Development of a Malaria Vaccine for Sub-Saharan African Children December 3, 2009 Lode Schuerman

Similar documents
Status of RTS,S/AS01 malaria vaccine candidate

BRIEFING ON RTS,S/AS01 MALARIA VACCINE FOR THE SEPTEMBER 2012 MEETING OF MPAC

RTS,S/AS01 Candidate Malaria vaccine Summary for the SAGE meeting

JTEG s Summary of RTS,S/ AS01 Clinical Trial Data

Enhancing Immunogenicity of Recombinant Vaccines: Chemical Conjugation

Overview of the Malaria Vaccine Implementation Programme (MVIP) Prof. Fred Were SAGE meeting 17 April, 2018

Ad35.CS.01 RTS,S/AS01 prime boost second generation malaria vaccine candidate

RTS,S malaria candidate vaccine reduces malaria by approximately one-third in African infants

New vaccine technologies: Promising advances may save more lives

Progress and Challenges in Malaria Vaccines

Malaria parasite vaccine development Strategies & Targets

Malaria. Dr. Salim Abdulla, Director Ifakara Health Institute, Dar-es-salaam, Tanzania

Fighting Harder and Smarter Against Malaria. Dr.Bernard Nahlen Deputy US Global Malaria Coordinator University of Georgia, February 23, 2010

Malaria Vaccine Pipeline

WHO Consultation on universal access to core malaria interventions in high burden countries: main conclusions and recommendations

Malaria vaccine development

Key Messages for World Malaria Day 2009

ASSESSMENT OF HUMORAL AND CELLULAR IMMUNE RESPONSES OF THE RTS,S/AS02D MALARIA VACCINE CANDIDATE ADMINISTERED TO

Ending Malaria in Nigeria: The WHO Agenda

Summary World Malaria Report 2010

WHO Global Malaria Programme. February 2009

Global landscape analysis and literature review of 2 nd Year of Life immunization platform

Economics of Vaccine Development A Vaccine Manufacturer s Perspective

Summary of the Eighth Meeting of the ITFDE (II) October 4, 2005

Roadmaps as a Vehicle for Addressing Large- Scale Public Health Challenges: Lessons from the Malaria Vaccine Technology Roadmap

Malaria in pregnancy programmes: challenges and priorities in antimalarial drug development for African pregnant women

The RTS,S Clinical Trials Partnership " * Abstract

Counting Malaria Out: Progress of Efforts in the Americas and More Stories of Innovations. Counting Malaria Out in the Americas

Implementing the Abuja Declaration and Plan of Action: the journey so far

APPENDICES BACKGROUND PAPER ON THE RTS,S/AS01 MALARIA VACCINE SEPTEMBER 2015

Malaria: A Global Perspective and Prospects for Elimination. Rima Shretta

Malaria and vaccination: an update. Robert Sauerwein Dept. of Medical Microbiology Radboud University Medical Center, Nijmegen, the Netherlands

PROGRESS REPORT ON CHILD SURVIVAL: A STRATEGY FOR THE AFRICAN REGION. Information Document CONTENTS

INTERVENTION MODEL OF MALARIA

Media centre Malaria. Key facts. Symptoms

Repellent Soap. The Jojoo Mosquito. Africa s innovative solution to Malaria prevention. Sapphire Trading Company Ltd

Long term Efficacy of a Pre-erythrocytic malaria vaccine and correlates of. protection in children residing in a malaria endemic country

Malaria. Population at Risk. Infectious Disease epidemiology BMTRY 713 (Lecture 23) Epidemiology of Malaria. April 6, Selassie AW (DPHS) 1

The Challenge of Malaria

Malaria Funding. Richard W. Steketee MACEPA, PATH. April World Malaria Day 2010, Seattle WA

Resource Allocation for Malaria Prevention. Bahar Yetis Kara

Republic of Kenya MINISTRY OF PUBLIC HEALTH & SANITATION ESSENTIAL. Malaria. Action Guide FOR KENYAN FAMILIES

A Research Agenda for Malaria Eradication: Vaccines

The President s Malaria Initiative (PMI) Indoor Residual Spraying (IRS) in Motion: Malaria Stories from the Field

26/06/ NIMR 2018 Conference - Malaria - a reality

Tanzania s Progress in Combating Malaria: Achievement and Challenges

Kath Maitland Imperial College London & KEMRI / Wellcome Trust Programme, Kilifi, Kenya

PURPOSE The purpose of the Malaria Control Strategic Plan 2005/ /10 is to provide a common platform and detailed description of interventions

Viral Hepatitis in Reproductive Health

Malaria DR. AFNAN YOUNIS

M A L A R I A. The Burden of Malaria: The Impact and Cost of Malaria:

Copyright White et al. Open Access article distributed under the terms of CC BY.

Vaccines on the Global Scale

Epidemiology and control profile of malaria in. Sierra Leone 2017 Supplement

Ph.D. Thesis: Protective immune response in P.falciparum malaria 2011 CHAPTER I: Introduction. S.D. Lourembam 16

Overview of Malaria Epidemiology in Ethiopia

Moderate efficacy malaria vaccines as part of comprehensive malaria control and elimination

Copenhagen, Denmark, September August Malaria

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

Selected vaccine introduction status into routine immunization

Global Consultation on Child and Adolescent Health and Development 13 March 2002 Stockholm, Sweden

From development to delivery: Decision-making for the introduction of a new vaccine

STATUS REPORT ON MALARIA

THE PRESIDENT S MALARIA INITIATIVE

Invest in the future, defeat malaria

Developing a novel Group B Streptococcus (GBS) Vaccine. Patrick Tippoo

abstract n engl j med 374;26 nejm.org June 30,

Malaria Vaccine Implementation Programme Framework for Policy Decision

Analysis of the demand for a malaria vaccine: outcome of a consultative study in eight countries

Aboubacar Kampo Chief of Health UNICEF Nigeria

Malaria. Edwin J. Asturias, MD

GSK VACCINES: KEY GROWTH DRIVERS

Overall presentation of IVR Strategy

A Quarterly Update on HIV Prevention Research. Vol. 8 No. 2

Prompt and Effective Treatment of Malaria through Integrated Services. Dr G.N Ntadom Case Management Branch, NMEP

GAVI S VACCINE INVESTMENT STRATEGY

EXECUTIVE SUMMARY THE PRESIDENT S MALARIA INITIATIVE

Addressing climate change driven health challenges in Africa

Chemoprophylaxis and intermittent treatment for preventing malaria in children (Review)

Maternal, Child and Reproductive Health Initiative

Malaria Elimination: Future Reality?

Task Force on Immunization (TFI) in Africa 14 th Annual Meeting. And. Africa Regional Inter-Agency Coordination Committee (ARICC) 13 th Annual Meeting

How to scale up delivery of malaria control interventions:

Bannie Hulsey MIT Holding, Inc.

LANTOS-HYDE UNITED STATES GOVERNMENT MALARIA STRATEGY

A Descriptive Analysis of Intervention Coverage Scale Up for. Malaria Prevention in Pregnancy and Equity of Services. Rebekah Sherman.

Stakeholders meeting on Malaria

ESCMID Online Lecture Library. by author

Adolescent vaccination strategies

HVTN P5 Vaccine Trials

Updated WHO position paper on pertussis vaccines. Geneva, Switzerland October 2010

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

Evaluation of 4 artemisinin-based combinations for treating uncomplicated malaria in African children Preliminary results

Vaccine Preventable Disease Surveillance: Overview. Thomas Cherian, WHO

DISEASE PREVENTION & ANTIMICROBIAL USE REDUCTION: IMPACT OF VACCINATION

Challenging the Current Malaria Vaccine Pipeline. Dr Odile LEROY Basel 6th December 2016

EDCTP Portfolio Evaluating clinical trials in Africa

The development and introduction of vaccines for the developing world

Efficacy of RTS,S malaria vaccines: individual-participant pooled analysis of phase 2 data

Regulatory Considerations for Determining Vaccine Efficacy U.S. FDA Perspective

Transcription:

Development of a Malaria Vaccine for Sub-Saharan African Children December 3, 2009 Lode Schuerman Copyright John-Michael Maas, Darby Communications

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

Millions of children still die from preventable infectious diseases Malaria 28% Yellow Fever, Diphteria, Polio, Hep B 0% Tetanus 5% Pertussis 7% TB 1% HIV 9% Meningococcus A/C, Japanese Encephalitis 1% Pneumococcus 17% Measles 13% Hib 9% Rotavirus 10% WHO World Health Report 2004

The intolerable burden of malaria 3.3 billion people at risk, 50% of the world s population 250 million malaria cases per year, 86% in Africa 109 endemic countries, 45 within WHO African region 1 million deaths per year, 90% in Africa Plasmodium falciparum is the most severe form Mostly children under 5 years Leading cause of death from a single infectious agent Cost US$12 billion and loss of 1.3% of economic growth annually in Africa

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

Fighting against malaria: tools available today Preventive Insecticide Treated bednets (ITNs) and Long-Lasting Insecticidal Nets (LLINs) Indoor Residual Spraying (IRS) and other Vector Controls Intermittent Preventive Treatment (IPT) in pregnancy (IPTp) in infancy (IPTi) or children (IPTc) Curative Anti-malarial Drug Treatment (ACT, Artemisinin Combinations Therapy) Improved Malaria Case Management (RDTs, Rapid Diagnostic Tests) RBM Partnership web site. Global Malaria Action Plan, GMAP. Available from: <http://www.rbm.who.int/gmap/index.html> [Accessed: 15 June 2009]

Recent trends in malaria incidence Zanzibar (Tanzania) What caused these trends? Usage of RDT? Implementation of LLIN? Treatment by ACT? Improved health care (training)? Rwanda Changes in rainfall pattern and climate (droughts)? Changes in health information systems (in Zanzibar for example all other causes of hospitalization also decreased)? In Rwanda, malaria incidences increased again in 2008-09

The need for a malaria vaccine Important malaria disease burden, but those with the greatest need can least afford current prevention and control measures Challenges to Malaria Control in the SSA setting: Parasite resistance to drugs Mosquito resistance to insecticides HIV co-infection Climate change increasing suitable mosquito habitats Inadequate infrastructure for delivery of control measures Low compliance to protective measures Additional tools (such as a malaria vaccine channeled through EPI) would help to meet public health policy goals and targets WHO IVR 2005, WHO 2008 malaria report, RBM GMAP 2008 A malaria vaccine will be an essential component of future malaria prevention and control measures

The development of a malaria vaccine Challenging Protozoan with a large genome: 14 chromosomes, 5-6000 genes Multistage life cycle with stage specific expression of proteins Allelic and antigenic variation Human immune response is complex and genetically variable but feasible Acquisition of natural immunity against disease in individuals living in endemic regions Protective immunity has been achieved in several malaria animal models (by active immunization as well as passive transfer of monoclonal antibodies and T cells) Passive transfer of protection by purified immunoglobulins obtained from immune adults Active immunization of mice and humans with radiation-treated sporozoites confers sterile immunity

Plasmodium falciparum life cycle Pre-erythrocytic stage Sexual blood stage Asexual blood or erythrocytic stage

Malaria Vaccine Pipeline RTS,S/AS01 Source: WHO 2004

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

Objectives of the RTS,S Malaria Vaccine Candidate Development Program Develop a vaccine that will protect infants and children residing in malaria endemic regions from clinical disease and severe malaria resulting from Plasmodium falciparum infection Safe and well tolerated Compatible with standard EPI vaccines (DTPw, HBV, Hib, OPV ) Implementable through existing delivery programs such as the EPI Complements existing malaria control measures

The RTS,S pre-erythrocytic antigen Circumsporozoite Protein: 9 Major surface protein of the sporozoite 9 Involved in binding of sporozoite to liver cells Sporozoites Liver-stage parasites SS RI Repeat RII Target of neutralizing antibodies GPI T cell epitopes Generation of RTS,S virus-like particles Co-expression of RTS (fusion protein) and HBS protein in Saccharomyces cerevisiae. Spontaneously assemble into mixed virus-like particles (VLP) HBsAg VLP Gordon et al. J Infect Dis 1995 R T S HBsAg + RTS,S VLP S HBsAg

The Adjuvant System Designed to induce strong antibody and Th-1 cell mediated immune responses Immunostimulants: QS21: Saponin extract of Quillaja saponaria MPL: Monophosphoryl Lipid A with: Oil-in-water emulsion (= AS02) or Liposome suspension (= AS01) Clinical development with both adjuvant systems in parallel select the best one for phase III

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

First Proof of Concept (PoC) for efficacy of the RTS,S vaccine against P. falciparum infection Human challenge model at the Walter Read Army Institute of Research Vaccine # Challenged # Infected None 6 6 RTS,S/AS04 8 7 RTS,S/AS03 7 5 RTS,S/AS02 7 1 The most efficacious formulation is the one that consistently induced the best humoral and CMI responses in preclinical testing Stoute JA et al. N. Engl. J. Med. 336(2), 86 91 (1997)

Antibodies against the CS protein of Plasmodium falciparum in vaccinated volunteers Different RTS,S vaccine formulations with different Adjuvant Systems 100 P<0.02 P<0.02 Antibody responses against tandem-repeat epitopes (ELISA with recombinant R32LR) lgg Antibody (μg/ml) 10 1 AS02 AS03 AS04 0.1 0 14 30 44 150 194 220 Day of Study Stoute JA et al. N. Engl. J. Med. 336 (2), 86 91 (1997)

Cell mediated immune responses in vaccinated volunteers IFN γ responses in volunteers immunized with different formulations of RTS,S and association with protective efficacy AS04 AS03 AS02 Number of ELISPOTs/10 6 cells 600 400 200 0 Subjects Protected Not protected Post dose 3 Preimmune Sun et al J of Immunol: 171 (2), 6961-7 (2003)

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

Efficacy of RTS,S/AS against malaria in African children (supported by PATH-MVI) Population (age) 1-4 years 1,2,3 5-17 months 4 10 weeks 5 8 weeks 6 (+EPI) Vaccine Efficacy Clinical malaria Severe malaria Hospitalized malaria All clinical episodes Severe malaria Clinical malaria All clinical episodes Clinical malaria Clinical malaria Efficacy 35% 49% 31% 26% 38% 53% 56% 66% 43% Significance (p-value) <0.001 0.02 0.032 <0.001 0.045 <0.001 <0.001 0.007 0.24 Duration of follow-up 18 months 18 months 18 months 42 months 42 months 8 months 8 months 3 months 6 months Unprecedented and significant reduction of clinical & severe malaria episodes, across different malaria transmission settings Clinical benefit extending over 42 months following vaccination Could have a major impact on burden of malaria 1. Alonso et al, Lancet 2004; 364: 1411-20 2. Alonso et al, Lancet 2005, 366: 2012-18 3. Sacarlal et al JID 2009, 200: 329-36 4. Bejon et al 2008 NEJM 359; 24: 2521-32 5. Aponte et al. Lancet 2007; 370: 1543-51 6. Abdulla et al. NEJM 2008; 359: 2533-44

RTS,S safety and tolerability profile Over 8,000 doses of RTS,S/AS02 or AS01 administered to more than 3,000 children/infants (6 wks to 6 yrs of age) Reactogenicity pattern comparable to control vaccines including routine EPI vaccines Laboratory safety monitoring: no apparent safety signal Favourable assessment of differences in frequency of SAEs (RTS,S vs control) 1. Alonso et al. Lancet 2004; 364: 1411-20 2. Alonso et al. Lancet 2005, 366: 2012-18 3. Sacarlal et al. JID 2009, 200: 329-36 4. Bejon et al 2008 NEJM 359; 24: 2521-32 5. Aponte et al. Lancet 2007; 370: 1543-51 6. Abdulla et al. NEJM 2008; 359: 2533-44 7. Agnandji et al MIM 2009

SAEs following RTS,S/AS01 in infants (N=341, Tanzania, Ghana & Gabon) Any SAE Gastroenteritis Pneumonia Anaemia P. falciparum infection URTI Impetigo RTS,S/AS01(012) + DTPw-HepB/Hib (N = 170) % (95% CI) 22.9 11.2 6.5 3.5 2.9 2.4 1.8 (17 30) (7 17) (3 11) (1 8) (1 7) (1 6) (0 5) DTPw-HepB/Hib (N = 171) % (95% CI) 21.1 (15 28) 8.2 (5 13) 5.8 (3 11) 8.2 (5 13) 9.4 (5 15) 3.5 (1 8) 2.3 (1 6) SAEs occurring within 8 months of follow-up, in at least 2% of subjects in any of the vaccine groups Agnandji et al MIM 2009

Anti-CS immune responses Strong antibody response to the P. falciparum circumsporozoite (CS) repeat domain (anti-cs) in all age groups Anti-CS antibodies consistently associated with protection against infection in the adult challenge model and in field trials with children and infants, but no correlate of protection could be defined. Anti-CS antibody levels wane over time, but remain significantly higher compared to control groups up to 42 months after the last vaccine dose Robust CS-specific CD4 T-cell responses induced in malaria naïve adult volunteers and associated with protection against infection in challenge model (Kester et al. JID 2009)

Summary of Phase 2 findings Unprecedented and consistent efficacy demonstrated in different transmission settings in Kenya, Mozambique and Tanzania Beneficial effect on clinical and severe disease up to 42 months after the last vaccine dose Trend toward higher efficacy against severe forms of disease Trend toward higher efficacy with the AS01 Adjuvant System (Kester et al. JID 2009; Bejon et al NEJM 2008) Favorable safety & reactogenicity profile Trend towards clinical benefit on all cause morbidity and mortality Can be co-administered within routine infant EPI immunizations (compatible in terms of safety, efficacy & immune responses) Induction of CS-specific humoral and cell mediated Immune responses shown to be associated with protection against infection «GO» FOR PHASE 3!

Agenda Malaria Disease burden Prevention and vaccine development RTS,S vaccine Vaccine design Phase I early development Phase II: overview of results Phase III study design Copyright John-Michael Maas, Darby Communications

Phase 3 multi-center efficacy trial 11 centers in 7 African countries Sites represent different malaria transmission settings Up to 16,000 children in 2 age categories: 6 weeks to 12 weeks in EPI co-ad 5 to 17 months Designed in collaboration with scientific community, with feedback of MALVAC*, WHO, FDA, EMEA and African National Regulatory Agencies Number of months of suitable climate No transmission in average year 1-3 months 4-6 months 7-12 months Epidemic or strongly seasonal Endemic and seasonal Endemic and perennial *Moorthy V. et al. Vaccine 2008 http://www.clinicaltrials.gov/ct2/show/nct00866619?term=malaria+efficacy&rank=2

Efficacy Objectives Co-primary objectives: Efficacy against clinical malaria disease over 1 year post dose 3 in: Children aged 5 to 17 months Infants aged 6 weeks at first dose (EPI co-administration) Secondary objectives: Efficacy against severe malaria disease Prevention of malaria hospitalization Prevention of anemia Efficacy against clinical malaria in different transmission settings Duration of efficacy to 2.5 years post dose 3 Requirement for a booster dose Efficacy against fatal malaria and all-cause mortality* Efficacy against other serious illness* All-cause hospitalization, sepsis and pneumonia http://www.clinicaltrials.gov/ct2/show/nct00866619?term=malaria+efficacy&rank=2 *uncertain power

Duration of efficacy Duration of efficacy up to 30 months post primary series Time periods: overall, pre and post booster Assessed as efficacy against first episodes and all episodes Evaluation of requirement for a booster at 18 months post primary Evaluated as efficacy post booster R R R R R R R C C C C C M1 M2 M3 M20 M30 R = RTS,S/AS01 C = Control vaccine http://www.clinicaltrials.gov/ct2/show/nct00866619?term=malaria+efficacy&rank=2

Conclusion from clinical studies to date The RTS,S vaccine is the first malaria vaccine candidate to demonstrate that young children and infants exposed to intense Plasmodium falciparum transmission can be protected from infection and malaria disease. If this vaccine is licensed (after the level of efficacy across various transmission settings has been clarified in the ongoing phase 3 study), it could have a major societal, economic and public health impact in malariaendemic regions in Sub-Saharan Africa. Abdulla et al. NEJM 2008;359:2533-44 Ally Olotu et al. MIM 2009 Alonso et al. Lancet 2004;364:1411-20 Alonso et al. Lancet 2005;366: 2012-8 Aponte et al. Lancet 2007;370:1543-51 Bejon et al 2008 NEJM 359; 24: 2521-32 Sacarlal et al. JID 2009;200:329-36

Clinical development conducted in partnership with PATH-Malaria Vaccine Initiative, GSK and: Institut de Recherche en Science de la Santé, Nanoro, Burkina Faso Kumasi Centre for Collaborative Research & School of Medical Sciences Kumasi, Ghana Kintampo Health Research Centre, Ghana Albert Schweitzer Hospital, Prince GabonLeopold Institute of Tropical Medicine, Belgium KEMRI-Walter Reed Project, University Kenya of Copenhagen, Denmark KEMRI-Wellcome Trust Research University Programme, of Tuebingen, KenyaGermany KEMRI/CDC Research and Public Bernhard Health Nocht Collaboration, Institute, Germany Kenya University of North Carolina University Project, Malawi of Barcelona, Spain Centro de Investigação em Swiss Saude Tropical de Manhiça, Institute, Mozambique Switzerland Ifakara Health Research Development London School Centre, of Hygiene Tanzania& Tropical Medicine, UK National Institute of Medical Center Research, for Disease Tanzania Control and Prevention, USA University of North Carolina at Chapel Hill, USA Walter Reed Army Institute of Research, USA In collaboration with the Malaria Clinical Trials Alliance

Copyright John-Michael Maas, Darby Communications