Immunogenicity and Efficacy of the COVID-19 Candidate Vector Vaccine MVA-SARS-2-S in Preclinical Vaccination

Total Page:16

File Type:pdf, Size:1020Kb

Immunogenicity and Efficacy of the COVID-19 Candidate Vector Vaccine MVA-SARS-2-S in Preclinical Vaccination Immunogenicity and efficacy of the COVID-19 candidate vector vaccine MVA-SARS-2-S in preclinical vaccination Alina Tschernea,b,1, Jan Hendrik Schwarza,1, Cornelius Rohdec,d,1, Alexandra Kupkec,d,1, Georgia Kalodimoua,b,1, Leonard Limpinsela, Nisreen M. A. Okbae, Berislav Bošnjakf, Inga Sandrockf, Ivan Odakf, Sandro Halwec,d, Lucie Sauerheringc,d, Katrin Brosinskia, Nan Lianglianga, Elke Duella,b, Sylvia Janya, Astrid Freudensteina, Jörg Schmidtc,d, Anke Wernerc,d, Michelle Gellhorn Serrac,d, Michael Klüverc,d, Wolfgang Guggemosg, Michael Seilmaierg, Clemens-Martin Wendtnerg, Reinhold Försterf,h,i, Bart L. Haagmanse, Stephan Beckerc,d, Gerd Suttera,b,2, and Asisa Volza,b,j aDivision of Virology, Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany; bDivision of Virology, Department of Veterinary Sciences, German Center for Infection Research, 80539 Munich, Germany; cInstitute of Virology, Philipps University Marburg, 35037 Marburg, Germany; dInstitute of Virology, Marburg, German Center for Infection Research, 35392 Giessen-Marburg-Langen, Germany; eDepartment of Viroscience, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; fInstitute of Immunology, Hannover Medical School, 30625 Hannover, Germany; gMunich Clinic Schwabing, Academic Teaching Hospital, Ludwig-Maximilians-Universität München, 80804 Munich, Germany; hInstitute of Immunology, German Center for Infection Research, 30625 Hannover, Germany; iCluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany; and jInstitute of Virology, University of Veterinary Medicine, Hannover, 30559 Hannover, Germany Edited by Peter Palese, Icahn School of Medicine at Mount Sinai, New York, NY, and approved May 23, 2021 (received for review December 20, 2020) Severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2) for SARS-CoV-2 cell entry. As a class I viral fusion protein, it has emerged as the infectious agent causing the pandemic coronavi- mediates virus interaction with the cellular receptor angiotensin- rus disease 2019 (COVID-19) with dramatic consequences for global converting enzyme 2 (ACE2), and fusion with the host cell mem- human health and economics. Previously, we reached clinical evalua- brane, both key steps in infection. Thus, infection can be prevented tion with our vector vaccine based on modified vaccinia virus Ankara by S-specific antibodies neutralizing the virus (6–9). (MVA) against the Middle East respiratory syndrome coronavirus Among the front-runner vaccines are new technologies such as MICROBIOLOGY (MERS-CoV), which causes an infection in humans similar to SARS messenger RNA (mRNA)-based vaccines and nonreplicating adeno- and COVID-19. Here, we describe the construction and preclinical char- virusvectorvaccines(10–13). First reports from these SARS-CoV-2- acterization of a recombinant MVA expressing full-length SARS-CoV-2 S–specific vaccines in phase 1/2 clinical studies demonstrated spike (S) protein (MVA-SARS-2-S). Genetic stability and growth char- acceptable safety and promising immunogenicity profiles, and by acteristics of MVA-SARS-2-S, plus its robust expression of S protein as now data from large phase 3 clinical trials show promising levels antigen, make it a suitable candidate vaccine for industrial-scale pro- of protective efficacy (4, 12–14). In December 2020, the first duction. Vaccinated mice produced S-specific CD8+ T cells and serum antibodies binding to S protein that neutralized SARS-CoV-2. Prime- Significance boost vaccination with MVA-SARS-2-S protected mice sensitized with a human ACE2-expressing adenovirus from SARS-CoV-2 infection. The highly attenuated vaccinia virus MVA is licensed as small- MVA-SARS-2-S is currently being investigated in a phase I clinical trial pox vaccine; as a vector it is a component of the approved as aspirant for developing a safe and efficacious vaccine against adenovirus-MVA–based prime-boost vaccine against Ebola vi- COVID-19. rus disease. Here, we provide results from testing the COVID-19 candidate vaccine MVA-SARS-2-S, a poxvirus-based vector vac- vaccine vector | vaccinia virus | poxvirus | nonclinical testing cine that proceeded to clinical evaluation. When administered by intramuscular inoculation, MVA-SARS-2-S expresses and safely evere acute respiratory syndrome coronavirus 2 (SARS-CoV-2), delivers the full-length SARS-CoV-2 S protein, inducing balanced Sthe causal agent of coronavirus disease 2019 (COVID-19), first SARS-CoV-2–specific cellular and humoral immunity, and protec- emerged in late 2019 in China (1). SARS-CoV-2 exhibits extremely tive efficacy in vaccinated mice. Substantial clinical experience efficient human-to-human transmission, the new pathogen rapidly has been gained with MVA vectors using homologous and het- spread worldwide, and within months it caused a global pan- erologous prime-boost applications, including the immunization demic, changing daily life for billions of people. The COVID-19 of children and immunocompromised individuals. Thus, MVA- case fatality rate of ∼2–5% makes the development of counter- SARS-2-S represents an important resource for developing fur- measures a global priority. In fact, the development of COVID- ther optimized COVID-19 vaccines. 19 vaccine candidates is advancing at an international level with unprecedented speed. About 1 y after the first known cases of Author contributions: G.S. and A.V. designed research; A.T., J.H.S., C.R., G.K., L.L., N.L., S.J., COVID-19, we can account for >80 SARS-CoV-2–specific vaccines A.F., G.S., and A.V. performed research; N.M.O., B.B., I.S., I.O., S.H., L.S., J.S., A.W., M.G.S., > M.K., W.G., M.S., C.-M.W., R.F., and B.L.H. contributed new reagents/analytic tools; A.T., in clinical evaluations and 10 candidate vaccines already in phase J.H.S., C.R., A.K., G.K., N.M.A.O., B.B., I.O., K.B., E.D., R.F., B.L.H., S.B., G.S., and A.V. ana- III trials (2–4). However, we still lack information on the key im- lyzed data; and A.T., J.H.S., C.R., A.K., S.B., G.S., and A.V. wrote the paper. mune mechanisms needed for protection against COVID-19. A The authors declare no competing interest. better understanding of the types of immune response elicited upon This article is a PNAS Direct Submission. natural SARS-CoV-2 infections has become an essential compo- This open access article is distributed under Creative Commons Attribution License 4.0 nent to assess the promise of various vaccination strategies (5). (CC BY). The SARS-CoV-2 spike (S) protein serves as the most impor- 1A.T., J.H.S., C.R., A.K., and G.K. contributed equally to this work. tant target antigen for vaccine development based on preclinical 2To whom correspondence may be addressed. Email: [email protected]. research on candidate vaccines against SARS-CoV or Middle East This article contains supporting information online at https://www.pnas.org/lookup/suppl/ respiratory syndrome coronavirus (MERS-CoV). The trimeric S doi:10.1073/pnas.2026207118/-/DCSupplemental. protein is a prominent structure at the virion surface and essential Published June 23, 2021. PNAS 2021 Vol. 118 No. 28 e2026207118 https://doi.org/10.1073/pnas.2026207118 | 1of9 Downloaded by guest on September 30, 2021 mRNA-based COVID-19 vaccines received emergency use au- antibody directed against the 9-aa HA-tag at the C terminus of thorization or conditional licensing by the US Food and Drug the recombinant SARS-2-S protein revealed highly specific staining in Administration and European Medicines Agency (11, 15, 16). By permeabilized cells corresponding to the expected intracellular local- March 2021, two adenovirus vector-based COVID-10 vaccines ization of the S-protein C-terminal end. A SARS-CoV-1-S–specific had been approved by regulatory authorities (17, 18). This is good monoclonal antibody showing cross-reactivity with SARS-CoV-2 (29) news because efficacious vaccines will provide a strategy to change in recognizing an epitope in the external domain of the SARS-CoV-2- SARS-CoV-2 transmission dynamics. In addition, multiple vac- S protein also allowed the specific staining of nonpermeabilized cine types will be advantageous to meet specific demands across MVA-SARS-2-S–infected Vero cells, suggesting that the SARS- different target populations. This includes the possibility of using 2-S protein was readily translocated to the plasma membrane heterologous immunization strategies depending on an individ- (Fig. 2A). ual’s health status, boosting capacities, and the need for balanced To examine the MVA-produced recombinant S protein in more humoral and Th1-directed cellular immune responses. detail, we prepared total lysates from MVA-SARS-2-S–infected MVA, a highly attenuated strain of vaccinia virus originating chicken embryonic fibroblasts (CEFs) or Vero cells for separation from growth selection on chicken embryo tissue cultures, shows a by SDS-PAGE and subsequent immunoblot analysis (Fig. 2). The characteristic replication defect in mammalian cells but allows mouse monoclonal antibody directed against the HA-tag at the C unimpaired production of heterologous proteins (19). At present, terminus of the recombinant SARS-2-S protein revealed two prom- MVA serves as an advanced vaccine technology platform for de- inent protein bands that migrated with molecular masses of ∼190 and veloping new vector vaccines against infectious disease including 90–100 kDa (Fig. 2B). As in the SDS-PAGE the detected protein emerging viruses and cancer (20). In response to the ongoing pan- bands migrated at molecular masses significantly higher than the demic, the MVA vector vaccine platform allows rapid generation of 145 kDa predicted for full-length SARS-CoV-2-S protein based experimental SARS-CoV-2–specific vaccines (21). Previous work on its amino acid sequence, we hypothesized that the proteins from our laboratory addressed the development of an MVA can- might be glycosylated.
Recommended publications
  • The Long Road to a Universal Influenza Virus Vaccine †
    Abstract The Long Road to a Universal Influenza Virus Vaccine † Peter Palese Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; [email protected] † Viruses 2020 - Novel Concepts in Virology, Barcelona, Spain, 5–7 February 2020. Published: 8 July 2020 Abstract: Seasonal and pandemic influenza virus infections can cause significant disease worldwide. Current vaccines only provide limited, short-lived protection, and antigenic drift/shift in the hemagglutinin (HA) surface glycoprotein necessitates their annual reformulation and re-administration. To overcome these limitations, universal influenza virus vaccine strategies aim at eliciting broadly protective antibodies to conserved epitopes of the HA. We have developed two approaches. (1) The first is based on “chimeric” HA constructs that retain the conserved stalk domain of the HA and have exotic HA heads. Vaccination and boosting with such constructs successfully redirects the immune system in animals and in humans towards the conserved immune sub-dominant domains of the HA stalks; this results in an antigenic silencing of the HA heads and a protective immune response facilitated by the conserved HA stalks. In mice and ferrets, such a strategy protects the animals against homo-subtypic and hetero-subtypic challenge with influenza A strains as well as against influenza B variants. It is hoped that vaccine constructs expressing three components (i.e., conserved group 1 HA stalks, conserved group 2 HA stalks, and conserved influenza B HA stalks) will be protective against all future seasonal and pandemic strains. (2) The “mosaic” HA approach is based on antigenic silencing of the major immunodominant antigenic sites of the HA heads by only replacing those epitopes with corresponding sequences of exotic avian HAs, yielding “mosaic” HAs.
    [Show full text]
  • Toward a Universal Influenza Virus (Revised)
    TOWARDS A UNIVERSAL INFLUENZA VIRUS VACCINE Peter Palese Icahn School of Medicine at Mount Sinai New York OPTIONS IX 8-26-16 ISIRV - Options IX for the Control of Influenza Peter Palese, PhD Professor and Chair Department of Microbiology Icahn School of Medicine, New York Mount Sinai has submitted patent applications for a universal influenza virus vaccine Work has been supported by the NIH, The Bill & Melinda Gates Foundation, GSK My presentation does not include discussion of off-label or investigational use. Similar variation for influenza, HIV and HCV F. Krammer EIGHTEEN SUBTYPES OF INFLUENZA A VIRUS HEMAGGLUTININS Group 2 Influenza A Influenza B Group 1 bat HAs Influenza viruses circulating in the human population B pH1N1 H3N2 (Group2) A ? H2N2 (Group1) H1N1 (Group1) 1918 1940 1960 1980 2000 AVIAN INFLUENZA VIRUSES INFECTING HUMANS H5N6 China 2016 H7N9 China 2015, 2014, 2013 H10N8 China 2013 H6N1 Taiwan 2013 H10N7 Australia,Egypt 2010,2004 H7N3 Mexico,UK,Canada,Italy 2012,2006,04,03 H7N2 UK,USA 2007,2003 H9N2 Hong Kong 1999 H5N1 Asia,Europe,Africa, Hong Kong 2015-2003 , 1997 H7N7 Netherlands,UK,USA,Austr.,USA 2003,96,80,77,59 INFLUENZA VIRUS VACCINES INACTIVATED LIFE ATTENUATED RECOMBINANT INFLUENZA VIRUS VACCINE STRAINS 2016-2017 A/California/7/2009 (H1N1)pdm09 A/Hong Kong/4801/2014 (H3N2) B/Phuket/3073/2013 B/Brisbane/60/2008 • INFLUENZA VIRUS VACCINES ARE UNIQUE. • THEY HAVE TO BE GIVEN ANNUALLY, BECAUSE NOVEL VACCINE FORMULATIONS HAVE TO BE PREPARED REFLECTING THE RAPID ANTIGENIC CHANGE OF THE VIRUS. Antigenic diversity: analysis
    [Show full text]
  • Sequence of the 1918 Pandemic Influenza Virus Nonstructural Gene (NS) Segment and Characterization of Recombinant Viruses Bearing the 1918 NS Genes
    Sequence of the 1918 pandemic influenza virus nonstructural gene (NS) segment and characterization of recombinant viruses bearing the 1918 NS genes Christopher F. Basler*†, Ann H. Reid*‡, Jody K. Dybing*§¶, Thomas A. Janczewski‡, Thomas G. Fanning‡, Hongyong Zheng†, Mirella Salvatore†, Michael L. Perdue§ʈ**, David E. Swayne§, Adolfo Garcı´a-Sastre†ʈ, Peter Palese†ʈ, and Jeffery K. Taubenberger‡ʈ †Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029; ‡Division of Molecular Pathology, Department of Cellular Pathology, Armed Forces Institute of Pathology, Washington, DC 20306; and §Southeast Poultry Research Laboratory, United States Department of Agriculture, Athens, GA 30605 Contributed by Peter Palese, December 5, 2000 The influenza A virus pandemic of 1918–1919 resulted in an generation of influenza viruses from cloned cDNAs allow re- estimated 20–40 million deaths worldwide. The hemagglutinin combinant influenza viruses bearing genes of the 1918 pandemic and neuraminidase sequences of the 1918 virus were previously virus to be constructed. The resulting viruses can be studied in determined. We here report the sequence of the A͞Brevig Mission͞ vitro or in animal models (12, 13). 1͞18 (H1N1) virus nonstructural (NS) segment encoding two pro- The full-length sequences of the two predominant surface teins, NS1 and nuclear export protein. Phylogenetically, these proteins of the 1918 virus, hemagglutinin (HA) and neuramin- genes appear to be close to the common ancestor of subsequent idase (NA), have recently been described (2, 9). These sequences human and classical swine strain NS genes. Recently, the influenza were determined first because influenza pandemics are thought A virus NS1 protein was shown to be a type I IFN antagonist that to result from the emergence of influenza virus strains with novel plays an important role in viral pathogenesis.
    [Show full text]
  • Plant-Based COVID-19 Vaccines: Current Status, Design, and Development Strategies of Candidate Vaccines
    Review Plant-Based COVID-19 Vaccines: Current Status, Design, and Development Strategies of Candidate Vaccines Puna Maya Maharjan 1 and Sunghwa Choe 2,3,* 1 G+FLAS Life Sciences, 123 Uiryodanji-gil, Osong-eup, Heungdeok-gu, Cheongju-si 28161, Korea; punamaya.maharjan@gflas.com 2 G+FLAS Life Sciences, 38 Nakseongdae-ro, Gwanak-gu, Seoul 08790, Korea 3 School of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Korea * Correspondence: [email protected] Abstract: The prevalence of the coronavirus disease 2019 (COVID-19) pandemic in its second year has led to massive global human and economic losses. The high transmission rate and the emergence of diverse SARS-CoV-2 variants demand rapid and effective approaches to preventing the spread, diagnosing on time, and treating affected people. Several COVID-19 vaccines are being developed using different production systems, including plants, which promises the production of cheap, safe, stable, and effective vaccines. The potential of a plant-based system for rapid production at a commercial scale and for a quick response to an infectious disease outbreak has been demonstrated by the marketing of carrot-cell-produced taliglucerase alfa (Elelyso) for Gaucher disease and tobacco- produced monoclonal antibodies (ZMapp) for the 2014 Ebola outbreak. Currently, two plant-based COVID-19 vaccine candidates, coronavirus virus-like particle (CoVLP) and Kentucky Bioprocessing (KBP)-201, are in clinical trials, and many more are in the preclinical stage. Interim phase 2 clinical Citation: Maharjan, P.M.; Choe, S. trial results have revealed the high safety and efficacy of the CoVLP vaccine, with 10 times more Plant-Based COVID-19 Vaccines: neutralizing antibody responses compared to those present in a convalescent patient’s plasma.
    [Show full text]
  • BANCOVID, the First D614G Variant Mrna-Based Vaccine Candidate Against SARS
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.29.319061; this version posted September 30, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1 BANCOVID, the first D614G variant mRNA-based vaccine candidate against SARS- 2 CoV-2 elicits neutralizing antibody and balanced cellular immune response 3 Juwel Chandra Baray, Md. Maksudur Rahman Khan, Asif Mahmud, Md. Jikrul Islam, Sanat 4 Myti, Md. Rostum Ali, Md. Enamul Haq Sarker, Samir Kumar, Md. Mobarak Hossain 5 Chowdhury, Rony Roy, Faqrul Islam, Uttam Barman, Habiba Khan, Sourav Chakraborty, Md. 6 Manik Hossain, Md. Mashfiqur Rahman Chowdhury, Polash Ghosh, Mohammad Mohiuddin, 7 Naznin Sultana*, Kakon Nag* 8 Globe Biotech Ltd., 3/Ka, Tejgaon I/A, Dhaka – 1208, Bangladesh, 9 *, to whom correspondence should be made. 10 E-mail: [email protected], [email protected] 11 12 13 Key words: COVID, Coronavirus, Lipid nanoparticle, LNP, Vaccination, Immunization, 14 15 16 Abstract 17 Effective vaccine against SARS-CoV-2 is the utmost importance in the current world. More 18 than 1 million deaths are accounted for relevant pandemic disease COVID-19. Recent data 19 showed that D614G genotype of the virus is highly infectious and responsible for almost all 20 infection for 2nd wave. Despite of multiple vaccine development initiatives, there are currently 21 no report that has addressed this critical variant D614G as vaccine candidate. Here we report 22 the development of an mRNA-LNP vaccine considering the D614G variant and 23 characterization of the vaccine in preclinical trial.
    [Show full text]
  • 1. Sars-Cov Nucleocapsid Protein Epitopes and Uses Thereof
    www.engineeringvillage.com Citation results: 500 Downloaded: 4/24/2020 1. SARS-COV NUCLEOCAPSID PROTEIN EPITOPES AND USES THEREOF KELVIN, David; PERSAD, Desmond; CAMERON, Cheryl; BRAY, Kurtis, R.; LOFARO, Lori, R.; JOHNSON, Camille; SEKALY, Rafick-Pierre; YOUNES, Souheil-Antoine; CHONG, Pele Assignee: UNIVERSITY HEALTH NETWORK; BECKMAN COULTER, INC.; UNIVERSITE DE MONTREAL; NATIONAL HEALTH RESEARCH INSTITUTES Publication Number: WO2005103259 Publication date: 11/03/2005 Kind: Patent Application Publication Database: WO Patents Compilation and indexing terms, 2020 LexisNexis Univentio B.V. Data Provider: Engineering Village 2. SARS-CoV-specific B-cell epitope and applications thereof Wu, Han-Chung; Liu, I-Ju; Chiu, Chien-Yu Assignee: National Taiwan University Publication Number: US20060062804 Publication date: 03/23/2006 Kind: Utility Patent Application Database: US Patents Compilation and indexing terms, 2020 LexisNexis Univentio B.V. Data Provider: Engineering Village 3. A RECOMBINANT SARS-COV VACCINE COMPRISING ATTENUATED VACCINIA VIRUS CARRIERS QIN, Chuan; WEI, Qiang; GAO, Hong; TU, Xinming; CHEN, Zhiwei; ZHANG, Linqi; HO, David, D. Assignee: INSTITUTE OF LABORATORY ANIMAL SCIENCE OF CHINESE ACADEMY OF MEDICAL SCIENCES; THE AARON DIAMOND AIDS RESEARCH CENTER; QIN, Chuan; WEI, Qiang; GAO, Hong; TU, Xinming; CHEN, Zhiwei; ZHANG, Linqi; HO, David, D. Publication Number: WO2006079290 Publication date: 08/03/2006 Kind: Patent Application Publication Database: WO Patents Compilation and indexing terms, 2020 LexisNexis Univentio B.V. Data
    [Show full text]
  • Mrna-Based Vaccines to Elicit CD8+ T Cell Immunity
    mRNA-based Vaccines to Elicit CD8+ T Cell Immunity Ans De Beuckelaer Thesis submitted in partial fulfilment of the requirements for the degree of Doctor in Sciences: Biochemistry and Biotechnology 2016 Promoter Prof. Dr. Johan Grooten Co-promoter Dr. Stefaan De Koker Laboratory of Molecular Immunology Department of Biomedical Molecular Biology Faculty of Sciences, Ghent University The research described in this doctoral thesis was performed at the Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University. Ans De Beuckelaer was supported by a personal fellowship of the IWT (Innovation by Science and Technology). No part of this thesis may be reproduced or published without prior permission of the author. © Ans De Beuckelaer 2016 mRNA- based Vaccines to Elicit CD8+ T cell Immunity Ans De Beuckelaer1 Academic year: 2015- 2016 Promoter: Prof. Dr. Johan Grooten1 Co-Promoter: Dr. Stefaan De Koker2 Examination committee Chair Prof. Dr. Rudi Beyaert1,3 Secretary Prof. Dr. Xavier Saelens1,4 Voting members Prof. Dr. Vanham5,6 Prof. Dr. Karine Breckpot7 Prof. Dr. Katrien Remaut8 1 Department of Biomedical Molecular Research, Ghent University, Ghent, Belgium 2 eTheRNA Immunotherapeutics NV, Niel, Belgium 3 Inflammation Research Center, VIB, UGhent, Ghent, Belgium 4 Medical Biotechnology, VIB, UGhent, Ghent, Belgium 5 Department of Biomedical Sciences, University of Antwerp, Anwterp, Belgium 6 Department of Biomedical Sciendes, Virology Unit, Institute of Tropical Medicine, Antwerp, Belgium 7 Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Univ. Brussel, Brussels, Belgium 8 Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharaceutical Sciences, UGhent, Ghent, Belgium TABLE OF CONTENT List of Abbreviations 8 Summary 13 Samenvatting 17 Introduction 21 CHAPTER 1.
    [Show full text]
  • Antigenic Sites in Influenza H1 Hemagglutinin Display Species- Specific Immunodominance
    Antigenic sites in influenza H1 hemagglutinin display species- specific immunodominance Sean T.H. Liu, … , Florian Krammer, Peter Palese J Clin Invest. 2018. https://doi.org/10.1172/JCI122895. Concise Communication In-Press Preview Immunology Virology Hemagglutination inhibition (HI) titers are a major correlate of protection for influenza-related illness. The influenza virus hemagglutinin possesses antigenic sites that are the targets of HI active antibodies. Here, a panel of mutant viruses each lacking a classically defined antigenic site was created to compare the species-specific immunodominance of the antigenic sites in a clinically relevant hemagglutinin. HI active antibodies of antisera from influenza-virus infected mice targeted sites Sb and Ca2. HI active antibodies of guinea pigs were not directed against any specific antigenic site, although trends were observed towards Sb, Ca2, and Sa. HI titers of antisera from infected ferrets were significantly affected by site Sa. HI active antibodies of adult humans followed yet another immunodominance pattern, where sites Sb and Sa were immunodominant. When comparing the HI profiles between different species by antigenic cartography, animals and humans grouped separately. This study provides characterizations of the antibody-mediated immune responses against the head domain of a recent H1 hemagglutinin in animals and humans. Find the latest version: https://jci.me/122895/pdf 1 Title 2 Antigenic sites in influenza H1 hemagglutinin display species-specific immunodominance 3 4 Sean T. H. Liu1,2, Mohammad Amin Behzadi1, †, Weina Sun1, †, Alec Freyn1, Wen-Chun Liu1, Felix Broecker1, 5 Randy A. Albrecht1,3, Nicole M. Bouvier1,2, Viviana Simon1,3, Raffael Nachbagauer1, Florian Krammer1, and 6 Peter Palese1 7 1Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
    [Show full text]
  • Adolfo García-Sastre Professor, Department of Microbiology
    Adolfo García-Sastre Professor, Department of Microbiology Fishberg Professor, Department of Medicine, Division of Infectious Diseases Director, Global Health and Emerging Pathogens Institute Icahn School of Medicine at Mount Sinai 1468 Madison Avenue, New York, NY 10029 [email protected]; Short Bio; Dr. Adolfo García-Sastre is Professor in the Departments of Microbiology and Medicine and Director of the Global Health and Emerging Pathogens Institute of Icahn School of Medicine at Mount Sinai in New York. He received his BSc and PhD from University of Salamanca, Spain and postdoctoral training at the Laboratory of Peter Palese at Mount Sinai School of medicine in New York City. Area of Research; For the past 30 years, his research interest has been focused on the molecular biology of influenza viruses and several other negative strand RNA viruses. During his post-doctoral training in the early 1990s, he developed, for the first time, novel strategies for expression of foreign antigens by a negative strand RNA virus, influenza virus. He has made major contributions to the influenza virus field, including 1) the development of reverse genetics techniques allowing the generation of recombinant influenza viruses from plasmid DNA, (studies in collaboration with Dr. Palese); 2) the generation and evaluation of negative strand RNA virus vectors as potential vaccine candidates against different infectious diseases, including malaria and AIDS, and 3) the identification of the biological role of the non structural protein NS1 of influenza virus during infection: the inhibition of the type I interferon (IFN) system. His studies provided the first description and molecular analysis of a viral- encoded IFN antagonist among negative strand RNA viruses.
    [Show full text]
  • Dynavax and Mount Sinai Announce Collaboration to Develop a Universal Influenza Vaccine Candidate with Cpg 1018 Adjuvant
    Dynavax and Mount Sinai Announce Collaboration to Develop a Universal Influenza Vaccine Candidate with CpG 1018 Adjuvant July 16, 2020 Development funded under contract award from NIAID/NIH as part of the CIVICs program Currently no approved universal influenza vaccine CDC estimates 35.5 million people were infected with influenza in U.S. during 2018-2019 season Collaboration will combine Dynavax’s CpG 1018 with influenza antigens designed to protect against all strains of influenza NEW YORK and EMERYVILLE, Calif., July 16, 2020 (GLOBE NEWSWIRE) -- Dynavax Technologies Corporation (NASDAQ: DVAX), a biopharmaceutical company focused on developing and commercializing novel vaccines, and the Icahn School of Medicine at Mount Sinai (“Mount Sinai”) today announced they have entered into a collaboration to develop a universal influenza (flu) vaccine. Mount Sinai’s current work in this area is funded under a contract award from the National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (NIH), as part of the Collaborative Influenza Vaccine Innovation Centers (CIVICs) program established by NIAID. The Mount Sinai CIVICs team will evaluate a novel approach they have developed called chimeric hemagglutinin (cHA) designed to protect against all strains of influenza in combination with Dynavax’s CpG 1018TM adjuvant. The development program will support an Investigational New Drug (IND) application for Phase I clinical trials. Drs. Peter Palese, PhD, Professor and Chair of the Department of Microbiology at Mount Sinai, Adolfo-Garcia-Sastre, PhD, Director of the Global Health and Emerging Pathogens Institute, and the Irene and Dr. Arthur M. Fishberg Professor of Microbiology and Medicine (Infectious Diseases) at Mount Sinai, and Florian Krammer, PhD, Professor of Microbiology at Mount Sinai will be leading the development of the program.
    [Show full text]
  • Mcmx: a Proposal for a Federal Authority to Enhance Speed, Scale and Access to Medical Countermeasures
    MCMx: A Proposal for a Federal Authority to Enhance Speed, Scale and Access to Medical Countermeasures Findings from the Program in Global Public Policy and Social Change Medical Countermeasures Taskforce: Partnering for Public Good Authors Kendall Hoyt, Geisel School of Medicine, Dartmouth College Margaret Bourdeaux, Harvard Medical School Annmarie Sasdi, Harvard Medical School May 28, 2021 PROGRAM IN GLOBAL PUBLIC POLICY AND SOCIAL CHANGE TASK FORCE MEMBERS Katrine Thor Andersen Deputy Director, Global Health, William and Melinda Gates Foundation Thomas J. Bollyky Director, Global Health Program, Council on Foreign Relations Rick Bright, PhD Senior Vice President, Pandemic Prevention & Response, Health Initiative, The Rockefeller Foundation Philip Dormitzer, MD, PhD Vice President and Chief Scientific Officer Viral Vaccines, Pfizer Rebecca Fish President, Hart Ledge Consulting Bruce Gellin, MD, MPH President, Global Immunization, Sabin Vaccine Institute Arpa (Shah) Garay President, Global Pharmaceuticals, Commercial Analytics, Digital Marketing, Merck & Co. Jonathan P. Gertler, MD Managing Partner and CEO Of Back Bay Life Science Advisors and Managing Director, Bioventures Investors Medtech Funds Robert V. House, PhD, FATS Senior Vice President, Government Contracts, Ology Bioservices Kendall Hoyt, PhD Assistant Professor, Geisel School of Medicine, Dartmouth College Heather Ignatius Director, U.S. and Global Policy and Advocacy, PATH Ryan Morhard Ginkgo Bioworks Jake Reder, PhD CEO, Celdara Medical James Robinson Vice Chair, The Coalition for Epidemic Preparedness Innovations Julia Barnes-Weise, J.D., CFP Executive Director, The Global Healthcare Innovation Alliance Accelerator (GHIAA) Charlie Weller Head of Vaccines, Wellcome Trust 2 PROGRAM IN GLOBAL PUBLIC POLICY AND SOCIAL CHANGE CONTENTS I. Introduction II. MCMx Mission III. MCMx Strategic Plan: Creating a Research and Development Blueprint for MCMs in Future Outbreaks A.
    [Show full text]
  • Making Better Influenza Virus Vaccines? Peter Palese*
    Making Better Influenza Virus Vaccines? Peter Palese* Killed and live influenza virus vaccines are effective in 361/2002 viruses. Changes in the HA of circulating virus- preventing and curbing the spread of disease, but new es (antigenic drift) require periodic replacement of the vac- technologies such as reverse genetics could be used to cine strains during interpandemic periods. The World improve them and to shorten the lengthy process of prepar- Health Organization publishes semiannual recommenda- ing vaccine seed viruses. By taking advantage of these tions for the strains to be included for the Northern and new technologies, we could develop live vaccines that would be safe, cross-protective against variant strains, and Southern Hemispheres (2). To allow sufficient time for require less virus per dose than conventional vaccines. manufacture, in the United States the US Food and Drug Furthermore, pandemic vaccines against highly virulent Administration (FDA) determines in February which vac- strains such as the H5N1 virus can only be generated by cine strains should be included in the following winter’s reverse genetics techniques. Other technologic break- vaccine. Unfortunately, FDA recommendations are not throughs should result in effective adjuvants for use with always optimal. For example, in 2003 FDA rejected the killed and live vaccines, increasing the number of available use of the most appropriate H3N2 strain, A/Fujian/411/ doses. Finally, universal influenza virus vaccines seem to 2002, and instead again used the same strain as in the 2002 be within reach. These new strategies will be successful if formulation. This decision was made primarily because the they are supported by regulatory agencies and if a robust market for influenza virus vaccines against interpandemic A/Fujian/411/2002 strain had first been isolated in Madin and pandemic threats is made and sustained.
    [Show full text]