1
|
Liu W, Lien YH, Lee PI, Chan TC, Wang LC, Yang CR, Ho MS, Chen JR, Ku CC, King CC. Impact of prior infection and repeated vaccination on post-vaccination antibody titers of the influenza A(H1N1)pdm09 strain in Taiwan schoolchildren: Implications for public health. Vaccine 2022; 40:3402-3411. [PMID: 35525727 DOI: 10.1016/j.vaccine.2022.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/01/2022] [Accepted: 03/18/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The objective of this study was to evaluate the effects of prior-infection and repeated vaccination on post-vaccination antibody titers. METHODS A(H1N1)pdm09 strain was included in 2009 pandemic monovalent, 2010-2011, and 2011-2012 trivalent influenza vaccines (MIVpdm09, TIV10/11, TIV11/12) in Taiwan. During the 2011-2012 influenza season, we conducted a prospective sero-epidemiological cohort study among schoolchildren from grades 1 - 6 in the two elementary schools in Taipei with documented A(H1N1)pdm09 vaccination records since 2009. Serum samples were collected at pre-vaccination, 1-month, and 4-months post-vaccination (T1, T2, T3). Anti-A(H1N1)pdm09 hemagglutination inhibition titers (HI-Ab-titers) were examined. We also investigated the impact of four vaccination histories [(1) no previous vaccination (None), (2) vaccinated in 2009-2010 season (09v), (3) vaccinated in 2010-2011 season (10v), and (4) vaccinated consecutively in 2009-2010 and 2010-2011 seasons (09v + 10v)] and pre-vaccination HI-Ab levels on post-vaccination HI-Ab responses as well as adjusted vaccine effectiveness (aVE) against serologically-defined infection from T2 to T3. RESULTS TIV11/12 had zero serious adverse events reported. A(H1N1)pdm09 strain in TIV11/12 elicited seroprotective Ab-titers in 98% of children and showed promising protection (aVE: 70.3% [95% confidence interval (CI): 51.0-82.1%]). Previously unvaccinated but infected children had a 3.96 times higher T2 geometric mean titer (T2-GMT) of HI-Ab than those naïve to A(H1N1)pdm09 (GMT [95% CI]: 1039.7[585.3-1845.9] vs. 262.5[65.9-1045], p = 0.046). Previously vaccinated children with seroprotective T1-Ab-titers had a higher T2-GMT and a greater aVE than those with non-seroprotective T1-Ab-titers. Repeatedly vaccinated children had lower T2-GMT than those receiving primary doses of TIV11/12. However, after controlling prior infection and T1-Ab-titers, differences in T2-GMT among the four vaccination histories became insignificant (p = 0.16). CONCLUSION This study supports the implementation of annual mass-vaccination with A(H1N1)pdm09 in schoolchildren for three consecutive influenza seasons when vaccine and circulating strains were well matched, and found that prior infection and pre-vaccination HI-Ab levels positively impacted post-vaccination HI-Ab responses.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University (NTU), Taipei 100, Taiwan, ROC
| | - Yu-Hui Lien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University (NTU), Taipei 100, Taiwan, ROC
| | - Ping-Ing Lee
- Department of Pediatrics, NTU Hospital and NTU College of Medicine, Taipei 100, Taiwan, ROC
| | - Ta-Chien Chan
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei 115, Taiwan, ROC
| | | | - Chin-Rur Yang
- Institute of Immunology, NTU College of Medicine, Taipei 100, Taiwan, ROC
| | - Mei-Shang Ho
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, ROC
| | | | - Chia-Chi Ku
- Institute of Immunology, NTU College of Medicine, Taipei 100, Taiwan, ROC.
| | - Chwan-Chuen King
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University (NTU), Taipei 100, Taiwan, ROC.
| |
Collapse
|
2
|
Neerukonda SN, Vassell R, Herrup R, Liu S, Wang T, Takeda K, Yang Y, Lin TL, Wang W, Weiss CD. Establishment of a well-characterized SARS-CoV-2 lentiviral pseudovirus neutralization assay using 293T cells with stable expression of ACE2 and TMPRSS2. PLoS One 2021; 16:e0248348. [PMID: 33690649 PMCID: PMC7946320 DOI: 10.1371/journal.pone.0248348] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Pseudoviruses are useful surrogates for highly pathogenic viruses because of their safety, genetic stability, and scalability for screening assays. Many different pseudovirus platforms exist, each with different advantages and limitations. Here we report our efforts to optimize and characterize an HIV-based lentiviral pseudovirus assay for screening neutralizing antibodies for SARS-CoV-2 using a stable 293T cell line expressing human angiotensin converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). We assessed different target cells, established conditions that generate readouts over at least a two-log range, and confirmed consistent neutralization titers over a range of pseudovirus input. Using reference sera and plasma panels, we evaluated assay precision and showed that our neutralization titers correlate well with results reported in other assays. Overall, our lentiviral assay is relatively simple, scalable, and suitable for a variety of SARS-CoV-2 entry and neutralization screening assays.
Collapse
Affiliation(s)
- Sabari Nath Neerukonda
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Russell Vassell
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Rachel Herrup
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Shufeng Liu
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Tony Wang
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Kazuyo Takeda
- US Food and Drug Administration, Office of Blood Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Ye Yang
- US Food and Drug Administration, Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Tsai-Lien Lin
- US Food and Drug Administration, Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Wei Wang
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| | - Carol D. Weiss
- US Food and Drug Administration, Office of Vaccine Research and Review, Center for Biologics Evaluation and Research and Review, Silver Spring, Maryland, United States of America
| |
Collapse
|
3
|
Wang W, Alvarado-Facundo E, Vassell R, Collins L, Colombo RE, Ganesan A, Geaney C, Hrncir D, Lalani T, Markelz AE, Maves RC, McClenathan B, Mende K, Richard SA, Schofield C, Seshadri S, Spooner C, Utz GC, Warkentien TE, Levine M, Coles CL, Burgess TH, Eichelberger M, Weiss CD. Comparison of A(H3N2) neutralizing antibody responses elicited by 2018-2019 season quadrivalent influenza vaccines derived from eggs, cells, and recombinant hemagglutinin. Clin Infect Dis 2020; 73:e4312-e4320. [PMID: 32898271 DOI: 10.1093/cid/ciaa1352] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Low vaccine effectiveness against A(H3N2) influenza in seasons with little antigenic drift has been attributed to substitutions in hemagglutinin (HA) acquired during vaccine virus propagation in eggs. Clinical trials comparing recombinant HA vaccine (rHA) and cell-derived inactivated influenza vaccine (IIV) to egg-derived IIVs provide opportunities to assess how egg-adaptive substitutions influence HA immunogenicity. METHODS Neutralization titers in pre- and post-immunization sera from 133 adults immunized with one of three types of influenza vaccines in a randomized, open-label trial during the 2018-2019 influenza season were measured against egg- and cell-derived A/Singapore/INFIMH-16-0019/2016-like and circulating A(H3N2) influenza viruses using HA-pseudoviruses. RESULTS All vaccines elicited neutralizing antibodies to all H3 vaccine antigens, but the rHA vaccine elicited the highest titers and seroconversion rates against all strains tested. Egg- and cell-derived IIVs elicited responses similar to each other. Pre-immunization titers against H3 HA-pseudoviruses containing egg-adaptive substitutions T160K and L194P were high, but lower against H3 HA-pseudoviruses without those substitutions. All vaccines boosted neutralization titers against HA-pseudoviruses with egg-adaptive substitutions, but poorly neutralized wildtype 2019-2020 A/Kansas/14/2017 (H3N2) HA-pseudoviruses. CONCLUSION Egg- and cell-derived 2018-2019 season influenza vaccines elicited similar neutralization titers and response rates, indicating that the cell-derived vaccine did not improve immunogenicity against the A(H3N2) viruses. The higher responses after rHA vaccination may be due to its higher HA content. All vaccines boosted titers to HA with egg-adaptive substitutions, suggesting boosting from past antigens or better exposure of HA epitopes. Studies comparing immunogenicity and effectiveness of different influenza vaccines across many seasons are needed.
Collapse
Affiliation(s)
- Wei Wang
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Russell Vassell
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Limone Collins
- Defense Health Agency- Immunization Healthcare Division, Arlington Boulevard, Falls Church, VA, USA.,Walter Reed National Military Medical Center, Rockville Pike, Bethesda, MD, USA
| | - Rhonda E Colombo
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA.,Madigan Army Medical Center, Fort Lewis, Washington, USA
| | - Anuradha Ganesan
- Walter Reed National Military Medical Center, Rockville Pike, Bethesda, MD, USA.,Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA
| | - Casey Geaney
- Walter Reed National Military Medical Center, Rockville Pike, Bethesda, MD, USA
| | - David Hrncir
- Defense Health Agency- Immunization Healthcare Division Regional Office, Wilford Hall Loop, Lackland AFB, TX, USA
| | - Tahaniyat Lalani
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA.,Naval Medical Center, John Paul Jones Circle, Portsmouth VA
| | | | - Ryan C Maves
- Division of Infectious Diseases, Naval Medical Center, San Diego, CA, USA
| | - Bruce McClenathan
- Defense Health Agency- Immunization Healthcare Division Regional Office, Ft. Bragg, NC, USA
| | - Katrin Mende
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA.,Brooke Army Medical Center, Roger Brooke Dr, JBSA Fort Sam Houston, TX, USA
| | - Stephanie A Richard
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA
| | - Christina Schofield
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Madigan Army Medical Center, Fort Lewis, Washington, USA
| | - Srihari Seshadri
- Defense Health Agency- Immunization Healthcare Division, Arlington Boulevard, Falls Church, VA, USA
| | - Christina Spooner
- Defense Health Agency- Immunization Healthcare Division, Arlington Boulevard, Falls Church, VA, USA
| | - Gregory C Utz
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA.,Division of Infectious Diseases, Naval Medical Center, San Diego, CA, USA
| | | | - Min Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta GA, USA
| | - Christian L Coles
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockledge Dr, Bethesda, MD, USA
| | - Timothy H Burgess
- Infectious Diseases Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Maryna Eichelberger
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Carol D Weiss
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
4
|
Wang W, Chen Q, Ford-Siltz LA, Katzelnick LC, Parra GI, Song HS, Vassell R, Weiss CD. Neutralizing Antibody Responses to Homologous and Heterologous H1 and H3 Influenza A Strains After Vaccination With Inactivated Trivalent Influenza Vaccine Vary With Age and Prior-year Vaccination. Clin Infect Dis 2020; 68:2067-2078. [PMID: 30256912 DOI: 10.1093/cid/ciy818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/21/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prior influenza immunity influences the homologous neutralizing antibody responses elicited by inactivated influenza vaccines (IIV), but neutralizing antibody responses to heterologous strains have not been extensively characterized. METHODS We analyzed neutralizing antibody titers in individuals aged 1-88 who received the 2009-2010 season IIV before infection by or vaccination against the 2009 pandemic H1N1 virus. Neutralization titers to homologous and heterologous past, recent, and advanced H1 and H3 strains, as well as H2, H5, and H7 strains, were measured using influenza hemagglutinin pseudoviruses. We performed exploratory analyses based on age, prior-year IIV, and prevaccination titer, without controlling for Type I errors. RESULTS IIV elicited neutralizing antibodies to past and advanced H1 and H3 strains, as well as to an H2 strain in individuals who were likely infected early in life. The neutralization of avian subtype viruses was rare, and there was no imprinting of neutralization responses to novel avian subtype viruses based on the influenza group. Compared to adults, children had higher seroresponse rates to homologous and heterologous strains, and their sera generated larger antigenic distances among strains. Seroresponse rates to homologous and heterologous strains were lower in subjects vaccinated with prior-year IIV, though postimmunization titers were generally high. CONCLUSIONS IIV elicited neutralizing antibodies to heterologous H1 and H3 strains in all ages groups, but titers and seroresponse rates were usually higher in children. Prior-year vaccination with the same strains tended to blunt IIV neutralization responses to all strains in young and old age groups, yet postimmunization titers were high.
Collapse
Affiliation(s)
- Wei Wang
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Qiong Chen
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Lauren A Ford-Siltz
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley
| | - Gabriel I Parra
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Hyo Sook Song
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Russell Vassell
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Carol D Weiss
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
5
|
Nath Neerukonda S, Vassell R, Weiss CD. Neutralizing Antibodies Targeting the Conserved Stem Region of Influenza Hemagglutinin. Vaccines (Basel) 2020; 8:E382. [PMID: 32664628 PMCID: PMC7563823 DOI: 10.3390/vaccines8030382] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022] Open
Abstract
Influenza continues to be a public health threat despite the availability of annual vaccines. While vaccines are generally effective at inducing strain-specific immunity, they are sub-optimal or ineffective when drifted or novel pandemic strains arise due to sequence changes in the major surface glycoprotein hemagglutinin (HA). The discovery of a large number of antibodies targeting the highly conserved stem region of HAs that are capable of potently neutralizing a broad range of virus strains and subtypes suggests new ways to protect against influenza. The structural characterization of HA stem epitopes and broadly neutralizing antibody paratopes has enabled the design of novel proteins, mini-proteins, and peptides targeting the HA stem, thus providing a foundation for the design of new vaccines. In this narrative, we comprehensively review the current knowledge about stem-directed broadly neutralizing antibodies and the structural features contributing to virus neutralization.
Collapse
Affiliation(s)
| | | | - Carol D. Weiss
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA; (S.N.N.); (R.V.)
| |
Collapse
|
6
|
Generation of a protective murine monoclonal antibody against the stem of influenza hemagglutinins from group 1 viruses and identification of resistance mutations against it. PLoS One 2019; 14:e0222436. [PMID: 31513662 PMCID: PMC6742228 DOI: 10.1371/journal.pone.0222436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/30/2019] [Indexed: 01/03/2023] Open
Abstract
Vaccines that elicit broadly cross-neutralizing antibodies, including antibodies that target the conserved stem of hemagglutinin (HA), are being developed as a strategy for next-generation influenza vaccines that protect against influenza across multiple years. However, efficient induction of cross-neutralizing antibodies remains a challenge, and potential escape mutations have not been well characterized. Here we elicited cross-neutralizing antibodies by immunizing animals with the hemagglutinins from H5 and H9 subtype influenza A viruses that are sensitive to neutralization by stem antibodies. We further isolated and characterized an HA stem monoclonal antibody 4C2 that broadly neutralizes group 1 influenza viruses and identified HA mutations that reduced sensitivity to stem antibodies. Our results offer insights for next-generation influenza vaccine strategies for inducing cross-neutralizing antibodies.
Collapse
|
7
|
Rohini K, Roy R, Ramanathan K, Shanthi V. E-pharmacophore hypothesis strategy to discover potent inhibitor for influenza treatment. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2019. [DOI: 10.1142/s0219633619500214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The surface protein of Influenza virus, Neuraminidase (NA), is believed to play a critical role in the release of new viral particle and thus spreads infection. It has been recognized as a valid drug target for anti-influenza therapy. Despite the number of available approved drugs for the influenza infection treatment, the emergence of resistant variants with novel mutations are the foremost challenges for the currently used NA inhibitors. Thus, the current investigation was carried out to ascertain potent inhibitors using computational strategies such as e-pharmacophore based virtual screening and docking approach. A three-dimensional e-pharmacophore hypothesis was generated based on the chemical features of complexes of the drugs and NA protein using PHASE module of Schrödinger suite. The generated hypothesis consisted of one hydrogen bond acceptor (A), two hydrogen bond donors (D), one negatively charged group (N) and one aromatic ring (R), ADDNR. The hypothesis was further evaluated for its integrity using enrichment analysis and used to filter out molecules with similar pharmacophoric features from approved, investigational and experimental subsets of DrugBank and ZINC database. In addition, ligand filtration was performed to curb down the molecules to an efficient collection of hit molecules by using Lipinski “rule of five and ADME analysis by using Qikprop module. Overall, the results from our analysis suggest that compound lisinopril and formoterol could serve as potent antiviral compounds for the treatment of influenza A virus infection. It is worth mentioning that the results correlate well with literature evidences.
Collapse
Affiliation(s)
- K. Rohini
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Roosha Roy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - K. Ramanathan
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - V. Shanthi
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
8
|
Conformational Stability of the Hemagglutinin of H5N1 Influenza A Viruses Influences Susceptibility to Broadly Neutralizing Stem Antibodies. J Virol 2018; 92:JVI.00247-18. [PMID: 29593038 PMCID: PMC5974491 DOI: 10.1128/jvi.00247-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/14/2018] [Indexed: 12/29/2022] Open
Abstract
Vaccines that elicit broadly neutralizing antibodies to the conserved stem of hemagglutinin (HA) are being developed as universal influenza vaccines that protect against influenza across multiple years. However, different influenza virus strains, even those in the same subtype with identical stem sequences, can vary in susceptibility to broadly neutralizing stem antibodies, and the reasons are not understood. Here we studied potential mechanisms underlying the differing sensitivities of a panel of H5N1 HA pseudoviruses to broadly neutralizing stem antibodies. We found that greater HA conformational stability, as measured by thermal inactivation and pH triggering of conformational changes, correlates with reduced neutralization sensitivity and antibody binding to HA under neutral- and low-pH conditions. Our data indicate that the conformational stability of HA is an important attribute of susceptibility to broadly neutralizing stem antibodies and is influenced by residues outside the stem antibody epitopes. IMPORTANCE The influenza virus surface glycoprotein hemagglutinin (HA) mediates virus attachment and membrane fusion between virus and host cells, allowing the viral core to enter the host cell cytoplasm for replication. Fusion occurs when HA undergoes low-pH-induced-conformational changes during endocytosis. Broadly neutralizing antibodies targeted to the conserved stem region of HA interfere with conformational changes required for fusion. Vaccines that elicit such antibodies are being developed as novel universal influenza vaccines for multiyear protection. We investigated why H5N1 HAs from different strains differ in their sensitivity to broadly neutralizing stem antibodies despite having conserved epitopes. We report that HA conformational stability due to residues outside the antibody binding site accounted for much of the variation in susceptibility to neutralization by stem antibodies. These findings highlight the importance of nonepitope residues in influencing neutralization sensitivity to stem antibodies and the complexities in developing universal vaccines targeting conserved epitopes in the HA stem.
Collapse
|
9
|
Meseda CA, Atukorale V, Soto J, Eichelberger MC, Gao J, Wang W, Weiss CD, Weir JP. Immunogenicity and Protection Against Influenza H7N3 in Mice by Modified Vaccinia Virus Ankara Vectors Expressing Influenza Virus Hemagglutinin or Neuraminidase. Sci Rep 2018; 8:5364. [PMID: 29599502 PMCID: PMC5876369 DOI: 10.1038/s41598-018-23712-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/14/2018] [Indexed: 12/18/2022] Open
Abstract
Influenza subtypes such as H7 have pandemic potential since they are able to infect humans with severe consequences, as evidenced by the ongoing H7N9 infections in China that began in 2013. The diversity of H7 viruses calls for a broadly cross-protective vaccine for protection. We describe the construction of recombinant modified vaccinia virus Ankara (MVA) vectors expressing the hemagglutinin (HA) or neuraminidase (NA) from three H7 viruses representing both Eurasian and North American H7 lineages – A/mallard/Netherlands/12/2000 (H7N3), A/Canada/rv444/2004 (H7N3), and A/Shanghai/02/2013 (H7N9). These vectors were evaluated for immunogenicity and protective efficacy against H7N3 virus in a murine model of intranasal challenge. High levels of H7-, N3-, and N9-specific antibodies, including neutralizing antibodies, were induced by the MVA-HA and MVA-NA vectors. Mice vaccinated with MVA vectors expressing any of the H7 antigens were protected, suggesting cross-protection among H7 viruses. In addition, MVA vectors expressing N3 but not N9 elicited protection against H7N3 virus challenge. Similar outcomes were obtained when immune sera from MVA vector-immunized mice were passively transferred to naïve mice prior to challenge with the H7N3 virus. The results support the further development of an MVA vector platform as a candidate vaccine for influenza strains with pandemic potential.
Collapse
Affiliation(s)
- Clement A Meseda
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Vajini Atukorale
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jackeline Soto
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Maryna C Eichelberger
- Laboratory of Respiratory Viral Diseases, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jin Gao
- Laboratory of Respiratory Viral Diseases, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Wei Wang
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Carol D Weiss
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Jerry P Weir
- Laboratory of DNA Viruses, Center for Biologics Evaluations and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
10
|
Khalenkov A, He Y, Reed JL, Kreil TR, McVey J, Norton M, Scott J, Scott DE. Characterization of source plasma from self-identified vaccinated or convalescent donors during the 2009 H1N1 pandemic. Transfusion 2018; 58:1108-1116. [DOI: 10.1111/trf.14530] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 12/27/2017] [Accepted: 12/27/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Alexey Khalenkov
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| | - Yong He
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| | - Jennifer L. Reed
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| | | | | | - Malgorzata Norton
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| | - John Scott
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| | - Dorothy E. Scott
- Center for Biologics Evaluation and Research; Food and Drug Administration; Silver Spring Maryland
| |
Collapse
|
11
|
Anderson CS, Ortega S, Chaves FA, Clark AM, Yang H, Topham DJ, DeDiego ML. Natural and directed antigenic drift of the H1 influenza virus hemagglutinin stalk domain. Sci Rep 2017; 7:14614. [PMID: 29097696 PMCID: PMC5668287 DOI: 10.1038/s41598-017-14931-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/18/2017] [Indexed: 12/02/2022] Open
Abstract
The induction of antibodies specific for the influenza HA protein stalk domain is being pursued as a universal strategy against influenza virus infections. However, little work has been done looking at natural or induced antigenic variability in this domain and the effects on viral fitness. We analyzed human H1 HA head and stalk domain sequences and found substantial variability in both, although variability was highest in the head region. Furthermore, using human immune sera from pandemic A/California/04/2009 immune subjects and mAbs specific for the stalk domain, viruses were selected in vitro containing mutations in both domains that partially contributed to immune evasion. Recombinant viruses encoding amino acid changes in the HA stalk domain replicated well in vitro, and viruses incorporating two of the stalk mutations retained pathogenicity in vivo. These findings demonstrate that the HA protein stalk domain can undergo limited drift under immune pressure and the viruses can retain fitness and virulence in vivo, findings which are important to consider in the context of vaccination targeting this domain.
Collapse
Affiliation(s)
- Christopher S Anderson
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States
| | - Sandra Ortega
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States
| | - Francisco A Chaves
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States
| | - Amelia M Clark
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States
| | - Hongmei Yang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States.
| | - Marta L DeDiego
- David H. Smith Center for Vaccine Biology and Immunology, and Department of Microbiology and Immunology, Rochester, NY, United States.
| |
Collapse
|
12
|
Lo CY, Strobl SL, Dunham K, Wang W, Stewart L, Misplon JA, Garcia M, Gao J, Ozawa T, Price GE, Navidad J, Gradus S, Bhattacharyya S, Viboud C, Eichelberger MC, Weiss CD, Gorski J, Epstein SL. Surveillance Study of Influenza Occurrence and Immunity in a Wisconsin Cohort During the 2009 Pandemic. Open Forum Infect Dis 2017; 4:ofx023. [PMID: 28730155 PMCID: PMC5510460 DOI: 10.1093/ofid/ofx023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/08/2017] [Indexed: 12/21/2022] Open
Abstract
Background Antibody and T-cell immunity to conserved influenza virus antigens can protect animals against infection with diverse influenza strains. Although immunity against conserved antigens occurs in humans, whether such responses provide cross-protection in humans and could be harnessed as the basis for universal influenza vaccines is controversial. The 2009 pandemic provided an opportunity to investigate whether pre-existing cross-reactive immunity affected susceptibility to infection. Methods In 2009, we banked sera and peripheral blood mononuclear cells (PBMC) from blood donors, then monitored them for pandemic influenza infection (pH1N1) by polymerase chain reaction or seroconversion. Antibodies to hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix 2 (M2), and HA-pseudotypes were measured in sera. T-cell inteferon-γ enzyme-linked immunospot responses were measured in PBMC. Results There were 13 infections in 117 evaluable donors. Pre-existing T-cell reactivity to pH1N1 was substantial (of 153 donors tested, 146 had >100 spot-forming cells/106 cells). Antibodies reactive with pH1N1 were common: anti-NP (all donors) and anti-M2 (44% of donors). Pseudotype-neutralizing antibodies to H1 were detected, but not to highly conserved HA epitopes. Unexpectedly, donors with symptomatic pH1N1 infection had sharp rises in HA pseudotype-neutralizing antibodies, not only pH1N1 but also against multiple seasonal H1s. In addition, an exploratory study of a T-cell marker (response to NP418-426) identified probable infection missed by standard criteria. Conclusions Although the number of infections was inadequate for conclusions about mechanisms of protection, this study documents the wide variety of pre-existing, cross-reactive, humoral and cellular immune responses to pandemic influenza virus antigens in humans. These responses can be compared with results of other studies and explored in universal influenza vaccine studies.
Collapse
Affiliation(s)
- Chia-Yun Lo
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Susan L Strobl
- Laboratory of Cell-Mediated Immunity, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Kimberly Dunham
- Laboratory of Cell-Mediated Immunity, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Wei Wang
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Julia A Misplon
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Mayra Garcia
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Jin Gao
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Tatsuhiko Ozawa
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Japan
| | - Graeme E Price
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Jose Navidad
- City of Milwaukee Health Department Laboratory, Wisconsin; and
| | - Steve Gradus
- City of Milwaukee Health Department Laboratory, Wisconsin; and
| | | | - Cecile Viboud
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland
| | - Maryna C Eichelberger
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Carol D Weiss
- Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Jack Gorski
- BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Suzanne L Epstein
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
13
|
Alvarado-Facundo E, Vassell R, Schmeisser F, Weir JP, Weiss CD, Wang W. Glycosylation of Residue 141 of Subtype H7 Influenza A Hemagglutinin (HA) Affects HA-Pseudovirus Infectivity and Sensitivity to Site A Neutralizing Antibodies. PLoS One 2016; 11:e0149149. [PMID: 26862918 PMCID: PMC4749315 DOI: 10.1371/journal.pone.0149149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
Human infections with H7 subtype influenza virus have been reported, including an H7N7 outbreak in Netherlands in 2003 and H7N9 infections in China in 2013. Previously, we reported murine monoclonal antibodies (mAbs) that recognize the antigenic site A of H7 hemagglutinin (HA). To better understand protective immunity of H7 vaccines and vaccine candidate selection, we used these mAbs to assess the antigenic relatedness among two H7 HA isolated from past human infections and determine residues that affect susceptibility to neutralization. We found that these mAbs neutralize pseudoviruses bearing HA of A/Shanghai/02/2013(H7N9), but not A/Netherlands/219/2003(H7N7). Glycosylation of the asparagine residue at position 141 (N141) (N133, H3 HA numbering) in the HA of A/Netherlands/219/2003 HA is responsible for this resistance, and it affects the infectivity of HA-pseudoviruses. The presence of threonine at position 143 (T135, H3 HA numbering) in the HA of A/Netherlands/219/2003, rather than an alanine found in the HA of A/Shanghai/02/2013(H7N9), accounts for these differences. These results demonstrate a key role for glycosylation of residue N141 in affecting H7 influenza HA-mediated entry and sensitivity to neutralizing antibodies, which have implications for candidate vaccine design.
Collapse
MESH Headings
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Enzyme-Linked Immunosorbent Assay
- Glycosylation
- HEK293 Cells
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Humans
- Influenza A Virus, H7N7 Subtype/immunology
- Influenza A Virus, H7N7 Subtype/pathogenicity
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/pathogenicity
- Influenza Vaccines/immunology
- Influenza, Human/virology
- Neutralization Tests
- Open Reading Frames
- Plasmids/metabolism
Collapse
Affiliation(s)
- Esmeralda Alvarado-Facundo
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
| | - Russell Vassell
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
| | - Falko Schmeisser
- Laboratory of DNA Viruses, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
| | - Jerry P. Weir
- Laboratory of DNA Viruses, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
| | - Carol D. Weiss
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
- * E-mail: (CDW); (WW)
| | - Wei Wang
- Laboratory of Immunoregulation, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland 20993, United States of America
- * E-mail: (CDW); (WW)
| |
Collapse
|
14
|
Intermonomer Interactions in Hemagglutinin Subunits HA1 and HA2 Affecting Hemagglutinin Stability and Influenza Virus Infectivity. J Virol 2015; 89:10602-11. [PMID: 26269180 DOI: 10.1128/jvi.00939-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/04/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Influenza virus hemagglutinin (HA) mediates virus entry by binding to cell surface receptors and fusing the viral and endosomal membranes following uptake by endocytosis. The acidic environment of endosomes triggers a large-scale conformational change in the transmembrane subunit of HA (HA2) involving a loop (B loop)-to-helix transition, which releases the fusion peptide at the HA2 N terminus from an interior pocket within the HA trimer. Subsequent insertion of the fusion peptide into the endosomal membrane initiates fusion. The acid stability of HA is influenced by residues in the fusion peptide, fusion peptide pocket, coiled-coil regions of HA2, and interactions between the surface (HA1) and HA2 subunits, but details are not fully understood and vary among strains. Current evidence suggests that the HA from the circulating pandemic 2009 H1N1 influenza A virus [A(H1N1)pdm09] is less stable than the HAs from other seasonal influenza virus strains. Here we show that residue 205 in HA1 and residue 399 in the B loop of HA2 (residue 72, HA2 numbering) in different monomers of the trimeric A(H1N1)pdm09 HA are involved in functionally important intermolecular interactions and that a conserved histidine in this pair helps regulate HA stability. An arginine-lysine pair at this location destabilizes HA at acidic pH and mediates fusion at a higher pH, while a glutamate-lysine pair enhances HA stability and requires a lower pH to induce fusion. Our findings identify key residues in HA1 and HA2 that interact to help regulate H1N1 HA stability and virus infectivity. IMPORTANCE Influenza virus hemagglutinin (HA) is the principal antigen in inactivated influenza vaccines and the target of protective antibodies. However, the influenza A virus HA is highly variable, necessitating frequent vaccine changes to match circulating strains. Sequence changes in HA affect not only antigenicity but also HA stability, which has important implications for vaccine production, as well as viral adaptation to hosts. HA from the pandemic 2009 H1N1 influenza A virus is less stable than other recent seasonal influenza virus HAs, but the molecular interactions that contribute to HA stability are not fully understood. Here we identify molecular interactions between specific residues in the surface and transmembrane subunits of HA that help regulate the HA conformational changes needed for HA stability and virus entry. These findings contribute to our understanding of the molecular mechanisms controlling HA function and antigen stability.
Collapse
|
15
|
Wang W, Alvarado-Facundo E, Chen Q, Anderson CM, Scott D, Vassell R, Weiss CD. Serum Samples From Middle-aged Adults Vaccinated Annually with Seasonal Influenza Vaccines Cross-neutralize Some Potential Pandemic Influenza Viruses. J Infect Dis 2015; 213:403-6. [PMID: 26243315 PMCID: PMC7313900 DOI: 10.1093/infdis/jiv407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/22/2015] [Indexed: 11/26/2022] Open
Abstract
We examined serum samples from adults ages 48–64 who received multiple seasonal influenza vaccines from 2004 to 2009 for cross-neutralizing antibodies to potential pandemic strains. Using pseudoviruses bearing various hemagglutinins (HA-pseudoviruses), we found serum neutralization titers (≥160) in 100% against A/Japan/305/1957 (H2N2), 53% against A/Hong Kong/1073/99 (H9N2), 56% against the H3N2 variant A/Indiana/08/11 (H3N2v), 11% against A/Hong Kong/G9/97 (H9N2), and 36% A/chicken/Hong Kong/SF4/01 (H6N1). None had titers >160 to A/Shanghai/2/13 (H7N9) or A/Netherlands/219/03 (H7N7). Thirty-six percent to 0% had neutralization titers to various H5N1 strains. Titers to H9, H6, and H5 HA-pseudoviruses correlated with each other, but not with H3N2v, suggesting group-specific cross-neutralization.
Collapse
Affiliation(s)
- Wei Wang
- Laboratory of Immunoregulation, Division of Viral Products
| | | | - Qiong Chen
- Laboratory of Immunoregulation, Division of Viral Products
| | - Christine M Anderson
- Division of Hematology, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Dorothy Scott
- Division of Hematology, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Carol D Weiss
- Laboratory of Immunoregulation, Division of Viral Products
| |
Collapse
|
16
|
Carnell GW, Ferrara F, Grehan K, Thompson CP, Temperton NJ. Pseudotype-based neutralization assays for influenza: a systematic analysis. Front Immunol 2015; 6:161. [PMID: 25972865 PMCID: PMC4413832 DOI: 10.3389/fimmu.2015.00161] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/25/2015] [Indexed: 12/02/2022] Open
Abstract
The use of vaccination against the influenza virus remains the most effective method of mitigating the significant morbidity and mortality caused by this virus. Antibodies elicited by currently licensed influenza vaccines are predominantly hemagglutination-inhibition (HI)-competent antibodies that target the globular head of hemagglutinin (HA) thus inhibiting influenza virus entry into target cells. These antibodies predominantly confer homosubtypic/strain specific protection and only rarely confer heterosubtypic protection. However, recent academia or pharma-led R&D toward the production of a “universal vaccine” has centered on the elicitation of antibodies directed against the stalk of the influenza HA that has been shown to confer broad protection across a range of different subtypes (H1–H16). The accurate and sensitive measurement of antibody responses elicited by these “next-generation” influenza vaccines is, however, hampered by the lack of sensitivity of the traditional influenza serological assays HI, single radial hemolysis, and microneutralization. Assays utilizing pseudotypes, chimeric viruses bearing influenza glycoproteins, have been shown to be highly efficient for the measurement of homosubtypic and heterosubtypic broadly neutralizing antibodies, making them ideal serological tools for the study of cross-protective responses against multiple influenza subtypes with pandemic potential. In this review, we will analyze and compare literature involving the production of influenza pseudotypes with particular emphasis on their use in serum antibody neutralization assays. This will enable us to establish the parameters required for optimization and propose a consensus protocol to be employed for the further deployment of these assays in influenza vaccine immunogenicity studies.
Collapse
Affiliation(s)
- George William Carnell
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Francesca Ferrara
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Keith Grehan
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| | - Craig Peter Thompson
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK ; Department of Zoology, University of Oxford , Oxford , UK ; The Jenner Institute Laboratories, University of Oxford , Oxford , UK
| | - Nigel James Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway , Chatham Maritime, Kent , UK
| |
Collapse
|
17
|
Comparative structural analysis of haemagglutinin proteins from type A influenza viruses: conserved and variable features. BMC Bioinformatics 2014; 15:363. [PMID: 25492298 PMCID: PMC4265342 DOI: 10.1186/s12859-014-0363-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023] Open
Abstract
Background Genome variation is very high in influenza A viruses. However, viral evolution and spreading is strongly influenced by immunogenic features and capacity to bind host cells, depending in turn on the two major capsidic proteins. Therefore, such viruses are classified based on haemagglutinin and neuraminidase types, e.g. H5N1. Current analyses of viral evolution are based on serological and primary sequence comparison; however, comparative structural analysis of capsidic proteins can provide functional insights on surface regions possibly crucial to antigenicity and cell binding. Results We performed extensive structural comparison of influenza virus haemagglutinins and of their domains and subregions to investigate type- and/or domain-specific variation. We found that structural closeness and primary sequence similarity are not always tightly related; moreover, type-specific features could be inferred when comparing surface properties of haemagglutinin subregions, monomers and trimers, in terms of electrostatics and hydropathy. Focusing on H5N1, we found that variation at the receptor binding domain surface intriguingly relates to branching of still circulating clades from those ones that are no longer circulating. Conclusions Evidence from this work suggests that integrating phylogenetic and serological analyses by extensive structural comparison can help in understanding the ‘functional evolution’ of viral surface determinants. In particular, variation in electrostatic and hydropathy patches can provide molecular evolution markers: intriguing surface charge redistribution characterizing the haemagglutinin receptor binding domains from circulating H5N1 clades 2 and 7 might have contributed to antigenic escape hence to their evolutionary success and spreading. Electronic supplementary material The online version of this article (doi:10.1186/s12859-014-0363-5) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Influenza virus M2 protein ion channel activity helps to maintain pandemic 2009 H1N1 virus hemagglutinin fusion competence during transport to the cell surface. J Virol 2014; 89:1975-85. [PMID: 25473053 DOI: 10.1128/jvi.03253-14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED The influenza virus hemagglutinin (HA) envelope protein mediates virus entry by first binding to cell surface receptors and then fusing viral and endosomal membranes during endocytosis. Cleavage of the HA precursor (HA0) into a surface receptor-binding subunit (HA1) and a fusion-inducing transmembrane subunit (HA2) by host cell enzymes primes HA for fusion competence by repositioning the fusion peptide to the newly created N terminus of HA2. We previously reported that the influenza virus M2 protein enhances pandemic 2009 influenza A virus [(H1N1)pdm09] HA-pseudovirus infectivity, but the mechanism was unclear. In this study, using cell-cell fusion and HA-pseudovirus infectivity assays, we found that the ion channel function of M2 was required for enhancement of HA fusion and HA-pseudovirus infectivity. The M2 activity was needed only during HA biosynthesis, and proteolysis experiments indicated that M2 proton channel activity helped to protect (H1N1)pdm09 HA from premature conformational changes as it traversed low-pH compartments during transport to the cell surface. While M2 has previously been shown to protect avian influenza virus HA proteins of the H5 and H7 subtypes that have polybasic cleavage motifs, this study demonstrates that M2 can protect HA proteins from human H1N1 strains that lack a polybasic cleavage motif. This finding suggests that M2 proton channel activity may play a wider role in preserving HA fusion competence among a variety of HA subtypes, including HA proteins from emerging strains that may have reduced HA stability. IMPORTANCE Influenza virus infects cells when the hemagglutinin (HA) surface protein undergoes irreversible pH-induced conformational changes after the virus is taken into the cell by endocytosis. HA fusion competence is primed when host cell enzymes cleave the HA precursor. The proton channel function of influenza virus M2 protein has previously been shown to protect avian influenza virus HA proteins that contain a polybasic cleavage site from pH-induced conformational changes during biosynthesis, but this effect is less well understood for human influenza virus HA proteins that lack polybasic cleavage sites. Using assays that focus on HA entry and fusion, we found that the M2 protein also protects (H1N1)pdm09 influenza A virus HA from premature conformational changes as it transits low-pH compartments during biosynthesis. This work suggests that M2 may play a wider role in preserving HA function in a variety of influenza virus subtypes that infect humans and may be especially important for HA proteins that are less stable.
Collapse
|
19
|
Ren J, Ellis J, Li J. Influenza A HA's conserved epitopes and broadly neutralizing antibodies: a prediction method. J Bioinform Comput Biol 2014; 12:1450023. [PMID: 25208658 DOI: 10.1142/s0219720014500231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A conserved epitope is an epitope retained by multiple strains of influenza as the key target of a broadly neutralizing antibody. Identification of conserved epitopes is of strong interest to help design broad-spectrum vaccines against influenza. Conservation score measures the evolutionary conservation of an amino acid position in a protein based on the phylogenetic relationships observed amongst homologous sequences. Here, Average Amino Acid Conservation Score (AAACS) is proposed as a method to identify HA's conserved epitopes. Our analysis shows that there is a clear distinction between conserved epitopes and nonconserved epitopes in terms of AAACS. This method also provides an excellent classification performance on an independent dataset. In contrast, alignment-based comparison methods do not work well for this problem, because conserved epitopes to the same broadly neutralizing antibody are usually not identical or similar. Location-based methods are not successful either, because conserved epitopes are located at both the less-conserved globular head (HA1) and the more-conserved stem (HA2). As a case study, two conserved epitopes on HA are predicted for the influenza A virus H7N9: One should match the broadly neutralizing antibodies CR9114 or FI6v3, while the other is new and requires validation by wet-lab experiments.
Collapse
Affiliation(s)
- Jing Ren
- Advanced Analytics Institute, Faculty of Engineering and Information Technology, University of Technology Sydney, P. O. Box 123, Broadway, NSW 2007, Australia
| | | | | |
Collapse
|
20
|
Li J, Arévalo MT, Chen Y, Chen S, Zeng M. T-cell-mediated cross-strain protective immunity elicited by prime-boost vaccination with a live attenuated influenza vaccine. Int J Infect Dis 2014; 27:37-43. [PMID: 25172265 DOI: 10.1016/j.ijid.2014.05.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/24/2014] [Accepted: 05/19/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Antigenic drift and shift of influenza viruses require frequent reformulation of influenza vaccines. In addition, seasonal influenza vaccines are often mismatched to the epidemic influenza strains. This stresses the need for a universal influenza vaccine. METHODS BALB/c mice were vaccinated with the trivalent live attenuated (LAIV; FluMist) or inactivated (TIV; FluZone) influenza vaccines and challenged with PR8 (H1N1), FM/47 (H1N1), or HK/68 (H3N2) influenza virus. Cytokines and antibody responses were tested by ELISA. Furthermore, different LAIV dosages were applied in BALB/c mice. LAIV vaccinated mice were also depleted of T-cells and challenged with PR8 virus. RESULTS LAIV induced significant protection against challenge with the non-vaccine strain PR8 influenza virus. Furthermore, protective immunity against PR8 was dose-dependent. Of note, interleukin 2 and interferon gamma cytokine secretion in the lung alveolar fluid were significantly elevated in mice vaccinated with LAIV. Moreover, T-cell depletion of LAIV vaccinated mice compromised protection, indicating that T-cell-mediated immunity is required. In contrast, passive transfer of sera from mice vaccinated with LAIV into naïve mice failed to protect against PR8 challenge. Neutralization assays in vitro confirmed that LAIV did not induce cross-strain neutralizing antibodies against PR8 virus. Finally, we showed that three doses of LAIV also provided protection against challenge with two additional heterologous viruses, FM/47 and HK/68. CONCLUSIONS These results support the potential use of the LAIV as a universal influenza vaccine under a prime-boost vaccination regimen.
Collapse
Affiliation(s)
- Junwei Li
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Maria T Arévalo
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Yanping Chen
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Shan Chen
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Mingtao Zeng
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA.
| |
Collapse
|
21
|
Magadán JG, Altman MO, Ince WL, Hickman HD, Stevens J, Chevalier A, Baker D, Wilson PC, Ahmed R, Bennink JR, Yewdell JW. Biogenesis of influenza a virus hemagglutinin cross-protective stem epitopes. PLoS Pathog 2014; 10:e1004204. [PMID: 24945804 PMCID: PMC4055778 DOI: 10.1371/journal.ppat.1004204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 05/05/2014] [Indexed: 11/18/2022] Open
Abstract
Antigenic variation in the globular domain of influenza A virus (IAV) hemagglutinin (HA) precludes effective immunity to this major human pathogen. Although the HA stem is highly conserved between influenza virus strains, HA stem-reactive antibodies (StRAbs) were long considered biologically inert. It is now clear, however, that StRAbs reduce viral replication in animal models and protect against pathogenicity and death, supporting the potential of HA stem-based immunogens as drift-resistant vaccines. Optimally designing StRAb-inducing immunogens and understanding StRAb effector functions require thorough comprehension of HA stem structure and antigenicity. Here, we study the biogenesis of HA stem epitopes recognized in cells infected with various drifted IAV H1N1 strains using mouse and human StRAbs. Using a novel immunofluorescence (IF)-based assay, we find that human StRAbs bind monomeric HA in the endoplasmic reticulum (ER) and trimerized HA in the Golgi complex (GC) with similar high avidity, potentially good news for producing effective monomeric HA stem immunogens. Though HA stem epitopes are nestled among several N-linked oligosaccharides, glycosylation is not required for full antigenicity. Rather, as N-linked glycans increase in size during intracellular transport of HA through the GC, StRAb binding becomes temperature-sensitive, binding poorly to HA at 4°C and well at 37°C. A de novo designed, 65-residue protein binds the mature HA stem independently of temperature, consistent with a lack of N-linked oligosaccharide steric hindrance due to its small size. Likewise, StRAbs bind recombinant HA carrying simple N-linked glycans in a temperature-independent manner. Chemical cross-linking experiments show that N-linked oligosaccharides likely influence StRAb binding by direct local effects rather than by globally modifying the conformational flexibility of HA. Our findings indicate that StRAb binding to HA is precarious, raising the possibility that sufficient immune pressure on the HA stem region could select for viral escape mutants with increased steric hindrance from N-linked glycans.
Collapse
Affiliation(s)
- Javier G. Magadán
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Meghan O. Altman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - William L. Ince
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James Stevens
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Aaron Chevalier
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Patrick C. Wilson
- Department of Medicine, Section of Rheumatology, Committee on Immunology, Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, Illinois, United States of America
| | - Rafi Ahmed
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jack R. Bennink
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
22
|
Shatzkes K, Teferedegne B, Murata H. A simple, inexpensive method for preparing cell lysates suitable for downstream reverse transcription quantitative PCR. Sci Rep 2014; 4:4659. [PMID: 24722424 PMCID: PMC3983595 DOI: 10.1038/srep04659] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/24/2014] [Indexed: 11/30/2022] Open
Abstract
Sample nucleic acid purification can often be rate-limiting for conventional quantitative PCR (qPCR) workflows. We recently developed high-throughput virus microneutralization assays using an endpoint assessment approach based on reverse transcription qPCR (RT-qPCR). The need for cumbersome RNA purification is circumvented in our assays by making use of a commercial reagent that can easily generate crude cell lysates amenable to direct analysis by one-step RT-qPCR. In the present study, we demonstrate that a simple buffer containing a non-ionic detergent can serve as an inexpensive alternative to commercially available reagents for the purpose of generating RT-qPCR-ready cell lysates from MDCK cells infected with influenza virus. We have found that addition of exogenous RNase inhibitor as a buffer component is not essential in order to maintain RNA integrity, even following stress at 37 °C incubation for 1-2 hours, in cell-lysate samples either freshly prepared or previously stored frozen at -80 °C.
Collapse
Affiliation(s)
- Kenneth Shatzkes
- Laboratory of DNA Viruses, Division of Viral Products, OVRR, CBER, FDA, Bethesda, MD 20892, USA
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, OVRR, CBER, FDA, Bethesda, MD 20892, USA
- These authors contributed equally to this work
- Current address: Graduate School of Biomedical Sciences, New Jersey Medical School and Rutgers School of Dental Medicine, Newark, New Jersey, USA
| | - Belete Teferedegne
- Laboratory of DNA Viruses, Division of Viral Products, OVRR, CBER, FDA, Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Haruhiko Murata
- Laboratory of DNA Viruses, Division of Viral Products, OVRR, CBER, FDA, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Prior contacts with the 2000–2003 seasonal vaccines extends the 2009 pandemic A/H1N1 vaccine-specific immune protection to non-humoral compartments. Eur Geriatr Med 2014. [DOI: 10.1016/j.eurger.2014.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Lutteke N, Sausy A, Black AP, Wildschutz F, Muller CP. Neutralizing antibodies against influenza A in pigs before and after the 2009 pandemic, Luxembourg. Vet Microbiol 2014; 169:96-101. [DOI: 10.1016/j.vetmic.2013.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 12/11/2013] [Accepted: 12/15/2013] [Indexed: 12/01/2022]
|
25
|
Lees WD, Moss DS, Shepherd AJ. Evolution in the influenza A H3 stalk - a challenge for broad-spectrum vaccines? J Gen Virol 2013; 95:317-324. [PMID: 24187015 DOI: 10.1099/vir.0.059410-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recently, a number of broad-spectrum human antibodies binding to the stalk region of influenza A haemagglutinin (HA) have been isolated. As this region tends to develop substitutions at a slower rate than other regions of HA, a vaccine eliciting such antibodies could have a longer effective life. But this begs a question: is the stalk resistant to change even in the face of evolutionary pressure? In this paper, we analysed the known epitopes in the H3 stalk and, utilizing a collection of 3440 sequences, present a novel approach for detecting putative B-cell epitopes in regions such as this, in which mutations occur infrequently. We concluded that there have been periods of activity in the stalk that are consistent with the evolution of antigenic escape. This work casts light on the presence of stalk-binding antibodies in the population as a whole and, through the analysis of antigenically active regions in the stalk, may contribute to the identification of epitopes that are refractive to change and hence useful for vaccine development.
Collapse
Affiliation(s)
- William D Lees
- Institute of Structural and Molecular Biology and Department of Biological Sciences, Birkbeck College, University of London, UK
| | - David S Moss
- Institute of Structural and Molecular Biology and Department of Biological Sciences, Birkbeck College, University of London, UK
| | - Adrian J Shepherd
- Institute of Structural and Molecular Biology and Department of Biological Sciences, Birkbeck College, University of London, UK
| |
Collapse
|
26
|
Athmaram T, Saraswat S, Sikarwar B, Verma SK, Singh AK, Boopathi M. Characterization of pandemic influenza A (H1N1) virus hemagglutinin specific polyclonal antibodies for biosensor applications. J Med Virol 2013; 86:363-71. [DOI: 10.1002/jmv.23753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2013] [Indexed: 11/06/2022]
Affiliation(s)
- T.N. Athmaram
- Defence Research and Development Establishment; Ministry of Defence; Gwalior MP India
| | - Shweta Saraswat
- Defence Research and Development Establishment; Ministry of Defence; Gwalior MP India
| | - Bhavna Sikarwar
- Defence Research and Development Establishment; Ministry of Defence; Gwalior MP India
| | | | - Anil K. Singh
- Defence Research and Development Establishment; Ministry of Defence; Gwalior MP India
| | - M. Boopathi
- Defence Research and Development Establishment; Ministry of Defence; Gwalior MP India
| |
Collapse
|
27
|
Comparative serological assays for the study of h5 and h7 avian influenza viruses. INFLUENZA RESEARCH AND TREATMENT 2013; 2013:286158. [PMID: 24163763 PMCID: PMC3791816 DOI: 10.1155/2013/286158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/16/2013] [Indexed: 01/20/2023]
Abstract
The nature of influenza virus to randomly mutate and evolve into new types is an important challenge in the control of influenza infection. It is necessary to monitor virus evolution for a better understanding of the pandemic risk posed by certain variants as evidenced by the highly pathogenic avian influenza (HPAI) viruses. This has been clearly recognized in Egypt following the notification of the first HPAI H5N1 outbreak. The continuous circulation of the virus and the mass vaccination programme undertaken in poultry have resulted in a progressive genetic evolution and a significant antigenic drift near the major antigenic sites. In order to establish if vaccination is sufficient to provide significant intra- and interclade cross-protection, lentiviral pseudotypes derived from H5N1 HPAI viruses (A/Vietnam/1194/04, A/chicken/Egypt-1709-01/2007) and an antigenic drift variant (A/chicken/Egypt-1709-06-2008) were constructed and used in pseudotype-based neutralization assays (pp-NT). pp-NT data obtained was confirmed and correlated with HI and MN assays. A panel of pseudotypes belonging to influenza Groups 1 and 2, with a combination of reporter systems, was also employed for testing avian sera in order to support further application of pp-NT as an alternative valid assay that can improve avian vaccination efficacy testing, vaccine virus selection, and the reliability of reference sera.
Collapse
|
28
|
Development of a neutralization assay for influenza virus using an endpoint assessment based on quantitative reverse-transcription PCR. PLoS One 2013; 8:e56023. [PMID: 23437084 PMCID: PMC3577804 DOI: 10.1371/journal.pone.0056023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 01/09/2013] [Indexed: 12/15/2022] Open
Abstract
A microneutralization assay using an ELISA-based endpoint assessment (ELISA-MN) is widely used to measure the serological response to influenza virus infection and vaccination. We have developed an alternative microneutralization assay for influenza virus using a quantitative reverse transcription PCR-based endpoint assessment (qPCR-MN) in order to improve upon technical limitations associated with ELISA-MN. For qPCR-MN, infected MDCK-London cells in 96-well cell-culture plates are processed with minimal steps such that resulting samples are amenable to high-throughput analysis by downstream one-step quantitative reverse transcription PCR (qRT-PCR; SYBR Green chemistry with primers targeting a conserved region of the M1 gene of influenza A viruses). The growth curves of three recent vaccine strains demonstrated that the qRT-PCR signal detected at 6 hours post-infection reflected an amplification of at least 100-fold over input. Using ferret antisera, we have established the feasibility of measuring virus neutralization at 6 hours post-infection, a duration likely confined to a single virus-replication cycle. The neutralization titer for qPCR-MN was defined as the highest reciprocal serum dilution necessary to achieve a 90% inhibition of the qRT-PCR signal; this endpoint was found to be in agreement with ELISA-MN using the same critical reagents in each assay. qPCR-MN was robust with respect to assay duration (6 hours vs. 12 hours). In addition, qPCR-MN appeared to be compliant with the Percentage Law (i.e., virus neutralization results appear to be consistent over an input virus dose ranging from 500 to 12,000 TCID50). Compared with ELISA-MN, qPCR-MN might have inherent properties conducive to reducing intra- and inter-laboratory variability while affording suitability for automation and high-throughput uses. Finally, our qRT-PCR-based approach may be broadly applicable to the development of neutralization assays for a wide variety of viruses.
Collapse
|
29
|
Schmeisser F, Friedman R, Besho J, Lugovtsev V, Soto J, Wang W, Weiss C, Williams O, Xie H, Ye Z, Weir JP. Neutralizing and protective epitopes of the 2009 pandemic influenza H1N1 hemagglutinin. Influenza Other Respir Viruses 2012; 7:480-90. [PMID: 23122228 PMCID: PMC5779835 DOI: 10.1111/irv.12029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
AIMS AND METHODS To facilitate antigenic characterization of the influenza A 2009 pandemic H1N1 [A(H1N1)pdm09] hemagglutinin (HA), we generated a panel of murine monoclonal antibodies (mAbs) using as the immunogen mammalian-derived virus-like particles containing the HA of the A/California/04/2009 virus. The antibodies were specific for the A/California/04/2009 HA, and individual mAbs suitable for use in several practical applications including ELISA, immunofluorescence, and Western blot analysis were identified. RESULTS AND CONCLUSIONS As the panel of mAbs included antibodies with hemagglutination inhibition (HI) and virus neutralizing activities, this allowed identification and characterization of potentially important antigenic and neutralizing epitopes of the A/California/04/2009 HA and comparison of those epitopes with the HAs of other influenza viruses including seasonal H1N1 viruses as well as the A/South Carolina/1918 and A/New Jersey/1976 H1N1 viruses. Three mAbs with the highest HI and neutralizing titers were able to provide passive protection against virus challenge. Two other mAbs without HI or neutralizing activities were able to provide partial protection against challenge. HA epitopes recognized by the strongest neutralizing mAbs in the panel were identified by isolation and selection of virus escape mutants in the presence of individual mAbs. Cloned viruses resistant to HI and antibody neutralization were sequenced to identify mutations, and two unique mutations (D127E and G155E) were identified, both near the antigenic site Sa. Using human post-vaccination sera, however, there were no differences in HI titer between A/California/04/2009 and either escape mutant, suggesting that these single mutations were not sufficient to abrogate a protective antibody response to the vaccine.
Collapse
Affiliation(s)
- Falko Schmeisser
- Division of Viral Products, Center for Biologics Evaluations and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Patel A, Kobinger GP. Evaluation of mismatched immunity against influenza viruses. Future Virol 2012. [DOI: 10.2217/fvl.12.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prior immunity against influenza A viruses generates sterilizing immunity against matched (homologous) viruses and varying levels of protection against mismatched (heterologous) viruses of the same or different subtypes. Natural immunity carries the risk of high morbidity and mortality, therefore immunization offers the best preventative measure. Antibody responses against the viral hemagglutinin protein correlate with protection in humans and evidence increasingly supports a role for robust cellular immune responses. By exploiting mismatched immunity, current conventional and experimental vaccine candidates can improve the generation of cross-protective immune responses against heterologous viruses. Experimental vaccines such as virus-like particles, DNA vectors, viral vectors and broadly neutralizing antibodies are able to expand cross-protection through mismatched B- and T-cell responses. However, the generation of mismatched immune responses can also have the opposite effect and impair protective immunity. This review discusses mismatched immunity in the context of natural infection and immunization. Additionally, we discuss strategies to exploit mismatched immunity in order to improve current conventional and experimental influenza A virus vaccines.
Collapse
Affiliation(s)
- Ami Patel
- San Raffaele-Telethon Institute of Gene Therapy (hSR-TIGET), Milan, Italy Division of Gene Therapy & Regenerative Medicine, via Olgettina 58, Milan, Italy, 20132
| | - Gary P Kobinger
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
- Special Pathogens Programme, National Microbiology Laboratory, Public Health Agency of Canada, Canadian Science Centre for Human & Animal Health, 1015 Arlington Street, Winnipeg, Manitoba, Canada, R3E 3R2
| |
Collapse
|
31
|
Miller MS, Tsibane T, Krammer F, Hai R, Rahmat S, Basler CF, Palese P. 1976 and 2009 H1N1 influenza virus vaccines boost anti-hemagglutinin stalk antibodies in humans. J Infect Dis 2012; 207:98-105. [PMID: 23087428 DOI: 10.1093/infdis/jis652] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Infection with pandemic H1N1 influenza A viruses (IAVs) containing hemagglutinin (HA) proteins with globular heads that differ substantially from seasonal strains results in a boost in broadly cross-reactive antibodies that bind to the HA stalk. Boosting these antibodies has become an attractive strategy for creating a universal IAV vaccine. Therefore, it was essential to determine whether vaccines containing H1N1 viruses whose head domains differ substantially compared to seasonal strains could also achieve this boost. METHODS Prospective samples of subjects who had received the A/New Jersey/1976 (NJ/76) vaccine and healthy, age-matched controls were assessed for the presence of anti-HA stalk antibodies before and after receiving the A/California/04/2009 (Cal/09) vaccine between October 2009 and January 2010. RESULTS Individuals who received either the NJ/76 vaccine or the Cal/09 vaccine experienced a robust boost in HA stalk-reactive, neutralizing antibodies similar to what has been observed in individuals infected with Cal/09. CONCLUSIONS These results demonstrate that vaccines containing viruses whose HA head domains that differ substantially from seasonal strains are capable of boosting titers of HA stalk antibodies. Furthermore, anti-HA stalk antibodies elicited by vaccination appear to be long-lived and therefore could be targeted for the generation of a universal IAV vaccine.
Collapse
Affiliation(s)
- Matthew S Miller
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Staneková Z, Adkins I, Kosová M, Janulíková J, Sebo P, Varečková E. Heterosubtypic protection against influenza A induced by adenylate cyclase toxoids delivering conserved HA2 subunit of hemagglutinin. Antiviral Res 2012; 97:24-35. [PMID: 23036818 DOI: 10.1016/j.antiviral.2012.09.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/31/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
The protective efficacy of currently available influenza vaccines is restricted to vaccine strains and their close antigenic variants. A new strategy to obtain cross-protection against influenza is based on conserved antigens of influenza A viruses (IAV), which are able to elicit a protective immune response. Here we describe a vaccination approach involving the conserved stem part of hemagglutinin, the HA2 subunit, shared by different HA subtypes of IAV. To increase its immunogenicity, a novel strategy of antigen delivery to antigen presenting cells (APCs) has been used. The HA2 segment (residues 23-185) was inserted into a genetically detoxified adenylate cyclase toxoid (CyaA-E5) which specifically targets and penetrates CD11b-expressing dendritic cells. The CyaA-E5-HA2 toxoid induced HA2(93-102), HA2(96-104) and HA2(170-178)-specific and Th1 polarized T-cell responses, and also elicited strong broadly cross-reactive HA2-specific antibody response. BALB/c mice immunized with three doses of purified CyaA-E5-HA2 without any adjuvant recovered from influenza infection 2days earlier than the control mock-immunized mice. More importantly, immunized mice were protected against a lethal challenge with 2LD(50) dose of a homologous virus (H3 subtype), as well as against the infection with a heterologous (H7 subtype) influenza A virus. This is the first report on heterosubtypic protection against influenza A infection mediated by an HA2-based vaccine that can induce both humoral and cellular immune responses without the need of adjuvant.
Collapse
Affiliation(s)
- Zuzana Staneková
- Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | | | | | | | | | | |
Collapse
|
33
|
Du L, Li Y, Gao J, Zhou Y, Jiang S. Potential strategies and biosafety protocols used for dual-use research on highly pathogenic influenza viruses. Rev Med Virol 2012; 22:412-9. [PMID: 22987727 DOI: 10.1002/rmv.1729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/29/2012] [Accepted: 07/20/2012] [Indexed: 12/16/2022]
Abstract
Influenza A viruses (IAVs), particularly the highly pathogenic avian influenza H5N1, have posed a substantial threat to public health worldwide. Although the laboratory generation of the mutant influenza virus H5N1 with airborne transmissibility among mammals, which has been considered as a dual-use research, may benefit the development of effective vaccines and therapeutics against the emerging infectious agents, it may also pose threats to national biosecurity, laboratory biosafety, and/or public health. This review introduces the classification and characterization of IAVs, pinpoints historic pandemics and epidemics caused by IAVs, emphasizes the significance and necessity of biosafety, summarizes currently established biosafety-related protocols for IAV research, and provides potential strategies to improve biosafety protocols for dual-use research on the highly pathogenic avian influenza viruses and other emerging infectious agents.
Collapse
Affiliation(s)
- Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | | | | | | | | |
Collapse
|
34
|
Ehrlich HJ, Müller M, Kollaritsch H, Pinl F, Schmitt B, Zeitlinger M, Loew-Baselli A, Kreil TR, Kistner O, Portsmouth D, Fritsch S, Maritsch F, Aichinger G, Pavlova BG, Barrett PN. Pre-vaccination immunity and immune responses to a cell culture-derived whole-virus H1N1 vaccine are similar to a seasonal influenza vaccine. Vaccine 2012; 30:4543-51. [PMID: 22475864 DOI: 10.1016/j.vaccine.2012.03.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 02/22/2012] [Accepted: 03/20/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND Immune responses to novel pandemic influenza vaccines may be influenced by previous exposure to antigenically similar seasonal strains. METHODS An open-label, randomized, phase I/II study was conducted to assess the immunogenicity and safety of a non-adjuvanted, inactivated whole-virus H1N1 A/California/07/2009 vaccine. 408 subjects were stratified by age (18-59 and >60 years) and randomized 1:1 to receive two vaccinations with either 3.75 or 7.5 μg hemagglutinin antigen 21 days apart. Safety, immunogenicity and the influence of seasonal influenza vaccination and antibody cross-reactivity with a seasonal H1N1 strain was assessed. RESULTS A single vaccination with either dose induced substantial increases in H1N1 A/California/07/2009 hemagglutination inhibition (HI) and neutralizing (MN) antibody titers in both adult and elderly subjects. A single 7.5 μg dose induced seroprotection rates of 86.9% in adults and 75.2% in elderly subjects. Two 7.5 μg vaccinations induced seroprotection rates in adult and elderly subjects of 90.9% and 89.1%, respectively. The robust immune response to vaccination was confirmed by analyses of neutralizing antibody titers. Both HI and MN antibodies persisted for ≥ 6 months post-vaccination. Between 34% and 49% of subjects had seroprotective levels of H1N1 A/California/07/2009 antibodies at baseline. Higher baseline HI titers were associated with receipt of the 2008-09 or 2009-10 seasonal influenza vaccine. High baseline A/California/07/2009 neutralizing antibody titers were also associated with high baseline titers against A/New Caledonia/20/99, a seasonal H1N1 strain which circulated and was included in the seasonal vaccine from 2000-01 to 2006-07. Pre-adsorption with A/H1N1/New Caledonia/20/99 antigen reduced A/H1N1/California/07/2009 baseline titers in 55% of tested sera. The vaccine was well tolerated with low rates of fever. CONCLUSIONS A whole-virus H1N1 A/California/07/2009 vaccine was safe and well tolerated and a single dose induced substantial immune responses similar to seasonal influenza vaccines, probably due to immunological priming by previous seasonal influenza vaccines or infections.
Collapse
|
35
|
Fang Y, Banner D, Kelvin AA, Huang SSH, Paige CJ, Corfe SA, Kane KP, Bleackley RC, Rowe T, Leon AJ, Kelvin DJ. Seasonal H1N1 influenza virus infection induces cross-protective pandemic H1N1 virus immunity through a CD8-independent, B cell-dependent mechanism. J Virol 2012; 86:2229-38. [PMID: 22130540 PMCID: PMC3302411 DOI: 10.1128/jvi.05540-11] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/22/2011] [Indexed: 02/05/2023] Open
Abstract
During the 2009 H1N1 influenza virus pandemic (pdmH1N1) outbreak, it was found that most individuals lacked antibodies against the new pdmH1N1 virus, and only the elderly showed anti-hemagglutinin (anti-HA) antibodies that were cross-reactive with the new strains. Different studies have demonstrated that prior contact with the virus can confer protection against strains with some degree of dissimilarity; however, this has not been sufficiently explored within the context of a pdmH1N1 virus infection. In this study, we have found that a first infection with the A/Brisbane/59/2007 virus strain confers heterologous protection in ferrets and mice against a subsequent pdmH1N1 (A/Mexico/4108/2009) virus infection through a cross-reactive but non-neutralizing antibody mechanism. Heterologous immunity is abrogated in B cell-deficient mice but maintained in CD8(-/-) and perforin-1(-/-) mice. We identified cross-reactive antibodies from A/Brisbane/59/2007 sera that recognize non-HA epitopes in pdmH1N1 virus. Passive serum transfer showed that cross-reactive sH1N1-induced antibodies conferred protection in naive recipient mice during pdmH1N1 virus challenge. The presence or absence of anti-HA antibodies, therefore, is not the sole indicator of the effectiveness of protective cross-reactive antibody immunity. Measurement of additional antibody repertoires targeting the non-HA antigens of influenza virus should be taken into consideration in assessing protection and immunization strategies. We propose that preexisting cross-protective non-HA antibody immunity may have had an overall protective effect during the 2009 pdmH1N1 outbreak, thereby reducing disease severity in human infections.
Collapse
Affiliation(s)
- Yuan Fang
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David Banner
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alyson A. Kelvin
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Stephen S. H. Huang
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | - Steven A. Corfe
- Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Kevin P. Kane
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - R. Chris Bleackley
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas Rowe
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alberto J. Leon
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David J. Kelvin
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- University di Sassari, Dipartimento di Scienze Biomediche, Sassari, Italy
| |
Collapse
|
36
|
Athmaram TN, Saraswat S, Santhosh SR, Singh AK, Suryanarayana WS, Priya R, Gopalan N, Parida M, Rao PVL, Vijayaraghavan R. Yeast expressed recombinant Hemagglutinin protein of novel H1N1 elicits neutralising antibodies in rabbits and mice. Virol J 2011; 8:524. [PMID: 22126628 PMCID: PMC3251546 DOI: 10.1186/1743-422x-8-524] [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] [Received: 09/09/2011] [Accepted: 11/29/2011] [Indexed: 11/25/2022] Open
Abstract
Currently available vaccines for the pandemic Influenza A (H1N1) 2009 produced in chicken eggs have serious impediments viz limited availability, risk of allergic reactions and the possible selection of sub-populations differing from the naturally occurring virus, whereas the cell culture derived vaccines are time consuming and may not meet the demands of rapid global vaccination required to combat the present/future pandemic. Hemagglutinin (HA) based subunit vaccine for H1N1 requires the HA protein in glycosylated form, which is impossible with the commonly used bacterial expression platform. Additionally, bacterial derived protein requires extensive purification and refolding steps for vaccine applications. For these reasons an alternative heterologous system for rapid, easy and economical production of Hemagglutinin protein in its glycosylated form is required. The HA gene of novel H1N1 A/California/04/2009 was engineered for expression in Pichia pastoris as a soluble secreted protein. The full length HA- synthetic gene having α-secretory tag was integrated into P. pastoris genome through homologous recombination. The resultant Pichia clones having multiple copy integrants of the transgene expressed full length HA protein in the culture supernatant. The Recombinant yeast derived H1N1 HA protein elicited neutralising antibodies both in mice and rabbits. The sera from immunised animals also exhibited Hemagglutination Inhibition (HI) activity. Considering the safety, reliability and also economic potential of Pichia expression platform, our preliminary data indicates the feasibility of using this system as an alternative for large-scale production of recombinant influenza HA protein in the face of influenza pandemic threat.
Collapse
Affiliation(s)
- T N Athmaram
- Division of Virology, Defence Research and Development Establishment, Ministry of Defence (Govt, of India), Gwalior, MP-474 002, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|