Active and Passive Immunization for Avian Influenza Virus Infections

Similar documents
H5N1 and H7 LAIV-IAV Prime-Boost Studies

Application of Reverse Genetics to Influenza Vaccine Development

Studying Repeated Immunization in an Animal Model. Kanta Subbarao Laboratory of Infectious Diseases, NIAID

A Live Attenuated H7N7 Candidate Vaccine Virus Induces Neutralizing Antibody That Confers Protection from Challenge in Mice, Ferrets, and Monkeys

Live Attenuated Influenza Vaccine. I. Background and Seasonal Vaccine

Min Levine, Ph. D. Influenza Division US Centers for Disease Control and Prevention. June 18, 2015 NIBSC

Clinical Trials of Pandemic Vaccines: Key Issues. John Treanor University of Rochester Rochester, NY

7/14/2014 VACCINE-INDUCED ANTI-HA2 ANTIBODIES PROMOTE VIRUS FUSION AND ENHANCE INFLUENZA VIRUS RESPIRATORY DISEASE (VAERD)

Development of live attenuated pediatric RSV vaccines

Cristina Cassetti, Ph.D.

Strategies for control of influenza by targeting broadly conserved viral features

(the change introduced is to add a risk assessment, missing from the previous version, for small-scale laboratory work with characterized CVV)

WHO biosafety risk assessment and guidelines for the production and quality control of human influenza pandemic vaccines: Update

Immunogenicity of Avian Influenza H7N9 Virus in Birds

24 26 January 2013, Hong Kong SAR, CHINA. TITLE from VIEW and SLIDE MASTER February 27, 2013

GSK s Adjuvanted Influenza Vaccines The Taming of the Flu

Influenza or flu is a

Broadly protective influenza vaccines for pandemic preparedness. Suresh Mittal Department of Comparative Pathobiology Purdue University

Avian influenza and pandemic threats

Nanoparticulate Vaccine Design: The VesiVax System

100 years of Influenza Pandemic and the prospects for new influenza vaccines

Public health relevant virological features of Influenza A(H7N9) causing human infection in China

Hemagglutinin-stalk specific antibodies: How to induce them and how to measure them

Update of WHO biosafety risk assessment and guidelines for the production and quality control of human influenza pandemic vaccines

Update on influenza monitoring and vaccine development

Gene Vaccine Dr. Sina Soleimani

Cover Page. The handle holds various files of this Leiden University dissertation

Supporting Information

Development of safe and immunogenic reassortant viruses with 5:3 genotype for live attenuated influenza vaccine

Biotechnology-Based Vaccines. Dr. Aws Alshamsan Department of Pharmaceutics Office: AA87 Tel:

PATH Influenza Vaccine Projects

VIROLOGY OF INFLUENZA. Subtypes: A - Causes outbreak B - Causes outbreaks C - Does not cause outbreaks

Innate-adaptive immunity duo as a regimen for conferring rapid-sustained-broad protection against pathogens

Heat-killed Lactobacillus casei

Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine

Pandemic Influenza influenza epidemic: realization of a worst-case scenario

Prophylactic and Therapeutic Efficacy of Human Monoclonal Antibodies against H5N1 Influenza

REVIEW Cell-mediated Immunity to Influenza Virus Infections: From the Perspective to the Vaccine Development against Highly Pathogenic Avian Influenza

Influenza: Ecology and Continuing Evolution

C E E Z A D. Rational Development of Influenza Vaccines: NDV-based influenza vaccines for poultry and livestock

Acute respiratory illness This is a disease that typically affects the airways in the nose and throat (the upper respiratory tract).

UNIVERSAL INFLUENZA VIRUS VACCINES Adolfo García Sastre. Icahn School of Medicine at Mount Sinai, New York

Influenza: Seasonal, Avian, and Otherwise

Patricia Fitzgerald-Bocarsly

The humoral immune responses to IBV proteins.

Vaccine 30 (2012) Contents lists available at SciVerse ScienceDirect. Vaccine. jou rn al h om epa ge:

Influenza: The past, the present, the (future) pandemic

Incorporating virologic data into seasonal and pandemic influenza vaccines

Current Strategies in HIV-1 Vaccine Development Using Replication-Defective Adenovirus as a Case Study

7/14/2014. Multiple immune effector mechanisms contribute to protection influenza. What is a correlate of protection?

1918 Influenza; Influenza A, H1N1. Basic agent information. Section I- Infectious Agent. Section II- Dissemination

Introduction to Avian Influenza

An Exploration to Determine if Fab Molecules are Efficacious in Neutralizing Influenza H1 and H3 Subtypes. Nick Poulton June September 2012

Nature Immunology: doi: /ni Supplementary Figure 1. Infection strategy.

Hemagglutinin (HA) and neuraminidase (NA), the major envelope

Quality, safety and standards for poliomyelitis vaccines

Meta Analysis: Blood Products for Spanish Influenza Pneumonia: A Future H5N1 Treatment?

The Impact of Pandemic Influenza on Public Health

Evaluation of Three Live Attenuated H2 Pandemic Influenza Vaccine Candidates in Mice and Ferrets

Risk Assessment of H3N2 Avian Origin Canine Influenza Viruses

Reverse genetic platform for inactivated and live-attenuated influenza vaccine

Combinatorial Vaccines for AIDS and other Infectious Diseases

The influenza A viruses of swine in Japan, Thailand, and Vietnam

Introduction. In the past 15 years, several technological advancements have open new perspectives and applications in the field of vaccinology.

Zheng, BJ; Du, LY; Zhao, GY; Lin, YP; Sui, HY; Chan, C; Ma, S; Guan, Y; Yuen, KY. Citation Hong Kong Medical Journal, 2008, v. 14 suppl. 4, p.

Supplemental Information Dose Response Parameters for Gain of Function Pathogens

Possible Modes of Transmission of Avian Viruses to People: Studies in Experimental Models

Potential Role of Exposure to Poultry Products and By-products for Human H5N1 infections

NASDAQ:NVAX Novavax, Inc. All rights reserved.

Global Pandemic Preparedness Research Efforts. Klaus Stöhr. WHO Global Influenza Programme. Today

Technology Overview. Summary

Original Article Development and Sequence Analysis of a Cold-Adapted Strain of Influenza A/New Caledonia/20/1999(H1N1) Virus

H5N1 Vaccine-Specific B Cell Responses in Ferrets Primed with Live Attenuated Seasonal Influenza Vaccines

Russian-backbone LAIV history and clinical development. L.Rudenko Institute of Experimental Medicine, St. Petersburg, Russia

ORTHOMYXOVIRUSES INFLUENZA VIRUSES. (A,B and C)

TOWARDS A UNIVERSAL INFLUENZA VIRUS VACCINE

Evaluation of Live Attenuated Influenza A Virus H6 Vaccines in Mice and Ferrets

Synthetic Genomics and Its Application to Viral Infectious Diseases. Timothy Stockwell (JCVI) David Wentworth (JCVI)

An avian live attenuated master backbone for potential use in epidemic and pandemic influenza vaccines

Viral vaccines. Lec. 3 أ.د.فائزة عبد هللا مخلص

SCIENTIFIC DISCUSSION

Pandemic Influenza Preparedness

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) POINTS TO CONSIDER ON THE DEVELOPMENT OF LIVE ATTENUATED INFLUENZA VACCINES

Review on vectored influenza vaccines. Sarah Gilbert Jenner Institute Oxford

INFLUENZA-2 Avian Influenza

Agricultural Outlook Forum Presented: February 16, 2006 THE CURRENT STATE OF SCIENCE ON AVIAN INFLUENZA

Ralf Wagner Paul-Ehrlich-Institut

For the control of avian influenza

Zoonotic potential of non-avian influenza A viruses

A Mouse Model for the Evaluation of Pathogenesis and Immunity to Influenza A (H5N1) Viruses Isolated from Humans

Universal Influenza Vaccine Development

OFFLU swine influenza virus meeting March 2017 FAO Headquarters, Rome, Italy. Amy Vincent USDA-ARS National Animal Disease Center

The A(H7N9) influenza outbreak in China

Regulatory Challenges to Production of Veterinary Influenza Vaccines in Human Facilities

Vaccine. Design and Manufacturing. Liting Bi.

Challenges and Solutions for the Next Generation of Vaccines: Jonathan Liu Luis Maranga Sachin Mani Richard Schwartz

Laboratory Diagnosis of Avian Influenza and Newcastle Disease

Experiences with Live Attenuated Avian Influenza Vaccine Trials in Thailand

Evolution of influenza

What is the role of animal models in studying protective titres and the need for establishing surrogates/correlates of protection?

Transcription:

NIAID Active and Passive Immunization for Avian Influenza Virus Infections Kanta Subbarao, MD, MPH Laboratory of Infectious Diseases NIAID, NIH

Immortalizing H5 HA-Specific Memory B Cells Collection of PBMC from 4 Vietnamese adults who recovered from H5N1 influenza infection >14 months after onset of illness Memory B cells were seeded 10-50 cells/well in a 96 U-bottom plate in the presence of CpG (polyclonal activator of memory B cells), EBV, and irradiated mononuclear cells IgG+ memory B cells were isolated by binding to CD22 microbeads, followed by depletion of IgM+, IgD+ and IgA+ cells by FACS Screening of culture supernatants for H5 HA-specific antibodies Cloning of positive cultures by limiting dilution Antibody purification by affinity chromatography

Cross-neutralizing activity of H5 hmabs against H5N1 viruses NEUTRALIZING AB TITER AGAINST ANTIBODIES H3N2 HK97 HK03 VN04 JPH05 INA05 A146 mab <10 <10 <10 <10 <10 <10 Sheep anti-sera <10 2032 2560 806 1613 806 FLA5.10 <10 127 4064 508 806 <10 FLA3.14 <10 403 508 226 508 508 FLD20.19 <10 905 5120 1613 6451 5120 FLD21.14 <10 32 >14882 5120 12902 <10 An undetectable titer is assigned a value of <10 Simmons PLoS Medicine 2007

Efficacy of Immunoprophylaxis: Survival

Immunoprophylaxis: Virus titers Mean virus titer (log 10 TCID 50 /g) *p<0.01 vs D2.2 ** p<0.001 vs D2.2 Lower limit of detection D2.2 FLA3.14 FLA5.10 D2.2 FLA3.14 FLA5.10 D 2 post-infection D 4 post-infection

Histopathology in mice challenged with H5N1 virus following immunoprophylaxis Irrelevant MAb D2.2 H5 MAb FLA5.10 MAb D2.2 H5 MAb FLA5.10 MAb D2.2 X100 H5 Mab FLA5.10 X100

Histopathological analysis of the lung Ab received Mouse # Normal bronchioles Bronchioles with Lesions Bronchiolar Necrosis D2.2 1 4/11 (36%) 7/11 (64%) 7/11 (64%) 2 3/12 (25%) 9/12 (75%) 9/12 (75%) FLA3.14 1 6/10 (60%) 4/10 (40%) 4/10 (40%) 2 13/13 (100%) 0/13 (0%) 0/13 (0%) FLA5.10 1 9/10 (90%) 1/10 (10%) 0/10 (0%) 2 8/12 (67%) 4/12 (33%) 4/12 (33%)

Efficacy of H5 Mabs administered 24, 48 and 72 h after infection with 5LD 50 VN/04

Summary of Studies on H5 HA MAbs The technology can be used to generate human MAbs against emerging pathogens In addition to the 4 MAbs described in detail, 7 additional MAbs have been selected The MAbs against the H5 HA had neutralizing activity in vitro and in vivo The MAbs were efficacious in prophylaxis in mice The MAbs were efficacious in therapy in mice against homologous and heterologous viruses, even when administered 72 h after virus infection

Implications and Future Studies A cocktail of MAbs could be used for post-exposure prophylaxis or as an adjunct to antiviral therapy Mapping the epitopes to which the MAbs bind Extend preclinical data Proceed with clinical development

Options for Vaccines for Pandemic Influenza Principle: Induction of a protective immune response against the hemagglutinin protein. Inactivated vaccine: a reassortant virus containing HA and NA from avian virus (or an antigenically similar Surrogate virus) and internal genes from A/PR/8/34 Live attenuated cold-adapted (ca) vaccine: a reassortant prepared with master ca strain A/Ann Arbor/6/60 used in FluMist Purified expressed HA protein HA expressed in a vectored vaccine e.g. adenovirus DNA vaccine encoding the HA and?other genes from the avian virus

Development of Vaccines against Avian Influenza Viruses with Pandemic Potential Correlates of protection are known Antibodies directed against the HA and?na are critical for protection Systemic immune response is strain specific Mucosal immune response provides broader cross protection Cellular immunity for viral clearance Human influenza vaccine experience suggests that the vaccine strain must closely match the circulating strain However, it is not possible to predict which avian influenza strain will cross the species barrier The significance of antigenic drift among avian influenza viruses is not known Avian HAs are poorly immunogenic in humans H5 and H7 HAs may bear a multibasic cleavage site that makes them lethal for poultry and chicken embryos

Challenges in the Development of Pandemic Influenza Vaccines Diversity of avian influenza viruses Different subtypes: H1-H16, N1-N9 HPAI vs LPAI Eurasian vs North American lineages among many HA subtypes Antigenic drift caused by natural infection or vaccine use Animal models: mice, ferrets, others? Evaluation of candidate vaccines (safety and immunogenicity are evaluated but efficacy is not) Assays for immunogenicity are not sensitive and not standardized and correlates of protection are not known Avian HAs appear to be poorly immunogenic; high doses or multiple doses of HA or adjuvant are needed Others Biosafety restrictions Lack of reagents for quality control Use of reverse genetics Subbarao & Joseph Nature Rev Imm 2007

Goal of a Pandemic Influenza Vaccine To prevent severe illness and death from pandemic influenza and it s complications. An ideal influenza vaccine will induce a rapid systemic and mucosal immune response directed at the HA, NA and conserved internal proteins of the virus protect against a broad range of influenza viruses, within a subtype and across subtypes In practical terms, our goal is to generate a vaccine that rapidly induces a cross-reactive neutralizing antibody response

Pandemic Influenza Vaccines Generated by Genetic Reassortment Vaccine donor virus with phenotype of attenuation or high growth in eggs PB2 PB1 PA HA NP NA M NS PB2 PB1 PA HA NP NA M NS Avian influenza virus PB2 PB1 PA HA NP NA M NS Reassortant vaccine virus with phenotype of attenuation or high growth in eggs

Live Attenuated Pandemic Influenza Vaccines ts Subtype (# evaluated) ca att Immunogenicity Efficacy Mice Ferrets Mice Ferrets H5 (3) H6 (3) H7 (1) H9 (1)

Attenuation In chickens: if HA derived from HPAI In mice: lethality (if relevant) virus replication in respiratory tract In ferrets: virus replication in the respiratory tract

The H5N1 ca Reassortant Viruses are not Highly Pathogenic for Chickens Virus # inoculated # died Mean time to death 1997, 2003 and 2004 H5N1 wt 8 8 1-2 days 1997 H5N1 ca 8 0-2003 H5N1 ca 8 0-2004 H5N1 ca 8 0-4-week-old SPF White Plymouth Rock chickens were inoculated intravenously with a 1:10 dilution of stock virus (10 8-8.75 /ml) and observed for 10 days.

The 2004 H5N1 ca vaccine candidate is attenuated for mice and does not spread to the brain 9 7 5 9 8 7 6 5 Lungs Nasal turbinates Brain Mean virus titer in log 10 TCID 50 /g 3 1 7 5 3 1 5 3 4 3 2 1 0 7 6 5 4 3 2 1 0 6 5 4 3 2 2 4 6 2 4 6 A/VN/2004 (H5N1) wt A/VN/2004 (H5N1) ca H1N1 Beijing/95 ca H3N2 Panama/99 ca 1 1 0 2 4 4 6 6-12 Days following virus administration Suguitan PLoS Medicine 2006

Immunogenicity In mice and in ferrets Following one dose or two doses Tested against homologous and heterologous viruses

Two doses of H5N1 ca vaccines are required to elicit serum neutralizing Ab titers in mice Immunizing virus Doses Geometric mean serum neutralizing Ab titers against indicated virus 1997 wt 2003 wt 2004 wt A/VN/2004 ca 1 10 10 10 2 160 528 388 A/HK/2003 ca 1 10 37 10 2 19 1056 61 Sera were collected before (prebleed) and 28 days following each dose of vaccine; an undetectable titer is assigned a value of 10

ELISA and Nt Ab response to 1 or 2 doses of the 2004 H5N1 ca vaccine # of doses of vaccine s Nt Ab titer vs homologous wt H5N1 virus ELISA Ab titer vs recombinant H5 HA 1 10 40,637 2 69 325,100 Two doses of 10 6 TCID 50 of H5N1 2004 ca virus was administered i.n. 4 weeks apart Sera were collected before (prebleed) and 28 days following each dose of vaccine; an undetectable titer was assigned a value of 10; prevaccination titers were 10

Efficacy In mice and in ferrets Following one dose or two doses Tested against homologous and heterologous viruses Efficacy against lethal challenge Protection from pulmonary replication or systemic spread of challenge virus

A Single Dose of H5N1 ca Vaccine Protects Mice from Lethal Challenge with 50, 500 or 5000 LD 50 of Homologous and Heterologous Wild-type H5N1 Viruses H5N1 1997 wt challenge H5N1 2004 wt challenge 100 90 1997 ca 2003 ca 2004 ca 100 90 1997 ca 2003 ca 2004 ca 80 80 Percent survival 70 60 50 40 30 20 70 60 50 40 30 20 10 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 Mock immunized 10 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 Mock immunized Days following administration of challenge virus Suguitan PLoS Medicine 2006

Complete Protection from Pulmonary Replication of wt H5N1 Challenge Viruses is Conferred by 2 doses of the 2004 H5N1 ca Vaccine 9 9.0 Single dose 7 8.0 7.0 5 6.0 5.0 Immunization: Two doses Mean virus titer (log 10 TCID 50 /g) 3 1 9 7 5 3 4.0 3.0 2.0 1.0 0.0 9 8 7 6 5 4 3 Mock H5N1 VN 2004/AA ca H5N1 2003 wt 2 1 1 0 1997 wt 2003 wt Challenge Virus 2004 wt Vaccine dose: 10 6 TCID 50 per dose 2004 H5N1 ca; Challenge virus dose: 10 5 TCID 50 of wt virus

2 Doses of H5N1 ca Vaccines Provide Complete Protection from Pulmonary Replication of Homologous and Heterologous wt H5N1 Viruses 8 Mean virus titer in lungs (log10 TCID50/g) 7 6 5 4 3 2 1 Mock H5N1 97 ca H5N1 2003 ca H5N1 2004 ca 0 1 2 3 4 HK/491/97 HK/213/2003 VN/1203/2004 Indo/5/2005 H5N1 wt Challenge: Homologous Virus Heterologous Virus Suguitan PLoS Medicine 2006

Goals of the Pandemic Influenza Vaccine Program Generate and evaluate a library of vaccines against viruses of each subtype (H2, H4-16) to protect humans against pandemic influenza Proceed to clinical trials to evaluate safety, infectivity and immunogenicity in healthy adults Bank sera from vaccinated volunteers to test against avian viruses that emerge in humans Determine the significance of antigenic differences among avian influenza viruses in humans Program: CRADA with MedImmune Vaccines, collaboration with CIR, Johns Hopkins Univ. Approach: Live attenuated vaccines Evaluation: In inpatients

Acknowledgements Laura, Amorsolo, Josie, Tomy, Catherine, Celia, and Kim (and Mike) Jack Vaccines: LID: Brian Murphy SEPRL, USDA: David Swayne and Joan Beck MedImmune: George Kemble, Hong Jin, Bin Lu CIR, JHU: Ruth Karron Comparative Medicine Branch, NIAID, NIH MAbs: Antonio Lanzavecchia and Nadia Bernasconi, IRB, Switzerland Cameron Simmons, Oxford Unit, Ho Chi Minh City, VietNam Jerrold Ward, Comparative Medicine Branch, NIAID, NIH Hana Golding and Surender Khurana, CBER, FDA