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

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

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

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

Enhancing Immunogenicity of Recombinant Vaccines: Chemical Conjugation

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

New vaccine technologies: Promising advances may save more lives

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

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

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

JTEG s RTS,S/AS01Candidate Policy Recommendations

The RTS,S Clinical Trials Partnership " * Abstract

Malaria parasite vaccine development Strategies & Targets

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

Malaria Vaccine Implementation Programme (MVIP) update and framework for policy decision. Mary J Hamel, WHO MPAC, 11 April 2018

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

Malaria Vaccine Implementation Programme Framework for Policy Decision

14/10/16. Background. Streptococcus pneumoniae (Sp) is an important cause for these diseases

Progress and Challenges in Malaria Vaccines

Malaria Vaccine Pipeline

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

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

Comparison of the cost effectiveness of LLINs, SMC, the RTS,S vaccine and RTS,S plus IPTi in African settings.

ASSESSING THE VALUE OF VACCINE BEYOND EFFICACY AND SAFETY

IMPORTANCE OF VACCINE PREVENTABLE DISEASE INCIDENCE. Bradford D. Gessner, MD Agence de Médecine Preventive

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

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

HVTN P5 Vaccine Trials

Study No.: Title: Rationale: Phase: Study Period: Study Design: Centers: Indication: Treatment: Objectives: Primary Outcome/Efficacy Variable:

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

Malaria vaccine development

Report to the Board June 2016

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

Modeling the public health impact of malaria vaccines for developers and policymakers

Gavi s Vaccine Investment Strategy

MALARIA VACCINE PILOTS

Cost-effectiveness of Influenza Vaccine

Global Advisory Committee on Vaccine Safety (GACVS) Report from the June 2017 meeting

Economics of Vaccine Development A Vaccine Manufacturer s Perspective

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

COMBINATION VACCINES: HOW AND WHY? LESSONS LEARNED.

IMPACT OF HPV IMMUNIZATION STRATEGIES & POTENTIAL FOR CERVICAL CANCER ELIMINATION

RTS,S/AS01 malaria vaccine efficacy and its interaction with seasonal precipitation:

GRADE tables to assist guideline development and recommendations. Plain Language Summary of Results

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

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

Summary of the Meeting of the Strategic Advisory Group of Experts on immunization, April 2018

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

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

SAGE evidence to recommendations framework i

Vaccination Decision Making: What Providers Need to Know

Study No.: Title: Rationale: Phase: Study Periods: Study Design: Centers: Indication: Treatment: Objectives:

HPV Vaccination: Myths and Misconceptions

Modelling the cost-effectiveness of introducing the RTS,S malaria vaccine relative to scaling up other malaria interventions in sub-saharan Africa

Can we improve the performance of live oral rotavirus vaccines?

Pneumococcal Vaccine Introductions Dr. Carsten Mantel WHO/FCH/IVB/EPI

Journal Club 3/4/2011

Introduction and overview of the program; new vaccine pipeline and prioritization process

Determining the size of a vaccine trial

To demonstrate that DTPa-HBV-IPV/Hib-MenC-TT co-administered with 10Pn, is non-inferior to DTPa-HBV-IPV/Hib coadministered

Global Immunization Vision and Strategies (GIVS) Vaccine Tender , Pretender Meeting, Copenhagen December 2008

Outsourcing in Clinical Trials 1-2 July 2015

New evidence for conquering malaria: Operations, Costs and Cost-Effectiveness: Five Insecticide-Treated Net Programmes

Rotavirus vaccines: Issues not fully addressed in efficacy trials

Overall presentation of IVR Strategy

Report to the Board. Typhoid vaccines: WHO Position paper. Weekly epidemiological record. 2008; 6: ibid

Taking vaccine effectiveness into public health decision making: The ProVac Example

Malaria Control. Umberto D Alessandro

UNIVAC decision support model A universal framework for evaluating vaccine policy options in low- and middle-income countries

GAVI ALLIANCE: UPDATE AND FUTURE DIRECTIONS FOR GLOBAL VACCINES AND IMMUNISATIONS

Schedules and strategies for HPV immunization Conclusions and proposed recommendations for SAGE

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

RTS,S Malaria Vaccine Implementation Programme A joint initiative of GMP & IVB

Protocol Synopsis. Administrative information

Access to vaccination in GAVI countries and at global level

Immunization Update 2017 Peds Clinical Learning Day. Tamara Sheffield, MD, MPA, MPH

EDCTP Portfolio Evaluating clinical trials in Africa

Prophylactic HPV Vaccines. Margaret Stanley Department of Pathology Cambridge

Population-level effectiveness & cost-effectiveness of the 9-valent HPV vaccine

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

Meningitis Outbreak Response intervention thresholds in sub-saharan Africa

Objectives. Immunity. Childhood Immunization Risk of Non-Vaccinated Children 12/22/2015

National Immunization Update

National Immunization Update

SCIENTIFIC DISCUSSION

Development of a Group A meningococcal conjugate vaccine for sub-saharan Africa: clinical trial results

Could a combination of OPV & IPV accelerate wild type poliovirus eradication? Nicholas Grassly

Malaria vaccines where are we now?

Malaria. Edwin J. Asturias, MD

Using a mathematical model to evaluate the impact of different PCV schedules:

Current Status of PCV Use and WHO Recommendations

Value of post-licensure data to assess public health value Example of rotavirus vaccines

Olivier JT Briët 1,2* and Melissa A Penny 1,2

Polio vaccines and polio immunization in the pre-eradication era: WHO position paper. Published in WER 4 June, 2010

VACCINE IMPACT AND VALUE OF VACCINATION LOOKING AT BENEFITS BEYOND EFFICACY AND SAFETY

GSK Medication: Study No.: Title: Rationale: Phase: Study Period Study Design: Centres: Indication: Treatment: Objectives:

Executive Summary SAGE October 2017, Pneumococcal Conjugate Vaccine Session

1) SO1: We would like to suggest that the indicator used to measure vaccine hesitancy be DTP 1 to measles first dose dropout.

Clinical overview of GSK s AS04 adjuvanted vaccine: data up to 6.4 years

Papillomavirus Rapid Interface for Modelling and Economics Tool. User Manual

Transcription:

JTEG s Summary of RTS,S/ AS01 Clinical Trial Data Peter Smith On behalf of the Joint Technical Expert Group (JTEG)

based on Cohen et al 2010 RTS,S/AS01 Malaria Vaccine l Based on large segment of P. falciparum circumsporozoite protein and hepatitis B viral surface protein (as carrier matrix) expressed in yeast cells and includes adjuvant (AS01) l Phase 2 trials shown consistent evidence of partial protection against clinical malaria l Pivotal Phase 3 multicentre trial started in 2009 N Circumsporozoite Protein (CS) Repeat: target of neutralizing antibodies T cell epitopes RTS protein S protein R T S S C + S HepB antigen HepB antigen co- expression of RTS (fusion protein) and HBS protein that assemble into mixed particles (in S. cerevisiae) RTS,S/AS01: 25ug of RTS,S adjuvanted with AS01, which is composed of liposomes and the immunomodulatory molecules 3-Odesacyl-f4-monophosphoryl lipid A (MPL) and QS21

Design of RTS,S/AS01 Phase 3 trial In both 5-17 months and 6-12 weeks at enrolment Study End (SE) (Rabies or Men C) (Men C) Median follow up to SE: 48 months for 5-17 mo 38 months for 6-12 wks

Pivotal Phase III RTS,S/AS01 malaria vaccine efficacy trial Randomized, controlled, double-blind trial conducted in 11 centres in 7 countries Over 15,000 children enrolled Wide range of malaria transmission intensities (0.01 to >2.0 clinical episodes per child per year) Efficacy measured in presence of other malaria control interventions: ITN coverage 86% in 6-12 weeks and 75% in 5-17 months Nanoro, Burkina Faso Kintampo, Ghana Study sites Unstable risk Pf Malaria free Country boundary Water bodies Agogo, Ghana Lambaréné, Gabon Lilongwe, Malawi Manhiça, Mozambique Siaya & Kombewa, Kenya Kilifi, Kenya Korogwe, Tanzania Bagamoyo, Tanzania 4

RTS,S/AS01 Trial Objectives l Co-primary objectives: efficacy over one year post-dose 3 against clinical malaria* when administered in each of the two age groups l Secondary objectives: Vaccine efficacy against severe malaria, anaemia, malaria hospitalization, fatal malaria, all-cause mortality Vaccine efficacy against clinical malaria by transmission setting Vaccine efficacy over time Effect of a fourth dose given at 18 months * primary case definition for clinical malaria was 5,000 parasites/ul with an axillary temperature of >37.5⁰C or a case that met the primary case definition for severe malaria

European Medicines Agency assessment The European Medicines Agency (EMA), under a process known as article 58, performed a scientific evaluation of this vaccine and has now issued what is called "a European scientific opinion". EMA s opinion was positive indicating that in their assessment the quality of the vaccine and the risk/ benefit is favourable from a regulator y perspective.

VACCINE EFFICACY

Vaccine efficacy (95%CI) against all episodes of clinical malaria and severe malaria Study period VE against clinical malaria 6-12 weeks VE against severe malaria VE against clinical malaria 5-17 months VE against severe malaria 2.5M- 14M 32.9% (26.3, 38.9) 38.5% (7.8, 59.0) 51.3% (47.5, 54.9) 44.5% (23.8, 59.6) 2.5M- 20M 26.6% (20.3, 32.4) 17.4% (- 16.2, 41.3) 45.7% (41.7, 49.5) 37.7% (18.0, 52.6) 2.5M- SE* (3 doses) 18.2% (11.4, 24.5) 16.0% (- 14.5, 38.4) 26.2% (20.8, 31.2) - 2.2% (- 31.3, 20.4) 2.5M- SE* (4 doses) 26.7% (20.5, 32.4) 20.5% (- 9.8, 42.5) 39.0% (34.3, 43.3) *median 38 months for 6-12 weeks; 48 months for 5-17 months 31.5% (9.3, 48.3) 8

Vaccine efficacy against clinical malaria stratified by time since entry in the 5-17 month group = Post 4 th dose 4 th dose CONFIDENTIAL; For Internal Use of the GSK- MVI Partnership 9

Vaccine efficacy against clinical malaria stratified by time since entry in the 6-12 week group = Post 4 th dose 4 th dose CONFIDENTIAL; For Internal Use of the GSK- MVI Partnership 10

VE against clinical malaria 5-17 months group, by site M2.5-SE 3-dose schedule M2.5-SE 4-dose schedule

VE against clinical malaria 6-12 weeks group, by site M2.5-SE 3-dose schedule M2.5-SE 4-dose schedule

VE against clinical malaria, 5-17 month group, by MONTH of 1 st dose

Vaccine efficacy against severe malaria by time interval 5-17 months 3- dose schedule 4- dose schedule M2.5- M8 70.1 (49.0, 82.5) M9- M14 20.5 (- 17.8, 46.4) M15-20 14.6 (- 41.0, 48.2) M21-32 M33- SE M2.5- SE - 47.9 (- 134.6, 6.8) - 74.2 (- 220.0, 5.2) - 2.2 (- 31.3, 20.4) - 6.0 (- 75.2, 35.9) - 22.7 (- 137.9, 36.8) 31.5 (9.3, 48.3) 6-12 weeks M2.5- M8 3- dose schedule 53.7 (18.7, 73.6) M9- M14 18.2 (- 43.8, 53.5) M15-20 M21-32 4.7 (- 52.8, 40.6) M33- SE 7.3 (- 113.0, 59.9) M2.5- SE 16.0 (- 14.5, 38.4) - 38.9 (- 143.2, 20.6) 4- dose schedule 37.7 (- 4.8, 63.0) 13.8 (- 91.6, 61.2) 20.5 14 (- 9.8, 42.5)

Incidence of severe malaria in 3-month periods since initial vaccination in children 5-17 months. Controls 3-dose 4-dose

Vaccine efficacy against other outcomes (M2.5- SE) Outcome 5-17 months 6-12 weeks 3- dose 4- dose 3- dose 4- dose Malaria hospitaliza[on 12.1 (- 5.0-26.4) 37.2 (23.6-48.5) 13.2 (- 9.2-31.1) 27.1 (7.1-42.9) Incident Severe anaemia 20.6 (- 32.7-52.9) 61.2 (26.5-80.6) 12.8 (- 50.9-49.9) 31.5 (- 23.1-62.6) All- cause hospitaliza[on 8.8 (- 2.9-19.3) 14.9 (3.6-24.8) 4.8 (- 8.3-16.4) 7.0 (- 6.0-18.4) All- cause mortality - 1.3 (- 79.5-42.8) - 17.8 (- 105-31.9) - 21.5 (- 108-28.5) - 15.6 (- 99.2-32.6) 16

IMMUNOGENICITY

Immune response following initial series and 4th dose 5-17 month group 4 th dose Children = 5-17 months Infants = 6-12 weeks 6-12 wk group 18

Relationship between immune response and VE to M32 (Tertiles of anti-cs antibody titre 1 month post dose 3) 5-17 months group Trend test p=0.6015 6-12 weeks group Trend test p=0.1172 19

SAFETY

Summary of Safety in EMA positive opinion The safety profile of this vaccine is acceptable and quite similar to others apart from a higher risk for febrile convulsions in the older age group within 7 days after a dose (mostly the third dose) of Mosquirix. There also is no safety signal from the supportive studies that might indicate a general problem with the antigens. All identified potential safety issues are addressed in the Risk Management Plan. Ongoing and planned studies will also provide new data for safety and especially following a possible rebound.

Serious Adverse Events: 5-17 months group 5-17 Month Age Group 4- dose schedule N=2976 3- dose schedule N=2972 Controls N=2974 n % n % n % At least one SAE 720 24.2 752 25.3 846 28.4 At least one SAE excluding malaria 673 22.6 704 23.7 784 26.4 Fatal SAE 61 2.0 51 1.7 46 1.5 At least one related SAE Meningi[s (any pathogen) 8 0.3 4 0.1 1 0.0 11 0.4 10 0.3 1 0.0 22

Serious Adverse Events: 6-12 weeks group 6-12 Week Age Group 4- dose schedule N=2976 3- dose schedule N=2178 Controls N=2179 n % n % n % At least one SAE 580 26.6 602 27.6 619 28.4 At least one SAE excluding malaria 562 25.8 582 26.7 591 27.1 Fatal SAE 51 2.3 55 2.5 42 1.9 At least one related SAE Meningi[s (any pathogen) 6 0.3 1 0.0 3 0.1 5 0.2 7 0.3 6 0.3 23

Meningi[s cases in 5-17 month group by study site 6 5 4 3 2 R3R R3C C3C 1 0 24

Meningitis cases in 5-17 month age group Months 0-20 4- dose schedule N=2976 3- dose schedule N=2972 Controls N=2974 MeningiCs 4 5 1 MeningiCs haemophilus 1 0 0 MeningiCs meningococcal 3 1 0 MeningiCs pneumococcal 0 1 0 MeningiCs viral 1 0 0 Meningi[s total 9 7 1 Months 21- SE 4- dose schedule N=2681 3- dose schedule N=2719 Controls N=2702 MeningiCs 1 0 0 MeningiCs haemophilus 0 2 0 MeningiCs meningococcal 0 1 0 MeningiCs tuberculous 1 0 0 Meningi[s total 25

Meningitis case in 5-17 months group by time since doses Post dose 1 Post dose 2 C3C R3C R3R Post dose 3 Post time of dose 4 C3C R3C R3R

Review of CNS infection/inflammation cases classified by experts assessment (both age categories) Ø Experts review 55 cases : 41 cases of meningitis as reported by the investigators and 14 cases of other CNS infection and inflammation 5-17 months 6-12 weeks A`er 3- doses R3R R3C C3C A`er 3- doses R3R R3C C3C Confirmed meningi[s 5 4 0 Confirmed meningi[s 3 4 1 No meningics 4 2 2 No meningics 2 2 3 Undetermined 4 2 3 Undetermined 2 0 0 A`er 4- doses R3R R3C C3C A`er 4- doses R3R R3C C3C Confirmed meningi[s 1 2 0 Confirmed meningi[s 0 1 2 No meningics 1 2 0 No meningics 0 0 1 Undetermined 1 0 0 Undetermined 0 1 0 Unassessed 0 0 1 CONFIDENTIAL 27

Ini[al series Febrile seizures in 7 days following vaccination : 5-17 months group n RTS,S N=17306 doses Rate per 1000 doses (95%CI) Control N=8728 doses n Rate per 1000 doses (95%CI) Febrile seizures within 7 days 18 1.04 (0.62, 1.64) 5 0.57 (0.19, 1.34) A`er [me of 4 th dose Febrile seizures within 7 days 4- dose schedule N=2447 doses n Rate per 1000 doses (95%CI) 3- dose schedule N=2472 doses n Rate per 1000 doses (95%CI) Control N=2473 doses n Rate per 1000 doses (95%CI) 6 2.5 (0.9, 5.3) 3 1.2 (0.3, 3.5) 1 0.4 (0.0, 2.3) 28

Febrile seizures in 7 days following vaccination : 6-12 weeks group Ini[al series n RTS,S N=12739 doses Rate per 1000 doses (95%CI) n Control N=6403 doses Rate per 1000 doses (95%CI) Febrile seizures within 7 days 2 0.16 (0.02, 0.57) 3 0.47 (0.10, 1.37) A`er [me of 4 th dose Febrile seizures within 7 days 4- dose schedule N=1825 n Rate per 1000 doses (95%CI) 3- dose schedule N=1837 n Rate per 1000 doses (95%CI) n Control N=1827 Rate per 1000 doses (95%CI) 4 2.2 (0.6, 5.6) 0 - (0, 2) 1 0.5 (0.0, 3.0) 29

Severe malaria in the 5-17 month age group Time Period M0- M20 Time period M21- SE Malaria Ini[al series N=5948 Control N=2974 Syndrome N Died N Died All Cases 205 6 158 2 Cerebral 16 3 5 1 Cerebral + 6 1 1 0 Anaemia Anaemia 25 0 29 1 Other 157 2 123 0 Missing 1 0 0 0 Malaria Syndrome 3- dose schedule N=2719 4- dose schedule N=2681 Control N=2702 N Died N Died N Died All Cases 103 6 76 3 76 2 Cerebral 9 4 11 2 2 0 Cerebral + 0 0 1 0 2 1 Anaemia Anaemia 18 1 11 0 17 0 Other 75 1 53 1 54 1

Cerebral malaria cases in the 5-17 month age category by site Site No. of subjects Cases of cerebral malaria Siaya 799 9 Kintampo 1002 14 Nanoro 600 8 Agogo 600 8 Manhica 1002 3 Lambarene 704 3 Kombewa 1000 4 Lilongwe 800 2 Bagamoyo 903 2 Korogwe 912 0 Kilifi 600 0 Total 8922 53 31

Severe malaria in the 6-12 week age group 32

VACCINE IMPACT

Cases of clinical malaria averted per 1000 vaccinees Up to Study month 5-17 months age group 3- dose schedule 21 721 (591, 847) 4- dose schedule 6-12 weeks age group 3- dose schedule 296 (179, 413) 4- dose schedule 33 855 (653, 1053) 1097 (894, 1295) 336 (103, 558) 583 (374, 798) SE 860 (534, 1166) 1239 (908, 1552) 368 (73, 638) 665 (407, 922) 34

Cases of clinical malaria averted (to SE) per 1000 vaccinees at each site 5-17 months 6-12 weeks 3-dose 4-dose 35

Summary of mathematical model comparison process l In 2010, WHO initiated an extensive comparison exercise of four mathematical models (Imperial College, Swiss Tropical and Public Health Institute, Intellectual Ventures and GlaxoSmithKline) l Developed to estimate the public health impact and costeffectiveness of RTS,S/AS01 in a range of scenarios. l Extensive comparison of intermediate outputs of epidemiological models without vaccination. This enabled better inter-group understanding of baseline models l Efficacy against infection fitted to reproduce Phase 3 trial clinical malaria efficacy by site

Outcomes of model comparisons l In all models, vaccination is predicted to lead to an age shift in malaria incidence. In high prevalence settings, and for more severe disease endpoints, this age shift is predicted to occur sooner after vaccine introduction than in low prevalence settings. This predicted effect is general to any preventive malaria intervention (including bed-nets and SMC). l The models that were designed to include indirect effects of vaccination predict little herd protection from RTS,S/AS01 vaccination when introduction is limited to this 5-9 month age group, given the role of infected individuals of all ages in contributing to transmission.

Conclusions of model comparisons l All models predict an overall beneficial impact of the vaccine on mortality but with a small excess of deaths in older age groups (a feature of malaria preventive interventions, not the vaccine, also predicted for SMC) l Consensus range is 10% to 28% reduction for under 5 malariarelated deaths among fully vaccinated children (with a 3 or 4 dose schedule) l Contrary to the results seen in the trial for severe malaria, the models were consistent for predicting an overall beneficial effect on severe malaria and mortality even in the absence of a 4th dose l Sensitivity analysis: Key drivers of cost-effectiveness are transmission intensity and vaccine price

Cost-effectiveness by prevalence level (assuming $5/dose) (Incremental cost-effectiveness ratio (ICER) in USD 2013) Parasite prevalence in 2-10 yr olds Cost per DALY averted decreases with transmission intensity, with plateau at prevalence 10%-65%, where cost per DALY averted is <$100, assuming $5/dose

Conclusions of comparative cost effectiveness work (Imperial College model only) RTS,S/AS01, under 3- and 4-dose schedule, compared to LLINs and SMC were examined across a range of transmission settings in Africa. In all settings LLINs are the most cost-effective initial intervention, followed by SMC in areas where recommended, and then RTS,S/AS01 RTS,S/AS01 remains cost-effective as a second/third intervention at levels of parasite prevalence > 10% (consistent with the wider modelling exercise)

Conclusions of comparative cost effectiveness work (Imperial College model only) The most cost-effective prevention package is to scale-up LLINs to between 60% and 75% usage before introducing the vaccine Data from 2011-2013 show rates below this in many countries (though additional progress made between 2013 and 2015) ITN usage in children <5y, based on most recent DHS/MIS surveys (2011-2013)

Indica[ve es[mates of cost- effec[veness of other vaccines Vaccine Rotavirus PCV7 HPV RTS,S 4- dose schedule Price $1.50-7.50 per dose $5 per dose $25 per course $5 per dose Cost per DALY averted $42 ($31- $64) $100 $400 ($200- $500) $60- $100 Care should be taken when making inter- vaccine comparisons as the cost- effec+veness of each vaccine is evaluated using different models and hence is based on different modelling assump+ons.

RISK-BENEFIT ASSESSMENT

Risk/benefit assessment over median 48 months follow- up in those aged 5-17 months BENEFITS RISKS UNCERTAINTIES 4- dose schedule VE clinical malaria: 39.0% (95%CI 34.3, 43.3) VE severe malaria: 31.5% (95%CI 9.3, 48.3) VE malaria hospitaliza[on: 37.2% (95%CI 23.6, 48.5) VE all- cause hospitaliza[on: 14.9% (95%CI 3.6, 24.8) VE incident severe anaemia: 61.2% (95%CI 26.5, 80.6) IdenCfied Risk Excess of febrile convulsion a\er any of the first three doses (0.5/1000 doses within 7 days of vaccinacon) a\er fourth dose (2.0/1000 doses within 7 days of vaccinacon) PotenCal Risk Meningi[s (numerical excess, no clear associacon with Cme since vaccinacon, biological model not well established, excess predominantly in only 2 of 11 sites) Uncertain overall proteccon against severe malaria beyond trial period Beneficial overall effect on severe malaria is dependent on delivery of fourth dose Relevance of imbalance of cerebral malaria cases, possibly due to chance 44

Risk/benefit assessment over median 38 months follow- up in those aged 6-12 weeks BENEFITS IDENTIFIED RISKS UNCERTAINTIES 4- dose schedule VE clinical malaria: 26.7% (95%CI 20.5, 32.4) VE malaria hospitaliza[on: 27.1% (95%CI 7.1, 42.9) Excess of febrile convulsion a\er fourth dose (1.6/1000 doses within 7 days of vaccinacon) NA 45

End of presentation 46