Skip to main content

Seasonal malaria chemoprevention: successes and missed opportunities

Abstract

Seasonal malaria chemoprevention (SMC) was recommended in 2012 for young children in the Sahel during the peak malaria transmission season. Children are given a single dose of sulfadoxine/pyrimethamine combined with a 3-day course of amodiaquine, once a month for up to 4 months. Roll-out and scale-up of SMC has been impressive, with 12 million children receiving the intervention in 2016. There is evidence of its overall benefit in routine implementation settings, and a meta-analysis of clinical trial data showed a 75% decrease in clinical malaria compared to placebo. SMC is not free of shortcomings. Its target zone includes many hard-to-reach areas, both because of poor infrastructure and because of political instability. Treatment adherence to a 3-day course of preventive treatment has not been fully documented, and could prove challenging. As SMC is scaled up, integration into a broader, community-based paradigm which includes other preventive and curative activities may prove beneficial, both for health systems and for recipients.

Background

Since its recommendation by the World Health Organization in 2012 [1], Seasonal malaria chemoprevention (SMC) has been introduced in 12 countries [2]. SMC targets children aged 3–59 months in the Sahel during the short, but intense, malaria transmission season. Children are given monthly courses of sulfadoxine-pyrimethamine (SP) and amodiaquine (AQ) for up to 4 months. Each month, AQ is given in daily doses for 3 days, and is given with single-dose of SP on the 1st day. The clinical trials that evaluated strategies similar to what has come to be called SMC used different treatment regimens (SP alone, SP with AQ, SP with artesunate, and artesunate-AQ) with different treatment intervals (monthly versus every 2 months). None gave treatment for more than 3 months of the year [3,4,5,6,7,8,9]. With a variety of drug regimens used, direct comparison is difficult, but meta-analysis of clinical trial data showed a 75% decrease in clinical malaria with SMC compared to placebo, a modest impact on the prevalence of anaemia but with high heterogeneity between studies, and no significant decrease in hospitalizations or all-cause mortality, though death rates in all trials were low [10]. The goal of treating all children in the community irrespective of disease is to clear parasites and to provide a prophylactic effect thereafter [11]. Prior to the scale-up of SMC, chemoprevention strategies have been used in health centers in the setting of antenatal visits (IPTp) and routine childhood vaccinations (IPTi) [12, 13].

Since 2012, the scale-up of SMC has been remarkable, with over 12 million children receiving the intervention in 2016. Much of the growth has been driven by a UNITAID-funded consortium, Access-SMC, which provided SMC to 4 million children in 2016 [2]. Other major funders include the World Bank and UNICEF. National Malaria Control Programmes (NMCP) have readily adopted donor-funded SMC, and many non-governmental organizations are supporting its implementation. Few data from programme-implementation settings have been published, but one recent observational study in Mali in 2014 showed that prevalence of parasitaemia at the end of the high transmission seasonal was lower in areas receiving SMC (18%) than in areas not receiving SMC (46%) [14]. For recipients, data from clinical trials suggested high levels of community acceptability when taken during the peak malaria season [15], and at least one study from a programme implementation setting has shown a reasonable cost-effectiveness of the strategy (approximately US $100 per case averted and $3300 per under-5 death averted) [16], but broader data have yet to be published.

Since SMC has been recommended, research on innovative strategies has continued. In an area with long seasonal transmission, children receiving 5 monthly distributions of SMC had approximately 50% lower malaria incidence than children receiving no SMC (or children receiving 3 monthly distributions/ [17]. A trial of azithromycin in addition to SP + AQ has been carried out, and a trial of SMC and seasonal vaccination with the RTS,S/AS01 malaria vaccine is underway [18].

Challenges for SMC

Despite its impact, SMC remains unsatisfying for several reasons, including its logistical burden, the use of preventive medications that require a 3-day course of therapy, and incomplete coverage of zones which could benefit most from SMC.

SMC distributions occur once per month, necessitating a large commitment of time and resources by NMCPs and their partners. Reaching millions of children every month, every rainy season, is a challenge in any setting, particularly so in the rural Sahel, where infrastructure is poor, and access is difficult especially during the rainy season. As with many preventive interventions, children who stand to benefit the most from SMC (those living farthest from health centres and who have the worst access to care) are also the hardest to reach month after month. With implementing partners facing “SMC fatigue”, it is a challenge to continue reaching these most isolated regions. One way forward could be decentralization, perhaps making one delivery per season, storing enough SP-AQ to cover all four rounds in remote villages, and transferring the responsibility to distribute the anti-malarials to community health workers. Such a strategy would be logistically less challenging but requires close, continued supervision in these areas.

Data from clinical trials have suggested high levels of adherence to the 3-day course required by SMC [19], but to date, there is little objective, quantitative data that confirm adherence (such as plasma drug levels) from areas where SMC is implemented as a routine programme. Poor adherence will accelerate the spread of resistance to SP and AQ, which may in turn lead to decreased protective effectiveness of SMC. Ideally, preventive treatments (and cures) would be single-dose, but no single-dose therapies are available for use in SMC, so ensuring good adherence to the multi-dose treatment is important, both now and for the future. Other regimens have been trialled, notably dihydroartemisinin-piperaquine, which was shown to be non-inferior to SP-AQ in Burkina Faso [20], but still require multiple doses and pose the same problems with adherence.

One of the biggest blank spots in SMC coverage is in northern Nigeria, where an estimated 11 million children live in areas eligible for SMC, but where only approximately 4 million were targeted to receive SMC in 2018. Weak infrastructure and ongoing armed conflicts make programme expansion difficult in this area despite the large potential benefit. One of the few strong systems in that area is related to polio eradication, and one possible entry for SMC into this region could be to repurpose already-existing human resources to also distribute SMC.

Opportunities

In the Sahel, immunization is administered as a vertical programme in the Expanded Programme on Immunization (EPI). In 2015, at a national level, measles-containing vaccine coverage was 88% in Burkina Faso, 76% in Mali, and 73% in Niger, all below target levels [21]. During the peak season of malnutrition (which frequently overlaps with the malaria season), therapeutic feeding programmes are run as independent vertical programmes by different departments of Ministries of Health. Coverage is chronically low, with only an estimated 7–13% of children suffering from severe acute malnutrition enrolled in a programme [22]. SMC is also managed as another vertical programme by NMCPs. Each of these three large vertical programmes (EPI, nutrition, and SMC) has its own funding streams, with little incentive for integration. A few local pilot programmes, often supported by international partners, have combined SMC with nutritional screening and catch-up vaccination, but these have not been scaled up. Integrating programmes is not only cost efficient for donors, it could also reach more children and increase adherence.

Ideally integrated programmes would be administered through community health workers responsible for many different activities. There is increasing evidence that the existence of a competent community health worker in a village who provides diagnosis and treatment is associated with a reduction of malaria incidence and the prevalence of Plasmodium falciparum infections in areas with low malaria endemicity [23]. Although implementing such programmes can be challenging in high endemicity environments [24], taking a more holistic approach to malaria control—and child health in general—in the Sahel could be beneficial. This sort of integrated model may also avoid future problems with sustainability that have been observed in other programmes that involve regular mass distributions of drugs, like those used in several neglected tropical disease programmes, where integrated models have proven both effective and sustainable [25].

Conclusions

SMC was never intended to be a “magic bullet” [2]. Given the multi-factorial and multi-sectorial nature of malaria control, focus on SMC by potential funders, and even programme participants, highlights the risk of perceiving SMC as a singular solution. As SMC expansion continues, efforts should be made to integrate the programme into a more comprehensive and sustainable community-based paradigm, that also includes other curative and preventive care.

Abbreviations

AQ:

amodiaquine

EPI:

Expanded Programme on Immunisation

IPTi:

Intermittent preventive treatment for malaria in infants

IPTp:

Intermittent preventive treatment for malaria in pregnant women

NMCP:

National malaria control programme

SMC:

seasonal malaria chemoprevention

SP:

sulfadoxine-pyrimethamine

References

  1. WHO Global Malaria Programme. WHO policy recommendation: Seasonal malaria chemoprevention for Plasmodium falciparum control in highly seasonal transmission areas of the Sahel sub-region in Africa. Geneva: World Health Organization; 2012.

    Google Scholar 

  2. York A. Seasonal malaria chemoprevention in the Sahel. Lancet Infect Dis. 2017;17:588.

    Article  PubMed  Google Scholar 

  3. Dicko A, Sagara I, Sissoko MS, Guindo O, Diallo AI, Kone M, et al. Impact of intermittent preventive treatment with sulphadoxine-pyrimethamine targeting the transmission season on the incidence of clinical malaria in children in Mali. Malar J. 2008;7:123.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Dicko A, Diallo AI, Tembine I, Dicko Y, Dara N, Sidibe Y, et al. 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:e1000407.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Kweku M, Liu D, Adjuik M, Binka F, Seidu M, Greenwood B, et al. Seasonal intermittent preventive treatment for the prevention of anaemia and malaria in Ghanaian children: a randomized, placebo controlled trial. PLoS ONE. 2008;3:e4000.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Konaté AT, Yaro JB, Ouédraogo AZ, Diarra A, Gansané A, Soulama I, et al. 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:e1000408.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Sesay S, Milligan P, Touray E, Sowe M, Webb EL, Greenwood BM, et al. A trial of intermittent preventive treatment and home-based management of malaria in a rural area of The Gambia. Malar J. 2011;10:2.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Tagbor H, Cairns M, Nakwa E, Browne E, Sarkodie B, Counihan H, et al. The clinical impact of combining intermittent preventive treatment with home management of malaria in children aged below 5 years: cluster randomised trial. Trop Med Int Health. 2011;16:280–9.

    Article  PubMed  Google Scholar 

  9. Cissé B, Sokhna C, Boulanger D, Milet J, Bâ EH, Richardson K, et al. Seasonal intermittent preventive treatment with artesunate and sulfadoxine-pyrimethamine prevents malaria in Senegalese children. Lancet. 2006;367:659–67.

    Article  PubMed  Google Scholar 

  10. Meremikwu Martin M, 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:CD003756.

    Google Scholar 

  11. Greenwood B. Anti-malarial drugs and the prevention of malaria in the population of malaria endemic areas. Malar J. 2010;9(Suppl 3):S2.

    Article  PubMed  PubMed Central  Google Scholar 

  12. WHO. Updated WHO policy recommendation: intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP). Geneva: World Health Organization; 2012.

    Google Scholar 

  13. WHO. Policy recommendation on intermittent preventive treatment during infancy with sulphadoxine-pyrimethamine (SP-IPTi) for Plasmodium falciparum malaria control in Africa. Geneva: World Health Organization; 2010.

    Google Scholar 

  14. Diawara F, Steinhardt LC, Mahamar A, Traore T, Kone DT, Diawara H, et al. Measuring the impact of seasonal malaria chemoprevention as part of routine malaria control in Kita, Mali. Malar J. 2017;16:325.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Pitt C, Diawara H, Ouédraogo DJ, Diarra S, Kaboré H, Kouéla K, et al. Intermittent preventive treatment of malaria in children: a qualitative study of community perceptions and recommendations in Burkina Faso and Mali. PLoS ONE. 2012;7:e32900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nonvignon J, Aryeetey GC, Issah S, Ansah P, Malm KL, Ofosu W, et al. Cost-effectiveness of seasonal malaria chemoprevention in upper west region of Ghana. Malar J. 2016;15:367.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Tagbor H, Antwi GD, Acheampong PR, Bart Plange C, Chandramohan D, Cairns M. Seasonal malaria chemoprevention in an area of extended seasonal transmission in Ashanti, Ghana: an individually randomised clinical trial. Trop Med Int Health. 2016;21:224–35.

    Article  PubMed  Google Scholar 

  18. Greenwood B, Dicko A, Sagara I, Zongo I, Tinto H, Cairns M, et al. Seasonal vaccination against malaria: a potential use for an imperfect malaria vaccine. Malar J. 2017;16:182.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Souares A, Lalou R, Sene I, Sow D, Le Hesran J-Y. Factors related to compliance to anti-malarial drug combination: example of amodiaquine/sulphadoxine-pyrimethamine among children in rural Senegal. Malar J. 2009;8:118.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zongo I, Milligan P, Compaore YD, Some AF, Greenwood B, Tarning J, et al. Randomized non-inferiority trial of dihydroartemisinin-piperaquine compared with sulfadoxine-pyrimethamine plus amodiaquine for seasonal malaria chemoprevention in Burkina Faso. Antimicrob Agents Chemother. 2015;59:4387–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. WHO. Vaccine preventable diseases: monitoring system. Geneva, World Health Organization, 2017. Global summary. http://apps.who.int/immunization_monitoring/globalsummary/. Accessed 19 June 2017.

  22. UNICEF. Coverage monitoring network, ACF international. The state of global SAM management coverage. New York and London; 2012.

  23. Malaria elimination task force. Malaria elimination task force activity report: May 2014 to December 2016. 2017. http://www.shoklo-unit.com/sites/default/files/reports/malaria-elimination-task-force/metf_report_may_14_dec_16.pdf. Accessed 1 Oct 2017.

  24. Sunguya BF, Mlunde LB, Ayer R, Jimba M. Towards eliminating malaria in high endemic countries: the roles of community health workers and related cadres and their challenges in integrated community case management for malaria: a systematic review. Malar J. 2017;16:10.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Linehan M, Hanson C, Weaver A, Baker M, Kabore A, Zoerhoff KL, et al. Integrated implementation of programs targeting neglected tropical diseases through preventive chemotherapy: proving the feasibility at national scale. Am J Trop Med Hyg. 2011;84:5–14.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Authors’ contributions

MEC wrote the first draft of the manuscript. LVS and RFG provided significant intellectual input and revised the manuscript. All authors read and approved the final the manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Funding

The preparation of this commentary was supported by Médecins Sans Frontières, which had no role in the writing of the manuscript.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Matthew E. Coldiron.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Coldiron, M.E., Von Seidlein, L. & Grais, R.F. Seasonal malaria chemoprevention: successes and missed opportunities. Malar J 16, 481 (2017). https://doi.org/10.1186/s12936-017-2132-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-017-2132-1

Keywords