The promise and potential challenges of intermittent preventive treatment for malaria in infants (IPTi)

Wendy Prudhomme O'Meara, Joel G Breman, F Ellis McKenzie, Wendy Prudhomme O'Meara, Joel G Breman, F Ellis McKenzie

Abstract

Intermittent preventive treatment (IPT) administers a full therapeutic course of an anti-malarial drug at predetermined intervals, regardless of infection or disease status. It is recommended by the World Health Organization (WHO) for protecting pregnant women from the adverse effects of malaria (IPTp) and shows great potential as a strategy for reducing illness from malaria during infancy (IPTi). Administered concurrently with standard immunizations, IPTi is expected to reduce the frequency of clinical disease, but to allow blood-stage infections to occur between treatments, thus allowing parasite-specific immunity to develop. While wide deployment of IPTi is being considered, it is important to assess other potential effects. Transmission conditions, drug choice and administration schedule will likely affect the possibility of post-treatment rebound in child morbidity and mortality and the increased spread of parasite drug resistance and should be considered when implementing IPTi.

Figures

Figure 1
Figure 1
Concentration of anti-malarial drug in bloodstream during IPT. Drug concentration initially rises after administration of a therapeutic drug regimen. The concentration declines gradually as the drug is cleared from the bloodstream. Parasites with increasing sensitivity to the drug will be able to grow as the drug concentration declines. Parasites are divided roughly into three categories; fully sensitive, partially resistant and fully resistant. The interval between the time at which the concentration falls below the threshold for partially sensitive parasites and the time at which it falls below the threshold for sensitive parasites is a window of selection for resistant parasites. The dotted curve represents the target blood concentrations during chemoprophylaxis.
Figure 2
Figure 2
Effect of transmission intensity on outcome of IPTi. Qualitative depiction of the effect of transmission intensity on the age-incidence pattern of malaria morbidity and mortality. In areas of high transmission (solid line), malaria mortality is concentrated in the first two years of life. In areas of low or unstable transmission (dashed line), children may be at risk until much later in life. The arrows represent the window of greatest risk or the age interval during which roughly 75% of childhood malaria episodes are experienced.
Figure 3
Figure 3
Effect of transmission intensity on outcome of IPTi. Effect of biting frequency (arrows represent infectious bites) on the development of malaria-specific immunity during IPTi. The frequency of infectious bites determines whether or not an infant is exposed to malaria parasites between doses of drug during IPTi.
Figure 4
Figure 4
Drug treatment and immune mechanisms act synergistically to eliminate parasites. (a) A patient with some pre-existing immunity clears infection and reduces the number and transmission of the parasites that survive treatment. (b) Without pre-existing immunity, the window for proliferation and differential transmission of resistant parasites is extended. White and black circles represent sensitive and resistant parasites, respectively.
Figure 5
Figure 5
Effect of IPT on parasite diversity. Blood concentration of drug during two treatments of IPT is shown. All of the sensitive parasites (white circles) initially present in the host at the start of IPT are killed by the drug, leaving only resistant parasites (black circles). While the drug concentration is high, new infections are prevented and the resistant parasites proliferate without competition. When the drug concentration falls below the threshold for sensitive parasites, an infectious bite can produce an infection with sensitive parasites. A subsequent bloodmeal may take up gametocytes from both sensitive (white crescents) and resistant strains (black crescents), thereby allowing out-crossing between them. At the next treatment, the sensitive parasites are killed and the resistant parasites continue to proliferate and may acquire new genetic material at the next cycle. Black arrows at the top represent continual biting by infectious mosquitoes.

References

    1. Verhoeff FH, Brabin BJ, Chimsuku L, Kazembe P, Russell WB, Broadhead RL. An evaluation of the effects of intermittent sulfadoxine-pyrimethamine treatment in pregnancy on parasite clearance and risk of low birthweight in rural Malawi. Ann Trop Med Parasitol. 1998;92:141–150. doi: 10.1080/00034989859979.
    1. Parise ME, Ayisi JG, Nahlen BL, Schultz LJ, Roberts JM, Misore A, Muga R, Oloo AJ, Steketee RW. Efficacy of sulfadoxlne-pyrimethamine for prevention of placental malaria in an area of Kenya with a high prevalence of malaria and human immunodeficiency virus infection. Am J Trop Med Hyg. 1998;59:813–822.
    1. Shulman CE, Dorman EK, Cutts F, Kawuondo K, Bulmer JN, Peshu N, Marsh K. Intermittent sulphadoxine-pyrimethamine to prevent severe anaemia secondary to malaria in pregnancy: a randomised placebo-controlled trial. Lancet. 1999;353:632–636. doi: 10.1016/S0140-6736(98)07318-8.
    1. Rogerson SJ, Chaluluka E, Kanjala M, Mkundika P, Mhango C, Molyneux ME. Intermittent sulfadoxine-pyrimethamine in pregnancy: effectiveness against malaria morbidity in Blantyre, Malawi, in 1997–99. Trans R Soc Trop Med Hyg. 2000;94:549–553. doi: 10.1016/S0035-9203(00)90083-X.
    1. van Eijk AM, Ayisi JG, ter Kuile FO, Otieno JA, Misore AO, Odondi JO, Rosen DH, Kager PA, Steketee RW, Nahlen BL. Effectiveness of intermittent preventive treatment with sulphadoxine-pyrimethamine for control of malaria in pregnancy in western Kenya: a hospital-based study. Trop Med Int Health. 2004;9:351–360. doi: 10.1111/j.1365-3156.2004.01196.x.
    1. Kayentao K, Kodio M, Newman RD, Maiga H, Doumtabe D, Ongoiba A, Coulibaly D, Keita AS, Maiga B, Mungai M, Parise ME, Doumbo O. Comparison of intermittent preventive treatment with chemoprophylaxis for the prevention of malaria during pregnancy in Mali. J Infect Dis. 2005;191:109–116. doi: 10.1086/426400.
    1. Bonnet S, Gouagna LC, Paul RE, Safeukui I, Meunier JY, Boudin C. Estimation of malaria transmission from humans to mosquitoes in two neighbouring villages in south Cameroon: evaluation and comparison of several indices. Trans R Soc Trop Med Hyg. 2003;97:53–59. doi: 10.1016/S0035-9203(03)90022-8.
    1. Akim NIJ, Drakeley C, Kingo T, Simon B, Senkoro K, Sauerwein RW. Dynamics of P. falciparum gametocytemia in symptomatic patients in an area of intense perennial transmission in Tanzania. Am J Trop Med Hyg. 2000;63:199–203.
    1. Drakeley CJ, Akim NIJ, Sauerwein RW, Greenwood BM, Targett GAT. Estimates of the infectious reservoir of Plasmodium falciparum malaria in The Gambia and in Tanzania. Trans R Soc Trop Med Hyg. 2000;94:472–476. doi: 10.1016/S0035-9203(00)90056-7.
    1. White NJ. Intermittent Presumptive Treatment for Malaria. PLoS Med. 2005;2:e3. doi: 10.1371/journal.pmed.0020003.
    1. Schellenberg D, Menendez C, Kahigwa E, Aponte J, Vidal J, Tanner M, Mshinda H, Alonso P. Intermittent treatment for malaria and anaemia control at time of routine vaccinations in Tanzanian infants: a randomised, placebo-controlled trial. Lancet. 2001;357:1471–1477. doi: 10.1016/S0140-6736(00)04643-2.
    1. Schellenberg D, Menendez C, Aponte JJ, Kahigwa E, Tanner M, Mshinda H, Alonso P. Intermittent preventive antimalarial treatment for Tanzanian infants: Follow-up to age 2 years of a randomised, placebo-controlled trial. Lancet. 2005;365:1481–1483. doi: 10.1016/S0140-6736(05)66418-5.
    1. Massaga JJ, Kitua AY, Lemnge MM, Akida JA, Malle LN, Ronn AM, Theander TG, Bygbjerg IC. Effect of intermittent treatment with amodiaquine on anaemia and malarial fevers in infants in Tanzania: a randomised placebo-controlled trial. Lancet. 2003;361:1853–1860. doi: 10.1016/S0140-6736(03)13504-0.
    1. Verhoef H, West CE, Nzyuko SM, de Vogel S, van der Valk R, Wanga MA, Kuijsten A, Veenemans J, Kok FJ. Intermittent administration of iron and sulfadoxine-pyrimethamine to control anaemia in Kenyan children: a randomised controlled trial. Lancet. 2002;360:908–914. doi: 10.1016/S0140-6736(02)11027-0.
    1. Desai MR, Mei JV, Kariuki SK, Wannemuehler KA, Phillips-Howard PA, Nahlen BL, Kager PA, Vulule JM, ter Kuile FO. Randomized, controlled trial of daily iron supplementation and intermittent sulfadoxine-pyrimethamine for the treatment of mild childhood anemia in western Kenya. J Infect Dis. 2003;187:658–666. doi: 10.1086/367986.
    1. Bloland PB, Boriga DA, Ruebush TK, McCormick JB, Roberts JM, Oloo AJ, Hawley W, Lal A, Nahlen B, Campbell CC. Longitudinal cohort study of the epidemiology of malaria infections in an area of intense malaria transmission II. Descriptive epidemiology of malaria infection and disease among children. Am J Trop Med Hyg. 1999;60:641–648.
    1. Menendez C, Kahigwa E, Hirt R, Vounatsou P, Aponte JJ, Font F, Acosta CJ, Schellenberg DM, Galindo CM, Kimario J, Urassa H, Brabin B, Smith TA, Kitua AY, Tanner M, Alonso PL. Randomised placebo-controlled trial of iron supplementation and malaria chemoprophylaxis for prevention of severe anaemia and malaria in Tanzanian infants. Lancet. 1997;350:844–850. doi: 10.1016/S0140-6736(97)04229-3.
    1. Greenwood BM, David PH, OtooForbes LN, Allen SJ, Alonso PL, Schellenberg JRA, Byass P, Hurwitz M, Menon A, Snow RW. Mortality and morbidity from malaria after stopping malaria chemoprophylaxis. Trans R Soc Trop Med Hyg. 1995;89:629–633. doi: 10.1016/0035-9203(95)90419-0.
    1. Saarinen M, Thoren E, Iyambo N, Carlstedt A, Shinyafa L, Fernanda M, Paajanen H, Paajanen K, Indongo I, Rombo L. Malaria prophylaxis with proguanil to Namibian refugee children in Angola. Trop Med Parasitol. 1988;39:40–42.
    1. Bjorkman A, Brohult J, Pehrson PO, Willcox M, Rombo L, Hedman P, Kollie E, Alestig K, Hanson A, Bengtsson E. Monthly antimalarial chemotherapy to children in a holoendemic area of Liberia. Ann Trop Med Parasitol. 1986;80:155–167.
    1. Oyediran AB, Tropley E, Osunkoya BO, Bamgboye A, Williams AIO, Ogunba EO. Severe morbidity among children in a trial of malaria chemoprophylaxis with pyrimethamine or chloroquine in Ibarapa, Nigeria. Afr J Med Sci. 1993;22:55–63.
    1. Greenwood BM, Bradley AK, Byass P, Greenwood AM, Snow RW, Hayes RJ, Njie ABH. Comparison of two strategies for control of malaria within a primary health-care programme in the Gambia. Lancet. 1988;1:1121–1127. doi: 10.1016/S0140-6736(88)91949-6.
    1. Geerligs PDP, Brabin BJ, Eggelte TA. Analysis of the effects of malaria chemoprophylaxis in children on haematological responses, morbidity and mortality. Bull World Health Organ. 2003;81:205–216.
    1. von Seidlein L, Greenwood BM. Mass administrations of antimalarial drugs. Trends in Parasitology. 2003;19:452–460. doi: 10.1016/j.pt.2003.08.003.
    1. von Seidlein L, Walraven G, Milligan PJM, Alexander N, Manneh F, Deen JL, Coleman R, Jawara M, Lindsay SW, Drakeley C, De Martin S, Olliaro P, Bennett S, van der Loeff MS, Okunoye K, Targett GAT, McAdam K, Doherty JF, Greenwood BM, Pinder M. The effect of mass administration of sulfadoxine-pyrimethamine combined with artesunate on malaria incidence: a double-blind, community-randomized, placebo-controlled trial in The Gambia. Trans R Soc Trop Med Hyg. 2003;97:217–225. doi: 10.1016/S0035-9203(03)90125-8.
    1. Coulibaly D, Diallo DA, Thera MA, Dicko A, Guindo AB, Kone AK, Cissoko Y, Coulibaly S, Djimde A, Lyke K, Doumbo OK, Plowe CV. Impact of preseason treatment on incidence of falciparum malaria and parasite density at a site for testing malaria vaccines in Bandiagara, Mali. Am J Trop Med Hyg. 2002;67:604–610.
    1. Omumbo JA, Guerra CA, Hay SI, Snow RW. The influence of urbanisation on measures of Plasmodium falciparum infection prevalence in East Africa. Acta Tropica. 2005;93:11–21. doi: 10.1016/j.actatropica.2004.08.010.
    1. Hay SI, Guerra CA, Tatem AJ, Atkinson PM, Snow RW. Urbanization, malaria transmission and disease burden in Africa. Nature Reviews Microbiology. 2005;3:81–90. doi: 10.1038/nrmicro1069.
    1. Wagner G, Koram K, McGuiness D, Bennett S, Nkrumah F, Riley E. High incidence of asymptomatic malaria infections in a birth cohort of children less than one year of age in Ghana, detected by multicopy gene polymerase chain reaction. Am J Trop Med Hyg. 1998;59:115–123.
    1. Cisse B, Sokhna C, Alexander N, Lines J, Trape J-F, Greenwood B. Intermittent preventive treatment (IPT) with artesunate plus sulfadoxine pyrimethamine to reduce under 5 malaria morbidiy in the Sahel. Am J Trop Med Hyg. 2004;71:1–79.
    1. Drakeley C, Schellenberg D, Kihonda J, Sousa CA, Arez AP, Lopes D, Lines J, Mshinda H, Lengeler C, Schellenberg JA, Tanner M, Alonso P. An estimation of the entomological inoculation rate for Ifakara: a semi-urban area in a region of intense malaria transmission in Tanzania. Trop Med Int Health. 2003;8:767–774. doi: 10.1046/j.1365-3156.2003.01100.x.
    1. Kun JFJ, Lehman LG, Lell B, Schmidt-Ott R, Kremsner PG. Low-dose treatment with sulfadoxine-pyrimethamine combinations selects for drug-resistant Plasmodium falciparum strains. Antimicrob Agents Chemother. 1999;43:2205–2208.
    1. Watkins WM, Mosobo M. Treatment of Plasmodium-falciparum Malaria with Pyrimethamine-Sulfadoxine – Selective Pressure for Resistance Is a Function of Long Elimination Half-Life. Trans R Soc Trop Med Hyg. 1993;87:75–78. doi: 10.1016/0035-9203(93)90431-O.
    1. Hastings IM. Modelling parasite drug resistance: lessons for management and control strategies. Trop Med Int Health. 2001;6:883–890. doi: 10.1046/j.1365-3156.2001.00800.x.
    1. Dicko A, Sagara S, Sissoko MS, Guindo O, Diallo ABI, Kone M, Thera MA, Sacko M, Doumbo OK. Impact of intermittent preventive treatment with sulfadoxine pyrimethamine targetting the transmission season on the incidence of clinical malaria in children of 6 months to 10 years in Kambila, Mali. Am J Trop Med Hyg. 2004;71:1–79.
    1. Beck HP, Felger I, Vounatsou P, Hirt R, Tanner M, Alonso P, Menendez C. The epidemiology of multiple Plasmodium falciparum infections – 8. Effect of iron supplementation and malaria prophylaxis in infants on Plasmodium falciparum genotypes and multiplicity of infection. Trans R Soc Trop Med Hyg. 1999;93 Suppl 1:41–45. doi: 10.1016/S0035-9203(99)90326-7.
    1. Ocana-Morgner C, Mota MM, Rodriguez A. Malaria blood stage suppression of liver stage immunity by dendritic cells. J Exp Med. 2003;197:143–151. doi: 10.1084/jem.20021072.
    1. Belnoue E, Costa FTM, Frankenberg T, Vigario AM, Voza T, Leroy N, Rodrigues MM, Landau I, Snounou G, Renia L. Protective T cell immunity against malaria liver stage after vaccination with live sporozoites under chloroquine treatment. J Immunol. 2004;172:2487–2495.
    1. Gill CJ, Sabin LL, Tham J, Hamer DH. Reconsidering empirical cotrimoxazole prophylaxis for infants exposed to HIV infection. Bull World Health Organ. 2004;82:290–297.
    1. Iyer JK, Milhous WK, Cortese JF, Kublin JG, Plowe CV. Plasmodium falciparum cross-resistance between trimethoprim and pyrimethamine. Lancet. 2001;358:1066–1067. doi: 10.1016/S0140-6736(01)06201-8.
    1. Rosen JB, Breman JG. Malaria intermittent preventive treatment in infants, chemoprophylaxis, and childhood vaccinations. Lancet. 2004;363:1386–1388. doi: 10.1016/S0140-6736(04)16052-2.

Source: PubMed

3
Abonnieren