51
|
Rocca A, Biagi C, Scarpini S, Dondi A, Vandini S, Pierantoni L, Lanari M. Passive Immunoprophylaxis against Respiratory Syncytial Virus in Children: Where Are We Now? Int J Mol Sci 2021; 22:3703. [PMID: 33918185 PMCID: PMC8038138 DOI: 10.3390/ijms22073703] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) represents the main cause of acute respiratory tract infections in children worldwide and is the leading cause of hospitalization in infants. RSV infection is a self-limiting condition and does not require antibiotics. However hospitalized infants with clinical bronchiolitis often receive antibiotics for fear of bacteria coinfection, especially when chest radiography is performed due to similar radiographic appearance of infiltrate and atelectasis. This may lead to unnecessary antibiotic prescription, additional cost, and increased risk of development of resistance. Despite the considerable burden of RSV bronchiolitis, to date, only symptomatic treatment is available, and there are no commercially available vaccines. The only licensed passive immunoprophylaxis is palivizumab. The high cost of this monoclonal antibody (mAb) has led to limiting its prescription only for high-risk children: infants with chronic lung disease, congenital heart disease, neuromuscular disorders, immunodeficiencies, and extreme preterm birth. Nevertheless, it has been shown that the majority of hospitalized RSV-infected children do not fully meet the criteria for immune prophylaxis. While waiting for an effective vaccine, passive immune prophylaxis in children is mandatory. There are a growing number of RSV passive immunization candidates under development intended for RSV prevention in all infants. In this review, we describe the state-of-the-art of palivizumab's usage and summarize the clinical and preclinical trials regarding the development of mAbs with a better cost-effectiveness ratio.
Collapse
Affiliation(s)
- Alessandro Rocca
- Pediatric Emergency Unit, Scientific Institute for Research and Healthcare (IRCCS), Sant’Orsola Hospital, 40138 Bologna, Italy; (A.R.); (C.B.); (A.D.); (L.P.); (M.L.)
| | - Carlotta Biagi
- Pediatric Emergency Unit, Scientific Institute for Research and Healthcare (IRCCS), Sant’Orsola Hospital, 40138 Bologna, Italy; (A.R.); (C.B.); (A.D.); (L.P.); (M.L.)
| | - Sara Scarpini
- Specialty School of Paediatrics—Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Arianna Dondi
- Pediatric Emergency Unit, Scientific Institute for Research and Healthcare (IRCCS), Sant’Orsola Hospital, 40138 Bologna, Italy; (A.R.); (C.B.); (A.D.); (L.P.); (M.L.)
| | - Silvia Vandini
- Pediatrics and Neonatology Unit, Imola Hospital, 40026 Imola, Italy;
| | - Luca Pierantoni
- Pediatric Emergency Unit, Scientific Institute for Research and Healthcare (IRCCS), Sant’Orsola Hospital, 40138 Bologna, Italy; (A.R.); (C.B.); (A.D.); (L.P.); (M.L.)
| | - Marcello Lanari
- Pediatric Emergency Unit, Scientific Institute for Research and Healthcare (IRCCS), Sant’Orsola Hospital, 40138 Bologna, Italy; (A.R.); (C.B.); (A.D.); (L.P.); (M.L.)
| |
Collapse
|
52
|
Domachowske JB, Anderson EJ, Goldstein M. The Future of Respiratory Syncytial Virus Disease Prevention and Treatment. Infect Dis Ther 2021; 10:47-60. [PMID: 33656652 PMCID: PMC7926075 DOI: 10.1007/s40121-020-00383-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of respiratory tract infections in infants, young children, and older or immunocompromised adults. Although aerosolized ribavirin was licensed for RSV treatment on the basis of data demonstrating a reduced need for supplemental oxygen, ribavirin use is limited because of issues with efficacy, safety, and cost. Currently, the treatment of RSV is primarily supportive. New antiviral treatments for RSV are in the early stages of development, but it will be years until any of these may be licensed by the US Food and Drug Administration (FDA). Palivizumab, an RSV monoclonal antibody [immunoprophylaxis (IP)], has demonstrated effectiveness in disease prevention and is the only licensed IP for RSV disease in specific high-risk pediatric populations. Although its efficacy is well established, some challenges that may interfere with its clinical use include cost, need for monthly injections, and changing policy for use by the American Academy of Pediatrics (AAP). Preventing RSV disease would be possible through RSV vaccine development (e.g., live-attenuated, vector-based subunit, or particle-based). Alternatively, new long-acting monoclonal antibodies have demonstrated promising results in early clinical trials. Despite scientific advances, until new agents become available, palivizumab should continue to be used to reduce RSV disease burden in high-risk patients for whom it is indicated.
Collapse
Affiliation(s)
| | - Evan J Anderson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Mitchell Goldstein
- Department of Pediatrics, Loma Linda University Children's Hospital, Loma Linda, CA, USA.
| |
Collapse
|
53
|
Therapeutic Antibodies for the Treatment of Respiratory Tract Infections-Current Overview and Perspectives. Vaccines (Basel) 2021; 9:vaccines9020151. [PMID: 33668613 PMCID: PMC7917879 DOI: 10.3390/vaccines9020151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Respiratorytract infections (RTIs) are frequent and life-threatening diseases, accounting for several millions of deaths worldwide. RTIs implicate microorganisms, including viruses (influenza virus, coronavirus, respiratory syncytial virus (RSV)), bacteria (Pseudomonas aeruginosa, Streptococcus pneumoniae, Staphylococcus aureus and Bacillus anthracis) and fungi (Pneumocystis spp., Aspergillus spp. and very occasionally Candida spp.). The emergence of new pathogens, like the coronavirus SARS-CoV-2, and the substantial increase in drug resistance have highlighted the critical necessity to develop novel anti-infective molecules. In this context, antibodies (Abs) are becoming increasingly important in respiratory medicine and may fulfill the unmet medical needs of RTIs. However, development of Abs for treating infectious diseases is less advanced than for cancer and inflammatory diseases. Currently, only three Abs have been marketed for RTIs, namely, against pulmonary anthrax and RSV infection, while several clinical and preclinical studies are in progress. This article gives an overview of the advances in the use of Abs for the treatment of RTIs, based on the analysis of clinical studies in this field. It describes the Ab structure, function and pharmacokinetics, and discusses the opportunities offered by the various Ab formats, Ab engineering and co-treatment strategies. Including the most recent literature, it finally highlights the strengths, weaknesses and likely future trends of a novel anti-RTI Ab armamentarium.
Collapse
|
54
|
Current State and Challenges in Developing Respiratory Syncytial Virus Vaccines. Vaccines (Basel) 2020; 8:vaccines8040672. [PMID: 33187337 PMCID: PMC7711987 DOI: 10.3390/vaccines8040672] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/01/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the main cause of acute respiratory tract infections in infants and it also induces significant disease in the elderly. The clinical course may be severe, especially in high-risk populations (infants and elderly), with a large number of deaths in developing countries and of intensive care hospitalizations worldwide. To date, prevention strategies against RSV infection is based on hygienic measures and passive immunization with humanized monoclonal antibodies, limited to selected high-risk children due to their high costs. The development of a safe and effective vaccine is a global health need and an important objective of research in this field. A growing number of RSV vaccine candidates in different formats (particle-based vaccines, vector-based vaccines, subunit vaccines and live-attenuated vaccines) are being developed and are now at different stages, many of them already being in the clinical stage. While waiting for commercially available safe and effective vaccines, immune prophylaxis in selected groups of high-risk populations is still mandatory. This review summarizes the state-of-the-art of the RSV vaccine research and its implications for clinical practice, focusing on the characteristics of the vaccines that reached the clinical stage of development.
Collapse
|
55
|
Aliprantis AO, Wolford D, Caro L, Maas BM, Ma H, Montgomery DL, Sterling LM, Hunt A, Cox KS, Vora KA, Roadcap BA, Railkar RA, Lee AW, Stoch SA, Lai E. A Phase 1 Randomized, Double-Blind, Placebo-Controlled Trial to Assess the Safety, Tolerability, and Pharmacokinetics of a Respiratory Syncytial Virus Neutralizing Monoclonal Antibody MK-1654 in Healthy Adults. Clin Pharmacol Drug Dev 2020; 10:556-566. [PMID: 33125189 DOI: 10.1002/cpdd.883] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infection and related morbidity and mortality in infants. Passive immunization with an RSV-neutralizing antibody can provide rapid protection to this vulnerable population. Proof-of-concept for this approach has been demonstrated by palivizumab; however, the use of this antibody is generally restricted to the highest-risk infants due to monthly dosing requirements and its cost. To address the large unmet medical need for most infants, we are evaluating MK-1654, a fully human RSV-neutralizing antibody with half-life extending mutations targeting site IV of the fusion protein. In this 2-part, placebo-controlled, double-blind, first-in-human study, 152 healthy adults were randomized 3:1 to receive a single dose of MK-1654 or placebo in 5 cohorts (100 or 300 mg as an intramuscular dose or 300, 1000, or 3000 mg as an intravenous dose). Safety, pharmacokinetics, antidrug antibodies, and RSV serum-neutralizing antibody titers were evaluated through 1 year. MK-1654 serum concentrations increased proportionally with dose and resulted in corresponding elevations in RSV serum-neutralizing antibody titers. The antibody displayed a half-life of 73 to 88 days and an estimated bioavailability of 69% at the 300-mg dose. The overall safety profile of MK-1654 was similar to placebo, and treatment-emergent antidrug antibodies were low (2.6%) with no associated adverse events. These data support the continued development of MK-1654 for the prevention of RSV disease in infants.
Collapse
Affiliation(s)
| | | | | | | | - Hua Ma
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | - Kara S Cox
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | | | | | - Eseng Lai
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
56
|
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
|
57
|
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
|
58
|
Schultheis K, Pugh HM, Oh J, Nguyen J, Yung B, Reed C, Cooch N, Chen J, Yan J, Muthumani K, Humeau LM, Weiner DB, Broderick KE, Smith TRF. Active immunoprophylaxis with a synthetic DNA-encoded monoclonal anti-respiratory syncytial virus scFv-Fc fusion protein confers protection against infection and durable activity. Hum Vaccin Immunother 2020; 16:2165-2175. [PMID: 32544376 PMCID: PMC7553682 DOI: 10.1080/21645515.2020.1748979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Respiratory Syncytial virus (RSV) is a major threat to many vulnerable populations. There are currently no approved vaccines, and RSV remains a high unmet global medical need. Here we describe the employment of a novel synthetic DNA-encoded antibody technology platform to develop and deliver an engineered human DNA-encoded monoclonal antibody (dMAbTM) targeting the fusion protein (F) of RSV as a new approach to prevention or therapy of at risk populations. In in vivo models, a single administration of synthetic DNA-encoding the single-chain fragment variable-constant fragment (scFv-Fc) RSV-F dMAb resulted in robust and durable circulating levels of a functional antibody systemically and in mucosal tissue. In cotton rats, which are the gold-standard animals to model RSV infection, we observed sustained scFv-Fc RSV-F dMAb in the sera and lung-lavage samples, demonstrating the potential for both long-lasting immunity to RSV and effective biodistribution. The scFv-Fc RSV-F dMAb harbored in the sera exhibited RSV antigen-specific binding and potent viral neutralizing activity. Importantly, in vivo delivery of synthetic DNA-encoding, the scFv-Fc RSV-F dMAb protected animals against viral challenge. Our findings support the significance of dMAbs as a potential platform technology for durable protection against RSV disease.
Collapse
Affiliation(s)
| | - Holly M Pugh
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Janet Oh
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | | | - Bryan Yung
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Charles Reed
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Neil Cooch
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Jing Chen
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Jian Yan
- Inovio Pharmaceuticals , Plymouth Meeting, PA, USA
| | - Kar Muthumani
- Vaccine & Immunotherapy Center, The Wistar Institute , Philadelphia, PA, USA
| | | | - David B Weiner
- Vaccine & Immunotherapy Center, The Wistar Institute , Philadelphia, PA, USA
| | | | | |
Collapse
|
59
|
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
|
60
|
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
|
61
|
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
|
62
|
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
|
63
|
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
|
64
|
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
|
65
|
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
|
66
|
Kernstock R, Sperinde G, Finco D, Davis R, Montgomery D. Clinical Immunogenicity Risk Assessment Strategy for a Low Risk Monoclonal Antibody. AAPS JOURNAL 2020; 22:60. [PMID: 32185565 DOI: 10.1208/s12248-020-00440-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/20/2020] [Indexed: 12/31/2022]
Abstract
This article provides a theoretical case-study risk assessment report for a low-risk monoclonal antibody (mAb) therapeutic. In terms of risk, there are considerations around risks to safety, but also risks regarding effects on pharmacokinetics (PK), pharmacodynamics (PD), and efficacy. Much of the discussion in this document is around the risk of immunogenicity incidence. A higher incidence of immunogenicity would necessitate a detailed review of the PK, efficacy and safety in anti-drug antibody (ADA) positive and ADA negative subjects, in order to evaluate potential effects. The publication is intended to provide a framework of some the current thought processes around assessing immunogenicity risk and for building strategies to mitigate those risks. For this example, we have created a hypothetical antibody, ABC-123, targeting a membrane protein on antigen presenting cells, for the treatment of rheumatoid arthritis (RA). This hypothetical antibody therapeutic is provided as an example for the purposes of risk assessment for a low risk molecule, although any application of similar approach would be case by case.
Collapse
Affiliation(s)
| | - Gizette Sperinde
- BioAnalytical Sciences, Genentech, Inc., South San Francisco, CA, USA
| | | | | | - Diana Montgomery
- Predictive and Clinical Immunogenicity, Merck & Co., Inc., West Point, PA, USA.
| |
Collapse
|
67
|
Liu H, Lu B, Tabor DE, Tovchigrechko A, Wilkins D, Jin H, Madhi SA, Soofie N, Esser MT, Nunes MC. Characterization of human respiratory syncytial virus (RSV) isolated from HIV-exposed-uninfected and HIV-unexposed infants in South Africa during 2015-2017. Influenza Other Respir Viruses 2020; 14:403-411. [PMID: 32126161 PMCID: PMC7298309 DOI: 10.1111/irv.12727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND RSV is a leading cause of lower respiratory tract infection in infants. Monitoring RSV glycoprotein sequences is critical for understanding RSV epidemiology and viral antigenicity in the effort to develop anti-RSV prophylactics and therapeutics. OBJECTIVES The objective is to characterize the circulating RSV strains collected from infants in South Africa during 2015-2017. METHODS A subset of 150 RSV-positive samples obtained in South Africa from HIV-unexposed and HIV-exposed-uninfected infants from 2015 to 2017, were selected for high-throughput next-generation sequencing of the RSV F and G glycoprotein genes. The RSV G and F sequences were analyzed by a bioinformatic pipeline and compared to the USA samples from the same three-year period. RESULTS Both RSV A and RSV B co-circulated in South Africa during 2015-2017, with a shift from RSV A (58%-61% in 2015-2016) to RSV B (69%) in 2017. RSV A ON1 and RSV B BA9 genotypes emerged as the most prevalent genotypes in 2017. Variations at the F protein antigenic sites were observed for both RSV A and B strains, with dominant changes (L172Q/S173L) at antigenic site V observed in RSV B strains. RSV A and B F protein sequences from South Africa were very similar to the USA isolates except for a higher rate of RSV A NA1 and RSV B BA10 genotypes in South Africa. CONCLUSION RSV G and F genes continue to evolve and exhibit both local and global circulation patterns in South Africa, supporting the need for continued national surveillance.
Collapse
Affiliation(s)
- Hui Liu
- AstraZeneca, South San Francisco, CA, USA
| | - Bin Lu
- AstraZeneca, South San Francisco, CA, USA
| | | | | | | | - Hong Jin
- AstraZeneca, South San Francisco, CA, USA
| | - Shabir A Madhi
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nasiha Soofie
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Marta C Nunes
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
68
|
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
|
69
|
Nguyen AW, DiVenere AM, Papin JF, Connelly S, Kaleko M, Maynard JA. Neutralization of pertussis toxin by a single antibody prevents clinical pertussis in neonatal baboons. SCIENCE ADVANCES 2020; 6:eaay9258. [PMID: 32076653 PMCID: PMC7002138 DOI: 10.1126/sciadv.aay9258] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/20/2019] [Indexed: 05/13/2023]
Abstract
Pertussis continues to cause considerable infant mortality world-wide, which could be addressed in part by passive immunization strategies. Antibody hu1B7 is a candidate therapeutic that potently neutralizes pertussis toxin in vitro, prevents leukocytosis in mice and treats established disease in weanling baboons as part of an antibody cocktail. Here, we evaluated the potential for hu1B7 and an extended half-life hu1B7 variant to prevent death, leukocytosis and other clinical symptoms in a newborn baboon model that mimics many aspects of human disease. We administered a single antibody dose to newborn baboons five weeks prior to experimental infection. While all animals were heavily colonized with Bordetella pertussis, prophylaxed animals showed significantly greater survival (P < 0.005), delayed and suppressed leukocytosis (P < 0.01) and enhanced clinical outcomes, including coughing (P < 0.01), as compared to controls. Together, this work demonstrates that a single neutralizing anti-PTx antibody is sufficient to prevent clinical pertussis symptoms.
Collapse
Affiliation(s)
- Annalee W. Nguyen
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea M. DiVenere
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - James F. Papin
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sheila Connelly
- Synthetic Biologics, 9605 Medical Center Dr., Suite 270, Rockville, MD 20850, USA
| | - Michael Kaleko
- Synthetic Biologics, 9605 Medical Center Dr., Suite 270, Rockville, MD 20850, USA
| | - Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
70
|
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
|
71
|
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
|
72
|
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
|
73
|
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
|
74
|
Sedeyn K, Saelens X. New antibody-based prevention and treatment options for influenza. Antiviral Res 2019; 170:104562. [PMID: 31323236 DOI: 10.1016/j.antiviral.2019.104562] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/16/2019] [Indexed: 12/23/2022]
Abstract
The antigenic diversity of human influenza viruses represents a challenge to the development of vaccines with durable immune protection. In addition, small molecule anti-influenza viral drugs can bring clinical relief to influenza patients but the emergence of drug resistant viruses can rapidly limit the effectiveness of such drugs. In the past decade, a number of human monoclonal antibodies have been described that can bind to and neutralize a broad range of influenza A and B viruses. Most of these monoclonal antibodies are directed against the viral hemagglutinin (HA) stalk and some have now been evaluated in early to mid-stage clinical trials. An important conclusion from these clinical studies is that hemagglutinin stalk-specific antibodies are safe and can reduce influenza symptoms. In addition, examples of bi- and multi-specific anti-influenza antibodies are discussed, although such antibodies have not yet progressed into clinical testing. In the future, antibody-based therapies might become part of our arsenal to prevent and treat influenza.
Collapse
Affiliation(s)
- Koen Sedeyn
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biochemistry and Microbiology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biochemistry and Microbiology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
| |
Collapse
|
75
|
Nyanguile O. Peptide Antiviral Strategies as an Alternative to Treat Lower Respiratory Viral Infections. Front Immunol 2019; 10:1366. [PMID: 31293570 PMCID: PMC6598224 DOI: 10.3389/fimmu.2019.01366] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/29/2019] [Indexed: 01/24/2023] Open
Abstract
Lower respiratory infection caused by human pathogens such as influenza and respiratory syncytial virus (RSV) is a significant healthcare burden that must be addressed. The preferred options to achieve this goal are usually to develop vaccines for prophylaxis and to develop antiviral small molecules to treat infected patients with convenient, orally administrable drugs. However, developing a vaccine against RSV poses special challenges with the diminished immune system of infants and the elderly, and finding a universal flu vaccine is difficult because the product must target a large array of viral strains. On the other hand, the use of small-molecule antivirals can result in the emergence of resistant viruses as it has well-been reported for HIV, influenza, and hepatitis C virus (HCV). This paper reviews peptide antiviral strategies as an alternative to address these challenges. The discovery of influenza and RSV peptidic fusion inhibitors will be discussed and compared to small molecules in view of escape mutations. The importance of constraining peptides into macrocycles to improve both their inhibitory activity and pharmacological properties will be highlighted.
Collapse
Affiliation(s)
- Origène Nyanguile
- HES-SO Valais-Wallis, Institute of Life Technologies, Sion, Switzerland
| |
Collapse
|
76
|
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
|
77
|
Saunders KO. Conceptual Approaches to Modulating Antibody Effector Functions and Circulation Half-Life. Front Immunol 2019; 10:1296. [PMID: 31231397 PMCID: PMC6568213 DOI: 10.3389/fimmu.2019.01296] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 05/21/2019] [Indexed: 12/31/2022] Open
Abstract
Antibodies and Fc-fusion antibody-like proteins have become successful biologics developed for cancer treatment, passive immunity against infection, addiction, and autoimmune diseases. In general these biopharmaceuticals can be used for blocking protein:protein interactions, crosslinking host receptors to induce signaling, recruiting effector cells to targets, and fixing complement. With the vast capability of antibodies to affect infectious and genetic diseases much effort has been placed on improving and tailoring antibodies for specific functions. While antibody:antigen engagement is critical for an efficacious antibody biologic, equally as important are the hinge and constant domains of the heavy chain. It is the hinge and constant domains of the antibody that engage host receptors or complement protein to mediate a myriad of effector functions and regulate antibody circulation. Molecular and structural studies have provided insight into how the hinge and constant domains from antibodies across different species, isotypes, subclasses, and alleles are recognized by host cell receptors and complement protein C1q. The molecular details of these interactions have led to manipulation of the sequences and glycosylation of hinge and constant domains to enhance or reduce antibody effector functions and circulating half-life. This review will describe the concepts being applied to optimize the hinge and crystallizable fragment of antibodies, and it will detail how these interactions can be tuned up or down to mediate a biological function that confers a desired disease outcome.
Collapse
Affiliation(s)
- Kevin O. Saunders
- Laboratory of Protein Expression, Departments of Surgery, Molecular Genetics and Microbiology, and Immunology, Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| |
Collapse
|
78
|
Aranda SS, Polack FP. Prevention of Pediatric Respiratory Syncytial Virus Lower Respiratory Tract Illness: Perspectives for the Next Decade. Front Immunol 2019; 10:1006. [PMID: 31134078 PMCID: PMC6524688 DOI: 10.3389/fimmu.2019.01006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/18/2019] [Indexed: 12/30/2022] Open
Abstract
The landscape of infant bronchiolitis and viral pneumonia may be altered by preventive interventions against respiratory syncytial virus under evaluation today. Pediatric wards in 2018 in developing countries may differ from those attended by future generation pediatricians who may not witness the packed emergency rooms, lack of available beds, or emergency situations that all physicians caring for children with RSV experience every year. In this review, we describe and discuss different prevention strategies under evaluation to protect pediatric patients. Then, we outline a number of potential challenges, benefits, and concerns that may result from successful interventions after licensure.
Collapse
|
79
|
Jares Baglivo S, Polack FP. The long road to protect infants against severe RSV lower respiratory tract illness. F1000Res 2019; 8. [PMID: 31105933 PMCID: PMC6498742 DOI: 10.12688/f1000research.18749.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2019] [Indexed: 12/12/2022] Open
Abstract
Severe respiratory syncytial virus (RSV) lower respiratory tract illness (LRTI) in infants has proven challenging to prevent. In the last 50 years, conceptually different approaches failed to evolve into viable preventive alternatives for routine use. Inactivated RSV vaccine (that is, formalin-inactivated RSV) elicited severe LRTI in RSV-infected toddlers pre-immunized as infants; early purified F protein approaches in pregnant women failed to elicit sufficient immunity more than a decade ago; a second-generation monoclonal antibody (mAb) of high potency against the virus (that is, motavizumab) caused severe adverse reactions in the skin, and owing to lack of efficacy against RSV subgroup B, an extended half-life mAb targeting site V in the RSV fusion protein (that is, REG2222) did not meet its primary endpoint. In the meantime, two protein F vaccines failed to prevent medically attended LRTI in the elderly. However, palivizumab and the recent results of the Novavax maternal immunization trial with ResVax demonstrate that severe RSV LRTI can be prevented by mAb and by maternal immunization (at least to a certain extent). In fact, disease prevention may also decrease the rates of recurrent wheezing and all-cause pneumonia for at least 180 days. In this review, we discuss the history of RSV vaccine development, previous and current vaccine strategies undergoing evaluation, and recent information about disease burden and its implications for the effects of successful preventive strategies.
Collapse
|
80
|
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
|
81
|
Paes B, Carbonell-Estrany X. Respiratory syncytial virus prophylaxis for children with chronic lung disease: have we got the criteria right? Expert Rev Anti Infect Ther 2019; 17:211-222. [DOI: 10.1080/14787210.2019.1581062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Bosco Paes
- Department of Pediatrics (Neonatal Division), McMaster Children’s Hospital, Hamilton, Ontario, Canada
| | - Xavier Carbonell-Estrany
- Hospital Clinic, Institut d’Investigacions Biomediques August Pi Suner (IDIBAPS), Barcelona, Spain
| |
Collapse
|
82
|
Chow TW, Wright MR, Hop CECA, Wong H. Evaluation of the predictive performance of physiologically based pharmacokinetic models for intramuscular injections of therapeutic proteins. Xenobiotica 2019; 49:1423-1433. [PMID: 30794022 DOI: 10.1080/00498254.2019.1571651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Several physiologically-based pharmacokinetic (PBPK) models have been reported for intravenous (IV) and subcutaneous (SC) injections, but there has been a paucity of work for intramuscular (IM) injections. The primary objective of this work was a wide-scale evaluation of the predictive performance of IM PBPK models of therapeutic proteins. PBPK models for all administration routes available in the literature have regarded muscle as the total muscle (TM) in the body; however, anatomically, the body is composed of discrete muscle groups. Clinically, IM is administered to a specific muscle (SM). We explored the predictive performance of IM PBPK models with an SM or TM dosing site. The plasma concentration-time profiles of seven therapeutic proteins after an IM dose in humans served as the clinically observed data for model evaluation - this was a diverse group ranging from 30 to 149 kDa from six protein classes. Pharmacokinetic parameters Cmax, tmax, AUC0-∞, and ka were estimated. SM and TM IM PBPK approaches were compared using Average Fold Error (AFE) and Pearson Chi-Square LineShape analyses. This work represents the first wide-scale validation of IM PBPK models and suggests that these models predict IM PBPK reasonably well. The SM and TM approach provided comparable performance.
Collapse
Affiliation(s)
- Timothy W Chow
- Faculty of Pharmaceutical Sciences, The University of British Columbia , Vancouver , BC , Canada
| | - Matthew R Wright
- Department of Drug Metabolism and Pharmacokinetics, Genentech , South San Francisco , CA , USA
| | - Cornelis E C A Hop
- Department of Drug Metabolism and Pharmacokinetics, Genentech , South San Francisco , CA , USA
| | - Harvey Wong
- Faculty of Pharmaceutical Sciences, The University of British Columbia , Vancouver , BC , Canada
| |
Collapse
|
83
|
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
|
84
|
Keeffe JR, Van Rompay KKA, Olsen PC, Wang Q, Gazumyan A, Azzopardi SA, Schaefer-Babajew D, Lee YE, Stuart JB, Singapuri A, Watanabe J, Usachenko J, Ardeshir A, Saeed M, Agudelo M, Eisenreich T, Bournazos S, Oliveira TY, Rice CM, Coffey LL, MacDonald MR, Bjorkman PJ, Nussenzweig MC, Robbiani DF. A Combination of Two Human Monoclonal Antibodies Prevents Zika Virus Escape Mutations in Non-human Primates. Cell Rep 2018; 25:1385-1394.e7. [PMID: 30403995 PMCID: PMC6268006 DOI: 10.1016/j.celrep.2018.10.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/15/2018] [Accepted: 10/05/2018] [Indexed: 11/17/2022] Open
Abstract
Zika virus (ZIKV) causes severe neurologic complications and fetal aberrations. Vaccine development is hindered by potential safety concerns due to antibody cross-reactivity with dengue virus and the possibility of disease enhancement. In contrast, passive administration of anti-ZIKV antibodies engineered to prevent enhancement may be safe and effective. Here, we report on human monoclonal antibody Z021, a potent neutralizer that recognizes an epitope on the lateral ridge of the envelope domain III (EDIII) of ZIKV and is protective against ZIKV in mice. When administered to macaques undergoing a high-dose ZIKV challenge, a single anti-EDIII antibody selected for resistant variants. Co-administration of two antibodies, Z004 and Z021, which target distinct sites on EDIII, was associated with a delay and a 3- to 4-log decrease in peak viremia. Moreover, the combination of these antibodies engineered to avoid enhancement prevented viral escape due to mutation in macaques, a natural host for ZIKV.
Collapse
Affiliation(s)
- Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA; Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Priscilla C Olsen
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Stephanie A Azzopardi
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | | | - Yu E Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jackson B Stuart
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Anil Singapuri
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Jodie Usachenko
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Marianna Agudelo
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Eisenreich
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Lark L Coffey
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| | - Davide F Robbiani
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
85
|
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
|
86
|
Kenmoe S, Bigna JJ, Well EA, Simo FBN, Penlap VB, Vabret A, Njouom R. Prevalence of human respiratory syncytial virus infection in people with acute respiratory tract infections in Africa: A systematic review and meta-analysis. Influenza Other Respir Viruses 2018; 12:793-803. [PMID: 29908103 PMCID: PMC6185896 DOI: 10.1111/irv.12584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2018] [Indexed: 12/31/2022] Open
Abstract
AIM The epidemiology of human respiratory syncytial virus (HRSV) infection has not yet been systematically investigated in Africa. This systematic review and meta-analysis are to estimate the prevalence of HRSV infections in people with acute respiratory tract infections (ARTI) in Africa. METHOD We searched PubMed, EMBASE, Africa Journal Online, and Global Index Medicus to identify observational studies published from January 1, 2000, to August 1, 2017. We used a random-effects model to estimate the prevalence across studies. Heterogeneity (I2 ) was assessed via the chi-square test on Cochran's Q statistic. Review registration: PROSPERO CRD42017076352. RESULTS A total of 67 studies (154 000 participants) were included. Sixty (90%), seven (10%), and no studies had low, moderate, and high risk of bias, respectively. The prevalence of HRSV infection varied widely (range 0.4%-60.4%). The pooled prevalence was 14.6% (95% CI 13.0-16.4, I2 = 98.8%). The prevalence was higher in children (18.5%; 95% CI 15.8-21.5) compared to adults (4.0%; 95% CI 2.2-6.1) and in people with severe respiratory tract infections (17.9%; 95% CI 15.8-20.1) compared to those with benign forms (9.4%; 95% CI 7.4-11.5); P-values <0.0001. The HRSV prevalence was not associated with sex, subregion in Africa, setting, altitude, latitude, longitude, and seasonality. CONCLUSION This study suggests a high prevalence of HRSV in people with ARTI in Africa, particularly among children and people with severe clinical form. All innovative strategies to curb the burden should first focus on children which present the highest HRSV-related burden.
Collapse
Affiliation(s)
- Sebastien Kenmoe
- Department of VirologyNational Influenza CenterCentre Pasteur of CameroonYaoundéCameroon
| | - Jean Joel Bigna
- Department of Epidemiology and Public HealthNational Influenza CenterCentre Pasteur of CameroonYaoundéCameroon
- School of Public HealthFaculty of MedicineUniversity of Paris SudLe Kremlin‐BicêtreFrance
| | | | - Fredy Brice N. Simo
- Department of BiochemistryFaculty of SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Véronique B. Penlap
- Department of BiochemistryFaculty of SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Astrid Vabret
- Normandie UniversitéCaenFrance
- Groupe de Recherche sur l'Adaptation Microbienne (GRAM)Université de CaenCaenFrance
- Laboratoire de VirologieCentre Hospitalo‐Universitaire de CaenCaenFrance
| | - Richard Njouom
- Department of VirologyNational Influenza CenterCentre Pasteur of CameroonYaoundéCameroon
| |
Collapse
|
87
|
Abstract
Respiratory syncytial virus (RSV) is the most common cause of infant hospitalization and causes a high burden of disease in the elderly, too. This enveloped negative-stranded RNA virus has been recently reclassified in the Pneumoviridae family. Infections of the respiratory cells happens when the two major surface glycoproteins, G and F, take contact with the cell receptor CX3CR1 and mediate entry by fusion, respectively. Viral mRNA transcription, genomic RNA synthesis and nucleocapsid formation occur in large cytoplasmic inclusion bodies to avoid recognition by the host innate immune response. Most progeny virions remain associated to the infected cell surface; fusion of infected with adjacent cells results in the formation of large multinucleated syncytia that eventually undergo apoptosis. Desquamated epithelial cells form the plugs that with mucus and fibrin may cause lower airway obstructions. Pathogenetic mechanism of severe RSV disease likely involve both the extent of viral replication and the host immune response. Regarding the latter, single nucleotide polymorphism analysis and genome-wide association studies showed that genetic susceptibility to severe RSV infection is likely a complex trait, in which many different host genetic variants contribute. Recent studies pointed to the fact that bronchiolitis severity depends more on the specific infecting RSV genotypes than on the amount of viral loads. A population-based surveillance system to better define RSV burden of disease would be of valuable help for implementing future vaccination programs.
Collapse
Affiliation(s)
- Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy -
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
88
|
Mas V, Nair H, Campbell H, Melero JA, Williams TC. Antigenic and sequence variability of the human respiratory syncytial virus F glycoprotein compared to related viruses in a comprehensive dataset. Vaccine 2018; 36:6660-6673. [PMID: 30292456 PMCID: PMC6203811 DOI: 10.1016/j.vaccine.2018.09.056] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/18/2018] [Accepted: 09/22/2018] [Indexed: 11/17/2022]
Abstract
A comprehensive analysis of sequence variation was carried out comparing the fusion (F) protein of human respiratory syncytial viruses (hRSV) from antigenic groups A and B with the prototype sequence of the A2 strain, also belonging to antigenic group A. The limited number of full bovine RSV F sequences available were included, as well as an extensive set of F sequences from the related human metapneumovirus (hMPV). The results were analysed in the context of the recently determined three dimensional F protein structures, with antigenic sites mapped to these. Although a high degree of sequence conservation in hRSV F exists, and sequence changes did not correlate with location of antigenic sites, preferential accumulation of amino acid changes in certain antigenic sites was noted. When the analysis was extended to hMPV F, a high number of changes was noticed, in agreement with the limited degree of sequence conservation. However, some conserved regions were noted, which may account for the limited number of cross-reactive monoclonal antibodies described between hRSV F and hMPV F. These results provide information about the degree of sequence and antigenic variation currently found in the F protein of circulating viruses. They highlight the importance of establishing a baseline dataset to monitor for future changes that might evolve should preventative immunological measures be made widely available.
Collapse
Affiliation(s)
- Vicente Mas
- Centro Nacional de Microbiología and CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Harish Nair
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, UK
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, UK
| | - Jose A Melero
- Centro Nacional de Microbiología and CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Thomas C Williams
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, UK.
| |
Collapse
|
89
|
Mazur NI, Higgins D, Nunes MC, Melero JA, Langedijk AC, Horsley N, Buchholz UJ, Openshaw PJ, McLellan JS, Englund JA, Mejias A, Karron RA, Simões EA, Knezevic I, Ramilo O, Piedra PA, Chu HY, Falsey AR, Nair H, Kragten-Tabatabaie L, Greenough A, Baraldi E, Papadopoulos NG, Vekemans J, Polack FP, Powell M, Satav A, Walsh EE, Stein RT, Graham BS, Bont LJ. The respiratory syncytial virus vaccine landscape: lessons from the graveyard and promising candidates. THE LANCET. INFECTIOUS DISEASES 2018; 18:e295-e311. [PMID: 29914800 DOI: 10.1016/s1473-3099(18)30292-5] [Citation(s) in RCA: 347] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 05/01/2018] [Indexed: 02/02/2023]
Abstract
The global burden of disease caused by respiratory syncytial virus (RSV) is increasingly recognised, not only in infants, but also in older adults (aged ≥65 years). Advances in knowledge of the structural biology of the RSV surface fusion glycoprotein have revolutionised RSV vaccine development by providing a new target for preventive interventions. The RSV vaccine landscape has rapidly expanded to include 19 vaccine candidates and monoclonal antibodies (mAbs) in clinical trials, reflecting the urgency of reducing this global health problem and hence the prioritisation of RSV vaccine development. The candidates include mAbs and vaccines using four approaches: (1) particle-based, (2) live-attenuated or chimeric, (3) subunit, (4) vector-based. Late-phase RSV vaccine trial failures highlight gaps in knowledge regarding immunological protection and provide lessons for future development. In this Review, we highlight promising new approaches for RSV vaccine design and provide a comprehensive overview of RSV vaccine candidates and mAbs in clinical development to prevent one of the most common and severe infectious diseases in young children and older adults worldwide.
Collapse
Affiliation(s)
- Natalie I Mazur
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands; Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Deborah Higgins
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Marta C Nunes
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit and Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa; Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands
| | - José A Melero
- Centro Nacional de Microbiología and CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III Majadahonda, Madrid, Spain
| | - Annefleur C Langedijk
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Nicole Horsley
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Ursula J Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Peter J Openshaw
- National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Janet A Englund
- Department of Pediatrics, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Asuncion Mejias
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Department of Pediatrics, Division of Infectious Diseases, Center for Vaccines and Immunity at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Departamento de Farmacología y Pediatria, Facultad de Medicina, Universidad de Malaga, Malaga, Spain
| | - Ruth A Karron
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Eric Af Simões
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA; Department of Epidemiology Center for Global Health, Colorado School of Public Health, Aurora, CO, USA
| | - Ivana Knezevic
- Norms and Standards for Biologicals, Department of Essential Medicines and Health Products, World Health Organization, Geneva, Switzerland
| | - Octavio Ramilo
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Department of Pediatrics, Division of Infectious Diseases, Center for Vaccines and Immunity at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA; Department of Molecular Virology and Microbiology, and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Helen Y Chu
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands
| | - Ann R Falsey
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Department of Medicine, University of Rochester and Rochester General Hospital, Rochester, NY, USA
| | - Harish Nair
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
| | - Leyla Kragten-Tabatabaie
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Julius Clinical, Zeist, Netherlands
| | - Anne Greenough
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, School of Life Course Sciences, King's College London, London, UK
| | - Eugenio Baraldi
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Women's and Children's Health Department, University of Padova, Padova, Italy
| | - Nikolaos G Papadopoulos
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Allergy Department, 2nd Paediatric Clinic, National Kapodistrian University of Athens, Athens, Greece; Division of Infection, Immunity & Respiratory Medicine, University of Manchester, Manchester, UK
| | - Johan Vekemans
- Initiative for Vaccine Research, World Health Organization, Geneva, Switzerland
| | - Fernando P Polack
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Fundacion INFANT, Buenos Aires, Argentina
| | - Mair Powell
- Licensing Division, Medicines and Healthcare Products Regulatory Agency (MHRA), London, UK
| | - Ashish Satav
- Mahatma Gandhi Tribal Hospital, Karmagram, Utavali, Tahsil, Dharni, India
| | - Edward E Walsh
- Department of Medicine, University of Rochester and Rochester General Hospital, Rochester, NY, USA
| | - Renato T Stein
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands; Pontificia Universidade Católica RGS (PUCRS), Porto Alegre, Brazil
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Louis J Bont
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands; Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Netherlands; Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, Netherlands.
| | | |
Collapse
|
90
|
Domachowske JB, Khan AA, Esser MT, Jensen K, Takas T, Villafana T, Dubovsky F, Griffin MP. Safety, Tolerability and Pharmacokinetics of MEDI8897, an Extended Half-life Single-dose Respiratory Syncytial Virus Prefusion F-targeting Monoclonal Antibody Administered as a Single Dose to Healthy Preterm Infants. Pediatr Infect Dis J 2018; 37:886-892. [PMID: 29373476 PMCID: PMC6133204 DOI: 10.1097/inf.0000000000001916] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND MEDI8897 is a recombinant human monoclonal antibody being developed for prophylaxis of serious respiratory syncytial virus (RSV) disease in all infants. METHODS In this phase 1b/2a dose-escalation study, healthy preterm infants with a gestational age of 32-35 weeks were randomized to receive a single intramuscular injection of MEDI8897 (10, 25 or 50 mg) or placebo. Safety, pharmacokinetics, RSV-neutralizing antibody and antidrug antibody (ADA) assessments were performed during the 360-day follow-up period. Infants who experienced medically attended lower respiratory tract infections (LRTIs) were tested for RSV. RESULTS MEDI8897 serum half-life ranged from 62.5-72.9 days. On day 151, 87% of infants in the 50 mg group had serum concentrations above the 90% effective concentration target level of 6.8 µg/mL, and 90% showed a ≥4-fold rise from baseline in serum RSV-neutralizing antibody levels. Adverse events (AEs) were reported in 17 of 18 (94.4%) placebo and 66 of 71 (93.0%) MEDI8897 recipients. Three MEDI8897 recipients experienced 5 serious AEs (3 LRTIs, 2 febrile seizures). ADA was detected at any time postbaseline in 28.2% of MEDI8897 recipients and at day 361 only in 26.5% of subjects. ADA response was not associated with AEs. Five (7%) MEDI8897 recipients experienced medically attended LRTIs through day 150; 1 tested positive for RSV (10 mg group). CONCLUSIONS MEDI8897 had a favorable safety profile in healthy preterm infants. The extended half-life of MEDI8897 and demonstrated RSV-neutralizing activity support protection from RSV for the duration of a typical 5-month season after a single 50 mg intramuscular (IM) dose.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Neutralizing/adverse effects
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/adverse effects
- Antibodies, Viral/pharmacology
- Antiviral Agents/adverse effects
- Antiviral Agents/pharmacokinetics
- Dose-Response Relationship, Drug
- Double-Blind Method
- Female
- Gestational Age
- Half-Life
- Humans
- Infant
- Infant, Premature
- Injections, Intramuscular
- Male
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus, Human/drug effects
- Respiratory Syncytial Virus, Human/isolation & purification
- Viral Fusion Proteins/immunology
Collapse
Affiliation(s)
- Joseph B. Domachowske
- From the Division of Infectious Diseases, Department of Pediatrics, SUNY Upstate Medical University, Syracuse, New York
| | | | | | | | | | | | | | | |
Collapse
|
91
|
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
|
92
|
Chaudhuri S, Symons JA, Deval J. Innovation and trends in the development and approval of antiviral medicines: 1987-2017 and beyond. Antiviral Res 2018; 155:76-88. [PMID: 29758235 PMCID: PMC7126013 DOI: 10.1016/j.antiviral.2018.05.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 01/05/2023]
Abstract
2017 marked the 30th anniversary of the approval of zidovudine (AZT) as the first HIV/AIDS therapy. Since then, more than eighty antiviral drugs have received FDA approval, half of which treat HIV infection. Here, we provide a retrospective analysis of approved antiviral drugs, including therapeutics against other major chronic infections such as hepatitis B and C, and herpes viruses, over the last thirty years. During this time, only a few drugs were approved to treat acute viral infections, mainly influenza. Analysis of these approved antiviral drugs based on molecular class and mode of action shows that a large majority are small molecules and direct-acting agents as opposed to proteins, peptides, or oligonucleotides and host-targeting therapies. In addition, approvals of combination therapies accelerated over the last five years. We also provide a prospective study of future potential antiviral therapies, based on current clinical research pipelines across the pharmaceutical industry. Comparing past drug approvals with current clinical candidates hints at the future evolution in antiviral therapies and reveals how antiviral medicines are often discovered. Overall, this work helps forecast future trends and innovation in the field of antiviral research and development. This review summarizes all approved antiviral drugs over the last thirty years. Most are small molecules and direct-acting agents over biologics, oligonucleotides, and host-targeting therapies. We provide a prospective study and trend analysis of future potential antivirals based on current clinical research.
Collapse
Affiliation(s)
- Shuvam Chaudhuri
- Alios BioPharma, Inc., A Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, CA, USA
| | - Julian A Symons
- Alios BioPharma, Inc., A Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, CA, USA
| | - Jerome Deval
- Alios BioPharma, Inc., A Janssen Pharmaceutical Company of Johnson & Johnson, South San Francisco, CA, USA.
| |
Collapse
|
93
|
Wagner EK, Maynard JA. Engineering therapeutic antibodies to combat infectious diseases. Curr Opin Chem Eng 2018; 19:131-141. [PMID: 29911002 DOI: 10.1016/j.coche.2018.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Serum therapy fell out of favor 80 years ago, but antibodies against infectious diseases are now experiencing a renaissance. With the evolution of antibiotic-resistant bacteria, the emergence of new pathogens, and a growing population of immunocompromised individuals coupled with improvements in antibody manufacturing and biological efficacy, antibodies are an increasingly attractive therapeutic option. In this review, we highlight successful clinical strategies and discuss recent applications of advanced antibody engineering approaches to combat infectious diseases. Case studies include antibody mixtures to neutralize Staphylococcus aureus; bispecific antibodies promoting Pseudomonas aeruginosa clearance; antibody-antibiotic conjugates to eradicate S. aureus from protected intracellular niches; and novel anti-RSV antibodies with extended serum half-life. These new designs are powerful strategies for targeting infectious diseases due to their abilities to target multiple antigens and induce novel clearance mechanisms.
Collapse
Affiliation(s)
- Ellen K Wagner
- Department of Chemical Engineering, The University of Texas at Austin, Austin TX USA, 78712
| | - Jennifer A Maynard
- Department of Chemical Engineering, The University of Texas at Austin, Austin TX USA, 78712
| |
Collapse
|
94
|
Simões EAF, Bont L, Manzoni P, Fauroux B, Paes B, Figueras-Aloy J, Checchia PA, Carbonell-Estrany X. Past, Present and Future Approaches to the Prevention and Treatment of Respiratory Syncytial Virus Infection in Children. Infect Dis Ther 2018; 7:87-120. [PMID: 29470837 PMCID: PMC5840107 DOI: 10.1007/s40121-018-0188-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION The REGAL (RSV Evidence - A Geographical Archive of the Literature) series has provided a comprehensive review of the published evidence in the field of respiratory syncytial virus (RSV) in Western countries over the last 20 years. This seventh and final publication covers the past, present and future approaches to the prevention and treatment of RSV infection among infants and children. METHODS A systematic review was undertaken of publications between January 1, 1995 and December 31, 2017 across PubMed, Embase and The Cochrane Library. Studies reporting data on the effectiveness and tolerability of prophylactic and therapeutic agents for RSV infection were included. Study quality and strength of evidence (SOE) were graded using recognized criteria. A further nonsystematic search of the published literature and Clinicaltrials.gov on antiviral therapies and RSV vaccines currently in development was also undertaken. RESULTS The systematic review identified 1441 studies of which 161 were included. Management of RSV remains centered around prophylaxis with the monoclonal antibody palivizumab, which has proven effective in reducing RSV hospitalization (RSVH) in preterm infants < 36 weeks' gestational age (72% reduction), children with bronchopulmonary dysplasia (65% reduction), and infants with hemodynamically significant congenital heart disease (53% reduction) (high SOE). Palivizumab has also shown to be effective in reducing recurrent wheezing following RSVH (high SOE). Treatment of RSV with ribavirin has conflicting success (moderate SOE). Antibodies with increased potency and extended half-life are currently entering phase 3 trials. There are approximately 15 RSV vaccines in clinical development targeting the infant directly or indirectly via the mother. CONCLUSION Palivizumab remains the only product licensed for RSV prophylaxis, and only available for high-risk infants. For the general population, there are several promising vaccines and monoclonal antibodies in various stages of clinical development, with the aim to significantly reduce the global healthcare impact of this common viral infection. FUNDING AbbVie.
Collapse
Affiliation(s)
- Eric A F Simões
- Center for Global Health, Colorado School of Public Health, University of Colorado School of Medicine, Aurora, CO, USA
| | - Louis Bont
- University Medical Center Utrecht, Utrecht, The Netherlands
- ReSViNET (Respiratory Syncytial Virus Network), Utrecht, The Netherlands
| | - Paolo Manzoni
- ReSViNET (Respiratory Syncytial Virus Network), Utrecht, The Netherlands
- Neonatology and NICU, Sant'Anna Hospital, Turin, Italy
| | - Brigitte Fauroux
- Necker University Hospital and Paris 5 University, Paris, France
| | - Bosco Paes
- Department of Paediatrics (Neonatal Division), McMaster University, Hamilton, Canada
| | - Josep Figueras-Aloy
- Hospital Clínic, Catedràtic de Pediatria, Universitat de Barcelona, Barcelona, Spain
| | - Paul A Checchia
- Baylor College of Medicine, Texas Children's Hospital Houston, Houston, TX, USA
| | - Xavier Carbonell-Estrany
- Hospital Clinic, Institut d'Investigacions Biomediques August Pi Suñer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
95
|
Goodwin E, Gilman MSA, Wrapp D, Chen M, Ngwuta JO, Moin SM, Bai P, Sivasubramanian A, Connor RI, Wright PF, Graham BS, McLellan JS, Walker LM. Infants Infected with Respiratory Syncytial Virus Generate Potent Neutralizing Antibodies that Lack Somatic Hypermutation. Immunity 2018; 48:339-349.e5. [PMID: 29396163 PMCID: PMC6005179 DOI: 10.1016/j.immuni.2018.01.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/03/2017] [Accepted: 12/29/2017] [Indexed: 11/17/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of infant mortality, and there are currently no licensed vaccines to protect this vulnerable population. A comprehensive understanding of infant antibody responses to natural RSV infection would facilitate vaccine development. Here, we isolated more than 450 RSV fusion glycoprotein (F)-specific antibodies from 7 RSV-infected infants and found that half of the antibodies recognized only two antigenic sites. Antibodies targeting both sites showed convergent sequence features, and structural studies revealed the molecular basis for their recognition of RSV F. A subset of antibodies targeting one of these sites displayed potent neutralizing activity despite lacking somatic mutations, and similar antibodies were detected in RSV-naive B cell repertoires, suggesting that expansion of these B cells in infants may be possible with suitably designed vaccine antigens. Collectively, our results provide fundamental insights into infant antibody responses and a framework for the rational design of age-specific RSV vaccines.
Collapse
Affiliation(s)
| | - Morgan S A Gilman
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Daniel Wrapp
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Man Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Joan O Ngwuta
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Syed M Moin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Patricia Bai
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | | - Ruth I Connor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Jason S McLellan
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.
| | | |
Collapse
|
96
|
Zhu Q, McLellan JS, Kallewaard NL, Ulbrandt ND, Palaszynski S, Zhang J, Moldt B, Khan A, Svabek C, McAuliffe JM, Wrapp D, Patel NK, Cook KE, Richter BWM, Ryan PC, Yuan AQ, Suzich JA. A highly potent extended half-life antibody as a potential RSV vaccine surrogate for all infants. Sci Transl Med 2018; 9:9/388/eaaj1928. [PMID: 28469033 DOI: 10.1126/scitranslmed.aaj1928] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/24/2017] [Indexed: 01/12/2023]
Abstract
Prevention of respiratory syncytial virus (RSV) illness in all infants is a major public health priority. However, no vaccine is currently available to protect this vulnerable population. Palivizumab, the only approved agent for RSV prophylaxis, is limited to high-risk infants, and the cost associated with the requirement for dosing throughout the RSV season makes its use impractical for all infants. We describe the development of a monoclonal antibody as potential RSV prophylaxis for all infants with a single intramuscular dose. MEDI8897*, a highly potent human antibody, was optimized from antibody D25, which targets the prefusion conformation of the RSV fusion (F) protein. Crystallographic analysis of Fab in complex with RSV F from subtypes A and B reveals that MEDI8897* binds a highly conserved epitope. MEDI8897* neutralizes a diverse panel of RSV A and B strains with >50-fold higher activity than palivizumab. At similar serum concentrations, prophylactic administration of MEDI8897* was ninefold more potent than palivizumab at reducing pulmonary viral loads by >3 logs in cotton rats infected with either RSV A or B subtypes. MEDI8897 was generated by the introduction of triple amino acid substitutions (YTE) into the Fc domain of MEDI8897*, which led to more than threefold increased half-life in cynomolgus monkeys compared to non-YTE antibody. Considering the pharmacokinetics of palivizumab in infants, which necessitates five monthly doses for protection during an RSV season, the high potency and extended half-life of MEDI8897 support its development as a cost-effective option to protect all infants from RSV disease with once-per-RSV-season dosing in the clinic.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA.
| | - Jason S McLellan
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, 7200 Vail Building, Hanover, NH 03755, USA
| | - Nicole L Kallewaard
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Nancy D Ulbrandt
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Susan Palaszynski
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Jing Zhang
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Brian Moldt
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Anis Khan
- Department of Clinical Pharmacology and Drug Metabolism and Pharmacokinetics, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Catherine Svabek
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Josephine M McAuliffe
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Daniel Wrapp
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, 7200 Vail Building, Hanover, NH 03755, USA
| | - Nita K Patel
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Kimberly E Cook
- Department of Antibody Discovery and Protein Engineering, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Bettina W M Richter
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Patricia C Ryan
- Biologics Safety Assessment, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Andy Q Yuan
- Department of Antibody Discovery and Protein Engineering, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - JoAnn A Suzich
- Department of Infectious Disease, MedImmune LLC, One MedImmune Way, Gaithersburg, MD 20878, USA.
| |
Collapse
|
97
|
Janet S, Broad J, Snape MD. Respiratory syncytial virus seasonality and its implications on prevention strategies. Hum Vaccin Immunother 2018; 14:234-244. [PMID: 29194014 PMCID: PMC5791579 DOI: 10.1080/21645515.2017.1403707] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/06/2017] [Indexed: 01/04/2023] Open
Abstract
With maternal and infant vaccines against respiratory syncytial virus (RSV) in development, it is timely to consider how the deployment of these vaccines might vary according to local RSV disease seasonality. In temperate regions RSV infection is predictably limited to a period of 3 to 5 months, while in tropical regions disease seasonality is often both more variable and more prolonged. Accordingly, in tropical regions a year-round immunisation schedule for both maternal and infant immunisation might be appropriate. In contrast, in temperate regions the benefit of year-round maternal immunisation would be heavily dependent on the duration of protection this provided, potentially necessitating a strategy directed at children due to be born in the months immediately prior to the RSV season. This review will consider the impact of seasonality on maternal and infant immunisation strategies against RSV, and the potential of an alternative approach of passive immunisation for all infants immediately prior to the RSV season.
Collapse
Affiliation(s)
- Sophie Janet
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jonathan Broad
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Matthew D. Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
98
|
Wong SK, Li A, Lanctôt KL, Paes B. Adherence and outcomes: a systematic review of palivizumab utilization. Expert Rev Respir Med 2017; 12:27-42. [DOI: 10.1080/17476348.2018.1401926] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sophie K. Wong
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Abby Li
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Krista L. Lanctôt
- Medical Outcomes and Research in Economics (MORE®) Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Bosco Paes
- Division of Neonatology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
99
|
Clinical Potential of Prefusion RSV F-specific Antibodies. Trends Microbiol 2017; 26:209-219. [PMID: 29054341 DOI: 10.1016/j.tim.2017.09.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/30/2017] [Accepted: 09/25/2017] [Indexed: 11/23/2022]
Abstract
Human respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in the very young. The RSV fusion protein (F) is essential for virus entry because it mediates viral and host membrane fusion. During this fusion process F is converted from a metastable prefusion conformation into an energetically favored postfusion state. Antibodies that target F can prevent viral entry and reduce disease caused by RSV. During recent years, many prefusion F-specific antibodies have been described. These antibodies typically have stronger RSV-neutralizing activity compared to those that also bind F in the postfusion conformation. Here, we describe how F-specific antibodies protect against RSV and why specifically targeting prefusion F could have great clinical potential.
Collapse
|
100
|
Shi T, McAllister DA, O'Brien KL, Simoes EAF, Madhi SA, Gessner BD, Polack FP, Balsells E, Acacio S, Aguayo C, Alassani I, Ali A, Antonio M, Awasthi S, Awori JO, Azziz-Baumgartner E, Baggett HC, Baillie VL, Balmaseda A, Barahona A, Basnet S, Bassat Q, Basualdo W, Bigogo G, Bont L, Breiman RF, Brooks WA, Broor S, Bruce N, Bruden D, Buchy P, Campbell S, Carosone-Link P, Chadha M, Chipeta J, Chou M, Clara W, Cohen C, de Cuellar E, Dang DA, Dash-Yandag B, Deloria-Knoll M, Dherani M, Eap T, Ebruke BE, Echavarria M, de Freitas Lázaro Emediato CC, Fasce RA, Feikin DR, Feng L, Gentile A, Gordon A, Goswami D, Goyet S, Groome M, Halasa N, Hirve S, Homaira N, Howie SRC, Jara J, Jroundi I, Kartasasmita CB, Khuri-Bulos N, Kotloff KL, Krishnan A, Libster R, Lopez O, Lucero MG, Lucion F, Lupisan SP, Marcone DN, McCracken JP, Mejia M, Moisi JC, Montgomery JM, Moore DP, Moraleda C, Moyes J, Munywoki P, Mutyara K, Nicol MP, Nokes DJ, Nymadawa P, da Costa Oliveira MT, Oshitani H, Pandey N, Paranhos-Baccalà G, Phillips LN, Picot VS, Rahman M, Rakoto-Andrianarivelo M, Rasmussen ZA, Rath BA, Robinson A, Romero C, Russomando G, Salimi V, Sawatwong P, Scheltema N, Schweiger B, et alShi T, McAllister DA, O'Brien KL, Simoes EAF, Madhi SA, Gessner BD, Polack FP, Balsells E, Acacio S, Aguayo C, Alassani I, Ali A, Antonio M, Awasthi S, Awori JO, Azziz-Baumgartner E, Baggett HC, Baillie VL, Balmaseda A, Barahona A, Basnet S, Bassat Q, Basualdo W, Bigogo G, Bont L, Breiman RF, Brooks WA, Broor S, Bruce N, Bruden D, Buchy P, Campbell S, Carosone-Link P, Chadha M, Chipeta J, Chou M, Clara W, Cohen C, de Cuellar E, Dang DA, Dash-Yandag B, Deloria-Knoll M, Dherani M, Eap T, Ebruke BE, Echavarria M, de Freitas Lázaro Emediato CC, Fasce RA, Feikin DR, Feng L, Gentile A, Gordon A, Goswami D, Goyet S, Groome M, Halasa N, Hirve S, Homaira N, Howie SRC, Jara J, Jroundi I, Kartasasmita CB, Khuri-Bulos N, Kotloff KL, Krishnan A, Libster R, Lopez O, Lucero MG, Lucion F, Lupisan SP, Marcone DN, McCracken JP, Mejia M, Moisi JC, Montgomery JM, Moore DP, Moraleda C, Moyes J, Munywoki P, Mutyara K, Nicol MP, Nokes DJ, Nymadawa P, da Costa Oliveira MT, Oshitani H, Pandey N, Paranhos-Baccalà G, Phillips LN, Picot VS, Rahman M, Rakoto-Andrianarivelo M, Rasmussen ZA, Rath BA, Robinson A, Romero C, Russomando G, Salimi V, Sawatwong P, Scheltema N, Schweiger B, Scott JAG, Seidenberg P, Shen K, Singleton R, Sotomayor V, Strand TA, Sutanto A, Sylla M, Tapia MD, Thamthitiwat S, Thomas ED, Tokarz R, Turner C, Venter M, Waicharoen S, Wang J, Watthanaworawit W, Yoshida LM, Yu H, Zar HJ, Campbell H, Nair H. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet 2017; 390:946-958. [PMID: 28689664 PMCID: PMC5592248 DOI: 10.1016/s0140-6736(17)30938-8] [Show More Authors] [Citation(s) in RCA: 1661] [Impact Index Per Article: 207.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/07/2017] [Accepted: 03/30/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND We have previously estimated that respiratory syncytial virus (RSV) was associated with 22% of all episodes of (severe) acute lower respiratory infection (ALRI) resulting in 55 000 to 199 000 deaths in children younger than 5 years in 2005. In the past 5 years, major research activity on RSV has yielded substantial new data from developing countries. With a considerably expanded dataset from a large international collaboration, we aimed to estimate the global incidence, hospital admission rate, and mortality from RSV-ALRI episodes in young children in 2015. METHODS We estimated the incidence and hospital admission rate of RSV-associated ALRI (RSV-ALRI) in children younger than 5 years stratified by age and World Bank income regions from a systematic review of studies published between Jan 1, 1995, and Dec 31, 2016, and unpublished data from 76 high quality population-based studies. We estimated the RSV-ALRI incidence for 132 developing countries using a risk factor-based model and 2015 population estimates. We estimated the in-hospital RSV-ALRI mortality by combining in-hospital case fatality ratios with hospital admission estimates from hospital-based (published and unpublished) studies. We also estimated overall RSV-ALRI mortality by identifying studies reporting monthly data for ALRI mortality in the community and RSV activity. FINDINGS We estimated that globally in 2015, 33·1 million (uncertainty range [UR] 21·6-50·3) episodes of RSV-ALRI, resulted in about 3·2 million (2·7-3·8) hospital admissions, and 59 600 (48 000-74 500) in-hospital deaths in children younger than 5 years. In children younger than 6 months, 1·4 million (UR 1·2-1·7) hospital admissions, and 27 300 (UR 20 700-36 200) in-hospital deaths were due to RSV-ALRI. We also estimated that the overall RSV-ALRI mortality could be as high as 118 200 (UR 94 600-149 400). Incidence and mortality varied substantially from year to year in any given population. INTERPRETATION Globally, RSV is a common cause of childhood ALRI and a major cause of hospital admissions in young children, resulting in a substantial burden on health-care services. About 45% of hospital admissions and in-hospital deaths due to RSV-ALRI occur in children younger than 6 months. An effective maternal RSV vaccine or monoclonal antibody could have a substantial effect on disease burden in this age group. FUNDING The Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Ting Shi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - Katherine L O'Brien
- Department of International Health, International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MS, USA
| | | | - Shabir A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Evelyn Balsells
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, UK
| | - Sozinho Acacio
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | | | - Asad Ali
- Department of Pediatrics and Child Health, Aga Khan University, Pakistan
| | - Martin Antonio
- Medical Research Council Unit The Gambia, Basse, The Gambia
| | - Shally Awasthi
- Department of Pediatrics, King George's Medical University, Lucknow (UP), India
| | - Juliet O Awori
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Eduardo Azziz-Baumgartner
- International Centre for Diarrhoeal Disease Research, Bangladesh; Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Henry C Baggett
- Global Disease Detection Center, Thailand Ministry of Public Health-US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand; Division of Global Health Protection, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Vicky L Baillie
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Alfredo Barahona
- Hospital Materno Infantil Jose Domingo de Obaldia, Ciudad De David, Chiriqui, Panama
| | - Sudha Basnet
- Center for International Health, University of Bergen, Norway; Department of Child Health, Tribhuvan University Institute of Medicine, Nepal
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; ISGlobal, Barcelona Ctr Int Health Res (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; ICREA, Pg Lluís Companys 23, 08010 Barcelona, Spain
| | - Wilma Basualdo
- Hospital General Pediátrico Niños de Acosta Ñu, Ministerio de Salud Pública y Bienestar Social, San Lorenzo, Paraguay
| | - Godfrey Bigogo
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Louis Bont
- Wilhelmina Children's Hospital, University Medical Center Utrecht, The Netherlands
| | | | - W Abdullah Brooks
- Department of International Health, International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MS, USA; International Centre for Diarrhoeal Disease Research, Bangladesh
| | - Shobha Broor
- All India Institute of Medical Sciences, New Delhi, India
| | - Nigel Bruce
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Dana Bruden
- Arctic Investigations Program, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centres for Disease Control and Prevention, Anchorage, AK, USA
| | - Philippe Buchy
- Institute Pasteur Cambodia, Children's Hospital Colorado, Aurora, CO, USA; GSK Vaccines Singapore, Children's Hospital Colorado, Aurora, CO, USA
| | - Stuart Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, UK
| | - Phyllis Carosone-Link
- Department of Pediatric Infectious Diseases, Children's Hospital Colorado, Aurora, CO, USA
| | | | | | - Monidarin Chou
- Rodolphe Merieux Laboratory, Faculty of Pharmacy, University of Health Sciences, Phnom Penh, Cambodia
| | - Wilfrido Clara
- Centers for Disease Control and Prevention, Central American Region, Guatemala City, Guatemala
| | - 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, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Duc-Anh Dang
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | | | - Maria Deloria-Knoll
- Department of International Health, International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MS, USA
| | - Mukesh Dherani
- Department of Public Health and Policy, University of Liverpool, Liverpool, UK
| | - Tekchheng Eap
- Department of Pneumology, National Pediatric Hospital, Phnom Penh, Cambodia
| | | | | | | | | | - Daniel R Feikin
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Luzhao Feng
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Angela Gentile
- Epidemiology Department, Austral University and Ricardo Gutiérrez Children Hospital, Argentina
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Doli Goswami
- Department of International Health, International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MS, USA; International Centre for Diarrhoeal Disease Research, Bangladesh
| | - Sophie Goyet
- Institute Pasteur Cambodia, Children's Hospital Colorado, Aurora, CO, USA
| | - Michelle Groome
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Nusrat Homaira
- International Centre for Diarrhoeal Disease Research, Bangladesh; School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, NSW, Australia
| | - Stephen R C Howie
- Medical Research Council Unit The Gambia, Basse, The Gambia; Department of Paediatrics, University of Auckland, Auckland, New Zealand; Centre for International Health, University of Otago, Dunedin, New Zealand
| | - Jorge Jara
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala
| | - Imane Jroundi
- ISGlobal, Barcelona Ctr Int Health Res (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Unit of Training and Research in Public Health, School of Medicine and Pharmacy of Rabat, University Mohamed V, Rabat, Morocco
| | | | | | - Karen L Kotloff
- Department of Pediatrics and Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anand Krishnan
- All India Institute of Medical Sciences, New Delhi, India
| | - Romina Libster
- Fundacion INFANT, Buenos Aires, Argentina; Vanderbilt University, Nashville, TN, USA; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Olga Lopez
- Hospital Dr Ernesto Torres Galdames, Iquique, Chile
| | - Marilla G Lucero
- Research Institute for Tropical Medicine, Muntinlupa, Philippines
| | - Florencia Lucion
- Epidemiology Department, Austral University and Ricardo Gutiérrez Children Hospital, Argentina
| | - Socorro P Lupisan
- Research Institute for Tropical Medicine-Department of Health, Philippines
| | - Debora N Marcone
- Centro de Educación Médica envestigaciones Clínicas "CEMIC", Argentina
| | - John P McCracken
- Center for Health Studies, Universidad del Valle de Guatemala, Guatemala
| | - Mario Mejia
- Ministry of Public Health and Social Welfare, Guatemala
| | | | - Joel M Montgomery
- Division of Global Health Protection, Center for Global Health, Centers for Disease Control and Prevention, Nairobi, Kenya
| | - David P Moore
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Cinta Moraleda
- ISGlobal, Barcelona Ctr Int Health Res (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - 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, University of the Witwatersrand, Johannesburg, South Africa
| | - Patrick Munywoki
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya; Pwani University, Kilifi, Kenya
| | | | - Mark P Nicol
- Division of Medical Microbiology, University of Cape Town and National Health Laboratory Services, South Africa
| | - D James Nokes
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya; School of Life Sciences, University of Warwick, Coventry, UK
| | | | | | - Histoshi Oshitani
- Tohoku University Graduate School of Medicine, Department of Virology, Miyagi Prefecture, Japan
| | - Nitin Pandey
- Department of Pediatrics, King George's Medical University, Lucknow (UP), India
| | - Gláucia Paranhos-Baccalà
- Emerging Pathofens Laboratory, Fondation Mérieux, Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| | - Lia N Phillips
- Emory University, Rollins School of Public Health, AT, USA
| | - Valentina Sanchez Picot
- Emerging Pathofens Laboratory, Fondation Mérieux, Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| | | | | | - Zeba A Rasmussen
- Fogarty International Center Division of International Epidemiology and Population Studies, NIH, Bethesda, MD, USA
| | - Barbara A Rath
- Department of Pediatrics, Charité University Medical Center, Berlin, Germany
| | | | - Candice Romero
- United States Naval Medical Research Unit No. 6, Callao, Peru
| | - Graciela Russomando
- Departamento de Biología Molecular y Genética, Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asuncion, Paraguay
| | - Vahid Salimi
- School of Public Health, Virology Department, Tehran University of Medical Sciences, Iran
| | - Pongpun Sawatwong
- Global Disease Detection Center, Thailand Ministry of Public Health-US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Nienke Scheltema
- Wilhelmina Children's Hospital, University Medical Center Utrecht, The Netherlands
| | | | - J Anthony G Scott
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya; London School of Hygiene & Tropical Medicine, London, UK
| | - Phil Seidenberg
- Department of Emergency Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Kunling Shen
- Key Laboratory of Major Diseases in Children and National Key Discipline of Pediatrics (Capital Medical University), Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Beijing, China
| | - Rosalyn Singleton
- Arctic Investigations Program, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centres for Disease Control and Prevention, Anchorage, AK, USA; Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | | | - Tor A Strand
- Center for International Health, University of Bergen, Norway; Department of Research, Innlandet Hospital Trust, Lillehammer, Norway
| | | | | | - Milagritos D Tapia
- Department of Pediatrics and Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Somsak Thamthitiwat
- Global Disease Detection Center, Thailand Ministry of Public Health-US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Elizabeth D Thomas
- Fogarty International Center Division of International Epidemiology and Population Studies, NIH, Bethesda, MD, USA
| | - Rafal Tokarz
- Centre for Infection and Immunity, Mailman School of Public Health, Columbia University, NY, USA
| | - Claudia Turner
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Marietjie Venter
- Centre for Viral Zoonosis, Department of Medical Virology, University of Pretoria, Pretoria, South Africa
| | - Sunthareeya Waicharoen
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Thailand
| | - Jianwei Wang
- MOH Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, IPB, CAMS-Fondation Mérieux, Institute of Pathogen Biology (IPB), Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Wanitda Watthanaworawit
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Hongjie Yu
- Division of Infectious Disease, Key Laboratory of Surveillance and Early-warning on Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Heather J Zar
- Department of Paediatrics and Child Heath, Red Cross War Memorial Children's Hospital and MRC Unit on Child & Adolescent Health, University of Cape Town, South Africa
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, UK
| | - Harish Nair
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, UK; Public Health Foundation of India, New Delhi, India.
| |
Collapse
|