RTSS-SMC: Seasonal Malaria Vaccination (RTS,S/AS01) and Seasonal Malaria Chemoprevention (SP/AQ)

Sponsor
London School of Hygiene and Tropical Medicine (Other)
Overall Status
Completed
CT.gov ID
NCT03143218
Collaborator
Malaria Research and Training Center, Bamako, Mali (Other), Institut de Recherche en Sciences de la Sante, Burkina Faso (Other)
5,920
2
3
35.4
2960
83.5

Study Details

Study Description

Brief Summary

A double-blind, individual randomised trial will be undertaken in 6000 children under the age of five years living in areas of Burkina Faso or Mali where the transmission of malaria is intense and highly seasonal to determine whether the malaria vaccine RTS,S/AS01 is (a) as effective as SMC with SP + AQ in preventing clinical malaria (b) provides additional, useful protection when given together with SMC. The primary trial end-point will be the incidence of clinical episodes of malaria detected by passive case detection.

Condition or Disease Intervention/Treatment Phase
  • Biological: RABIPUR®
  • Biological: RTS,S/AS01
  • Drug: SMC with SP+AQ
  • Drug: SMC placebo
Phase 3

Detailed Description

The RTS,S/AS01 malaria vaccine is a recombinant protein vaccine in which the fusion protein RTS (containing parts of the circumsporozoite protein (CSP) of Plasmodium falciparum fused to hepatitis B surface antigen (HBsAg)) is co-expressed in yeast together with free HBsAg (S) to form a virus like particle (RTS,S); it is given with the powerful adjuvant AS01. RTS,S/AS01 induces a strong antibody response to the P. falciparum CSP and high titres of anti-CSP antibody are associated with protection. Following a long process of development, a phase 3 study of RTS,S/AS01 conducted in 15,439 children in 7 countries in Africa showed that three doses of RTS,S/AS01 given with a one month interval between doses, followed by a fourth dose 18 months post dose 3, gave 36.5 % [95% CI 31,41%] protection against clinical attacks of malaria when given to young children aged 5-17 months who were followed for 48 months; efficacy was less when given to infants at the age of 6-12 weeks. RTS,S/AS01 provides a high level of protection during the first three months after vaccination, modelled to be about 70% in the phase 3 trial, a level of initial efficacy similar to that observed in an earlier phase 2 trial in Gambian adults. However, efficacy wanes progressively over the following months. A subsequent dose given 18 months after the primary series restores some but not all of the efficacy seen immediately after the primary series. In July 2015, the European Medicines Agency reviewed efficacy and safety data on RTS.S/AS01 and concluded that the risk benefit balance favoured the vaccine and gave a positive opinion on its use in children aged 6 weeks to 17 months. The World Health Organization (WHO) Strategic Advisory Group of Experts (SAGE) committee reviewed the vaccine's efficacy and safety in October 2015 and made a number of recommendations on its further evaluation. These included the pilot implementation of RTS,S/AS01 in children aged 5-17 months in 3-5 settings with moderate-to-high malaria transmission intensity, with a preference for areas where SMC is not being delivered, and evaluation of alternative approaches to deployment of the vaccine. Recent evidence from challenge studies conducted in American adult volunteers suggests that a higher level of protection can be obtained when the third dose of the priming schedule is reduced to one fifth of the usual amount and delayed until approximately 6 months post dose 2, and when a reduced dose is used for boosting. In these studies, a vaccine efficacy of 86% was achieved three weeks following priming and 90% efficacy following boosting with a fractional dose. This encouraging result is now being followed in further studies.

SMC involves monthly administration of an antimalarial drug or drug combination in a full therapeutic course to children on three of four occasions during the period of highest risk of malaria infection. Studies undertaken in several countries in West Africa, including Burkina Faso and Mali, have shown that SMC with sulphadoxine/pyrimethamine (SP) and amodiaquine (AQ) is highly effective in areas where the transmission of malaria is markedly seasonal, reducing the incidence of severe and uncomplicated malaria by up to 80%. SMC with a combination of SP and AQ is safe, with no serious drug related adverse event being reported after administration of over 800,000 courses in Senegal. Recent studies have defined the areas where SMC would be an appropriate intervention based on the seasonality and incidence of malaria. These include most of the Sahel and sub-Sahel, population approximately 200 million, and possibly other areas in southern and eastern Africa. A Technical Expert Group of the WHO reviewed all the available evidence on the efficacy and safety of SMC in May 2011 and recommended SMC with SP+AQ in areas of the Sahel and sub-Sahel with highly seasonal transmission. This recommendation was endorsed by the WHO Malaria Policy Advisory Committee (MPAC) in February 2012. Most countries in the Sahel and sub-Sahel region have incorporated SMC, along with other malaria control interventions in their strategic malaria control plan and the implementation of SMC at scale is in progress in many countries in this region through the UNITAID supported SMC ACCESS programme and the support of other major donor organisations. Preliminary evaluation suggests that SMC is providing about 50% protection against clinical malaria when delivered through a national programme (http://www.malariaconsortium.org/pages/access-smc.htm).

SMC is effective but its delivery is demanding on the recipient and provider, requiring four contacts each malaria transmission season if anti-malarials are given to mothers to administer at home and 12 contacts if directly observed treatment is employed. In addition, SMC is threatened by the emergence of resistance to SP and AQ and there are currently no other combinations of licensed antimalarials that could be used to replace them. It is likely to be 5-10 years before novel antimalarials under development could be deployed for SMC. In contrast to SMC, seasonal vaccination with RTS,S/AS01 would require only one visit each transmission season after priming. RTS,S/AS01 may be a little less effective than SMC during the malaria transmission season but this may be balanced by provision of protection during the dry season, when some malaria transmission still occurs and when SMC would provide no benefit. There is, therefore, a need for a comparative study of these two interventions. In some areas where SMC is currently being deployed, and other malaria control interventions such as long-lasting insecticide treated nets used widely, the incidence of malaria in young children remains high (0.4 episodes per year in children under the age of five years in SMC recipients in Burkina Faso). Thus, determining whether RTS,S/AS01 would provide added, useful protection to SMC in such situations is also important. It might also be able to protect some children who, because of side effects, are unable or unwilling to take SMC.

Although the European Medicines Agency has given a positive opinion on RTS,S/AS01, it is not yet certain how this partially effective malaria vaccine can be used most effectively. Three, large-scale pilot implementation studies are being planned by WHO but it is unlikely that, following WHO recommendations, any of these will be conducted in a country where SMC is being delivered. The WHO recommendations on RTS,S/AS01 indicate the need for research on alternative approaches to the delivery of this vaccine. Exploration of the potential of the vaccine to prevent seasonal malaria, taking advantage of its high but rapidly waning efficacy, meets this recommendation and is, therefore, timely.

Study Design

Study Type:
Interventional
Actual Enrollment :
5920 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Intervention Model Description:
A double-blind, individually randomised trial to compare the incidence of clinical episodes of malaria across three study arms: Seasonal vaccination with the malaria vaccine RTS,S/AS01 Seasonal malaria chemoprevention with SP/AQ Combination of these two interventionsA double-blind, individually randomised trial to compare the incidence of clinical episodes of malaria across three study arms:Seasonal vaccination with the malaria vaccine RTS,S/AS01 Seasonal malaria chemoprevention with SP/AQ Combination of these two interventions
Masking:
Triple (Participant, Care Provider, Investigator)
Primary Purpose:
Prevention
Official Title:
A Phase IIIB Comparative Trial of Seasonal Vaccination With the Malaria Vaccine RTS,S/AS01, Seasonal Malaria Chemoprevention and of the Two Interventions Combined
Actual Study Start Date :
Apr 17, 2017
Actual Primary Completion Date :
Mar 31, 2020
Actual Study Completion Date :
Mar 31, 2020

Arms and Interventions

Arm Intervention/Treatment
Active Comparator: SMC with SP+AQ

Administration of RABIPUR® in Year 1 and Hepatitis A vaccine in Year 2 and 3, followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3.

Biological: RABIPUR®
Year 1 (2017) Three doses of rabies vaccine (April, May, June) Year 2 and 3 (2018/19) One dose of Hepatitis A vaccine (June)

Drug: SMC with SP+AQ
Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses.

Active Comparator: RTS,S/AS01

Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with placebo in Year 1,2 and 3.

Biological: RTS,S/AS01
Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June)

Drug: SMC placebo
Year 1, 2 and 3(2017/18/19) Four cycles of SMC placebo during the malaria transmission season

Active Comparator: RTS,S/AS01 PLUS SMC with SP+AQ

Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3.

Biological: RTS,S/AS01
Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June)

Drug: SMC with SP+AQ
Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses.

Outcome Measures

Primary Outcome Measures

  1. Incidence of Clinical Episodes of Malaria [Passive surveillance of clinical episodes of malaria within the study area starting from the date of the first dose of study vaccines (April/May 2017) until 31st March 2020- a total of 36 months.]

    Passive surveillance to detect episode of fever (temperature > 37.5 C), or a history of fever within the past 48 hours, that is severe enough to require treatment at a health centre and which is accompanied by a positive blood film with a parasite density of 5,000 per µl or more

Secondary Outcome Measures

  1. Clinical Episodes of Uncomplicated Febrile Illness [Passive surveillance in all health centers within the study area, active surveillance in a sub set of study children starting July 2017 till April 2020.]

    Passive and active surveillance to detect cases with temperature > 37.5o C), or a history of fever within the past 48 hours, with a positive blood film (any level of asexual parasitaemia) or a positive rapid diagnostic test (RDT) for malaria

  2. Hospital Admissions With Malaria, Including Severe Malaria [Through study completion (30 months), each child admitted in a study hospital will be treated and monitored until complete cure or death (a period of 3 years). Documentation of each hospital admission according to ICH-GCP.]

    Hospital admissions with malaria, including cases of severe malaria which meet WHO criteria for a diagnosis of severe malaria.

  3. Prevalence of Malaria Infection Not Severe Enough to Warrant a Clinic Visit [Weekly home visits through study completion from July 2017 - April 2020 to screen study children for malaria.]

    Active surveillance of malaria at household level to assess the prevalence of malaria infection not severe enough to warrant a clinic visit detected in a subset of randomly selected children.

  4. Prevalence of Malaria Parasitaemia, Including Gametocytaemia and the Prevalence of Moderate and Severe Anemia and Malnutrition [Blood sample collection during 2-week cross sectional survey at the end of each malaria transmission season.]

    The prevalence of malaria parasitaemia, including gametocytaemia, moderate and severe anaemia and malnutrition at the end of the malaria transmission season

  5. Serious Adverse Events (SAEs) [Through study completion (for 30 months), each SAE will be treated and documented according to ICH-GCP.]

    Serious adverse events (SAEs), including any deaths, occurring at any time during the study with special reference to any cases of meningitis and cerebral malaria (WHO case definition)

  6. Immune Response to the Vaccine (Anti-CSP Antibody Concentrations) [Blood sample collection prior to 1st dose of vaccine and 1 month after 3rd dose of the primary series of vaccination. In years 2 and 3 blood will be collected before the booster dose and 1 month after administration of the 4th (and 5th) vaccine dose.]

    After priming and after each booster dose, determined in a sub-sample of children

  7. Drug Resistance to SP and AQ [Blood sample collection during the 2-week cross sectional survey conducted at the end of malaria transmission season in 2019.]

    The presence of molecular markers of resistance to SP and AQ in parasite positive samples

  8. Prevalence of Malaria Parasitaemia in School Aged Children [Blood sample collection during the 2-week cross sectional survey at the end of the malaria transmission season in Year 2 and 3 (November 2018/19).]

    The prevalence of malaria parasitaemia at the end of the malaria transmission season in school-age children resident in the study areas, to determine overall malaria transmission

  9. SP+AQ Drug Sensitivity [Children with asymptomatic malaria parasitaemia identified during the final cross-sectional survey (November 2019), treated with a full course of SP+AQ over 3 days and followed for 28 days.]

    The 28-day treatment outcome in children with asymptomatic malaria parasitaemia treated with SP+AQ.

Other Outcome Measures

  1. Acceptability of RTS,S and SMC [Data collection in Year 3]

    The acceptability of the two interventions (separately and combined) to the health care deliverers and to the study communities (standardized questionnaires)

  2. Feasibility of Introducing Two Malaria Control Strategies Simultaneously [Data collection in Year 3]

    The feasibility of introducing two malaria control strategies simultaneously from the health system perspective (structured observations and interviews with health system officials)

Eligibility Criteria

Criteria

Ages Eligible for Study:
5 Months to 17 Months
Sexes Eligible for Study:
All
Accepts Healthy Volunteers:
No
Inclusion Criteria:
  • The child is a permanent resident of the study area and likely to remain a resident for the duration of the trial

  • The child is 5 - 17 months of age at the time of first vaccination

  • A parent or legally recognised guardian provides informed consent for the child to join the trial

Exclusion Criteria:
  • The child is a transient resident in the study area

  • The child is in care

  • The age of the child is outside the stipulated range

  • The child has a history of an adverse reaction to SP or AQ

  • The child has a serious underlying illness, including known HIV infection, unless this is well controlled by treatment, or severe malnutrition (weight for age or mid arm circumference Z scores < 3 SD)

  • The child is known to have an immune deficiency disease or is receiving an immunosuppressive drug

  • The child has previously received a malaria vaccine.

  • The child is enrolled in another malaria intervention trial

  • The parents or guardians do not provide informed consent

Contacts and Locations

Locations

Site City State Country Postal Code
1 Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest Ouagadougou Burkina Faso
2 Malaria Research & Training Center Bamako Mali

Sponsors and Collaborators

  • London School of Hygiene and Tropical Medicine
  • Malaria Research and Training Center, Bamako, Mali
  • Institut de Recherche en Sciences de la Sante, Burkina Faso

Investigators

  • Study Director: Alassane Dicko, Professor, Malaria Research & Training Center, Bamako
  • Study Director: Jean Bosco Ouedraogo, Professor, Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest (IRSS-DRO)

Study Documents (Full-Text)

More Information

Publications

Responsible Party:
Brian Greenwood, Professor, London School of Hygiene and Tropical Medicine
ClinicalTrials.gov Identifier:
NCT03143218
Other Study ID Numbers:
  • ITDCZJ29 - Greenwood
First Posted:
May 8, 2017
Last Update Posted:
Apr 7, 2022
Last Verified:
Mar 1, 2022
Individual Participant Data (IPD) Sharing Statement:
Yes
Plan to Share IPD:
Yes
Studies a U.S. FDA-regulated Drug Product:
No
Studies a U.S. FDA-regulated Device Product:
No
Keywords provided by Brian Greenwood, Professor, London School of Hygiene and Tropical Medicine
Additional relevant MeSH terms:

Study Results

Participant Flow

Recruitment Details
Pre-assignment Detail
Arm/Group Title SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Arm/Group Description Administration of RABIPUR® in Year 1 and Hepatitis A vaccine in Year 2 and 3, followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RABIPUR®: Year 1 (2017) Three doses of rabies vaccine (April, May, June) Year 2 and 3 (2018/19) One dose of Hepatitis A vaccine (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses. Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with placebo in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC placebo: Year 1, 2 and 3(2017/18/19) Four cycles of SMC placebo during the malaria transmission season Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses.
Period Title: Overall Study
STARTED 1965 1988 1967
COMPLETED 1716 1734 1740
NOT COMPLETED 249 254 227

Baseline Characteristics

Arm/Group Title SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ Total
Arm/Group Description Administration of RABIPUR® in Year 1 and Hepatitis A vaccine in Year 2 and 3, followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RABIPUR®: Year 1 (2017) Three doses of rabies vaccine (April, May, June) Year 2 and 3 (2018/19) One dose of Hepatitis A vaccine (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses. Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with placebo in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC placebo: Year 1, 2 and 3(2017/18/19) Four cycles of SMC placebo during the malaria transmission season Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses. Total of all reporting groups
Overall Participants 1965 1988 1967 5920
Age, Customized (Count of Participants)
Under 8 months
471
24%
448
22.5%
463
23.5%
1382
23.3%
8-11 months
532
27.1%
527
26.5%
531
27%
1590
26.9%
12-15 months
486
24.7%
552
27.8%
537
27.3%
1575
26.6%
16 months or older
476
24.2%
461
23.2%
436
22.2%
1373
23.2%
Sex: Female, Male (Count of Participants)
Female
950
48.3%
956
48.1%
948
48.2%
2854
48.2%
Male
1015
51.7%
1032
51.9%
1019
51.8%
3066
51.8%
Race and Ethnicity Not Collected (Count of Participants)
Count of Participants [Participants]
0
0%
Region of Enrollment (participants) [Number]
Burkina Faso
936
47.6%
911
45.8%
930
47.3%
2777
46.9%
Mali
1029
52.4%
1077
54.2%
1037
52.7%
3143
53.1%
LLIN use the night before the 2017 census (Count of Participants)
Yes
1593
81.1%
1580
79.5%
1577
80.2%
4750
80.2%
No
360
18.3%
391
19.7%
380
19.3%
1131
19.1%
Missing
12
0.6%
17
0.9%
10
0.5%
39
0.7%
PCV vaccination (Count of Participants)
Yes
1650
84%
1646
82.8%
1642
83.5%
4938
83.4%
No
82
4.2%
99
5%
74
3.8%
255
4.3%
Card not available
172
8.8%
175
8.8%
171
8.7%
518
8.8%
Card available but incomplete
61
3.1%
67
3.4%
79
4%
207
3.5%
Missing
0
0%
1
0.1%
1
0.1%
2
0%
Penta vaccination (Count of Participants)
Yes
1650
84%
1647
82.8%
1650
83.9%
4947
83.6%
No
87
4.4%
95
4.8%
78
4%
260
4.4%
Card not available
169
8.6%
179
9%
167
8.5%
515
8.7%
Card available but incomplete
58
3%
66
3.3%
67
3.4%
191
3.2%
Missing
1
0.1%
1
0.1%
5
0.3%
7
0.1%
MenAfriVac vaccination (Count of Participants)
Yes
468
23.8%
477
24%
478
24.3%
1423
24%
No
1245
63.4%
1251
62.9%
1251
63.6%
3747
63.3%
Missing
252
12.8%
260
13.1%
238
12.1%
750
12.7%

Outcome Measures

1. Primary Outcome
Title Incidence of Clinical Episodes of Malaria
Description Passive surveillance to detect episode of fever (temperature > 37.5 C), or a history of fever within the past 48 hours, that is severe enough to require treatment at a health centre and which is accompanied by a positive blood film with a parasite density of 5,000 per µl or more
Time Frame Passive surveillance of clinical episodes of malaria within the study area starting from the date of the first dose of study vaccines (April/May 2017) until 31st March 2020- a total of 36 months.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Arm/Group Description Administration of RABIPUR® in Year 1 and Hepatitis A vaccine in Year 2 and 3, followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RABIPUR®: Year 1 (2017) Three doses of rabies vaccine (April, May, June) Year 2 and 3 (2018/19) One dose of Hepatitis A vaccine (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses. Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with placebo in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC placebo: Year 1, 2 and 3(2017/18/19) Four cycles of SMC placebo during the malaria transmission season Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses.
Measure Participants 1965 1988 1967
Number (95% Confidence Interval) [No. of events/1000 person years at risk]
304.8
278.2
113.3
2. Secondary Outcome
Title Clinical Episodes of Uncomplicated Febrile Illness
Description Passive and active surveillance to detect cases with temperature > 37.5o C), or a history of fever within the past 48 hours, with a positive blood film (any level of asexual parasitaemia) or a positive rapid diagnostic test (RDT) for malaria
Time Frame Passive surveillance in all health centers within the study area, active surveillance in a sub set of study children starting July 2017 till April 2020.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
3. Secondary Outcome
Title Hospital Admissions With Malaria, Including Severe Malaria
Description Hospital admissions with malaria, including cases of severe malaria which meet WHO criteria for a diagnosis of severe malaria.
Time Frame Through study completion (30 months), each child admitted in a study hospital will be treated and monitored until complete cure or death (a period of 3 years). Documentation of each hospital admission according to ICH-GCP.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
4. Secondary Outcome
Title Prevalence of Malaria Infection Not Severe Enough to Warrant a Clinic Visit
Description Active surveillance of malaria at household level to assess the prevalence of malaria infection not severe enough to warrant a clinic visit detected in a subset of randomly selected children.
Time Frame Weekly home visits through study completion from July 2017 - April 2020 to screen study children for malaria.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
5. Secondary Outcome
Title Prevalence of Malaria Parasitaemia, Including Gametocytaemia and the Prevalence of Moderate and Severe Anemia and Malnutrition
Description The prevalence of malaria parasitaemia, including gametocytaemia, moderate and severe anaemia and malnutrition at the end of the malaria transmission season
Time Frame Blood sample collection during 2-week cross sectional survey at the end of each malaria transmission season.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
6. Secondary Outcome
Title Serious Adverse Events (SAEs)
Description Serious adverse events (SAEs), including any deaths, occurring at any time during the study with special reference to any cases of meningitis and cerebral malaria (WHO case definition)
Time Frame Through study completion (for 30 months), each SAE will be treated and documented according to ICH-GCP.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
7. Secondary Outcome
Title Immune Response to the Vaccine (Anti-CSP Antibody Concentrations)
Description After priming and after each booster dose, determined in a sub-sample of children
Time Frame Blood sample collection prior to 1st dose of vaccine and 1 month after 3rd dose of the primary series of vaccination. In years 2 and 3 blood will be collected before the booster dose and 1 month after administration of the 4th (and 5th) vaccine dose.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
8. Secondary Outcome
Title Drug Resistance to SP and AQ
Description The presence of molecular markers of resistance to SP and AQ in parasite positive samples
Time Frame Blood sample collection during the 2-week cross sectional survey conducted at the end of malaria transmission season in 2019.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
9. Secondary Outcome
Title Prevalence of Malaria Parasitaemia in School Aged Children
Description The prevalence of malaria parasitaemia at the end of the malaria transmission season in school-age children resident in the study areas, to determine overall malaria transmission
Time Frame Blood sample collection during the 2-week cross sectional survey at the end of the malaria transmission season in Year 2 and 3 (November 2018/19).

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
10. Secondary Outcome
Title SP+AQ Drug Sensitivity
Description The 28-day treatment outcome in children with asymptomatic malaria parasitaemia treated with SP+AQ.
Time Frame Children with asymptomatic malaria parasitaemia identified during the final cross-sectional survey (November 2019), treated with a full course of SP+AQ over 3 days and followed for 28 days.

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
11. Other Pre-specified Outcome
Title Acceptability of RTS,S and SMC
Description The acceptability of the two interventions (separately and combined) to the health care deliverers and to the study communities (standardized questionnaires)
Time Frame Data collection in Year 3

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description
12. Other Pre-specified Outcome
Title Feasibility of Introducing Two Malaria Control Strategies Simultaneously
Description The feasibility of introducing two malaria control strategies simultaneously from the health system perspective (structured observations and interviews with health system officials)
Time Frame Data collection in Year 3

Outcome Measure Data

Analysis Population Description
[Not Specified]
Arm/Group Title
Arm/Group Description

Adverse Events

Time Frame Serious adverse event data were collected over a 3 year period, from 1st April 2017 to 31st March 2020.
Adverse Event Reporting Description The number of participants at risk for other (non-serious) adverse events is zero because non-serious adverse events were not collected or assessed as part of this study.
Arm/Group Title SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Arm/Group Description Administration of RABIPUR® in Year 1 and Hepatitis A vaccine in Year 2 and 3, followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RABIPUR®: Year 1 (2017) Three doses of rabies vaccine (April, May, June) Year 2 and 3 (2018/19) One dose of Hepatitis A vaccine (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses. Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with placebo in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC placebo: Year 1, 2 and 3(2017/18/19) Four cycles of SMC placebo during the malaria transmission season Administration of the malaria vaccine RTS,S/AS01 followed by 4 cycles of SMC with sulphadoxine/pyrimethamine plus amodiaquine in Year 1,2 and 3. RTS,S/AS01: Year 1 (2017) Three doses of RTSS/AS01 (April, May, June) Year 2 and 3 (2018/19) One booster dose of RTSS/AS01 (June) SMC with SP+AQ: Year 1, 2 and 3(2017/18/19) Four cycles of SMC (SP+AQ) during the malaria transmission season One cycle of SMC for children above one year of age consisting of sulphadoxine - pyrimethamin (SP) 500mg/25 mg, and amodiaquine (AQ) 150mg on day 1, and AQ 150mg on days 2 and 3. Infants will receive half of these doses.
All Cause Mortality
SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 32/1965 (1.6%) 27/1988 (1.4%) 15/1967 (0.8%)
Serious Adverse Events
SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 97/1965 (4.9%) 94/1988 (4.7%) 71/1967 (3.6%)
General disorders
Non-fatal SAEs 65/1965 (3.3%) 67/1988 (3.4%) 56/1967 (2.8%)
Fatal SAEs 32/1965 (1.6%) 27/1988 (1.4%) 15/1967 (0.8%)
Other (Not Including Serious) Adverse Events
SMC With SP+AQ RTS,S/AS01 RTS,S/AS01 PLUS SMC With SP+AQ
Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
Total 0/0 (NaN) 0/0 (NaN) 0/0 (NaN)

Limitations/Caveats

[Not Specified]

More Information

Certain Agreements

All Principal Investigators ARE employed by the organization sponsoring the study.

There is NOT an agreement between Principal Investigators and the Sponsor (or its agents) that restricts the PI's rights to discuss or publish trial results after the trial is completed.

Results Point of Contact

Name/Title Professor Brian Greenwood
Organization London School of Hygiene and Tropical Medicine
Phone +442072994707
Email brian.greenwood@lshtm.ac.uk
Responsible Party:
Brian Greenwood, Professor, London School of Hygiene and Tropical Medicine
ClinicalTrials.gov Identifier:
NCT03143218
Other Study ID Numbers:
  • ITDCZJ29 - Greenwood
First Posted:
May 8, 2017
Last Update Posted:
Apr 7, 2022
Last Verified:
Mar 1, 2022