Effectiveness of seasonal malaria chemoprevention at scale in west and central Africa: an observational study

ACCESS-SMC Partnership, Ebenezer Baba, Prudence Hamade, Harriet Kivumbi, Maddy Marasciulo, Kolawole Maxwell, Diego Moroso, Arantxa Roca-Feltrer, Adama Sanogo, Joanna Stenstrom Johansson, James Tibenderana, Rahila Abdoulaye, Patrice Coulibaly, Eric Hubbard, Huja Jah, Eugene Kaman Lama, Lantorina Razafindralambo, Suzanne Van Hulle, George Jagoe, André-Marie Tchouatieu, David Collins, Colin Gilmartin, Gladys Tetteh, Yacine Djibo, Fara Ndiaye, Momodou Kalleh, Balla Kandeh, Bala Audu, Godwin Ntadom, Alice Kiba, Yacouba Savodogo, Kodbesse Boulotigam, Djiddi Ali Sougoudi, Timothee Guilavogui, Moussa Keita, Diakalidia Kone, Hadiza Jackou, Ibrahim Ouba, Emile Ouedraogo, Halimatou Alassana Messan, Fatou Jah, Markieu Janneh Kaira, Mariama Sire Sano, Mamadou Chérif Traore, Nadine Ngarnaye, Aishatu Yinusa Cassandra Elagbaje, Christine Halleux, Corinne Merle, Noha Iessa, Shanthi Pal, Houda Sefiani, Rachida Souleymani, Ibrahim Laminou, Daugla Doumagoum, Hamit Kesseley, Matt Coldiron, Rebecca Grais, Musa Kana, Jean Bosco Ouedraogo, Issaka Zongo, Tony Eloike, Sonny Johnbull Ogboi, Jane Achan, Kalifa Bojang, Serign Ceesay, Alassane Dicko, Abdoulaye Djimde, Issaka Sagara, Abdoulaye Diallo, Jean Louis NdDiaye, Kovana Marcel Loua, Khalid Beshir, Matt Cairns, Yolanda Fernandez, Sham Lal, Raoul Mansukhani, Julian Muwanguzi, Susana Scott, Paul Snell, Colin Sutherland, Rhosyn Tuta, Paul Milligan, ACCESS-SMC Partnership, Ebenezer Baba, Prudence Hamade, Harriet Kivumbi, Maddy Marasciulo, Kolawole Maxwell, Diego Moroso, Arantxa Roca-Feltrer, Adama Sanogo, Joanna Stenstrom Johansson, James Tibenderana, Rahila Abdoulaye, Patrice Coulibaly, Eric Hubbard, Huja Jah, Eugene Kaman Lama, Lantorina Razafindralambo, Suzanne Van Hulle, George Jagoe, André-Marie Tchouatieu, David Collins, Colin Gilmartin, Gladys Tetteh, Yacine Djibo, Fara Ndiaye, Momodou Kalleh, Balla Kandeh, Bala Audu, Godwin Ntadom, Alice Kiba, Yacouba Savodogo, Kodbesse Boulotigam, Djiddi Ali Sougoudi, Timothee Guilavogui, Moussa Keita, Diakalidia Kone, Hadiza Jackou, Ibrahim Ouba, Emile Ouedraogo, Halimatou Alassana Messan, Fatou Jah, Markieu Janneh Kaira, Mariama Sire Sano, Mamadou Chérif Traore, Nadine Ngarnaye, Aishatu Yinusa Cassandra Elagbaje, Christine Halleux, Corinne Merle, Noha Iessa, Shanthi Pal, Houda Sefiani, Rachida Souleymani, Ibrahim Laminou, Daugla Doumagoum, Hamit Kesseley, Matt Coldiron, Rebecca Grais, Musa Kana, Jean Bosco Ouedraogo, Issaka Zongo, Tony Eloike, Sonny Johnbull Ogboi, Jane Achan, Kalifa Bojang, Serign Ceesay, Alassane Dicko, Abdoulaye Djimde, Issaka Sagara, Abdoulaye Diallo, Jean Louis NdDiaye, Kovana Marcel Loua, Khalid Beshir, Matt Cairns, Yolanda Fernandez, Sham Lal, Raoul Mansukhani, Julian Muwanguzi, Susana Scott, Paul Snell, Colin Sutherland, Rhosyn Tuta, Paul Milligan

Abstract

Background: Seasonal malaria chemoprevention (SMC) aims to prevent malaria in children during the high malaria transmission season. The Achieving Catalytic Expansion of SMC in the Sahel (ACCESS-SMC) project sought to remove barriers to the scale-up of SMC in seven countries in 2015 and 2016. We evaluated the project, including coverage, effectiveness of the intervention, safety, feasibility, drug resistance, and cost-effectiveness.

Methods: For this observational study, we collected data on the delivery, effectiveness, safety, influence on drug resistance, costs of delivery, impact on malaria incidence and mortality, and cost-effectiveness of SMC, during its administration for 4 months each year (2015 and 2016) to children younger than 5 years, in Burkina Faso, Chad, The Gambia, Guinea, Mali, Niger, and Nigeria. SMC was administered monthly by community health workers who visited door-to-door. Drug administration was monitored via tally sheets and via household cluster-sample coverage surveys. Pharmacovigilance was based on targeted spontaneous reporting and monitoring systems were strengthened. Molecular markers of resistance to sulfadoxine-pyrimethamine and amodiaquine in the general population before and 2 years after SMC introduction was assessed from community surveys. Effectiveness of monthly SMC treatments was measured in case-control studies that compared receipt of SMC between patients with confirmed malaria and neighbourhood-matched community controls eligible to receive SMC. Impact on incidence and mortality was assessed from confirmed outpatient cases, hospital admissions, and deaths associated with malaria, as reported in national health management information systems in Burkina Faso and The Gambia, and from data from selected outpatient facilities (all countries). Provider costs of SMC were estimated from financial costs, costs of health-care staff time, and volunteer opportunity costs, and cost-effectiveness ratios were calculated as the total cost of SMC in each country divided by the predicted number of cases averted.

Findings: 12 467 933 monthly SMC treatments were administered in 2015 to a target population of 3 650 455 children, and 25 117 480 were administered in 2016 to a target population of 7 551 491. In 2015, among eligible children, mean coverage per month was 76·4% (95% CI 74·0-78·8), and 54·5% children (95% CI 50·4-58·7) received all four treatments. Similar coverage was achieved in 2016 (74·8% [72·2-77·3] treated per month and 53·0% [48·5-57·4] treated four times). In 779 individual case safety reports over 2015-16, 36 serious adverse drug reactions were reported (one child with rash, two with fever, 31 with gastrointestinal disorders, one with extrapyramidal syndrome, and one with Quincke's oedema). No cases of severe skin reactions (Stevens-Johnson or Lyell syndrome) were reported. SMC treatment was associated with a protective effectiveness of 88·2% (95% CI 78·7-93·4) over 28 days in case-control studies (2185 cases of confirmed malaria and 4370 controls). In Burkina Faso and The Gambia, implementation of SMC was associated with reductions in the number of malaria deaths in hospital during the high transmission period, of 42·4% (95% CI 5·9 to 64·7) in Burkina Faso and 56·6% (28·9 to 73·5) in The Gambia. Over 2015-16, the estimated reduction in confirmed malaria cases at outpatient clinics during the high transmission period in the seven countries ranged from 25·5% (95% CI 6·1 to 40·9) in Nigeria to 55·2% (42·0 to 65·3) in The Gambia. Molecular markers of resistance occurred at low frequencies. In individuals aged 10-30 years without SMC, the combined mutations associated with resistance to amodiaquine (pfcrt CVIET haplotype and pfmdr1 mutations [86Tyr and 184Tyr]) had a prevalence of 0·7% (95% CI 0·4-1·2) in 2016 and 0·4% (0·1-0·8) in 2018 (prevalence ratio 0·5 [95% CI 0·2-1·2]), and the quintuple mutation associated with resistance to sulfadoxine-pyrimethamine (triple mutation in pfdhfr and pfdhps mutations [437Gly and 540Glu]) had a prevalence of 0·2% (0·1-0·5) in 2016 and 1·0% (0·6-1·6) in 2018 (prevalence ratio 4·8 [1·7-13·7]). The weighted average economic cost of administering four monthly SMC treatments was US$3·63 per child.

Interpretation: SMC at scale was effective in preventing morbidity and mortality from malaria. Serious adverse reactions were rarely reported. Coverage varied, with some areas consistently achieving high levels via door-to-door campaigns. Markers of resistance to sulfadoxine-pyrimethamine and amodiaquine remained uncommon, but with some selection for resistance to sulfadoxine-pyrimethamine, and the situation needs to be carefully monitored. These findings should support efforts to ensure high levels of SMC coverage in west and central Africa.

Funding: Unitaid.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
SMC implementation areas Areas where SMC was implemented in 2015 are shown in blue, via the ACCESS-SMC project (dark blue) and other implementers (light blue). These areas continued SMC in 2016. Green shading shows the additional areas where SMC started in 2016, via the ACCESS SMC project (dark green) and other implementers (light green). In 2015, SMC programmes in nine countries reached about 7 million children, 3 million of them via ACCESS-SMC, and in 2016, programmes in 12 countries reached about 15 million children, 7 million via ACCESS-SMC (appendix p 3). SMC=seasonal malaria chemoprevention. ACCESS-SMC=Achieving Catalytic Expansion of SMC in the Sahel.
Figure 2
Figure 2
Effectiveness of SMC (A) Case-control estimates of the effectiveness of SMC treatments. Datapoints are the percentage reduction in malaria incidence in 28 days since the start of treatment, and 29–42 days post-treatment, compared with the incidence in children who had not received SMC within the last 42 days (underlying incidence data reported previously; appendix pp 6, 36). Error bars show 95% CIs (lower confidence limits of pfdhfr, pfdhps-436Ala, and pfdhps-4367Gly variants were too narrow to display). Results in children younger than 5 years are given in the appendix (p 15). SMC=seasonal malaria chemoprevention. pfcrt=Plasmodium falciparum chloroquine resistance transporter. pfmdr1=P falciparum multidrug resistance 1. pfdhfr=P falciparum dihydrofolate reductase. pfdhps=P falciparum dihydropteroate synthetase. pfcrt-CVIET=amino acid positions 72–76.
Figure 3
Figure 3
Examples of the effect of SMC on malaria rates (A) Numbers of confirmed cases of malaria in outpatient clinics, hospital inpatients admitted with a primary diagnosis of malaria, and deaths in hospital attributed to malaria, among children younger than 5 years and individuals aged 5 years and older, during transmission periods before and after SMC introduction for children younger than 5 years in the Upper River and Central River regions of The Gambia. (B) Numbers of confirmed cases of malaria among children younger than 5 years and individuals aged 5 years and older, in Kadiolo health centre, Sikasso region, Mali, each month before and after introduction of SMC for children younger than 5 years. SMC=seasonal malaria chemoprevention.

References

    1. WHO . World Health Organization; Geneva: 2019. World malaria report 2018.
    1. Wilson AL. A systematic review and meta-analysis of the efficacy and safety of intermittent preventive treatment of malaria in children (IPTc) PLoS One. 2011;6
    1. Meremikwu MM, Donegan S, Sinclair D, Esu E, Oringanje C. Intermittent preventive treatment for malaria in children living in areas with seasonal transmission. Cochrane Database Syst Rev. 2012;2
    1. Cissé B, Sokhna C, Boulanger D. Seasonal intermittent preventive treatment with artesunate and sulfadoxine-pyrimethamine for prevention of malaria in Senegalese children: a randomised, placebo-controlled, double-blind trial. Lancet. 2006;367:659–667.
    1. Sokhna C, Cissé B, Ba EH. A trial of the efficacy, safety and impact on drug resistance of four drug regimens for seasonal intermittent preventive treatment in Senegalese children. PLoS One. 2008;3
    1. Dicko A, Diallo AI, Tembine I. Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in Mali: a randomised, double-blind, placebo-controlled trial. PLoS Med. 2011;8
    1. Konaté AT, Yaro JB, Ouédraogo AZ. Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in Burkina Faso: a randomised, double-blind, placebo-controlled trial. PLoS Med. 2011;8
    1. Cissé B, Ba EH, Sokhna C. Effectiveness of seasonal malaria chemoprevention in children under ten years of age in Senegal: a stepped-wedge cluster-randomised trial. PLoS Med. 2016;13
    1. Sinclair D, Meremikwu MM, Garner P. Seasonal malaria chemoprevention for preventing malaria morbidity in children aged less than 5 years living in areas of marked seasonal transmission: GRADE tables to assist guideline development and recommendations. Oct 26, 2011.
    1. WHO Report of the technical consultation on seasonal malaria chemoprevention. 2012.
    1. WHO WHO policy recommendation: seasonal malaria chemoprevention (SMC) for Plasmodium falciparum malaria control in highly seasonal transmission areas of the Sahel sub-region in Africa. March, 2012.
    1. WHO . World Health Organization; Geneva: 2012. Seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in children. A field guide.
    1. Lasry E. A novel prevention programme has dramatically reduced malaria cases in Mali and Chad. Sept 24, 2012.
    1. Médecins Sans Frontières Malaria: MSF provides preventive treatment to children in the Sahel region. Nov 12, 2014.
    1. Koscolacova A. Chemical prevention of seasonal malaria in Niger. Executive summary. February, 2015.
    1. WHO Global Malaria Programme Minutes of the Technical Expert Group on Drug Efficacy and Response, 1–2 June 2017. 5.1 Monitoring efficacy of seasonal malaria chemoprevention in the ACCESS-SMC project. October, 2017.
    1. Pal SN, Duncombe C, Falzon D, Olsson S. WHO strategy for collecting safety data in public health programmes: complementing spontaneous reporting systems. Drug Saf. 2013;2013:75–81.
    1. Gilmartin C, Nonvignon J, Cairns M, et al. Seasonal malaria chemoprevention in the Sahel subregion of Africa: a cost-effectiveness and cost-savings analysis. Lancet Glob Health (in press).
    1. Winskill P, Slater HC, Griffin JT, Ghani AC, Walker PGT. The US President's malaria initiative, Plasmodium falciparum transmission and mortality: a modelling study. PLoS Med. 2017;14
    1. Scott S, Cairns M, Lal S. London School of Hygiene & Tropical Medicine; London: 2018. Seasonal malaria chemoprevention coverage in seven West African countries, 2015–2016. Report for UNITAID.
    1. Cairns M, Sagara I, Zongo I. London School of Hygiene & Tropical Medicine; London: 2018. Assessment of the protective efficacy of seasonal malaria chemoprevention with sulfadoxine-pyrimethamine plus amodiaquine in Burkina Faso, Chad, The Gambia, Mali and Nigeria, 2015–2016. Report for UNITAID.
    1. Druetz T, Corneau-Tremblay N, Millogo T. Impact evaluation of seasonal malaria chemoprevention under routine program implementation: a quasi-experimental study in Burkina Faso. Am J Trop Med Hyg. 2018;98:524–533.
    1. Diawara F, Steinhardt LC, Mahamar A. Measuring the impact of seasonal malaria chemoprevention as part of routine malaria control in Kita, Mali. Malar J. 2017;16:325.
    1. Nonvignon J, Aryeetey GC, Issah S. Cost-effectiveness of seasonal malaria chemoprevention in upper west region of Ghana. Malar J. 2016;15:367.
    1. NDiaye JL, Diallo I, NDiaye Y. Evaluation of two strategies for community-based safety monitoring during seasonal malaria chemoprevention campaigns in Senegal, compared with the national spontaneous reporting system. Pharmaceut Med. 2018;32:189–200.
    1. NDiaye JL, Cissé B, Ba EH. Safety of seasonal malaria chemoprevention (SMC) with sulfadoxine–pyrimethamine plus amodiaquine when delivered to children under 10 years of age by district health services in Senegal: results from a stepped-wedge cluster randomized trial. PLoS One. 2016;11
    1. WHO . Integrating pharmacovigilance in seasonal malaria chemoprevention: the story so far. In: WHO, editor. WHO pharmaceuticals newsletter no. 4. World Health Organization; Geneva: 2017. pp. 33–36.
    1. WHO . Fourteenth meeting of the WHO Advisory Committee on Safety of Medicinal Products (ACSoMP) In: WHO, editor. WHO pharmaceuticals newsletter no. 4. World Health Organization; Geneva: 2017. pp. 26–32.
    1. Greenwood BM, David PH, Otoo-Forbes LN. Mortality and morbidity from malaria after stopping malaria chemoprophylaxis. Trans R Soc Trop Med Hyg. 1995;89:629–633.
    1. NDiaye JLA, NDiaye Y, Ba MS. Seasonal malaria chemoprevention combined with community case management of malaria in children under 10 years of age, over 5 months, in southern Senegal: a cluster-randomized trial. PLoS Med. 2019;16

Source: PubMed

3
S'abonner