1
|
Virus reduction neutralization test and LI-COR microneutralization assay bridging and WHO international standard calibration studies for respiratory syncytial virus. Bioanalysis 2022; 14:1067-1079. [PMID: 36125040 DOI: 10.4155/bio-2022-0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Respiratory syncytial virus (RSV) vaccine is an unmet medical need. The virus reduction neutralization test (VRNT) was developed to replace the LI-COR microneutralization assay to measure RSV neutralization titers. Methods: A bridging study using selected V171 phase I samples and calibration studies using the WHO international standard antiserum to RSV were performed to compare VRNT and LI-COR. Results: From the bridging study, we showed good concordance between VRNT and LI-COR titers, and similar post-/pre-vaccination titer ratios. From the calibration studies, we can convert VRNT and LI-COR titers into similar IU/ml. Conclusion: The VRNT and LI-COR microneutralization assay correlate well and the titers can be standardized as similar IU/ml, enabling direct comparison of titers from different assays.
Collapse
|
2
|
Sun D, Hsu A, Quiroz J, He X, Whiteman MC, Gurney KB, Dellatore S. Development and comparison of three cell-based potency assays for anti-respiratory syncytial virus monoclonal antibody. Biologicals 2021; 74:1-9. [PMID: 34716091 DOI: 10.1016/j.biologicals.2021.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/21/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
There is an increasing demand for monoclonal antibody (mAb) therapies to confer passive immunity against viral diseases. Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis, lower respiratory tract infections, and hospitalization in infants. Currently, there is no RSV vaccine but a humanized mAb available for high risk infants. MK-1654 is a fully human mAb with YTE mutation in the fragment crystallizable (Fc) region to extend the half-life in circulation. It binds to a highly conserved epitope of RSV Fusion protein with high affinity and neutralizes RSV infection. A functional cell-based assay is a regulatory requirement for clinical development, commercial release, and stability testing of MK-1654. In this study, we have evaluated three RSV neutralization assays to test the potency of MK-1654, including an imaging-based virus reduction neutralization test (VRNT) and two reporter virus-based assays (RSV-GFP and RSV-NLucP). All three methods showed good dose response curves of MK-1654 with similar EC50 values. RSV-NLucP method was chosen for further development because it is simple and can be easily adapted to quality control testing laboratories. After optimization, the RSV-NLucP assay was pre-qualified with good linearity, relative accuracy, intermediate precision, and specificity, therefore suitable for a cell-based potency assay.
Collapse
Affiliation(s)
- Dengyun Sun
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA.
| | - Amy Hsu
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jorge Quiroz
- Research Chemistry Manufacturing & Controls Statistics, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Xi He
- Infectious Disease and Vaccines, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Melissa C Whiteman
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Kevin B Gurney
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Shara Dellatore
- Analytical Research & Development (AR&D), MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|
3
|
Viguria N, Navascués A, Juanbeltz R, Echeverría A, Ezpeleta C, Castilla J. Effectiveness of palivizumab in preventing respiratory syncytial virus infection in high-risk children. Hum Vaccin Immunother 2021; 17:1867-1872. [PMID: 33502928 PMCID: PMC8115746 DOI: 10.1080/21645515.2020.1843336] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/24/2020] [Indexed: 12/30/2022] Open
Abstract
To date, there is no consensus regarding palivizumab prophylaxis for respiratory syncytial virus infection. The purpose of this study is to assess the effectiveness of palivizumab prophylaxis to prevent respiratory syncytial virus-related infection consultations and hospitalizations in high-risk children <2 y. We studied children <2 y of age with risk factors who had indication of palivizumab prophylaxis over eight epidemic seasons (2011-2012 to 2018-2019) in Navarra, Spain. Children positives for respiratory syncytial virus by reverse-transcription polymerase chain reaction were compared to negative testers. Palivizumab was indicated in 1,214 children <2 y of age with risk factors during 2011-2012 to 2018-2019 seasons. A total of 142 high-risk children tested for respiratory syncytial virus were included in the study. From the 35 respiratory syncytial virus-positive confirmed cases, 20 (57%) had received palivizumab versus 82 (77%) from the 107 negative controls. The effectiveness of prophylactic palivizumab was 70% (95% CI, 19%-90%) in preventing confirmed clinical infection and 82% (95% CI, 29%-96%) in preventing hospitalized cases. Our results show that palivizumab is notably effective for preventing laboratory-confirmed cases of respiratory syncytial virus and hospitalization in high-risk children <2 y of age. For children who have received palivizumab, the risk of getting sick remains high; thus, other preventive measures are necessary.
Collapse
Affiliation(s)
- Natividad Viguria
- Department of Paediatrics, Complejo Hospitalario de Navarra – IdiSNA, Pamplona, Spain
| | - Ana Navascués
- Department of Clinical Microbiology, Complejo Hospitalario de Navarra – IdiSNA, Pamplona, Spain
| | - Regina Juanbeltz
- Department of Hospital Pharmacy, Complejo Hospitalario de Navarra – IdiSNA, Pamplona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | | | - Carmen Ezpeleta
- Department of Clinical Microbiology, Complejo Hospitalario de Navarra – IdiSNA, Pamplona, Spain
| | - Jesús Castilla
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain
- Instituto de Salud Pública de Navarra-IdiSNA, Pamplona, Spain
| |
Collapse
|
4
|
Development and qualification of a fast, high-throughput and robust imaging-based neutralization assay for respiratory syncytial virus. J Immunol Methods 2021; 494:113054. [PMID: 33845088 DOI: 10.1016/j.jim.2021.113054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 01/22/2023]
Abstract
Respiratory syncytial virus (RSV) is a common pathogen causing severe respiratory illness in infants and elder adults. The development of an effective RSV vaccine is an important unmet medical need and an area of active research. The traditional method for testing neutralizing antibodies against RSV in clinical trials is the plaque reduction neutralization test (PRNT), which uses 24-well plates and needs several days post infection to develop viral plaques. In this study, we have developed a virus reduction neutralization test (VRNT), which allows the number of RSV infected cells to be automatically counted by an imaging cytometer at one day post infection in 96-well plates. VRNT was found robust to cell seeding density, detection antibody concentration, virus input and infection time. By testing twenty human sera, we have shown good correlation between VRNT50 and PRNT50 titers for multiple RSV strains: A2, Long and 18537 (serotype B). To understand the VRNT performance, eight human serum samples with high, medium and low neutralization titers were selected for VRNT qualification. We have demonstrated that VRNT had good specificity, precision, linearity and relative accuracy. In conclusion, VRNT is a better alternative to PRNT in serum neutralization test for RSV vaccine candidates.
Collapse
|
5
|
Batista JDL, Ferreira MAP, Xavier CDS, Souza ITAD, Cruz LN, Polanczyk CA. A post-incorporation study on the use of palivizumab in the Brazilian public health system. Rev Inst Med Trop Sao Paulo 2021; 63:e5. [PMID: 33533808 PMCID: PMC7845933 DOI: 10.1590/s1678-9946202163005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/26/2020] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the main cause of lower respiratory disease in infants and children under five years of age. As there is no specific treatment for RSV infections, prophylaxis with the specific monoclonal antibody palivizumab (PVZ) has been widely recommended for high-risk cases during the RSV season. The present study aimed to evaluate the effectiveness of a public prophylaxis program with palivizumab on the incidence of hospitalizations for lower respiratory tract infections and RSV in children at high risk for severe RSV infections. A retrospective cohort study was carried out with preterm children or children under two years of age with chronic lung disease or hemodynamically significant congenital heart disease; the children were selected on the basis of their exposure status, which was defined as the prophylactic use of palivizumab during the RSV season. Children were enrolled retrospectively in two hospitals located in Southern Brazil, from May 2009 to August 2016. In a sample of 129 children, 69 (53.5%) received palivizumab and adherence to three or more doses was observed in 78%; 60 (46.5%) children did not receive palivizumab. PVZ prophylaxis was independently associated with a 66% reduction in hospitalizations for any cause (26/69 - 37.7%) in the PVZ group and 34/60 (56.7%) in the control group). A 52% reduction in hospitalizations due to lower respiratory tract infection was observed in the PVZ group (15/69 -21.7%) and 25/60 (41.7%) in the control group. These findings suggest that, for the group of studied patients, the adoption of an RSV prophylaxis scheme reached the same effectiveness as those described in previous clinical trials.
Collapse
Affiliation(s)
- Joanna d'Arc Lyra Batista
- Universidade Federal da Fronteira Sul, Chapecó, Santa Catarina, Brazil.,Instituto de Avaliação de Tecnologia em Saúde, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria Angélica Pires Ferreira
- Instituto de Avaliação de Tecnologia em Saúde, Porto Alegre, Rio Grande do Sul, Brazil.,Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | - Luciane Nascimento Cruz
- Instituto de Avaliação de Tecnologia em Saúde, Porto Alegre, Rio Grande do Sul, Brazil.,Hospital Moinhos de Vento, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carisi Anne Polanczyk
- Instituto de Avaliação de Tecnologia em Saúde, Porto Alegre, Rio Grande do Sul, Brazil.,Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Hospital Moinhos de Vento, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
6
|
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
|
7
|
Cross-sectional survey of viral testing on nasopharyngeal aspirates by laboratories in the UK in infants – is targeted testing the way forward? J Hosp Infect 2020; 105:329-331. [DOI: 10.1016/j.jhin.2019.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/17/2019] [Indexed: 11/18/2022]
|
8
|
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: 49] [Impact Index Per Article: 8.2] [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
|
9
|
Malekshahi SS, Razaghipour S, Samieipoor Y, Hashemi FB, Manesh AAR, Izadi A, Faghihloo E, Ghavami N, Mokhtari-Azad T, Salimi V. Molecular characterization of the glycoprotein and fusion protein in human respiratory syncytial virus subgroup A: Emergence of ON-1 genotype in Iran. INFECTION GENETICS AND EVOLUTION 2019; 71:166-178. [PMID: 30946992 DOI: 10.1016/j.meegid.2019.03.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/05/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022]
Abstract
HRSV is a principle cause of infant hospitalization, childhood wheezing and a common pathogen in the elderly. Limited information exists regarding HRSV genotypes in Iran. In order to better understand HRSV strain diversity, we performed an in-depth evaluation of the genetic variability of the HRSV F protein detected in children under two years of age that, presented with acute respiratory symptoms during 2015-2016 in Tehran. A total of 180 nasopharyngeal swabs were evaluated. The HRSV positive samples were genotyped for G and F gene sequences using RT-PCR and sequencing methods. Phylogenetic analysis was performed using the neighbor-joining and maximum likelihood methods. Genetic and antigenic characteristics of the F gene, nucleotide and amino acids in significant positions and immune system binding regions, as well as the p-distance, positive/negative selection site, linear epitopes and glycosylation sites were investigated in all selected sequences. Among the 83 HRSV positive samples, the Fifty-five cases were successfully sequenced. All of them were classified as subgroup A and belonged to the ON-1 genotype, which possessed 72-nt duplication in the G gene. This study is the first report on the emergence of ON-1 in Iran. ON-1 Iranian sequences clustered in three lineages according to virus fusion (F) gene variations. F gene sequence analysis showed that all genetic changes in the isolates from Iran were base substitutions and no deletion/insertions were identified. The low dN/dS ratio and lack of positively selected sites showed that the fusion genes found in the strains from Iran are not under host selective pressure. Continuing and long-term molecular epidemiological surveys for early detection of circulating and newly emerging genotypes are necessary to gain a better understanding of their epidemic potential.
Collapse
Affiliation(s)
| | - Shaghayegh Razaghipour
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yazdan Samieipoor
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Anahita Izadi
- Bahrami Children Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nastaran Ghavami
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Schlake T, Thess A, Thran M, Jordan I. mRNA as novel technology for passive immunotherapy. Cell Mol Life Sci 2019; 76:301-328. [PMID: 30334070 PMCID: PMC6339677 DOI: 10.1007/s00018-018-2935-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/13/2018] [Accepted: 10/03/2018] [Indexed: 12/17/2022]
Abstract
While active immunization elicits a lasting immune response by the body, passive immunotherapy transiently equips the body with exogenously generated immunological effectors in the form of either target-specific antibodies or lymphocytes functionalized with target-specific receptors. In either case, administration or expression of recombinant proteins plays a fundamental role. mRNA prepared by in vitro transcription (IVT) is increasingly appreciated as a drug substance for delivery of recombinant proteins. With its biological role as transient carrier of genetic information translated into protein in the cytoplasm, therapeutic application of mRNA combines several advantages. For example, compared to transfected DNA, mRNA harbors inherent safety features. It is not associated with the risk of inducing genomic changes and potential adverse effects are only temporary due to its transient nature. Compared to the administration of recombinant proteins produced in bioreactors, mRNA allows supplying proteins that are difficult to manufacture and offers extended pharmacokinetics for short-lived proteins. Based on great progress in understanding and manipulating mRNA properties, efficacy data in various models have now demonstrated that IVT mRNA constitutes a potent and flexible platform technology. Starting with an introduction into passive immunotherapy, this review summarizes the current status of IVT mRNA technology and its application to such immunological interventions.
Collapse
Affiliation(s)
- Thomas Schlake
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany.
| | - Andreas Thess
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Moritz Thran
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Ingo Jordan
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| |
Collapse
|
11
|
Lee SJ, Park SK, Kim JH, Cho SM. Bronchiolitis severity according to the infected viruses. ALLERGY ASTHMA & RESPIRATORY DISEASE 2018. [DOI: 10.4168/aard.2018.6.1.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Su Jin Lee
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Sang Kyu Park
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Sung Min Cho
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| |
Collapse
|
12
|
Hashimoto K, Hosoya M. Neutralizing epitopes of RSV and palivizumab resistance in Japan. Fukushima J Med Sci 2017; 63:127-134. [PMID: 28867684 PMCID: PMC5792496 DOI: 10.5387/fms.2017-09] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/20/2017] [Indexed: 01/26/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is one of the most important viral pathogen related to acute lower respiratory infection in young children. The virus surface envelope contains the G, F, and SH proteins as spike proteins. The F protein is considered to be a major antigenic target for the neutralizing (NT) epitope as only the F protein is essential for cell infection among the three viral envelope proteins, and it is more highly conserved than the G protein. Recently, four antigenic targets related to NT activity have been reported;site I, site II, site IV, and site zero (0). Site II is the target for palivizumab used throughout the world to suppress severe RSV infection as passive immunity in high-risk children since 1998. Under the recent conditions in which indications for palivizumab administered subjects are being expanded, palivizumab-resistant mutations have been confirmed overseas in children with RSV infection, although they remain infrequent. Therefore, continuous genetic analysis of the palivizumab-binding region of the F protein is necessary. In addition, as vaccine development progresses, RSV infection control is expected to improve greatly over the next decade.
Collapse
Affiliation(s)
- Koichi Hashimoto
- Department of Pediatrics, School of Medicine, Fukushima Medical University
| | - Mitsuaki Hosoya
- Department of Pediatrics, School of Medicine, Fukushima Medical University
| |
Collapse
|
13
|
Malekshahi SS, Salimi V, Arefian E, Fatemi-Nasab G, Adjaminejad-Fard S, Yavarian J, Mokhtari-Azad T. Inhibition of Respiratory Syncytial Virus Replication by Simultaneous Targeting of mRNA and Genomic RNA Using Dual-Targeting siRNAs. Mol Biotechnol 2017; 58:767-775. [PMID: 27766578 DOI: 10.1007/s12033-016-9976-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We attempted to generate siRNAs with two active strands, which can simultaneously knock down the expression of mRNA and viral genomic RNA. In this study, short hairpin RNAs (shRNAs) against N and F genes were used. Expression of F and N mRNA transcripts as well as genomic RNA was determined with relative real-time RT-PCR. The RSV load in infected cell culture supernatant was determined by absolute quantitative real-time PCR. We found that (i) in the presence of shRNA-N, a greater reduction in viral genomic RNA was found; (ii) the level of expression at MOI 0.01 was reduced more than MOI 0.1; (iii) reduction in N transcript was greater than F; and (iv) finally, in combination pre-treatment with two shRNAs, the reduction was not significant as compared to single shRNA transfection. shRNAs also inhibited the production of RSV progeny as shown by viral load in infected HEp-2 cells. (i) Virus load reduction was greater at MOI 0.01 than 0.1 and (ii) significant load reduction was not seen with combination shRNA pre-treatment. The antiviral potency was also confirmed by plaque assay and western blot analysis. Our results provided further evidence that RNAi could be a powerful treatment option against respiratory viruses.
Collapse
Affiliation(s)
| | - Vahid Salimi
- Virology Department, School of Public Health, Tehran University of Medical Science, Porsina Ave, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ghazal Fatemi-Nasab
- Virology Department, School of Public Health, Tehran University of Medical Science, Porsina Ave, Tehran, Iran
| | - Sarvin Adjaminejad-Fard
- Virology Department, School of Public Health, Tehran University of Medical Science, Porsina Ave, Tehran, Iran
| | - Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Science, Porsina Ave, Tehran, Iran
| | - Talat Mokhtari-Azad
- Virology Department, School of Public Health, Tehran University of Medical Science, Porsina Ave, Tehran, Iran.
| |
Collapse
|
14
|
Durigon EL, Botosso VF, de Oliveira DBL. Human Respiratory Syncytial Virus: Biology, Epidemiology, and Control. HUMAN VIROLOGY IN LATIN AMERICA 2017. [PMCID: PMC7121549 DOI: 10.1007/978-3-319-54567-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
15
|
Monteiro AIMP, Bellei NCJ, Sousa AR, dos Santos AMN, Weckx LY. Respiratory infections in children up to two years of age on prophylaxis with palivizumab. ACTA ACUST UNITED AC 2016; 32:152-8. [PMID: 25119744 PMCID: PMC4183017 DOI: 10.1590/0103-0582201432214813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/27/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To identify the viruses involved in acute respiratory tract infections and to analyze the rates of hospitalization and death in children on palivizumab prophylaxis. METHODS Prospective cohort of 198 infants up to one year old who were born before 29 weeks of gestational age and infants under two years old with hemodynamically unstable cardiopathy or chronic pulmonary disease who received prophylactic palivizumab against severe respiratory syncytial virus infections in 2008. During the study period, in each episode of acute respiratory tract infection, nasopharyngeal aspirate was collected to identify respiratory syncytial virus, adenovirus, parainfluenza 1, 2 and 3, influenza A and B by direct immunofluorescence, rhinovirus and metapneumovirus by polymerase chain reaction preceded by reverse transcription. Data regarding hospitalization and deaths were monitored. RESULTS Among the 198 studied infants, 117 (59.1%) presented acute respiratory tract infections, with a total of 175 episodes. Of the 76 nasopharyngeal aspirates collected during respiratory tract infections, 37 were positive, as follow: rhinovirus (75.7%), respiratory syncytial virus (18.9%), parainfluenza (8.1%), adenovirus 2 (2.7%), metapneumovirus (2.7%) and three samples presented multiple agents. Of the 198 children, 48 (24.4%) were hospitalized: 30 (15.2%) for non-infectious etiology and 18 (9.1%) for respiratory causes. Among these 18 children, one case of respiratory syncytial virus was identified. Two deaths were reported, but respiratory syncytial virus was not identified. CONCLUSIONS During the prophylaxis period, low frequency of respiratory syncytial virus infections and low rates of hospitalization were observed, suggesting the benefit of palivizumab prophylaxis.
Collapse
Affiliation(s)
| | | | | | | | - Lily Yin Weckx
- Escola Paulista de Medicina, Unifesp, São Paulo, SP, Brasil
| |
Collapse
|
16
|
Yasui Y, Yamaji Y, Sawada A, Ito T, Nakayama T. Cell fusion assay by expression of respiratory syncytial virus (RSV) fusion protein to analyze the mutation of palivizumab-resistant strains. J Virol Methods 2016; 231:48-55. [PMID: 26794681 DOI: 10.1016/j.jviromet.2016.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/11/2015] [Accepted: 01/09/2016] [Indexed: 11/18/2022]
Abstract
Respiratory syncytial virus (RSV) consists of fusion (F), glyco (G), and small hydrophobic (SH) proteins as envelope proteins, and infects through cell fusion. F protein is expressed on the surface of infected cells, and induces cell fusion. In the present report, expression plasmids of the F, G and SH proteins were constructed and cell fusion activity was investigated under T7 RNA polymerase. F protein alone induced cell fusion at a lower concentration than previously reported, and co-expression of F and SH proteins induced cell fusion more efficiently than F protein alone. Palivizumab is the only prophylactic agent against RSV infection. Palivizumab-resistant strains having mutations of the F protein of K272E and S275F were reported. These mutations were introduced into an F-expression plasmid, and exhibited no inhibition of cell fusion with palivizumab. Among the RSV F protein mutants, N276S has been reported to have partial resistance against palivizumab, but the F expression plasmid with the N276S mutation showed a reduction in cell fusion in the presence of palivizumab, showing no resistance to palivizumab. The present expression system was useful for investigating the mechanisms of RSV cell fusion.
Collapse
Affiliation(s)
- Yosuke Yasui
- Keio University, Health Center, 4-1-1 Hiyoshi, Kouhoku, Yokohama, Kanagawa, Japan; Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, Japan.
| | - Yoshiaki Yamaji
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, Japan.
| | - Akihito Sawada
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, Japan.
| | - Takashi Ito
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, Japan.
| | - Tetsuo Nakayama
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, Japan.
| |
Collapse
|
17
|
al-Alaiyan S, Pollack P, Notario GF. Safety and pharmacokinetics of extended use of palivizumab in Saudi Arabian infants and children. Drugs Context 2015; 4:212270. [PMID: 25767550 PMCID: PMC4353192 DOI: 10.7573/dic.212270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The peak season of respiratory syncytial virus (RSV) infections in warmer climates may extend beyond the typical five-month RSV season of temperate regions. Additional monthly doses of palivizumab may be necessary in warmer regions to protect children at high risk for serious infection by the RSV. METHODS In a Phase II, single-arm, single-center, non-comparative, open-label, prospective study conducted in Saudi Arabia, children at high risk for RSV infection received up to seven monthly injections of palivizumab (15 mg/kg) during the 2000-2001 RSV season. Key enrollment criteria were no previous exposure to palivizumab and gestational age ≤35 weeks, ≤6 months of age at enrollment, or chronic lung disease and ≤24 months of age at enrollment. We wished to assess the safety, immunogenicity, and pharmacokinetics of palivizumab as an extended seven-dose regimen. RESULTS Of 18 enrolled patients, 17 patients received seven palivizumab injections. Seven adverse events (AEs) occurred in five patients. Bronchiolitis was the most commonly reported AE. Six serious AEs occurred in four patients. No AEs were considered related to palivizumab. Trough levels of palivizumab in serum were >40 μg/mL in most patients after the first injection and in 16/18 and 14/17 patients after the fourth and sixth injections, respectively. Except for one patient at one visit, the anti-palivizumab titer was <1:10 at all visits. CONCLUSION These data suggest that an extended palivizumab regimen of up to seven monthly doses during the RSV season exhibited an acceptable safety profile in children at high risk for RSV infection in Saudi Arabia.
Collapse
Affiliation(s)
- Saleh al-Alaiyan
- Neonatology Section, Department of Pediatrics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Paul Pollack
- GI Immunology Medical Affairs, Global Pharmaceutical and Research Division, AbbVie Inc., North Chicago, IL, USA
| | - Gerard F Notario
- Virology Global Project Team, Global Pharmaceutical Research and Development, AbbVie Inc., North Chicago, IL, USA
| |
Collapse
|
18
|
Oliveira DBL, Iwane MK, Prill MM, Weinberg GA, Williams JV, Griffin MR, Szilagyi PG, Edwards KM, Staat MA, Hall CB, Durigon EL, Erdman DD. Molecular characterization of respiratory syncytial viruses infecting children reported to have received palivizumab immunoprophylaxis. J Clin Virol 2015; 65:26-31. [PMID: 25766983 PMCID: PMC7172275 DOI: 10.1016/j.jcv.2015.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/17/2015] [Accepted: 01/23/2015] [Indexed: 11/08/2022]
Abstract
Palivizumab (PZ) immunoprophylaxis of high-risk children reduces RSV hospitalizations. PZ resistance mutations can result in RSV breakthrough infections in PZ recipients. PZ resistance mutations were identified in 10.2% of children reportedly receiving PZ. PZ resistance mutations did not account for the majority of PZ failures.
Background Respiratory syncytial virus (RSV) is a major cause of respiratory infections in children. Palivizumab (PZ) is the only RSV-specific immunoprophylaxis approved by the U.S. Food and Drug Administration. Mutations leading to amino acid substitutions in the PZ binding site of the RSV F protein have been associated with breakthrough RSV infections in patients receiving PZ. Objective To detect PZ resistance conferring mutations in RSV strains from children who received PZ. Study design Children aged ≤24 months on October 31 who were hospitalized or had outpatient visits for respiratory illness and/or fever during October–May 2001–2008 in 3 US counties were included. PZ receipt was obtained from parent interviews and medical records among children subsequently infected with RSV. Archived nasal/throat swab specimens were tested for RSV by real-time RT-PCR. The coding region of the PZ binding site of the RSV F protein was sequenced using both Sanger and pyrosequencing methods. Results Of 8762 enrolled children, 375 (4.3%) were tested for RSV and had a history of PZ receipt, of which 56 (14.9%) were RSV-positive and 45 of these had available archived specimens. Molecular typing identified 42 partial F gene sequences in specimens from 39 children: 19 single RSV subgroup A, 17 subgroup B and 3 mixed infections. Nucleotide substitutions were identified in 12/42 (28.6%) RSV strains. PZ resistance mutations were identified in 4 (10.2%) of the 39 children, of which one had documented PZ receipt. Conclusions Although RSV PZ resistance mutations were infrequent, most RSV-associated illnesses in children with a history of PZ receipt were not due to strain resistance.
Collapse
Affiliation(s)
| | - Marika K Iwane
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mila M Prill
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | | | - Mary A Staat
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Caroline B Hall
- University of Rochester School of Medicine, Rochester, NY, USA
| | - Edison L Durigon
- Institute of Biomedical Science, University of São Paulo, Brazil
| | - Dean D Erdman
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
19
|
Zhan X, Slobod KS, Jones BG, Sealy RE, Takimoto T, Boyd K, Surman S, Russell CJ, Portner A, Hurwitz JL. Sendai virus recombinant vaccine expressing a secreted, unconstrained respiratory syncytial virus fusion protein protects against RSV in cotton rats. Int Immunol 2014; 27:229-36. [PMID: 25477211 DOI: 10.1093/intimm/dxu107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 11/27/2014] [Indexed: 11/14/2022] Open
Abstract
The respiratory syncytial virus (RSV) is responsible for as many as 199000 annual deaths worldwide. Currently, there is no standard treatment for RSV disease and no vaccine. Sendai virus (SeV) is an attractive pediatric vaccine candidate because it elicits robust and long-lasting virus-specific B cell and T cell activities in systemic and mucosal tissues. The virus serves as a gene delivery system as well as a Jennerian vaccine against its close cousin, human parainfluenza virus type 1. Here we describe the testing of a recombinant SeV (SeVRSV-Fs) that expresses an unconstrained, secreted RSV-F protein as a vaccine against RSV in cotton rats. After a single intranasal immunization of cotton rats with SeVRSV-Fs, RSV-specific binding and neutralizing antibodies were generated. These antibodies exhibited cross-reactivity with both RSV A and B isolates. RSV-F-specific IFN-γ-producing T cells were also activated. The SeVRSV-Fs vaccine conferred protection against RSV challenge without enhanced immunopathology. In total, results showed that an SeV recombinant that expresses RSV F in an unconstrained, soluble form can induce humoral and cellular immunity that protects against infection with RSV.
Collapse
Affiliation(s)
- Xiaoyan Zhan
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Present Address: Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Karen S Slobod
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Present Address: Department of Technical Development, Novartis Vaccines, Cambridge, MA 02139, USA
| | - Bart G Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Robert E Sealy
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Toru Takimoto
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Present Address: Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kelli Boyd
- Animal Resource Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Present Address: Division of Animal Care, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sherri Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Allen Portner
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Julia L Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
20
|
Updated guidance for palivizumab prophylaxis among infants and young children at increased risk of hospitalization for respiratory syncytial virus infection. Pediatrics 2014; 134:e620-38. [PMID: 25070304 DOI: 10.1542/peds.2014-1666] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Guidance from the American Academy of Pediatrics (AAP) for the use of palivizumab prophylaxis against respiratory syncytial virus (RSV) was first published in a policy statement in 1998. Guidance initially was based on the result from a single randomized, placebo-controlled clinical trial conducted in 1996-1997 describing an overall reduction in RSV hospitalization rate from 10.6% among placebo recipients to 4.8% among children who received prophylaxis. The results of a second randomized, placebo-controlled trial of children with hemodynamically significant heart disease were published in 2003 and revealed a reduction in RSV hospitalization rate from 9.7% in control subjects to 5.3% among prophylaxis recipients. Because no additional controlled trials regarding efficacy were published, AAP guidance has been updated periodically to reflect the most recent literature regarding children at greatest risk of severe disease. Since the last update in 2012, new data have become available regarding the seasonality of RSV circulation, palivizumab pharmacokinetics, the changing incidence of bronchiolitis hospitalizations, the effects of gestational age and other risk factors on RSV hospitalization rates, the mortality of children hospitalized with RSV infection, and the effect of prophylaxis on wheezing and palivizumab-resistant RSV isolates. These data enable further refinement of AAP guidance to most clearly focus on those children at greatest risk.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antiviral Agents/administration & dosage
- Antiviral Agents/pharmacokinetics
- Antiviral Agents/therapeutic use
- Child, Preschool
- Comorbidity
- Cystic Fibrosis/epidemiology
- Down Syndrome/epidemiology
- Gestational Age
- Hospitalization/statistics & numerical data
- Humans
- Immunocompromised Host
- Indians, North American/statistics & numerical data
- Infant
- Infant, Premature
- Injections, Intramuscular
- Neuromuscular Diseases/epidemiology
- Palivizumab
- Respiratory Sounds
- Respiratory Syncytial Virus Infections/epidemiology
- Respiratory Syncytial Virus Infections/ethnology
- Respiratory Syncytial Virus Infections/prevention & control
- Risk Factors
- Seasons
Collapse
|
21
|
Jones BG, Sealy RE, Surman SL, Portner A, Russell CJ, Slobod KS, Dormitzer PR, DeVincenzo J, Hurwitz JL. Sendai virus-based RSV vaccine protects against RSV challenge in an in vivo maternal antibody model. Vaccine 2014; 32:3264-73. [PMID: 24721531 DOI: 10.1016/j.vaccine.2014.03.088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/08/2014] [Accepted: 03/26/2014] [Indexed: 01/08/2023]
Abstract
Respiratory syncytial virus (RSV) is the cause of significant morbidity and mortality among infants, and despite decades of research there remains no licensed vaccine. SeVRSV is a Sendai virus (SeV)-based live intranasal vaccine that expresses the full length RSV fusion (F) gene. SeV is the murine counterpart of human parainfluenza virus type 1. Given that the target population of SeVRSV is young infants, we questioned whether maternal antibodies typical of this age group would inhibit SeVRSV vaccine efficacy. After measuring SeV- and RSV-specific serum neutralizing antibody titers in human infants, we matched these defined titers in cotton rats by the passive transfer of polyclonal or monoclonal antibody products. Animals were then vaccinated with SeVRSV followed by a 3 month rest period to allow passively transferred antibodies to wane. Animals were finally challenged with RSV to measure the de novo vaccine-induced immune responses. Despite the presence of passively-transferred serum neutralizing antibodies at the time of vaccination, SeVRSV induced immune responses that were protective against RSV challenge. The data encourage advancement of SeVRSV as a candidate vaccine for the protection of children from morbidity and mortality caused by RSV.
Collapse
Affiliation(s)
- Bart G Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA
| | - Robert E Sealy
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA
| | - Sherri L Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA
| | - Allen Portner
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA
| | - Charles J Russell
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | - John DeVincenzo
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA; Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, TN, USA; Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Julia L Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, USA; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
22
|
Genetic diversity and molecular evolution of the major human metapneumovirus surface glycoproteins over a decade. J Clin Virol 2013; 58:541-7. [DOI: 10.1016/j.jcv.2013.08.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/19/2013] [Accepted: 08/26/2013] [Indexed: 11/24/2022]
|
23
|
Monoclonal antibodies for prophylactic and therapeutic use against viral infections. ACTA ACUST UNITED AC 2013; 88:T15-T23. [PMID: 32287402 PMCID: PMC7111719 DOI: 10.1016/j.pepo.2013.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/15/2013] [Indexed: 11/21/2022]
Abstract
Neutralizing antibodies play an essential part in antiviral immunity and are instrumental in preventing or modulating viral diseases. Polyclonal antibody preparations are increasingly being replaced by highly potent monoclonal antibodies (mAbs). Cocktails of mAbs and bispecific constructs can be used to simultaneously target multiple viral epitopes and to overcome issues of neutralization escape. Advances in antibody engineering have led to a large array of novel mAb formats, while deeper insight into the biology of several viruses and increasing knowledge of their neutralizing epitopes has extended the list of potential targets. In addition, progress in developing inexpensive production platforms will make antiviral mAbs more widely available and affordable.
Collapse
|
24
|
Both L, Banyard AC, van Dolleweerd C, Wright E, Ma JKC, Fooks AR. Monoclonal antibodies for prophylactic and therapeutic use against viral infections. Vaccine 2013; 31:1553-9. [PMID: 23370150 PMCID: PMC7115371 DOI: 10.1016/j.vaccine.2013.01.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 01/01/2013] [Accepted: 01/15/2013] [Indexed: 12/27/2022]
Abstract
Neutralizing antibodies play an essential part in antiviral immunity and are instrumental in preventing or modulating viral diseases. Polyclonal antibody preparations are increasingly being replaced by highly potent monoclonal antibodies (mAbs). Cocktails of mAbs and bispecific constructs can be used to simultaneously target multiple viral epitopes and to overcome issues of neutralization escape. Advances in antibody engineering have led to a large array of novel mAb formats, while deeper insight into the biology of several viruses and increasing knowledge of their neutralizing epitopes has extended the list of potential targets. In addition, progress in developing inexpensive production platforms will make antiviral mAbs more widely available and affordable.
Collapse
Affiliation(s)
- Leonard Both
- The Hotung Molecular Immunology Unit, Division of Clinical Sciences, St George's, University of London, London, UK
- Animal Health and Veterinary Laboratories Agency (AHVLA), Wildlife Zoonoses and Vector-borne Diseases Research Group, Department of Virology, Weybridge, Surrey, UK
| | - Ashley C. Banyard
- Animal Health and Veterinary Laboratories Agency (AHVLA), Wildlife Zoonoses and Vector-borne Diseases Research Group, Department of Virology, Weybridge, Surrey, UK
| | - Craig van Dolleweerd
- The Hotung Molecular Immunology Unit, Division of Clinical Sciences, St George's, University of London, London, UK
| | - Edward Wright
- School of Life Sciences, University of Westminster, London, UK
| | - Julian K.-C. Ma
- The Hotung Molecular Immunology Unit, Division of Clinical Sciences, St George's, University of London, London, UK
| | - Anthony R. Fooks
- Animal Health and Veterinary Laboratories Agency (AHVLA), Wildlife Zoonoses and Vector-borne Diseases Research Group, Department of Virology, Weybridge, Surrey, UK
- National Consortium for Zoonosis Research, University of Liverpool, Leahurst, Neston, South Wirral CH64 7TE, UK
- Corresponding author at: Animal Health and Veterinary Laboratories Agency (AHVLA), Wildlife Zoonoses and Vector-borne Diseases Research Group, Department of Virology, Weybridge, Surrey KT15 3NB, UK. Tel.: +44 01932 357840; fax: +44 01932 357239.
| |
Collapse
|
25
|
Papenburg J, Carbonneau J, Hamelin MÈ, Isabel S, Bouhy X, Ohoumanne N, Déry P, Paes BA, Corbeil J, Bergeron MG, De Serres G, Boivin G. Molecular evolution of respiratory syncytial virus fusion gene, Canada, 2006-2010. Emerg Infect Dis 2012; 18:120-4. [PMID: 22264682 PMCID: PMC3310097 DOI: 10.3201/eid1801.110515] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
To assess molecular evolution of the respiratory syncytial virus (RSV) fusion gene, we analyzed RSV-positive specimens from 123 children in Canada who did or did not receive RSV immunoprophylaxis (palivizumab) during 2006-2010. Resistance-conferring mutations within the palivizumab binding site occurred in 8.7% of palivizumab recipients and none of the nonrecipients.
Collapse
|
26
|
Selection and characterization of human respiratory syncytial virus escape mutants resistant to a polyclonal antiserum raised against the F protein. Arch Virol 2012; 157:1071-80. [PMID: 22411099 DOI: 10.1007/s00705-012-1274-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 02/02/2012] [Indexed: 10/28/2022]
Abstract
A human respiratory syncytial virus (HRSV) neutralization escape mutant was obtained after 56 serial passages in the presence of a polyclonal antiserum raised against the F protein. Nucleotide sequence analysis of this escape mutant virus revealed two amino acid substitutions: Asn268Ile and Val533Met. When this virus was allowed to grow in the absence of the anti-F polyclonal serum, only the mutation Asn268Ile was stably maintained. Both the double and single escape mutant viruses lost reactivity with mAbs belonging to antigenic site II of the fusion protein of RSV. Mutation Asn268Ile has already been reported in RS viruses that are resistant to mAbs 47F and 11 and palivizumab (PZ). We have thus identified a novel mutation (Val533Met) in the transmembrane domain of the F protein that was selected under immune pressure.
Collapse
|
27
|
Zhu Q, Patel NK, McAuliffe JM, Zhu W, Wachter L, McCarthy MP, Suzich JA. Natural polymorphisms and resistance-associated mutations in the fusion protein of respiratory syncytial virus (RSV): effects on RSV susceptibility to palivizumab. J Infect Dis 2011; 205:635-8. [PMID: 22184728 DOI: 10.1093/infdis/jir790] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Specific mutations in respiratory syncytial virus (RSV) fusion protein can cause palivizumab resistance. We assessed the incidence of sequence polymorphisms and palivizumab resistance in clinical RSV isolates collected from immunoprophylaxis-naive subjects. Polymorphisms were identified at low frequency, and only polymorphic mutations in antigenic site A (<1% of all polymorphisms) conferred palivizumab resistance.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Infectious Disease-Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Zhu Q, McAuliffe JM, Patel NK, Palmer-Hill FJ, Yang CF, Liang B, Su L, Zhu W, Wachter L, Wilson S, MacGill RS, Krishnan S, McCarthy MP, Losonsky GA, Suzich JA. Analysis of respiratory syncytial virus preclinical and clinical variants resistant to neutralization by monoclonal antibodies palivizumab and/or motavizumab. J Infect Dis 2011; 203:674-82. [PMID: 21208913 PMCID: PMC3072724 DOI: 10.1093/infdis/jiq100] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 11/22/2010] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Palivizumab is a US Food and Drug Administration-approved monoclonal antibody for the prevention of respiratory syncytial virus (RSV) lower respiratory disease in high-risk infants. Motavizumab, derived from palivizumab with enhanced antiviral activity, has recently been tested in humans. Although palivizumab escape mutants have been generated in the laboratory, the development of resistant RSV in patients receiving palivizumab has not been reported previously. METHODS We generated palivizumab and motavizumab escape mutants in vitro and examined the development of resistant mutants in RSV-breakthrough patients receiving immunoprophylaxis. The effect of these mutations on neutralization by palivizumab and motavizumab and in vitro fitness was studied. RESULTS Antibody-resistant RSV variants selected in vitro had mutations at position 272 of the fusion protein, from lysine to asparagine, methionine, threonine, glutamine, or glutamate. Variants containing mutations at positions 272 and 275 were detected in breakthrough patients. All these variants were resistant to palivizumab, but only the glutamate variant at position 272 demonstrated resistance to motavizumab. Mixtures of wild-type and variant RSV soon lost the resistant phenotype in the absence of selection. CONCLUSIONS Resistant RSV variants were detected in a small subset (∼ 5%) of RSV breakthrough cases. The fitness of these variants was impaired, compared to wild-type RSV.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Infectious Disease-Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ochoa Sangrador C, González de Dios J. [Consensus conference on acute bronchiolitis (II): epidemiology of acute bronchiolitis. Review of the scientific evidence]. An Pediatr (Barc) 2010; 72:222.e1-222.e26. [PMID: 20153707 PMCID: PMC7105046 DOI: 10.1016/j.anpedi.2009.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Accepted: 11/30/2009] [Indexed: 11/08/2022] Open
Abstract
A review of the evidence on epidemiology, risk factors, etiology and clinical-etiological profile of acute bronchiolitis is presented. The frequency estimates are very heterogeneous; in the population under two years the frequency of admission for bronchiolitis is between 1 and 3.5%, primary care consultations between 4 and 20% and emergency visits between 1 and 2%. The frequency of admissions for respiratory infection by respiratory syncytial virus in the risk population is: in premature infants < or =32 weeks of gestation between 4.4 and 18%, in patients with bronchopulmonary dysplasia between 7.3 and 42%, and in infants with congenital heart disease between 1.6 and 9.8%. The main risk factors are: prematurity, chronic lung disease or bronchopulmonary dysplasia, congenital heart disease and age less than 3-6 months at onset of the epidemic. Other factors are: older siblings or day care attendance, male gender, exposure to smoking, breastfeeding for less than 1-2 months and variables associated with lower socioeconomic status. Respiratory syncytial virus is the dominant etiological agent, constituting just over half the cases (median 56%; interval 27% to 73%). Other viruses implicated, in descending order of frequency, are rhinovirus, adenovirus, metapneumovirus, influenza viruses, parainfluenza, enterovirus and bocavirus. In studies with genomic detection techniques, between 20 and 25% of cases the virus involved is not identified and between 9% and 27% of cases have viral co-infection. Although respiratory syncytial virus bronchiolitis shows more wheezing and retractions, longer duration of respiratory symptoms and oxygen therapy and are associated with lower use of antibiotics. This pattern is associated with the younger age of the patients and does not help us to predict the etiology. In general, the etiological identification is not useful for the management of patients. However, in young infants (<3 months) with febrile bronchiolitis in the hospital environment, conservative management may help these patients and avoid diagnostic and therapeutic procedures.
Collapse
|
30
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2008. [DOI: 10.1002/pds.1492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Asenjo A, González-Armas JC, Villanueva N. Phosphorylation of human respiratory syncytial virus P protein at serine 54 regulates viral uncoating. Virology 2008; 380:26-33. [PMID: 18706669 DOI: 10.1016/j.virol.2008.06.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 06/27/2008] [Accepted: 06/27/2008] [Indexed: 11/29/2022]
Abstract
The human respiratory syncytial virus (HRSV) structural P protein, phosphorylated at serine (S) and threonine (T) residues, is a co-factor of viral RNA polymerase. The phosphorylation of S54 is controlled by the coordinated action of two cellular enzymes: a lithium-sensitive kinase, probably glycogen synthetase kinase (GSK-3) beta and protein phosphatase 2A (PP2A). Inhibition of lithium-sensitive kinase, soon after infection, blocks the viral growth cycle by inhibiting synthesis and/or accumulation of viral RNAs, proteins and extracellular particles. P protein phosphorylation at S54 is required to liberate viral ribonucleoproteins (RNPs) from M protein, during the uncoating process. Kinase inhibition, late in infection, produces a decrease in genomic RNA and infectious viral particles. LiCl, intranasally applied to mice infected with HRSV A2 strain, reduces the number of mice with virus in their lungs and the virus titre. Administration of LiCl to humans via aerosol should prevent HRSV infection, without secondary effects.
Collapse
Affiliation(s)
- Ana Asenjo
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Ctra, Majadahonda, Madrid, Spain
| | | | | |
Collapse
|