 2020; 69:2208-2211. [PMID: 30963178 DOI: 10.1093/cid/ciz288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/04/2019] [Indexed: 11/14/2022] Open
Abstract
From 2011 through 2016, we conducted surveillance for severe respiratory illness in infants. Human immunodeficiency virus exposure significantly increased the risk of respiratory syncytial virus (RSV)-associated hospitalization in infants aged <5 months. More than 60% of RSV-associated hospitalizations occurred in the first 4 months of life and may be preventable through maternal vaccination or birth-dose monoclonal antibody.
Collapse
Affiliation(s)
- Meredith L McMorrow
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa.,US Public Health Service, Rockville, Maryland
| | - Stefano Tempia
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa.,Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,Mass Genics, Duluth, Georgia
| | - Sibongile Walaza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, Johannesburg, South Africa
| | - Florette K Treurnicht
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jocelyn Moyes
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, Johannesburg, South Africa
| | - Adam L Cohen
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia.,Influenza Program, Centers for Disease Control and Prevention, Pretoria, South Africa.,US Public Health Service, Rockville, Maryland.,Expanded Programme on Immunization, Department of Immunization, Vaccines and Biological, World Health Organization, Geneva
| | - Marthi Pretorius
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Orienka Hellferscee
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nicole Wolter
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Arthemon Nguweneza
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Johanna M McAnerney
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Fathima Naby
- Department of Paediatrics, Pietermaritzburg Metropolitan Hospitals, University of KwaZulu-Natal
| | - Omphile Mekgoe
- Department of Paediatrics, Klerksdorp Hospital, North West Province
| | - Marietjie Venter
- Emerging Vectorborne and Respiratory Virus Program, Center for Viral Zoonoses, Department of Medical Virology, University of Pretoria
| | - Shabir A Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Cheryl Cohen
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.,School of Public Health, Faculty of Health Sciences, Johannesburg, South Africa
| |
Collapse
|
104
|
Ahangarzadeh S, Payandeh Z, Arezumand R, Shahzamani K, Yarian F, Alibakhshi A. An update on antiviral antibody-based biopharmaceuticals. Int Immunopharmacol 2020; 86:106760. [PMID: 32645633 PMCID: PMC7336121 DOI: 10.1016/j.intimp.2020.106760] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 02/08/2023]
Abstract
Due to the vastness of the science virology, it is no longer an offshoot solely of the microbiology. Viruses have become as the causative agents of major epidemics throughout history. Many therapeutic strategies have been used for these microorganisms, and in this way the recognizing of potential targets of viruses is of particular importance for success. For decades, antibodies and antibody fragments have occupied a significant body of the treatment approaches against infectious diseases. Because of their high affinity, they can be designed and engineered against a variety of purposes, mainly since antibody fragments such as scFv, nanobody, diabody, and bispecific antibody have emerged owing to their small size and interesting properties. In this review, we have discussed the antibody discovery and molecular and biological design of antibody fragments as inspiring therapeutic and diagnostic agents against viral targets.
Collapse
Affiliation(s)
- Shahrzad Ahangarzadeh
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghaye Arezumand
- Department of Medical Biotechnology and Molecular Science, North Khorasan University of Medical Science, Bojnurd, Iran
| | - Kiana Shahzamani
- Isfahan Gastroenterology and Hepatology Research Center (IGHRC), Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Yarian
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Alibakhshi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
105
|
Kalergis AM, Soto JA, Gálvez NMS, Andrade CA, Fernandez A, Bohmwald K, Bueno SM. Pharmacological management of human respiratory syncytial virus infection. Expert Opin Pharmacother 2020; 21:2293-2303. [PMID: 32808830 DOI: 10.1080/14656566.2020.1806821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Human respiratory syncytial virus (hRSV) is the primary viral cause of respiratory diseases, leading to bronchiolitis and pneumonia in vulnerable populations. The only current treatment against this virus is palliative, and no efficient and specific vaccine against this pathogen is available. AREAS COVERED The authors describe the disease symptoms caused by hRSV, the economic and social impact of this infection worldwide, and how this infection can be modulated using pharmacological treatments, preventing and limiting its dissemination. The authors discuss the use of antibodies as prophylactic tools -such as palivizumab- and the use of nonspecific drugs to decrease the symptoms associated with the infection -such as bronchodilators, corticoids, and antivirals. They also discuss current vaccine candidates, new prophylactic treatments, and new antivirals options, which are currently being tested. EXPERT OPINION Today, many researchers are focused on developing different strategies to modulate the symptoms induced by hRSV. However, to achieve this, understanding how current treatments are working and their shortcomings needs to be further elucidated.
Collapse
Affiliation(s)
- 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
| | - Jorge 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
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Ayleen Fernandez
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Susan 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
| |
Collapse
|
106
|
Prediction of Clearance of Monoclonal and Polyclonal Antibodies and Non-Antibody Proteins in Children: Application of Allometric Scaling. Antibodies (Basel) 2020; 9:antib9030040. [PMID: 32764408 PMCID: PMC7551666 DOI: 10.3390/antib9030040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/09/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
Allometric scaling can be used for the extrapolation of pharmacokinetic parameters from adults to children. The objective of this study was to predict clearance of therapeutic proteins (monoclonal and polyclonal antibodies and non-antibody proteins) allometrically in preterm neonates to adolescents. There were 13 monoclonal antibodies, seven polyclonal antibodies, and nine therapeutic proteins (non-antibodies) in the study. The clearance of therapeutic proteins was predicted using the age dependent exponents (ADE) model and then compared with the observed clearance values. There were in total 29 therapeutic proteins in this study with 75 observations. The number of observations with ≤30%, ≤50%, and >50% prediction error was 60 (80%), 72 (96%), and 3 (4%), respectively. Overall, the predicted clearance values of therapeutic proteins in children was good. The allometric method proposed in this manuscript can be used to select first-in-pediatric dose of therapeutic proteins in pediatric clinical trials.
Collapse
|
107
|
Pecetta S, Finco O, Seubert A. Quantum leap of monoclonal antibody (mAb) discovery and development in the COVID-19 era. Semin Immunol 2020; 50:101427. [PMID: 33277154 PMCID: PMC7670927 DOI: 10.1016/j.smim.2020.101427] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
In recent years the global market for monoclonal antibodies (mAbs) became a multi-billion-dollar business. This success is mainly driven by treatments in the oncology and autoimmune space. Instead, development of effective mAbs against infectious diseases has been lagging behind. For years the high production cost and limited efficacy have blocked broader application of mAbs in the infectious disease space, which instead has been dominated for almost a century by effective and cheap antibiotics and vaccines. Only very few mAbs against RSV, anthrax, Clostridium difficile or rabies have reached the market. This is about to change. The development of urgently needed and highly effective mAbs as preventive and therapeutic treatments against a variety of pathogens is gaining traction. Vast advances in mAb isolation, engineering and production have entirely shifted the cost-efficacy balance. MAbs against devastating diseases like Ebola, HIV and other complex pathogens are now within reach. This trend is further accelerated by ongoing or imminent health crises like COVID-19 and antimicrobial resistance (AMR), where antibodies could be the last resort. In this review we will retrace the history of antibodies from the times of serum therapy to modern mAbs and lay out how the current run for effective treatments against COVID-19 will lead to a quantum leap in scientific, technological and health care system innovation around mAb treatments for infectious diseases.
Collapse
|
108
|
Griffin MP, Yuan Y, Takas T, Domachowske JB, Madhi SA, Manzoni P, Simões EAF, Esser MT, Khan AA, Dubovsky F, Villafana T, DeVincenzo JP. Single-Dose Nirsevimab for Prevention of RSV in Preterm Infants. N Engl J Med 2020; 383:415-425. [PMID: 32726528 DOI: 10.1056/nejmoa1913556] [Citation(s) in RCA: 431] [Impact Index Per Article: 86.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection in infants, and a need exists for prevention of RSV in healthy infants. Nirsevimab is a monoclonal antibody with an extended half-life that is being developed to protect infants for an entire RSV season with a single intramuscular dose. METHODS In this trial conducted in both northern and southern hemispheres, we evaluated nirsevimab for the prevention of RSV-associated lower respiratory tract infection in healthy infants who had been born preterm (29 weeks 0 days to 34 weeks 6 days of gestation). We randomly assigned the infants in a 2:1 ratio to receive nirsevimab, at a dose of 50 mg in a single intramuscular injection, or placebo at the start of an RSV season. The primary end point was medically attended RSV-associated lower respiratory tract infection through 150 days after administration of the dose. The secondary efficacy end point was hospitalization for RSV-associated lower respiratory tract infection through 150 days after administration of the dose. RESULTS From November 2016 through November 2017, a total of 1453 infants were randomly assigned to receive nirsevimab (969 infants) or placebo (484 infants) at the start of the RSV season. The incidence of medically attended RSV-associated lower respiratory tract infection was 70.1% lower (95% confidence interval [CI], 52.3 to 81.2) with nirsevimab prophylaxis than with placebo (2.6% [25 infants] vs. 9.5% [46 infants]; P<0.001) and the incidence of hospitalization for RSV-associated lower respiratory tract infection was 78.4% lower (95% CI, 51.9 to 90.3) with nirsevimab than with placebo (0.8% [8 infants] vs. 4.1% [20 infants]; P<0.001). These differences were consistent throughout the 150-day period after the dose was administered and across geographic locations and RSV subtypes. Adverse events were similar in the two trial groups, with no notable hypersensitivity reactions. CONCLUSIONS A single injection of nirsevimab resulted in fewer medically attended RSV-associated lower respiratory tract infections and hospitalizations than placebo throughout the RSV season in healthy preterm infants. (Funded by AstraZeneca and Sanofi Pasteur; ClinicalTrials.gov number, NCT02878330.).
Collapse
MESH Headings
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antiviral Agents/administration & dosage
- Antiviral Agents/adverse effects
- Female
- Hospitalization/statistics & numerical data
- Humans
- Incidence
- Infant
- Infant, Newborn
- Infant, Premature
- Injections, Intramuscular
- Kaplan-Meier Estimate
- Male
- Poisson Distribution
- Respiratory Syncytial Virus Infections/epidemiology
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus, Human
- Respiratory Tract Infections/epidemiology
- Respiratory Tract Infections/prevention & control
- Respiratory Tract Infections/virology
- Viral Fusion Proteins/antagonists & inhibitors
Collapse
Affiliation(s)
- M Pamela Griffin
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Yuan Yuan
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Therese Takas
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Joseph B Domachowske
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Shabir A Madhi
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Paolo Manzoni
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Eric A F Simões
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Mark T Esser
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Anis A Khan
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Filip Dubovsky
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - Tonya Villafana
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| | - John P DeVincenzo
- From AstraZeneca, Gaithersburg, MD (M.P.G., Y.Y., T.T., M.T.E., A.A.K., F.D., T.V.); SUNY Upstate Medical University, Syracuse, NY (J.B.D.); Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, and Department of Science and Technology/National Research Foundation South African Research Chair, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (S.A.M.); the Division of Pediatrics and Neonatology, Department of Maternal, Neonatal, and Infant Medicine, Nuovo Ospedale Degli Infermi, Biella, and Neonatology and NICU, Sant'Anna Hospital, AOU Città della Salute e della Scienza, Turin - both in Italy (P.M.); the University of Colorado School of Medicine, Aurora (E.A.F.S.); and the Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN (J.P.D.V.)
| |
Collapse
|
109
|
Finelli L, Choi Y, Goldstein E. Number needed to immunize to prevent RSV with extended half-life monoclonal antibody. Vaccine 2020; 38:5474-5479. [PMID: 32600912 PMCID: PMC8684408 DOI: 10.1016/j.vaccine.2020.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is one of the most important respiratory pathogens in young children. Infants <6 months of age and infants and young children with extreme pre-term birth, and cardiac and pulmonary co-morbidities experience the highest incidence of severe RSV disease. There are no licensed vaccines; immunoprophylaxis is recommended for the highest risk children. Extended half-life RSV monoclonal antibodies (EHL-mAbs) are under development intended for immunization of all infants and high-risk children <2 years of age. We modeled the anticipated public health benefits of RSV EHL-mAb immunization using the number needed to immunize (NNI). METHODS We combined RSV hospitalization, outpatient and outpatient lower respiratory tract infection (LRI) incidence estimates and a range of immunization efficacies to estimate the annual NNI. We calculated the absolute incidence rate reduction (ARR) by multiplying the incidence rates by immunization efficacy. NNI was calculated as the reciprocal of the ARR. RESULTS For an RSV EHL-mAb with 70% efficacy, 6-18 infants would need to be immunized to prevent one RSV-associated outpatient visit, and 13-33 infants would need to be immunized to prevent one RSV-associated LRI outpatient visit. To prevent one RSV-associated hospitalization, 37-85 infants 0-5 months of age, and 107-280 infants 6-11 months of age would need to be immunized. CONCLUSIONS Public health benefits, such as disease cases averted due to immunization, are essential elements in consideration of candidate vaccines for a national immunization program. An RSV EHL-mAb of moderate efficacy could have high impact. These data provide an additional perspective for public health decision making.
Collapse
Affiliation(s)
- Lyn Finelli
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Yoonyoung Choi
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Edward Goldstein
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
110
|
Kamau E, Otieno JR, Murunga N, Oketch JW, Ngoi JM, de Laurent ZR, Mwema A, Nyiro JU, Agoti CN, Nokes DJ. Genomic epidemiology and evolutionary dynamics of respiratory syncytial virus group B in Kilifi, Kenya, 2015-17. Virus Evol 2020; 6:veaa050. [PMID: 32913665 PMCID: PMC7474930 DOI: 10.1093/ve/veaa050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) circulates worldwide, occurring seasonally in communities, and is a leading cause of acute respiratory illness in young children. There is paucity of genomic data from purposively sampled populations by which to investigate evolutionary dynamics and transmission patterns of RSV. Here we present an analysis of 295 RSV group B (RSVB) genomes from Kilifi, coastal Kenya, sampled from individuals seeking outpatient care in nine health facilities across a defined geographical area (∼890 km2), over two RSV epidemics between 2015 and 2017. RSVB diversity was characterized by multiple virus introductions into the area and co-circulation of distinct genetic clusters, which transmitted and diversified locally with varying frequency. Increase in relative genetic diversity paralleled seasonal virus incidence. Importantly, we identified a cluster of viruses that emerged in the 2016/17 epidemic, carrying distinct amino-acid signatures including a novel nonsynonymous change (K68Q) in antigenic site ∅ in the Fusion protein. RSVB diversity was additionally marked by signature nonsynonymous substitutions that were unique to particular genomic clusters, some under diversifying selection. Our findings provide insights into recent evolutionary and epidemiological behaviors of RSVB, and highlight possible emergence of a novel antigenic variant, which has implications on current prophylactic strategies in development.
Collapse
Affiliation(s)
- Everlyn Kamau
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - James R Otieno
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Nickson Murunga
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - John W Oketch
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Joyce M Ngoi
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Zaydah R de Laurent
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anthony Mwema
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Joyce U Nyiro
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Charles N Agoti
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,School of Health and Human Sciences, Pwani University, Kilifi, Kenya
| | - D James Nokes
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,School of Life Sciences and Zeeman Institute (SBIDER), University of Warwick, Coventry, UK
| |
Collapse
|
111
|
Arvin AM, Fink K, Schmid MA, Cathcart A, Spreafico R, Havenar-Daughton C, Lanzavecchia A, Corti D, Virgin HW. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature 2020; 584:353-363. [DOI: 10.1038/s41586-020-2538-8] [Citation(s) in RCA: 359] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
|
112
|
Ramilo O, Rodriguez-Fernandez R, Peeples ME, Mejias A. Advanced Live Attenuated Vaccines for the Prevention of Respiratory Syncytial Virus Infections in Young Children. J Infect Dis 2020; 222:4-6. [PMID: 31549154 PMCID: PMC7296839 DOI: 10.1093/infdis/jiz409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Octavio Ramilo
- Center for Vaccines and Immunity, Columbus
- Division of Pediatric Infectious Diseases, Nationwide Children’s Hospital, Columbus
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus
| | - Rosa Rodriguez-Fernandez
- Center for Vaccines and Immunity, Columbus
- Department of Pediatrics Hospital Gregorio Marañón & Instituto de Investigacion Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mark E Peeples
- Center for Vaccines and Immunity, Columbus
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Columbus
- Division of Pediatric Infectious Diseases, Nationwide Children’s Hospital, Columbus
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus
- Departamento de Farmacología y Pediatria, Facultad de Medicina, Universidad de Malaga, Spain
| |
Collapse
|
113
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
114
|
Fu K, March K, Alexaki A, Fabozzi G, Moysi E, Petrovas C. Immunogenicity of Protein Therapeutics: A Lymph Node Perspective. Front Immunol 2020; 11:791. [PMID: 32477334 PMCID: PMC7240201 DOI: 10.3389/fimmu.2020.00791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
The continuous development of molecular biology and protein engineering technologies enables the expansion of the breadth and complexity of protein therapeutics for in vivo administration. However, the immunogenicity and associated in vivo development of antibodies against therapeutics are a major restriction factor for their usage. The B cell follicular and particularly germinal center areas in secondary lymphoid organs are the anatomical sites where the development of antibody responses against pathogens and immunogens takes place. A growing body of data has revealed the importance of the orchestrated function of highly differentiated adaptive immunity cells, including follicular helper CD4 T cells and germinal center B cells, for the optimal generation of these antibody responses. Understanding the cellular and molecular mechanisms mediating the antibody responses against therapeutics could lead to novel strategies to reduce their immunogenicity and increase their efficacy.
Collapse
Affiliation(s)
- Kristy Fu
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kylie March
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aikaterini Alexaki
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Giulia Fabozzi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
115
|
Priorización de nuevas vacunas e innovación al servicio de estrategias de vacunación. REVISTA MÉDICA CLÍNICA LAS CONDES 2020. [DOI: 10.1016/j.rmclc.2020.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
116
|
Antiviral Activity of a Llama-Derived Single-Domain Antibody against Enterovirus A71. Antimicrob Agents Chemother 2020; 64:AAC.01922-19. [PMID: 32152074 DOI: 10.1128/aac.01922-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/19/2020] [Indexed: 12/28/2022] Open
Abstract
In the past few decades, enterovirus A71 (EVA71) has caused devastating outbreaks in the Asia-Pacific region, resulting in serious sequelae in infected young children. No preventive or therapeutic interventions are currently available for curing EVA71 infection, highlighting a great unmet medical need for this disease. Here, we showed that one novel single-domain antibody (sdAb), F1, isolated from an immunized llama, could alleviate EVA71 infection both in vitro and in vivo We also confirmed that the sdAb clone F1 recognizes EVA71 through a novel conformational epitope comprising the highly conserved region of VP3 capsid protein by using competitive-binding and overlapping-peptide enzyme-linked immunosorbent assays (ELISAs). Because of the virion's icosahedral structure, we reasoned that adjacent epitopes must be clustered within molecular ranges that may be simultaneously bound by an engineered antibody with multiple valency. Therefore, two single-domain binding modules (F1) were fused to generate an sdAb-in-tandem design so that the capture of viral antigens could be further increased by valency effects. We showed that the tetravalent construct F1×F1-hFc, containing two sdAb-in-tandem on a fragment crystallizable (Fc) scaffold, exhibits more potent neutralization activity against EVA71 than does the bivalent sdAb F1-hFc by at least 5.8-fold. We also demonstrated that, using a human scavenger receptor class B member 2 (hSCARB2) transgenic mouse model, a half dose of the F1×F1-hFc provided better protection against EVA71 infection than did the F1-hFc. Thus, our study furnishes important insights into multivalent sdAb engineering against viral infection and provides a novel strategic deployment approach for preparedness of emerging infectious diseases such as EVA71.
Collapse
|
117
|
Soto JA, Gálvez NMS, Pacheco GA, Bueno SM, Kalergis AM. Antibody development for preventing the human respiratory syncytial virus pathology. Mol Med 2020; 26:35. [PMID: 32303184 PMCID: PMC7164255 DOI: 10.1186/s10020-020-00162-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is the most important etiological agent causing hospitalizations associated with respiratory diseases in children under 5 years of age as well as the elderly, newborns and premature children are the most affected populations. This viral infection can be associated with various symptoms, such as fever, coughing, wheezing, and even pneumonia and bronchiolitis. Due to its severe symptoms, the need for mechanical ventilation is not uncommon in clinical practice. Additionally, alterations in the central nervous system -such as seizures, encephalopathy and encephalitis- have been associated with cases of hRSV-infections. Furthermore, the absence of effective vaccines or therapies against hRSV leads to elevated expenditures by the public health system and increased mortality rates for the high-risk population. Along these lines, vaccines and therapies can elicit different responses to this virus. While hRSV vaccine candidates seek to promote an active immune response associated with the achievement of immunological memory, other therapies -such as the administration of antibodies- provide a protective environment, although they do not trigger the activation of the immune system and therefore do not promote an immunological memory. An interesting approach to vaccination is the use of virus-neutralizing antibodies, which inhibit the entry of the pathogen into the host cells, therefore impairing the capacity of the virus to replicate. Currently, the most common molecule targeted for antibody design against hRSV is the F protein of this virus. However, other molecular components of the virus -such as the G or the N hRSV proteins- have also been explored as potential targets for the control of this disease. Currently, palivizumab is the only monoclonal antibody approved for human use. However, studies in humans have shown a protective effect only after the administration of at least 3 to 5 doses, due to the stability of this vaccine. Furthermore, other studies suggest that palivizumab only has an effectiveness close to 50% in high-risk infants. In this work, we will review different strategies addressed for the use of antibodies in a prophylactic or therapeutic context and their ability to prevent the symptoms caused by hRSV infection of the airways, as well as in other tissues such as the CNS.
Collapse
Affiliation(s)
- Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, 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, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile.
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
118
|
Boyoglu-Barnum S, Tripp RA. Up-to-date role of biologics in the management of respiratory syncytial virus. Expert Opin Biol Ther 2020; 20:1073-1082. [PMID: 32264720 DOI: 10.1080/14712598.2020.1753696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in young children and a substantial contributor to respiratory tract disease throughout life. Despite RSV being a high priority for vaccine development, there is currently no safe and effective vaccine available. There are many challenges to developing an RSV vaccine and there are limited antiviral drugs or biologics available for the management of infection. In this article, we review the antiviral treatments, vaccination strategies along with alternative therapies for RSV. AREAS COVERED This review is a summary of the current antiviral and RSV vaccination approaches noting strategies and alternative therapies that may prevent or decrease the disease severity in RSV susceptible populations. EXPERT OPINION This review discusses anti-RSV strategies given that no safe and efficacious vaccines are available, and therapeutic treatments are limited. Various biologicals that target for RSV are considered for disease intervention, as it is likely that it may be necessary to develop separate vaccines or therapeutics for each at-risk population.
Collapse
Affiliation(s)
- Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, Animal Health Research Center, University of Georgia , Athens, GA, USA
| |
Collapse
|
119
|
Kumar R, Shrivastava T, Samal S, Ahmed S, Parray HA. Antibody-based therapeutic interventions: possible strategy to counter chikungunya viral infection. Appl Microbiol Biotechnol 2020; 104:3209-3228. [PMID: 32076776 PMCID: PMC7223553 DOI: 10.1007/s00253-020-10437-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022]
Abstract
Chikungunya virus (CHIKV), a mosquito-transmitted disease that belongs to the genus Alphaviruses, has been emerged as an epidemic threat over the last two decades, and the recent co-emergence of this virus along with other circulating arboviruses and comorbidities has influenced atypical mortality rate up to 10%. Genetic variation in the virus has resulted in its adaptability towards the new vector Aedes albopictus other than Aedes aegypti, which has widen the horizon of distribution towards non-tropical and non-endemic areas. As of now, no licensed vaccines or therapies are available against CHIKV; the treatment regimens for CHIKV are mostly symptomatic, based on the clinical manifestations. Development of small molecule drugs and neutralizing antibodies are potential alternatives of worth investigating until an efficient or safe vaccine is approved. Neutralizing antibodies play an important role in antiviral immunity, and their presence is a hallmark of viral infection. In this review, we describe prospects for effective vaccines and highlight importance of neutralizing antibody-based therapeutic and prophylactic applications to combat CHIKV infections. We further discuss about the progress made towards CHIKV therapeutic interventions as well as challenges and limitation associated with the vaccine development. Furthermore this review describes the lesson learned from chikungunya natural infection, which could help in better understanding for future development of antibody-based therapeutic measures.
Collapse
Affiliation(s)
- Rajesh Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, 121001, India.
| | - Tripti Shrivastava
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, 121001, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, 121001, India
| | - Shubbir Ahmed
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, 121001, India
| | - Hilal Ahmad Parray
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, 121001, India
| |
Collapse
|
120
|
Brand SP, Munywoki P, Walumbe D, Keeling MJ, Nokes DJ. Reducing respiratory syncytial virus (RSV) hospitalization in a lower-income country by vaccinating mothers-to-be and their households. eLife 2020; 9:47003. [PMID: 32216871 PMCID: PMC7556875 DOI: 10.7554/elife.47003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/26/2020] [Indexed: 01/15/2023] Open
Abstract
Respiratory syncytial virus is the leading cause of lower respiratory tract infection among infants. RSV is a priority for vaccine development. In this study, we investigate the potential effectiveness of a two-vaccine strategy aimed at mothers-to-be, thereby boosting maternally acquired antibodies of infants, and their household cohabitants, further cocooning infants against infection. We use a dynamic RSV transmission model which captures transmission both within households and communities, adapted to the changing demographics and RSV seasonality of a low-income country. Model parameters were inferred from past RSV hospitalisations, and forecasts made over a 10-year horizon. We find that a 50% reduction in RSV hospitalisations is possible if the maternal vaccine effectiveness can achieve 75 days of additional protection for newborns combined with a 75% coverage of their birth household co-inhabitants (~7.5% population coverage).
Collapse
Affiliation(s)
- Samuel Pc Brand
- Zeeman Institute of Systems Biology and Infectious Disease Research (SBIDER), University of Warwick, Warwick, United Kingdom.,School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Patrick Munywoki
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - David Walumbe
- Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Matthew J Keeling
- Zeeman Institute of Systems Biology and Infectious Disease Research (SBIDER), University of Warwick, Warwick, United Kingdom.,School of Life Sciences, University of Warwick, Coventry, United Kingdom.,Mathematics Institute, University of Warwick, Coventry, United Kingdom
| | - David James Nokes
- Zeeman Institute of Systems Biology and Infectious Disease Research (SBIDER), University of Warwick, Warwick, United Kingdom.,School of Life Sciences, University of Warwick, Coventry, United Kingdom.,Epidemiology and Demography Department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| |
Collapse
|
121
|
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
|
122
|
The journey to a respiratory syncytial virus vaccine. Ann Allergy Asthma Immunol 2020; 125:36-46. [PMID: 32217187 DOI: 10.1016/j.anai.2020.03.017] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The high burden associated with respiratory syncytial virus (RSV) has made the development of RSV vaccine(s) a global health high priority. This review summarizes the journey to an RSV vaccine, the different strategies and challenges associated with the development of preventive strategies for RSV, and the diverse products that are undergoing clinical testing. DATA SOURCES Studies on RSV biology, immunology, epidemiology, and monoclonal antibodies (mAbs) and vaccines were searched using MEDLINE. We also searched PATH.org and ClinicalTrials.gov for updated information regarding the status of RSV vaccines and mAbs undergoing clinical trials. STUDY SELECTIONS We selected relevant studies conducted in infants and young children, pregnant women, and elderly population for the prevention of RSV infection. RESULTS Identification of a safe and immunogenic vaccine has been an important but elusive initiative for more than 60 years for different reasons, including the legacy of formalin-inactivated vaccine, our limited understanding of the immune response to RSV and how it relates to clinical disease severity, or the need for different end points according to the different vaccine platforms. Nevertheless, there are currently 39 vaccines and mAbs under development and 19 undergoing clinical trials. CONCLUSION Over the past decade, there have been significant advances in our knowledge of RSV molecular and structural biology and in understanding the human immune response to RSV. Despite the barriers, there are several promising mAbs and RSV vaccines undergoing clinical trials that hope to offer protection to the most vulnerable populations.
Collapse
|
123
|
Drysdale SB, Barr RS, Rollier CS, Green CA, Pollard AJ, Sande CJ. Priorities for developing respiratory syncytial virus vaccines in different target populations. Sci Transl Med 2020; 12:eaax2466. [PMID: 32188721 PMCID: PMC7613568 DOI: 10.1126/scitranslmed.aax2466] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/25/2019] [Indexed: 01/13/2023]
Abstract
The development of an effective vaccine against respiratory syncytial virus (RSV) has been hampered by major difficulties that occurred in the 1960s when a formalin-inactivated vaccine led to increased severity of RSV disease after acquisition of the virus in the RSV season after vaccination. Recent renewed efforts to develop a vaccine have resulted in about 38 candidate vaccines and monoclonal antibodies now in clinical development. The target populations for effective vaccination are varied and include neonates, young children, pregnant women, and older adults. The reasons for susceptibility to infection in each of these groups may be different and, therefore, could require different vaccine types for induction of protective immune responses, adding a further challenge for vaccine development. Here, we review the current knowledge of RSV vaccine development for these target populations and propose a view and rationale for prioritizing RSV vaccine development.
Collapse
Affiliation(s)
- Simon B Drysdale
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK.
- Institute of Infection and Immunity, St George's, University of London, London SW17 0RE, UK
| | - Rachael S Barr
- Taunton and Somerset NHS Foundation Trust, Taunton TA1 5DA, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Christopher A Green
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Charles J Sande
- Oxford Vaccine Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7LE, UK.
- KEMRI-Wellcome Trust Research Programme, Kilifi 80108, Kenya
| |
Collapse
|
124
|
Bergeron HC, Tripp RA. Emerging small and large molecule therapeutics for respiratory syncytial virus. Expert Opin Investig Drugs 2020; 29:285-294. [PMID: 32096420 DOI: 10.1080/13543784.2020.1735349] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Respiratory syncytial virus (RSV) causes lower respiratory tract infections and can lead to morbidity and mortality in the infant, elderly and immunocompromised. There is no vaccine and therapeutic interventions are limited. RSV disease research has yielded the development of several prophylactic and therapeutic treatments. Several promising candidates are currently under investigation.Areas covered: Small and large molecule approaches to RSV treatment were examined and categorized by their mechanism of action using data from PubMed, clinicaltrials.gov, and from the sponsoring organizations publicly available pipeline information. These results are prefaced by an overview of RSV to provide the context for rational therapy development.Expert opinion: While small molecule drugs show promise for RSV treatment, we believe that large molecule therapy using anti-RSV G and F protein monoclonal antibodies (mAbs) will most efficaciously and safely ameliorate RSV disease.
Collapse
Affiliation(s)
- Harrison C Bergeron
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| |
Collapse
|
125
|
Roy CJ, Van Slyke G, Ehrbar D, Bornholdt ZA, Brennan MB, Campbell L, Chen M, Kim D, Mlakar N, Whaley KJ, Froude JW, Torres-Velez FJ, Vitetta E, Didier PJ, Doyle-Meyers L, Zeitlin L, Mantis NJ. Passive immunization with an extended half-life monoclonal antibody protects Rhesus macaques against aerosolized ricin toxin. NPJ Vaccines 2020; 5:13. [PMID: 32128254 PMCID: PMC7018975 DOI: 10.1038/s41541-020-0162-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/13/2020] [Indexed: 02/07/2023] Open
Abstract
Inhalation of ricin toxin (RT), a Category B biothreat agent, provokes an acute respiratory distress syndrome marked by pro-inflammatory cytokine and chemokine production, neutrophilic exudate, and pulmonary edema. The severity of RT exposure is attributed to the tropism of the toxin's B subunit (RTB) for alveolar macrophages and airway epithelial cells, coupled with the extraordinarily potent ribosome-inactivating properties of the toxin's enzymatic subunit (RTA). While there are currently no vaccines or treatments approved to prevent RT intoxication, we recently described a humanized anti-RTA IgG1 MAb, huPB10, that was able to rescue non-human primates (NHPs) from lethal dose RT aerosol challenge if administered by intravenous (IV) infusion within hours of toxin exposure. We have now engineered an extended serum half-life variant of that MAb, huPB10-LS, and evaluated it as a pre-exposure prophylactic. Five Rhesus macaques that received a single intravenous infusion (25 mg/kg) of huPB10-LS survived a lethal dose aerosol RT challenge 28 days later, whereas three control animals succumbed to RT intoxication within 48 h. The huPB10-LS treated animals remained clinically normal in the hours and days following toxin insult, suggesting that pre-existing antibody levels were sufficient to neutralize RT locally. Moreover, pro-inflammatory markers in sera and BAL fluids collected 24 h following RT challenge were significantly dampened in huPB10-LS treated animals, as compared to controls. Finally, we found that all five surviving animals, within days after RT exposure, had anti-RT serum IgG titers against epitopes other than huPB10-LS, indicative of active immunization by residual RT and/or RT-immune complexes.
Collapse
Affiliation(s)
- Chad J. Roy
- Tulane National Primate Research Center, Covington, LA 70433 USA
| | - Greta Van Slyke
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY 12208 USA
| | - Dylan Ehrbar
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY 12208 USA
| | | | | | | | - Michelle Chen
- Mapp Biopharmaceutical, Inc, San Diego, CA 92121 USA
| | - Do Kim
- Mapp Biopharmaceutical, Inc, San Diego, CA 92121 USA
| | - Neil Mlakar
- Mapp Biopharmaceutical, Inc, San Diego, CA 92121 USA
| | | | - Jeffrey W. Froude
- Clinical Pharmacology Branch, Walter Reed Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910 USA
- Present Address: Vaccines and Therapeutics Division, Defense Threat Reduction Agency, 8725 John J. Kingman Rd., Fort Belvoir, VA 22060 USA
| | - Fernando J Torres-Velez
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY 12208 USA
| | - Ellen Vitetta
- Departments of Immunology and Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Peter J. Didier
- Tulane National Primate Research Center, Covington, LA 70433 USA
| | | | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc, San Diego, CA 92121 USA
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY 12208 USA
| |
Collapse
|
126
|
Respiratory syncytial virus. MATERNAL IMMUNIZATION 2020. [PMCID: PMC7149541 DOI: 10.1016/b978-0-12-814582-1.00011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of hospitalization and mortality associated with lower respiratory tract illness in infants and young children worldwide. The World Health Organization recognizes the need to develop and implement prevention strategies to reduce the impact of RSV in early life as a global health priority. RSV vaccination during pregnancy is a feasible strategy to achieve this goal. Vaccines for maternal immunization against RSV are in active development and could be implemented in the near future within existing maternal-child health platforms. In addition, infant protection may be achieved through either passive antibody administration or active immunization, depending on infant health status, given that options for these complementary interventions are being developed in parallel to vaccines for maternal immunization.
Collapse
|
127
|
Hooft van Huijsduijnen R, Kojima S, Carter D, Okabe H, Sato A, Akahata W, Wells TNC, Katsuno K. Reassessing therapeutic antibodies for neglected and tropical diseases. PLoS Negl Trop Dis 2020; 14:e0007860. [PMID: 31999695 PMCID: PMC6991954 DOI: 10.1371/journal.pntd.0007860] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In the past two decades there has been a significant expansion in the number of new therapeutic monoclonal antibodies (mAbs) that are approved by regulators. The discovery of these new medicines has been driven primarily by new approaches in inflammatory diseases and oncology, especially in immuno-oncology. Other recent successes have included new antibodies for use in viral diseases, including HIV. The perception of very high costs associated with mAbs has led to the assumption that they play no role in prophylaxis for diseases of poverty. However, improvements in antibody-expression yields and manufacturing processes indicate this is a cost-effective option for providing protection from many types of infection that should be revisited. Recent technology developments also indicate that several months of protection could be achieved with a single dose. Moreover, new methods in B cell sorting now enable the systematic identification of high-quality antibodies from humanized mice, or patients. This Review discusses the potential for passive immunization against schistosomiasis, fungal infections, dengue, and other neglected diseases.
Collapse
Affiliation(s)
| | | | - Dee Carter
- School of Life and Environmental Sciences and The Marie Bashir Institute, University of Sydney, NSW, Australia
| | | | | | - Wataru Akahata
- VLP Therapeutics, Gaithersburg, Maryland, United States of America
| | | | - Kei Katsuno
- Global Health Innovative Technology Fund, Tokyo, Japan
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Nagasaki University School of Tropical Medicine and Global Health, Nagasaki, Japan
| |
Collapse
|
128
|
Huang J, Diaz D, Mousa JJ. Antibody Epitopes of Pneumovirus Fusion Proteins. Front Immunol 2019; 10:2778. [PMID: 31849961 PMCID: PMC6895023 DOI: 10.3389/fimmu.2019.02778] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/13/2019] [Indexed: 11/13/2022] Open
Abstract
The pneumoviruses respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are two widespread human pathogens that can cause severe disease in the young, the elderly, and the immunocompromised. Despite the discovery of RSV over 60 years ago, and hMPV nearly 20 years ago, there are no approved vaccines for either virus. Antibody-mediated immunity is critical for protection from RSV and hMPV, and, until recently, knowledge of the antibody epitopes on the surface glycoproteins of RSV and hMPV was very limited. However, recent breakthroughs in the recombinant expression and stabilization of pneumovirus fusion proteins have facilitated in-depth characterization of antibody responses and structural epitopes, and have provided an enormous diversity of new monoclonal antibody candidates for therapeutic development. These new data have primarily focused on the RSV F protein, and have led to a wealth of new vaccine candidates in preclinical and clinical trials. In contrast, the major structural antibody epitopes remain unclear for the hMPV F protein. Overall, this review will cover recent advances in characterizing the antigenic sites on the RSV and hMPV F proteins.
Collapse
Affiliation(s)
- Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Darren Diaz
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Jarrod J. Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
129
|
Kang TH, Jung ST. Boosting therapeutic potency of antibodies by taming Fc domain functions. Exp Mol Med 2019; 51:1-9. [PMID: 31735912 PMCID: PMC6859160 DOI: 10.1038/s12276-019-0345-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 01/12/2023] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most widely used drug platforms for infectious diseases or cancer therapeutics because they selectively target pathogens, infectious cells, cancerous cells, and even immune cells. In this way, they mediate the elimination of target molecules and cells with fewer side effects than other therapeutic modalities. In particular, cancer therapeutic mAbs can recognize cell-surface proteins on target cells and then kill the targeted cells by multiple mechanisms that are dependent upon a fragment crystallizable (Fc) domain interacting with effector Fc gamma receptors, including antibody-dependent cell-mediated cytotoxicity and antibody-dependent cell-mediated phagocytosis. Extensive engineering efforts have been made toward tuning Fc functions by either reinforcing (e.g. for targeted therapy) or disabling (e.g. for immune checkpoint blockade therapy) effector functions and prolonging the serum half-lives of antibodies, as necessary. In this report, we review Fc engineering efforts to improve therapeutic potency, and propose future antibody engineering directions that can fulfill unmet medical needs. Fine-tuning the function of monoclonal antibodies (mAbs) holds promise for developing new therapeutic agents. Antibodies bind to pathogens or cancer cells, flagging them with Fc (fragment crystallizable) domain for destruction by the immune system. mAbs attached only to specific target cells enable lower side effect than other conventional drugs. Sang Taek Jung at Korea University and Tae Hyun Kang at Kookmin University, both in Seoul, reviewed recent developments in engineering therapeutic potency of mAbs. They report that mAbs can be engineered to activate effective immune cell types to treat a particular disease. Engineering can also increase mAbs’ persistence in the blood, enabling less frequent administration. Antibodies engineered to bind to two different antigens at once can also improve therapeutic efficacy. Applying these techniques could help developing new treatments against cancer, and infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Tae Hyun Kang
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
130
|
Malik PRV, Edginton AN. Integration of Ontogeny Into a Physiologically Based Pharmacokinetic Model for Monoclonal Antibodies in Premature Infants. J Clin Pharmacol 2019; 60:466-476. [DOI: 10.1002/jcph.1540] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Paul R. V. Malik
- School of PharmacyUniversity of Waterloo Kitchener Ontario Canada
| | | |
Collapse
|
131
|
Lee CH, Kang TH, Godon O, Watanabe M, Delidakis G, Gillis CM, Sterlin D, Hardy D, Cogné M, Macdonald LE, Murphy AJ, Tu N, Lee J, McDaniel JR, Makowski E, Tessier PM, Meyer AS, Bruhns P, Georgiou G. An engineered human Fc domain that behaves like a pH-toggle switch for ultra-long circulation persistence. Nat Commun 2019; 10:5031. [PMID: 31695028 PMCID: PMC6834678 DOI: 10.1038/s41467-019-13108-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
The pharmacokinetic properties of antibodies are largely dictated by the pH-dependent binding of the IgG fragment crystallizable (Fc) domain to the human neonatal Fc receptor (hFcRn). Engineered Fc domains that confer a longer circulation half-life by virtue of more favorable pH-dependent binding to hFcRn are of great therapeutic interest. Here we developed a pH Toggle switch Fc variant containing the L309D/Q311H/N434S (DHS) substitutions, which exhibits markedly improved pharmacokinetics relative to both native IgG1 and widely used half-life extension variants, both in conventional hFcRn transgenic mice and in new knock-in mouse strains. engineered specifically to recapitulate all the key processes relevant to human antibody persistence in circulation, namely: (i) physiological expression of hFcRn, (ii) the impact of hFcγRs on antibody clearance and (iii) the role of competing endogenous IgG. DHS-IgG retains intact effector functions, which are important for the clearance of target pathogenic cells and also has favorable developability.
Collapse
Affiliation(s)
- Chang-Han Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Tae Hyun Kang
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Department of Applied Chemistry, Kookmin University, Seoul, Republic of Korea
| | - Ophélie Godon
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - Makiko Watanabe
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Caitlin M Gillis
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - Delphine Sterlin
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France
| | - David Hardy
- Experimental Neuropathology Unit, Infection and Epidemiology Department, Institut Pasteur, 25, rue du Docteur Roux, 75015, Paris, France
| | | | | | | | - Naxin Tu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Jonathan R McDaniel
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Emily Makowski
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERMF-75015, Paris, France.
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA.
- Department of Molecular Bioscience, University of Texas at Austin, Austin, TX, USA.
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
132
|
Dibo M, Battocchio EC, dos Santos Souza LM, da Silva MDV, Banin-Hirata BK, Sapla MM, Marinello P, Rocha SP, Faccin-Galhardi LC. Antibody Therapy for the Control of Viral Diseases: An Update. Curr Pharm Biotechnol 2019; 20:1108-1121. [DOI: 10.2174/1389201020666190809112704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/22/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
The epidemiological impact of viral diseases, combined with the emergence and reemergence of some viruses, and the difficulties in identifying effective therapies, have encouraged several studies to develop new therapeutic strategies for viral infections. In this context, the use of immunotherapy for the treatment of viral diseases is increasing. One of the strategies of immunotherapy is the use of antibodies, particularly the monoclonal antibodies (mAbs) and multi-specific antibodies, which bind directly to the viral antigen and bring about activation of the immune system. With current advancements in science and technology, several such antibodies are being tested, and some are already approved and are undergoing clinical trials. The present work aims to review the status of mAb development for the treatment of viral diseases.
Collapse
Affiliation(s)
- Miriam Dibo
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Eduardo C. Battocchio
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lucas M. dos Santos Souza
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | | | - Bruna K. Banin-Hirata
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Milena M.M. Sapla
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Poliana Marinello
- Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Sérgio P.D. Rocha
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| | - Lígia C. Faccin-Galhardi
- Department of Microbiology, Biological Sciences Center, State University of Londrina, Parana, Brazil
| |
Collapse
|
133
|
Ison MG, Hirsch HH. Community-Acquired Respiratory Viruses in Transplant Patients: Diversity, Impact, Unmet Clinical Needs. Clin Microbiol Rev 2019; 32:e00042-19. [PMID: 31511250 PMCID: PMC7399564 DOI: 10.1128/cmr.00042-19] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Patients undergoing solid-organ transplantation (SOT) or allogeneic hematopoietic cell transplantation (HCT) are at increased risk for infectious complications. Community-acquired respiratory viruses (CARVs) pose a particular challenge due to the frequent exposure pre-, peri-, and posttransplantation. Although influenza A and B viruses have a top priority regarding prevention and treatment, recent molecular diagnostic tests detecting an array of other CARVs in real time have dramatically expanded our knowledge about the epidemiology, diversity, and impact of CARV infections in the general population and in allogeneic HCT and SOT patients. These data have demonstrated that non-influenza CARVs independently contribute to morbidity and mortality of transplant patients. However, effective vaccination and antiviral treatment is only emerging for non-influenza CARVs, placing emphasis on infection control and supportive measures. Here, we review the current knowledge about CARVs in SOT and allogeneic HCT patients to better define the magnitude of this unmet clinical need and to discuss some of the lessons learned from human influenza virus, respiratory syncytial virus, parainfluenzavirus, rhinovirus, coronavirus, adenovirus, and bocavirus regarding diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Michael G Ison
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hans H Hirsch
- Transplantation & Clinical Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
- Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
134
|
Abstract
Human respiratory syncytial virus (RSV) belongs to the recently defined Pneumoviridae family, Orthopneumovirus genus. It is a negative sense, single stranded RNA virus that results in epidemics of respiratory infections that typically peak in the winter in temperate climates and during the rainy season in tropical climates. Generally, one of the two genotypes (A and B) predominates in a single season, alternating annually, although regional variation occurs. RSV is a cause of disease and death in children, older people, and immunocompromised patients, and its clinical effect on adults admitted to hospital is clarified with expanded use of multiplex molecular assays. Among adults, RSV produces a wide range of clinical symptoms including upper respiratory tract infections, severe lower respiratory tract infections, and exacerbations of underlying disease. Here we discuss the latest evidence on the burden of RSV related disease in adults, especially in those with immunocompromise or other comorbidities. We review current therapeutic and prevention options, as well as those in development.
Collapse
Affiliation(s)
- Hannah H Nam
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael G Ison
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
135
|
Zhu Q, Lu B, McTamney P, Palaszynski S, Diallo S, Ren K, Ulbrandt ND, Kallewaard N, Wang W, Fernandes F, Wong S, Svabek C, Moldt B, Esser MT, Jing H, Suzich JA. Prevalence and Significance of Substitutions in the Fusion Protein of Respiratory Syncytial Virus Resulting in Neutralization Escape From Antibody MEDI8897. J Infect Dis 2019; 218:572-580. [PMID: 29617879 DOI: 10.1093/infdis/jiy189] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/28/2018] [Indexed: 11/13/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection among infants and young children. To date, no vaccine is approved for the broad population of healthy infants. MEDI8897, a potent anti-RSV fusion antibody with extended serum half-life, is currently under clinical investigation as a potential passive RSV vaccine for all infants. As a ribonucleic acid virus, RSV is prone to mutation, and the possibility of viral escape from MEDI8897 neutralization is a potential concern. Methods We generated RSV monoclonal antibody (mAb)-resistant mutants (MARMs) in vitro and studied the effect of the amino acid substitutions identified on binding and viral neutralization susceptibility to MEDI8897. The impact of resistance-associated mutations on in vitro growth kinetics and the prevalence of these mutations in currently circulating strains of RSV in the United States was assessed. Results Critical residues identified in MARMs for MEDI8897 neutralization were located in the MEDI8897 binding site defined by crystallographic analysis. Substitutions in these residues affected the binding of mAb to virus, without significant impact on viral replication in vitro. The frequency of natural resistance-associated polymorphisms was low. Conclusions Results from this study provide insights into the mechanism of MEDI8897 escape and the complexity of monitoring for emergence of resistance.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Bin Lu
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Patrick McTamney
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Susan Palaszynski
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Seme Diallo
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Kuishu Ren
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Nancy D Ulbrandt
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Nicole Kallewaard
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Weijia Wang
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Fiona Fernandes
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Steve Wong
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Catherine Svabek
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Brian Moldt
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Mark T Esser
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - Hong Jing
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| | - JoAnn A Suzich
- Department of Infectious Diseases-Vaccines, MedImmune, LLC, Gaithersburg, Maryland
| |
Collapse
|
136
|
Immunological Lessons from Respiratory Syncytial Virus Vaccine Development. Immunity 2019; 51:429-442. [DOI: 10.1016/j.immuni.2019.08.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/30/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022]
|
137
|
Andreano E, Seubert A, Rappuoli R. Human monoclonal antibodies for discovery, therapy, and vaccine acceleration. Curr Opin Immunol 2019; 59:130-134. [PMID: 31450054 DOI: 10.1016/j.coi.2019.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/08/2019] [Accepted: 07/18/2019] [Indexed: 01/24/2023]
Abstract
Screening of single B cells from convalescent or vaccinated people allows the discovery of novel targets for infectious diseases and rapid production of engineered human monoclonal antibodies (mAbs) that can prevent or control infections by passive immunization. Here we propose that the development of human mAbs can also significantly accelerate vaccine development by anticipating some of the key biological and regulatory questions.
Collapse
Affiliation(s)
| | | | - Rino Rappuoli
- vAMRes Lab, Toscana Life Sciences, Siena, Italy; GSK, Siena, Italy; Imperial College, London, United Kingdom.
| |
Collapse
|
138
|
Jones HG, Battles MB, Lin CC, Bianchi S, Corti D, McLellan JS. Alternative conformations of a major antigenic site on RSV F. PLoS Pathog 2019; 15:e1007944. [PMID: 31306469 PMCID: PMC6658013 DOI: 10.1371/journal.ppat.1007944] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/25/2019] [Accepted: 06/25/2019] [Indexed: 01/18/2023] Open
Abstract
The respiratory syncytial virus (RSV) fusion (F) glycoprotein is a major target of neutralizing antibodies arising from natural infection, and antibodies that specifically bind to the prefusion conformation of RSV F generally demonstrate the greatest neutralization potency. Prefusion-stabilized RSV F variants have been engineered as vaccine antigens, but crystal structures of these variants have revealed conformational differences in a key antigenic site located at the apex of the trimer, referred to as antigenic site Ø. Currently, it is unclear if flexibility in this region is an inherent property of prefusion RSV F or if it is related to inadequate stabilization of site Ø in the engineered variants. Therefore, we set out to investigate the conformational flexibility of antigenic site Ø, as well as the ability of the human immune system to recognize alternative conformations of this site, by determining crystal structures of prefusion RSV F bound to neutralizing human-derived antibodies AM22 and RSD5. Both antibodies bound with high affinity and were specific for the prefusion conformation of RSV F. Crystal structures of the complexes revealed that the antibodies recognized distinct conformations of antigenic site Ø, each diverging at a conserved proline residue located in the middle of an α-helix. These data suggest that antigenic site Ø exists as an ensemble of conformations, with individual antibodies recognizing discrete states. Collectively, these results have implications for the refolding of pneumovirus and paramyxovirus fusion proteins and should inform development of prefusion-stabilized RSV F vaccine candidates.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigen-Antibody Complex/chemistry
- Antigen-Antibody Complex/immunology
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Binding Sites/genetics
- Crystallography, X-Ray
- Humans
- Models, Molecular
- Proline/chemistry
- Protein Conformation
- Respiratory Syncytial Virus, Human/chemistry
- Respiratory Syncytial Virus, Human/genetics
- Respiratory Syncytial Virus, Human/immunology
- Viral Fusion Proteins/chemistry
- Viral Fusion Proteins/genetics
- Viral Fusion Proteins/immunology
Collapse
Affiliation(s)
- Harrison G. Jones
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Michael B. Battles
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Chun-Chi Lin
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Siro Bianchi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
139
|
Behzadi MA, Leyva-Grado VH. Overview of Current Therapeutics and Novel Candidates Against Influenza, Respiratory Syncytial Virus, and Middle East Respiratory Syndrome Coronavirus Infections. Front Microbiol 2019; 10:1327. [PMID: 31275265 PMCID: PMC6594388 DOI: 10.3389/fmicb.2019.01327] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/28/2019] [Indexed: 01/26/2023] Open
Abstract
Emergence and re-emergence of respiratory virus infections represent a significant threat to global public health, as they occur seasonally and less frequently (such as in the case of influenza virus) as pandemic infections. Some of these viruses have been in the human population for centuries and others had recently emerged as a public health problem. Influenza viruses have been affecting the human population for a long time now; however, their ability to rapidly evolve through antigenic drift and antigenic shift causes the emergence of new strains. A recent example of these events is the avian-origin H7N9 influenza virus outbreak currently undergoing in China. Human H7N9 influenza viruses are resistant to amantadines and some strains are also resistant to neuraminidase inhibitors greatly limiting the options for treatment. Respiratory syncytial virus (RSV) may cause a lower respiratory tract infection characterized by bronchiolitis and pneumonia mainly in children and the elderly. Infection with RSV can cause severe disease and even death, imposing a severe burden for pediatric and geriatric health systems worldwide. Treatment for RSV is mainly supportive since the only approved therapy, a monoclonal antibody, is recommended for prophylactic use in high-risk patients. The Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly emerging respiratory virus. The virus was first recognized in 2012 and it is associated with a lower respiratory tract disease that is more severe in patients with comorbidities. No licensed vaccines or antivirals have been yet approved for the treatment of MERS-CoV in humans. It is clear that the discovery and development of novel antivirals that can be used alone or in combination with existing therapies to treat these important respiratory viral infections are critical. In this review, we will describe some of the novel therapeutics currently under development for the treatment of these infections.
Collapse
Affiliation(s)
- Mohammad Amin Behzadi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Victor H Leyva-Grado
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
140
|
Abstract
Respiratory syncytial virus remains a major cause of infantile respiratory illness globally. Infants in the developing world experience the highest burden of mortality and morbidity. Risk factors associated with respiratory syncytial virus infection and progression to severe disease include household crowding, preterm birth and low birth weight. Maternally-derived antibody has a role in protection of infants through transplacental antibody transfer and breast milk antibody. Promising prevention strategies that are under development include vaccination during pregnancy and monoclonal antibody administration at birth.
Collapse
|
141
|
Goldeck D, Perry DM, Hayes JWP, Johnson LPM, Young JE, Roychoudhury P, McLuskey EL, Moffat K, Bakker AQ, Kwakkenbos MJ, Frossard JP, Rowland RRR, Murtaugh MP, Graham SP. Establishment of Systems to Enable Isolation of Porcine Monoclonal Antibodies Broadly Neutralizing the Porcine Reproductive and Respiratory Syndrome Virus. Front Immunol 2019; 10:572. [PMID: 30972067 PMCID: PMC6445960 DOI: 10.3389/fimmu.2019.00572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/04/2019] [Indexed: 02/01/2023] Open
Abstract
The rapid evolution of porcine reproductive and respiratory syndrome viruses (PRRSV) poses a major challenge to effective disease control since available vaccines show variable efficacy against divergent strains. Knowledge of the antigenic targets of virus-neutralizing antibodies that confer protection against heterologous PRRSV strains would be a catalyst for the development of next-generation vaccines. Key to discovering these epitopes is the isolation of neutralizing monoclonal antibodies (mAbs) from immune pigs. To address this need, we sought to establish systems to enable the isolation of PRRSV neutralizing porcine mAbs. We experimentally produced a cohort of immune pigs by sequential challenge infection with four heterologous PRRSV strains spanning PRRSV-1 subtypes and PRRSV species. Whilst priming with PRRSV-1 subtype 1 did not confer full protection against a subsequent infection with a PRRSV-1 subtype 3 strain, animals were protected against a subsequent PRRSV-2 infection. The infection protocol resulted in high serum neutralizing antibody titers against PRRSV-1 Olot/91 and significant neutralization of heterologous PRRSV-1/-2 strains. Enriched memory B cells isolated at the termination of the study were genetically programmed by transduction with a retroviral vector expressing the Bcl-6 transcription factor and the anti-apoptotic Bcl-xL protein, a technology we demonstrated efficiently converts porcine memory B cells into proliferating antibody-secreting cells. Pools of transduced memory B cells were cultured and supernatants containing PRRSV-specific antibodies identified by flow cytometric staining of infected MARC-145 cells and in vitro neutralization of PRRSV-1. Collectively, these data suggest that this experimental system may be further exploited to produce a panel of PRRSV-specific mAbs, which will contribute both to our understanding of the antibody response to PRRSV and allow epitopes to be resolved that may ultimately guide the design of immunogens to induce cross-protective immunity.
Collapse
Affiliation(s)
| | - Dana M Perry
- The Pirbright Institute, Pirbright, United Kingdom.,School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jack W P Hayes
- The Pirbright Institute, Pirbright, United Kingdom.,School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Luke P M Johnson
- The Pirbright Institute, Pirbright, United Kingdom.,School of Veterinary Science, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jordan E Young
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Parimal Roychoudhury
- The Pirbright Institute, Pirbright, United Kingdom.,College of Veterinary Science and Animal Husbandry, Central Agricultural University, Aizawl, India
| | - Elle L McLuskey
- The Pirbright Institute, Pirbright, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Katy Moffat
- The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Jean-Pierre Frossard
- Department of Virology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Raymond R R Rowland
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Michael P Murtaugh
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Simon P Graham
- The Pirbright Institute, Pirbright, United Kingdom.,School of Veterinary Science, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
142
|
Bin Lu, Liu H, Tabor DE, Tovchigrechko A, Qi Y, Ruzin A, Esser MT, Jin H. Emergence of new antigenic epitopes in the glycoproteins of human respiratory syncytial virus collected from a US surveillance study, 2015-17. Sci Rep 2019; 9:3898. [PMID: 30846850 PMCID: PMC6405860 DOI: 10.1038/s41598-019-40387-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of lower respiratory tract infection in infants and elderly. To understand the evolution of neutralizing epitopes on the RSV glycoprotein (G) and fusion (F) proteins, we conducted a multi-year surveillance program (OUTSMART-RSV) in the US. Analysis of 1,146 RSV samples from 2015-2017 revealed a slight shift in prevalence from RSV A (58.7%) to B (53.7%) between the two seasons. RSV B was more prevalent in elderly (52.9% and 73.4%). Approximately 1% of the samples contained both RSV A and B viruses. All RSV A isolates were ON1 and almost all the B isolates were BA9 genotypes. Compared with the 2013 reference sequences, changes at the F antigenic sites of RSV B were greater than RSV A, which mainly occurred at antigenic sites V (L172Q/S173L at 99.6%), Ø (I206M/Q209K at 18.6%) and IV (E463D at 7%) of RSV B F. Sequence diversities in the G protein second hypervariable region were observed in the duplicated regions for RSV A and B, and at the G stop codon resulting in extension of 7 amino acids (22.1%) for RSV B in 2016-17. Thus, RSV surface glycoproteins are continuously evolving, and continued surveillance is important for the clinical evaluation of immunoprophylactic products.
Collapse
Affiliation(s)
- Bin Lu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | - Hui Liu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Yanping Qi
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Hong Jin
- MedImmune/AstraZeneca, South San Francisco, CA, USA.
| |
Collapse
|
143
|
Introduction of the YTE mutation into the non-immunogenic HIV bnAb PGT121 induces anti-drug antibodies in macaques. PLoS One 2019; 14:e0212649. [PMID: 30785963 PMCID: PMC6437720 DOI: 10.1371/journal.pone.0212649] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/06/2019] [Indexed: 11/21/2022] Open
Abstract
Recombinant antibodies play increasingly important roles as immunotherapeutic
treatments for human cancers as well as inflammatory and infectious diseases and
have revolutionized their management. In addition, their therapeutic potential
may be enhanced by the introduction of defined mutations in the crystallizable
fragment (Fc) domains eg YTE (M252Y/S254T/T256E) and LS (M428L/N434S), as a
consequence of increased half-lives and prolonged duration of protection.
However, the functional properties of any biologic may be compromised by
unanticipated immunogenicity in humans, rendering them ineffective. Several
potent broadly neutralizing HIV monoclonal antibodies (bnAbs) have been
identified that protect against SHIV challenge in macaque models and reduce HIV
viremia in HIV-infected individuals. In the present study, the pharmacokinetics
and immunogenicity of one or more 5mg/kg subcutaneous (SC) injections in naïve
macaques of the HIV bnAb PGT121 and its PGT121-YTE mutant, both produced in
plants, have been compared towards prolonging efficacy. Induction of
anti-drug/anti-idiotypic antibodies (ADA, anti-id) has been monitored using both
binding ELISAs and more functional inhibition of virus neutralization (ID50)
assays. Timing of the anti-Id responses and their impact on pharmacokinetic
profiles (clearance) and efficacy (protection) have also been assessed. The
results indicate that ADA induction in naïve macaques may result both from
injection of the previously non-immunogenic PGT121 into pre-primed animals and
also by the introduction of the YTE mutation. Binding ADA antibody levels,
induced in 7/10 macaques within two weeks of a first or second PGT121-YTE
injection, were closely associated with both reduced pharmacokinetic profiles
and loss of protection. However no correlation was observed with inhibitory ADA
activity. These studies provide insights into both the structural features of
bnAb and the immune status of the host which may contribute to the development
of ADA in macaques and describe possible YTE-mediated changes in
structure/orientation of HIV bnAbs that trigger such responses.
Collapse
|
144
|
Gomi R, Sharma A, Wu W, Worgall S. Neonatal Genetic Delivery of Anti-Respiratory Syncytial Virus (RSV) Antibody by Non-Human Primate-Based Adenoviral Vector to Provide Protection against RSV. Vaccines (Basel) 2018; 7:vaccines7010003. [PMID: 30597977 PMCID: PMC6466083 DOI: 10.3390/vaccines7010003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/17/2018] [Accepted: 12/22/2018] [Indexed: 12/17/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of lower respiratory tract infection in infants. Immunoprophylaxis with the anti-RSV monoclonal antibody, palivizumab, reduces the risk for RSV-related hospitalizations, but its use is restricted to high-risk infants due to the high costs. In this study, we investigated if genetic delivery of anti-RSV antibody to neonatal mice by chimpanzee adenovirus type 7 expressing the murine form of palivizumab (AdC7αRSV) can provide protection against RSV. Intranasal and intramuscular administration of AdC7αRSV to adult mice resulted in similar levels of anti-RSV IgG in the serum. However, only intranasal administration resulted in detectable levels of anti-RSV IgG in the bronchoalveolar lavage fluid. Intranasal administration of AdC7αRSV provided protection against subsequent RSV challenge. Expression of the anti-RSV antibody was prolonged following intranasal administration of AdC7αRSV to neonatal mice. Protection against RSV was confirmed at 6 weeks of age. These data suggest that neonatal genetic delivery of anti-RSV antibody by AdC7αRSV can provide protection against RSV.
Collapse
Affiliation(s)
- Rika Gomi
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Anurag Sharma
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Wenzhu Wu
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA.
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
145
|
Improvement of pharmacokinetic properties of therapeutic antibodies by antibody engineering. Drug Metab Pharmacokinet 2018; 34:25-41. [PMID: 30472066 DOI: 10.1016/j.dmpk.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAbs) have become an important therapeutic option for several diseases. Since several mAbs have shown promising efficacy in clinic, the competition to develop mAbs has become severe. In efforts to gain a competitive advantage over other mAbs and provide significant benefits to patients, innovations in antibody engineering have aimed at improving the pharmacokinetic properties of mAbs. Because engineering can provide therapeutics that are more convenient, safer, and more efficacious for patients in several disease areas, it is an attractive approach to provide significant benefits to patients. Further advances in engineering mAbs to modulate their pharmacokinetics were driven by the increase of total soluble target antigen concentration that is often observed after injecting a mAb, which then requires a high dosage to antagonize. To decrease the required dosage, several antibody engineering techniques have been invented that reduce the total concentration of soluble target antigen. Here, we review the various ways that antibody engineering can improve the pharmacokinetic properties of mAbs.
Collapse
|
146
|
Bidmos FA, Siris S, Gladstone CA, Langford PR. Bacterial Vaccine Antigen Discovery in the Reverse Vaccinology 2.0 Era: Progress and Challenges. Front Immunol 2018; 9:2315. [PMID: 30349542 PMCID: PMC6187972 DOI: 10.3389/fimmu.2018.02315] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/17/2018] [Indexed: 11/13/2022] Open
Abstract
The ongoing, and very serious, threat from antimicrobial resistance necessitates the development and use of preventative measures, predominantly vaccination. Polysaccharide-based vaccines have provided a degree of success in limiting morbidity from disseminated bacterial infections, including those caused by the major human obligate pathogens, Neisseria meningitidis, and Streptococcus pneumoniae. Limitations of these polysaccharide vaccines, such as partial coverage and induced escape leading to persistence of disease, provide a compelling argument for the development of protein vaccines. In this review, we briefly chronicle approaches that have yielded licensed vaccines before highlighting reverse vaccinology 2.0 and its potential application in the discovery of novel bacterial protein vaccine candidates. Technical challenges and research gaps are also discussed.
Collapse
Affiliation(s)
- Fadil A Bidmos
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Sara Siris
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Paul R Langford
- Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
147
|
Ruzin A, Pastula ST, Levin-Sparenberg E, Jiang X, Fryzek J, Tovchigrechko A, Lu B, Qi Y, Liu H, Jin H, Yu L, Hackett J, Villafana T, Esser MT. Characterization of circulating RSV strains among subjects in the OUTSMART-RSV surveillance program during the 2016-17 winter viral season in the United States. PLoS One 2018; 13:e0200319. [PMID: 30040837 PMCID: PMC6057637 DOI: 10.1371/journal.pone.0200319] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 06/22/2018] [Indexed: 01/10/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is an established cause of serious lower respiratory disease in infants, elderly and high-risk populations. The OUTSMART surveillance program aims to characterize patient populations and currently circulating RSV strains, and monitor temporal and geographic evolution of RSV F and G proteins in the U.S. Methods The OUTSMART 2016–17 study collected RSV-positive samples from 25 RSVAlert® laboratories from 4 U.S. regions and Puerto Rico during November 2016 through March 2017. Frequencies of A and B subtypes and genotypes were determined for several demographic and geographic variables. To gauge the representativeness of the OUTSMART patients, results were compared to discharge data from the NEDS and NIS databases. Results A total of 1,041 RSV-positive samples with associated demographic data were obtained and the RSV F gene and second variable region of the G gene were sequenced. The majority of samples (76.0%) came from children under 2 years old: <1 year (48.4%), 1–2 years (27.6%). The OUTSMART patient sample was similar to NEDS and NIS for age, gender, and geographic location. Both OUTSMART and national RSV cases peaked in January. Of OUTSMART samples, 45.3% were subtype A, 53.7% were subtype B and 1.0% were mixed A and B. The percentage of RSV B cases increased with increasing age. Hospitalization (length of hospital stay, LOS, >24 hrs) occurred in 29.0% of patients of which 52.0% had RSV B. Outpatients (LOS <24 hrs) were 64.4% of total of which 73.3% were diagnosed in the ER and discharged, while only 6% were diagnosed in other outpatient settings. Conclusions The OUTSMART 2016–17 study was representative of the U.S. RSV experience. Geographic and temporal information from the RSV surveillance program will be used to establish a molecular baseline of RSV F and G sequence variability and to help inform development of novel agents for RSV prophylaxis and treatment.
Collapse
Affiliation(s)
- Alexey Ruzin
- AstraZeneca/MedImmune, Gaithersburg, Maryland, United States of America
- * E-mail:
| | - Susan T. Pastula
- Epidstat Institute, Ann Arbor, Michigan, United States of America
| | | | - Xiaohui Jiang
- Epidstat Institute, Ann Arbor, Michigan, United States of America
| | - Jon Fryzek
- Epidstat Institute, Ann Arbor, Michigan, United States of America
| | | | - Bin Lu
- AstraZeneca/MedImmune, Mountain View, California, United States of America
| | - Yanping Qi
- AstraZeneca/MedImmune, Mountain View, California, United States of America
| | - Hui Liu
- AstraZeneca/MedImmune, Mountain View, California, United States of America
| | - Hong Jin
- AstraZeneca/MedImmune, Mountain View, California, United States of America
| | - Li Yu
- AstraZeneca/MedImmune, Gaithersburg, Maryland, United States of America
| | - Judith Hackett
- AstraZeneca/MedImmune, Gaithersburg, Maryland, United States of America
| | - Tonya Villafana
- AstraZeneca/MedImmune, Gaithersburg, Maryland, United States of America
| | - Mark T. Esser
- AstraZeneca/MedImmune, Gaithersburg, Maryland, United States of America
| |
Collapse
|