58
Views
0
CrossRef citations to date
0
Altmetric
Rapid Communication

Malaria situation and elimination in Africa with specific focus on Zimbabwe: a systematic review

, &
Article: 2376945 | Received 21 Mar 2023, Accepted 02 Jul 2024, Published online: 16 Jul 2024

References

  • Abamecha, A.,Yilma, D.,Addisu, W.,El-Abid, H.,Ibenthal, A.,Noedl, H.,Yewhalaw, D.,Moumni, M., &Abdissa, A. (2020). Therapeutic efficacy of artemether-lumefantrine in the treatment of uncomplicated Plasmodium falciparum malaria in Chewaka District, Ethiopia. Malaria Journal, 19(240), 1–10. 32650784.
  • Abiodun, A., Andersen, H., Mamo, L. T., & Sisay, O. B. (2021). Vaccine manufacturing in Africa: what it takes and why it matters. Tony Blair Institute for Global Change.
  • African Union. (2021). World malaria report (2021), progress towards the African Union’s goal of eliminating malaria in Africa by 2030. African Union.
  • Ahorlu, C. S., Adongo, P., Koenker, H., Zigirumugabe, S., Sika-Bright, S., Koka, E., Tabong, P. T.-N., Piccinini, D., Segbaya, S., Olapeju, B., & Monroe, A. (2019). Understanding the gap between access and use: A qualitative study on barriers and facilitators to insecticide-treated net use in Ghana. Malaria Journal, 18(1), 417. https://doi.org/10.1186/s12936-019-3051-0
  • Allcock, S. H., Young, E. H., & Sandhu, M. S. (2018). A cross-sectional analysis of ITN and IRS coverage in Namibia in 2013. Malaria Journal, 17(1), 264. https://doi.org/10.1186/s12936-018-2417-z
  • Asingizwe, D., Poortvliet, P. M., Koenraadt, C. J., Van Vliet, A. J., Ingabire, C. M., Mutesa, L., & Leeuwis, C. (2019). Role of individual perceptions in the consistent use of malaria preventive measures: mixed methods evidence from rural Rwanda. Malaria Journal, 18(1), 270. https://doi.org/10.1186/s12936-019-2904-x
  • Balakrishnan, V. S. (2022). A new strategy is required for malaria elimination in Africa. The Lancet-Infectious Diseases, 22(2), 170–171. https://doi.org/10.1016/S1473-3099(22)00012-3
  • Batra, C. P., Adak, T., Sharma, V. P., & Mittal, P. K. (2001). Impact of urbanization on bionomics of An. culicifacies and An. stephensi in Delhi. Indian Journal of Malariology, 38(3–4), 61–75.
  • Beavogui, A. H., Camara, A., Delamou, A., Diallo, M. S., Doumbouya, A., Kourouma, K., Bouedouno, P., Guilavogui, T., Souza, S., Kelley, J., Talundzic, E., Fofana, A., & Plucinski, M. M. (2020). Efficacy and safety of artesunate – amodiaquine and artemether – lumefantrine and prevalence of molecular markers associated with resistance, Guinea: An open‑label two‑arm randomised controlled trial. Malaria Journal, 19(1), 223. https://doi.org/10.1186/s12936-020-03290-w
  • Bekono, B. D., Kang, F. N., Onguéné, P. A., Lifongo, L. L., Sippl, W., Fester, K., & Owono, L. C. O. (2020). The potential of anti‑malarial compounds derived from African medicinal plants : A review of pharmacological evaluations from 2013 to 2019. Malaria Journal, 19(1), 183. https://doi.org/10.1186/s12936-020-03231-7
  • Berthe, S., Harvey, S. A., Lynch, M., Koenker, H., Jumbe, V., Kaunda-Khangamwa, B., & Mathanga, D. P. (2019). Poverty and food security: Drivers of insecticide-treated mosquito net misuse in Malawi. Malaria Journal, 18(1), 320. https://doi.org/10.1186/s12936-019-2952-2
  • Bhagavathula, A. S., Elnour, A. A., & Shehab, A. (2016). Alternatives to currently used antimalarial drugs: in search of a magic bullet. Infectious Diseases of Poverty, 5(1), 103. https://doi.org/10.1186/s40249-016-0196-8
  • Bin Dajem, S. M., & Al-Qahtani, A. (2010). Analysis of gene mutations involved in chloroquine resistance in Plasmodium falciparum parasites isolated from patients in the southwest of Saudi Arabia. Annals of Saudi Medicine, 30(3), 187–192. https://doi.org/10.4103/0256-4947.62826
  • Carraz, M., Jossang, A., Franetich, J.-F., Siau, A., Ciceron, L., Hannoun, L., Sauerwein, R., Frappier, F., Rasoanaivo, P., Snounou, G., & Mazier, D. (2006). A plant-derived morphinan as a novel lead compound active against malaria liver stages. PLOS Medicine, 3(12), e513. https://doi.org/10.1371/journal.pmed.0030513
  • Casimiro, S., Coleman, M., Hemingway, J., & Sharp, B. (2006). Insecticide resistance in Anopheles arabiensis and Anopheles gambiae from Mozambique. Journal of Medical Entomology, 43(2), 276–282. https://doi.org/10.1093/jmedent/43.2.276
  • Chanda, E., Hemingway, J., Kleinschmidt, I., Rehman, A. M., Ramdeen, V., Phiri, F. N., Coetzer, S., Mthembu, D., Shinondo, C. J., Chizema-Kawesha, E., Kamuliwo, M., Mukonka, V., Baboo, K. S., & Coleman, M. (2011). Insecticide resistance and the future of malaria control in Zambia. PLOS One, 6(9), e24336. https://doi.org/10.1371/journal.pone.0024336
  • Chanda, J., Saili, K., Phiri, F., Stevenson, J. C., Mwenda, M., Chishimba, S., Mulube, C., Mambwe, B., Lungu, C., Earle, D., Bennett, A., Eisele, T. P., Kamuliwo, M., Steketee, R. W., Keating, J., Miller, J. M., & Sikaala, C. H. (2020). Pyrethroid and carbamate resistance in Anopheles funestus Giles along Lake Kariba in southern Zambia. The American Journal of Tropical Medicine and Hygiene, 103(2_Suppl), 90–97. https://doi.org/10.4269/ajtmh.19-0664
  • Chapu, G., & Mgocheki, N. (2017). A survey on traditional and modern prophylactic methods of malaria management in a resettlement area in the Southern Lowveld of Zimbabwe. International Journal of tropical disease & Health, 21(1), 1–17. https://doi.org/10.9734/IJTDH/2017/30433
  • Chinsembu, K. C. (2015). Plants as antimalarial agents in Sub-Saharan Africa. Acta Tropica, 152, 32–48. https://doi.org/10.1016/j.actatropica.2015.08.009
  • Choi, K. S., Christian, R., Nardini, L., Wood, O. R., Agubuzo, E., Muleba, M., Munyati, S., Makuwaza, A., Koekemoer, L. L., Brooke, B. D., Hunt, R. H., & Coetzee, M. (2014). Insecticide resistance and role in malaria transmission of Anopheles funestus populations from Zambia and Zimbabwe. Parasites & Vectors, 7(1), 464. https://doi.org/10.1186/s13071-014-0464-z
  • Cohen, M. K. (2024). World malaria day report (pp. 1–2). Centers for Disease Control and Prevention.
  • Coleman, M., Coleman, M., Mabuza, A. M., Kok, G., Coetzee, M., & Durrheim, D. N. (2009). Using the SaTScan method to detect local malaria clusters for guiding malaria control programmes. Malaria Journal, 8(1), 68. https://doi.org/10.1186/1475-2875-8-68
  • Conrad, M. D., LeClair, N., Arinaitwe, E., Wanzira, H., Kakuru, A., Bigira, V., Muhindo, M., Kamya, M. R., Tappero, J. W., Greenhouse, B., Dorsey, G., & Rosenthal, P. J. (2014). Comparative impacts over 5 years of artemisinin-based combination therapies on Plasmodium falciparum polymorphisms that modulate drug sensitivity in Ugandan children. The Journal of Infectious Diseases, 210(3), 344–353. https://doi.org/10.1093/infdis/jiu141
  • Conrad, M. D., & Rosenthal, P. J. (2019). Antimalarial drug resistance in Africa: The calm before the storm? The Lancet. Infectious Diseases, 19(10), e338–e351. https://doi.org/10.1016/S1473-3099(19)30261-0
  • Cuamba, N., Morgan, J. C., Irving, H., Steven, A., & Wondji, C. S. (2010). High level of pyrethroid resistance in an Anopheles funestus population of the Chokwe district in Mozambique. PLOS One, 5(6), e11010. https://doi.org/10.1371/journal.pone.0011010
  • Datoo, M. S., Dicko, A., Tinto, H., Ouédraogo, J.-B., Hamaluba, M., Olotu, A., Beaumont, E., Ramos Lopez, F., Natama, H. M., Weston, S., Chemba, M., Compaore, Y. D., Issiaka, D., Salou, D., Some, A. M., Omenda, S., Lawrie, A., Bejon, P., Rao, H., … Hill, A. V. S, R21/Matrix-M Phase 3 Trial Group. (2024). Safety and efficacy of malaria vaccine candidate R21/Matrix-M in African children: A multicentre, double-blind, randomised, phase 3 trial. Lancet, 403(10426), 533–544. https://doi.org/10.1016/S0140-6736(23)02511-4
  • Datoo, M. S., Natama, H. M., Somé, A., Bellamy, D., Traoré, O., Rouamba, T., Tahita, M. C., Ido, N. F. A., Yameogo, P., Valia, D., Millogo, A., Ouedraogo, F., Soma, R., Sawadogo, S., Sorgho, F., Derra, K., Rouamba, E., Ramos-Lopez, F., Cairns, M., … Tinto, H. (2022). Efficacy and immunogenicity of R21/Matrix-M vaccine against clinical malaria after 2 years’ follow-up in children in Burkina Faso: a phase 1/2b randomised controlled trial. The Lancet. Infectious Diseases, 22(12), 1728–1736. https://doi.org/10.1016/S1473-3099(22)00442-X
  • Datoo, M. S., Natama, M. H., Somé, A., Traoré, O., Rouamba, T., Bellamy, D., Yameogo, P., Valia, D., Tegneri, M., Ouedraogo, F., Soma, R., Sawadogo, S., Sorgho, F., Derra, K., Rouamba, E., Orindi, B., Ramos Lopez, F., Flaxman, A., Cappuccini, F., … Tinto, H. (2021). Efficacy of a low-dose candidate malaria vaccine, R21 in adjuvant Matrix-M, with seasonal administration to children in Burkina Faso: A randomised controlled trial. Lancet, 397(10287), 1809–1818. https://doi.org/10.1016/S0140-6736(21)00943-0
  • Desai, M., Gutman, J., Taylor, S. M., Wiegand, R. E., Khairallah, C., Kayentao, K., Ouma, P., Coulibaly, S. O., Kalilani, L., Mace, K. E., Arinaitwe, E., Mathanga, D. P., Doumbo, O., Otieno, K., Edgar, D., Chaluluka, E., Kamuliwo, M., Ades, V., Skarbinski, J., … Ter Kuile, F. O. (2015). Impact of sulfadoxine-pyrimethamine resistance on effectiveness of intermittent preventive therapy for malaria in pregnancy at clearing infections and preventing low birth weight. Clinical Infectious Diseases, 62(3), 323–333. https://doi.org/10.1093/cid/civ881
  • Desai, M., Hill, J., Fernandes, S., Walker, P., Pell, C., Gutman, J., Kayentao, K., Gonzalez, R., Webster, J., Greenwood, B., Cot, M., & Ter Kuile, F. O. (2018). Prevention of malaria in pregnancy. The Lancet-Infectious Diseases, 18(4), e119–e132. https://doi.org/10.1016/S1473-3099(18)30064-1
  • Diema, K. K., Dodam, K. K., Aarah-Bapuah, M., & Asibi, A. J. (2017). Barriers to sustained use of the insecticide treated bed net in the upper east region of Ghana. International Journal of Community Medicine and Public Health, 4(2), 500–505. https://doi.org/10.18203/2394-6040.ijcmph20170280
  • Dimala, C. A., Kika, B. T., Kadia, B. M., & Blencowe, H. (2018). Current challenges and proposed solutions to the effective implementation of the RTS, S/AS01 malaria vaccine program in sub-Saharan Africa: A systematic review. PLOS One, 13(12), e0209744. https://doi.org/10.1371/journal.pone.0209744
  • Dube, B., Mberikunashe, J., Dhliwayo, P., Tangwena, A., Shambira, G., Chimusoro, A., Madinga, M., & Gambinga, B. (2019). How far is the journey before malaria is knocked out in Zimbabwe: Results of the malaria indicator survey 2016. Malaria Journal, 18(1), 196. https://doi.org/10.1186/s12936-019-2823-x
  • Dwomoh, D., Adu, B., Dodoo, D., Theisen, M., Iddi, S., & Gerds, T. A. (2020). Evaluating the predictive performance of malaria antibodies and FCGR3B gene polymorphisms on Plasmodium falciparum infection outcome: A prospective cohort study. Malaria Journal, 19(1), 307. https://doi.org/10.1186/s12936-020-03381-8
  • Ehrlich, H. Y., Jones, J., & Parikh, S. (2020). Molecular surveillance of antimalarial partner drug resistance in sub-Saharan Africa: A spatial-temporal evidence mapping study. The Lancet-Microbe, 1(5), e209–e217. https://doi.org/10.1016/s2666-5247(20)30094-x
  • Ekström, A. M., Tomson, G., Wanyenze, R. K., Bhutta, Z. A., Kyobutungi, C., Binagwaho, A., & Ottersen, O. P. (2021). Addressing production gaps for vaccines in African countries. Bulletin of the World Health Organization, 99(12), 910–912. https://doi.org/10.2471/BLT.21.287381
  • Esu, E.,Effa, E. E.,Opie, O. N.,Uwaoma, A., &Meremikwu, M. M. (2014). Artemether for severe malaria. The Cochrane Database of Systematic Reviews, 2014(9), CD010678. https://doi.org/10.1002/14651858.
  • Flegg, J. A., Metcalf, C. J. E., Gharbi, M., Venkatesan, M., Shewchuk, T., Hopkins Sibley, C., & Guerin, P. J. (2013). Trends in antimalarial drug use in Africa. The American Journal of Tropical Medicine and Hygiene, 89(5), 857–865. https://doi.org/10.4269/ajtmh.13-0129
  • Foy, B. D., Some, A., Magalhaes, T., Gray, L., Rao, S., Sougue, E., Jackson, C. L., Kittelson, J., Slater, H. C., Bousema, T., Da, O., Coulidiaty, A. G. V., Colt, M., Wade, M., Richards, K., Some, A. F., Dabire, R. K., & Parikh, S. (2023). Repeat ivermectin mass drug administrations for malaria control II: Protocol for a double-blind, cluster-randomized, placebo-controlled trial for the integrated control of malaria. JMIR Research Protocols, 12(1), e41197. https://doi.org/10.2196/41197
  • Frosch, A. E., Venkatesan, M., & Laufer, M. K. (2011). Patterns of chloroquine use and resistance in sub-Saharan Africa: A systematic review of household survey and molecular data. Malaria Journal, 10(1), 116. https://doi.org/10.1186/1475-2875-10-116
  • Gao, L., Shi, Q., Liu, Z., Li, Z., & Dong, X. (2023). Impact of the COVID-19 pandemic on malaria control in Africa: A preliminary analysis. Tropical Medicine and Infectious Disease, 8(1), 67. https://doi.org/10.3390/tropicalmed8010067
  • Gavi, S., Tapera, O., Mberikunashe, J., & Kanyangarara, M. (2021). Malaria incidence and mortality in Zimbabwe during the COVID-19 pandemic: Analysis of routine surveillance data. Malaria Journal, 20(1), 233. https://doi.org/10.1186/s12936-021-03770-7
  • Getawen, S. K., Ashine, T., Massebo, F., Woldeyes, D., & Lindtjørn, B. (2018). Exploring the impact of house screening intervention on entomological indices and incidence of malaria in Arba Minch town, southwest Ethiopia: A randomized control trial. Acta Tropica, 181, 84–94. https://doi.org/10.1016/j.actatropica.2018.02.009
  • Gimnig, J. E., & Slutsker, L. (2009). House screening for malaria control. Lancet, 374(9694), 954–955. https://doi.org/10.1016/S0140-6736(09)61078-3
  • Global Technical Strategy for Malaria 2016–2030. (2016). World Health Organisation.
  • Gogue, C., Wagman, J., Tynuv, K., Saibu, A., Yihdego, Y., Malm, K., Mohamed, W., Akplu, W., Tagoe, T., Ofosu, A., Williams, I., Asiedu, S., Richardson, J., Fornadel, C., Slutsker, L., & Robertson, M. (2020). An observational analysis of the impact of indoor residual spraying in Northern, Upper East, and Upper West Regions of Ghana : 2014 Through 2017. Malaria Journal, 19(1), 242. https://doi.org/10.1186/s12936-020-03318-1
  • González, R., Manun’Ebo, M. F., Meremikwu, M., Rabeza, V. R., Sacoor, C., Figueroa-Romero, A., Arikpo, I., Macete, E., Mbombo Ndombe, D., Ramananjato, R., LIach, M., Pons-Duran, C., Sanz, S., Ramírez, M., Cirera, L., Maly, C., Roman, E., Pagnoni, F., & Menéndez, C. (2023). The impact of community delivery of intermittent preventive treatment of malaria in pregnancy on its coverage in four sub-Saharan African countries (Democratic Republic of the Congo, Madagascar, Mozambique, and Nigeria): A quasi-experimental multicentre evaluation. The Lancet-Global Health, 11(4), e566–e574. https://doi.org/10.1016/S2214-109X(23)00051-7
  • Green, C. A. (1981). Malaria epidemiology and anopheline cytogenetics. In R. Pal, J. B. Kitzmiller, & T. Kanda Kodansha (Eds.), Cytogenetics and genetics of vectors: Proceedings of a symposium of the XVIth International Congress of Entomology (p. c1981). Elsevier Biomedical Press.
  • Gunda, R., Chimbari, M. J., & Mukaratirwa, S. (2016). Assessment of burden of malaria in gwanda District, Zimbabwe, using the disability adjusted life years. International Journal of Environmental Research and Public Health, 13(2), 244. https://doi.org/10.3390/ijerph13020244
  • Gutman, J. R., Stephens, D. K., Tiendrebeogo, J., Badolo, O., Dodo, M., Burke, D., Williamson, J., Vibbert, K., Youll, S. J., Savadogo, Y., & Brieger, W. R. (2020). A cluster randomized trial of delivery of intermittent preventive treatment of malaria in pregnancy at the community level in Burkina Faso. Malaria Journal, 19(1), 282. https://doi.org/10.1186/s12936-020-03356-9
  • Gwitira, I., Mukonoweshuro, M., Mapako, G., Shekede, M. D., Chirenda, J., & Mberikunashe, J. (2020). Spatial and spatio-temporal analysis of malaria cases in Zimbabwe. Infectious Diseases of Poverty, 9(1), 146. https://doi.org/10.1186/s40249-020-00764-6
  • Gwitira, I., Murwira, A., Mberikunashe, J., & Masocha, M. (2018). Spatial overlaps in the distribution of HIV/AIDS and malaria in Zimbabwe. BMC Infectious Diseases, 18(1), 598. https://doi.org/10.1186/s12879-018-3513-y
  • Hemming-Schroeder, E., Umukoro, E., Lo, E., Fung, B., Tomás-Domingo, P., Zhou, G., Zhong, D., Dixit, A., Atieli, H., Githeko, A., Vardo-Zalik, A., & Yan, G. (2018). Impacts of antimalarial drugs on Plasmodium falciparum drug resistance markers, Western Kenya, 2003–2015. The American Journal of Tropical Medicine and Hygiene, 98(3), 692–699. https://doi.org/10.4269/ajtmh.17-0763
  • Hiben, M. G., Sibhat, G. G., Fanta, B. S., Gebrezgi, H. D., & Tesema, S. B. (2016). Evaluation of Senna singueana leaf extract as an alternative or adjuvant therapy for malaria. Journal of Traditional and Complementary Medicine, 6(1), 112–117. https://doi.org/10.1016/j.jtcme.2014.11.014
  • Ippolito, M. M., Gebhardt, M. E., Ferriss, E., Schue, J. L., Kobayashi, T., Chaponda, M., Kabuya, J.-B., Muleba, M., Mburu, M., Matoba, J., Musonda, M., Katowa, B., Lubinda, M., Hamapumbu, H., Simubali, L., Mudenda, T., Wesolowski, A., Shields, T. M., Hackman, A., … Moss, W. J, Southern and Central Africa International Center of Excellence for Malaria Research. (2022). Scientific findings of the Southern and Central Africa International Center of Excellence for malaria research: ten years of malaria control impact assessments in hypo-, meso-, and holoendemic transmission zones in Zambia and Zimbabwe. The American Journal of Tropical Medicine and Hygiene, 107(4_Suppl), 55–67. https://doi.org/10.4269/ajtmh.21-1287
  • Kanyangarara, M., Hamapumbu, H., Mamini, E., Lupiya, J., Stevenson, J. C., Mharakurwa, S., Chaponda, M., Thuma, P. E., Gwanzura, L., Munyati, S., Mulenga, M., Norris, D. E., & Moss, W. J, Southern Africa International Centers of Excellence for Malaria Research. (2018). Malaria knowledge and bed net use in three transmission settings in southern Africa. Malaria Journal, 17(1), 41. https://doi.org/10.1186/s12936-018-2178-8
  • Kanyangarara, M., Mamini, E., Mharakurwa, S., Munyati, S., Gwanzura, L., Kobayashi, T., Shields, T., Mullany, L. C., Mutambu, S., Mason, P. R., Curriero, F. C., & Moss, W. J, Southern Africa International Centers of Excellence for Malaria Research. (2016). Individual-and household-level risk factors associated with malaria in Mutasa District, Zimbabwe: A serial cross-sectional study. The American Journal of Tropical Medicine and Hygiene, 95(1), 133–140. https://doi.org/10.4269/ajtmh.15-0847
  • Kanyangarara, M., Mamini, E., Mharakurwa, S., Munyati, S., Gwanzura, L., Kobayashi, T., Shields, T., Mullany, L. C., Mutambu, S., Mason, P. R., Curriero, F. C., … & Moss, W. J, Southern Africa International Centers of Excellence for Malaria Research. (2016). Reduction in malaria incidence following indoor residual spraying with Actellic 300 CS in a setting with pyrethroid resistance: Mutasa District, Zimbabwe. PLOS One, 11(3), e0151971. https://doi.org/10.1371/journal.pone.0151971
  • Kayentao, K., Garner, P., van Eijk, A. M., Naidoo, I., Roper, C., Mulokozi, A., MacArthur, J. R., Luntamo, M., Ashorn, P., Doumbo, O. K., & ter Kuile, F. O. (2013). Intermittent preventive therapy for malaria during pregnancy using 2 vs. 3 or more doses of sulfadoxine-pyrimethamine and risk of low birth weight in Africa: Systematic review and meta-analysis. JAMA, 309(6), 594–604. https://doi.org/10.1001/jama.2012.216231
  • Kemibala, E. E., Neto, A. M., Saroli, J., Silva, R., Philbert, A., Ng, K., Foster, W. A., Dekker, T., & Mboera, L. E. G. (2020). Is Anopheles gambiae attraction to floral and human skin–based odours and their combination modulated by previous blood meal experience ? Malaria Journal, 19(1), 318. https://doi.org/10.1186/s12936-020-03395-2
  • Kesteman, T., Randrianarivelojosia, M., & Rogier, C. (2017). The protective effectiveness of control interventions for malaria prevention: A systematic review of the literature. F1000Research, 6, 1932. https://doi.org/10.12688/f1000research.12952.1
  • Kgoroebutswe, T. K., Makate, N., Fillinger, U., Mpho, M., Segoea, G., Sangoro, P. O., Mutero, C. M., Chanda, E., Ntebela, D., Mogopa, M., Mosweunyane, T., & Nkya, T. E. (2020). Vector control for malaria elimination in Botswana : Progress, gaps and opportunities. Malaria Journal, 19(1), 301. https://doi.org/10.1186/s12936-020-03375-6
  • Klooster, V., Jong, D., Ngarivhume, T., & Van, C. I. E. A. (2015). UvA-DARE (Digital Academic Repository) medicinal plants used by traditional healers for the treatment of malaria in the Chipinge district in Zimbabwe Medicinal plants used by traditional healers for the treatment of malaria in the Chipinge district in Zimbabwe. Journal of Ethnopharmacology, 159, 224–237. https://doi.org/10.1016/j.jep.2014.11.011
  • Knox, T. B., Juma, E. O., Ochomo, E. O., Pates Jamet, H., Ndungo, L., Chege, P., Bayoh, N. M., N’Guessan, R., Christian, R. N., Hunt, R. H., & Coetzee, M. (2014). An online tool for mapping insecticide resistance in major Anopheles vectors of human malaria parasites and review of resistance status for the Afrotropical region. Parasites & Vectors, 7(1), 76. https://doi.org/10.1186/1756-3305-7-76
  • Kolawole, E. O., Ayeni, E. T., Abolade, S. A., Ugwu, S. E., Awoyinka, T. B., Ofeh, A. S., & Okolo, B. O. (2023). Malaria endemicity in Sub-Saharan Africa: Past and present issues in public health. Microbes and Infectious Diseases, 4(1), 242–251.
  • Kremsner, P. G., Adegnika, A. A., Hounkpatin, A. B., Zinsou, J. F., Taylor, T. E., Chimalizeni, Y., Liomba, A., Kombila, M., Bouyou-Akotet, M. K., Mawili Mboumba, D. P., Agbenyega, T., Ansong, D., Sylverken, J., Ogutu, B. R., Otieno, G. A., Wangwe, A., Bojang, K. A., Okomo, U., Sanya-Isijola, F., … Krishna, S. (2016). Intramuscular artesunate for severe malaria in African children: A multicenter randomized controlled trial. PLOS Medicine, 13(1), e1001938. https://doi.org/10.1371/journal.pmed.1001938
  • Krettli, A. U., Andrade-Neto, V. F., Brandão, G. L., & Ferrari, W. M. S. (2001). The search for new antimalarial drugs from plants used to treat fever and malaria or plants randomly selected: A Review. Mem Inst Oswald Cuz, Rio de Janeiro, 96(November 2001), 1033–1042.
  • Lima, R. B. S., Rocha, L. F., Melo, M. R. S., Costa, J. S., Picanço, N. S., Lima, E. S., Vasconcellos, M. C., Boleti, A. P. A., Santos, J. M. P., Amorim, R. C. N., Chaves, F. C. M., Coutinho, J. P., Tadei, W. P., Krettli, A. U., & Pohlit, A. M. (2015). In vitro and in vivo anti‑malarial activity of plants from the Brazilian Amazon. Malaria Journal, 14, 508. https://doi.org/10.1186/s12936-015-0999-2
  • Lu, F., Zhang, M., Culleton, R. L., Xu, S., Tang, J., Zhou, H., Zhu, G., Gu, Y., Zhang, C., Liu, Y., Wang, W., Cao, Y., Li, J., He, X., Cao, J., & Gao, Q. (2017). Return of chloroquine sensitivity to Africa? Surveillance of African Plasmodium falciparum chloroquine resistance through malaria imported to China. Parasites & Vectors, 10(1), 355. https://doi.org/10.1186/s13071-017-2298-y
  • Lukwa, N., Sande, S., Makuwaza, A., Chiwade, T., Netsa, M., Asamoa, K., Vazquez-Prokopec, G., Reithinger, R., & Williams, J. (2014). Nationwide assessment of insecticide susceptibility in Anopheles gambiae populations from Zimbabwe. Malaria Journal, 13(1)2014, 408. https://doi.org/10.1186/1475-2875-13-408
  • Mabaso, M. L. H., Sharp, B., & Lengeler, C. (2004). Historical review of malaria control in southern Africa with emphasis on the use of indoor residual house-spraying. Tropical Medicine & International Health, 9(8), 846–856. https://doi.org/10.1111/j.1365-3156.2004.01263.x
  • Magaço, A., Botão, C., Nhassengo, P., Saide, M., Ubisse, A., Chicumbe, S., & Zulliger, R. (2019). Community knowledge and acceptance of indoor residual spraying for malaria prevention in Mozambique: A qualitative study. Malaria Journal, 18(1), 27. https://doi.org/10.1186/s12936-019-2653-x
  • Mahajan, N. N., Gajbhiye, R. K., Bahirat, S., Lokhande, P. D., Mathe, A., Rathi, S., Warty, N., Mahajan, K. N., Srivastava, V., Kuppusamy, P., & Mohite, S. C. (2021). Co-infection of malaria and early clearance of SARS-CoV-2 in healthcare workers. Journal of Medical Virology, 93(4), 2431–2438. https://doi.org/10.1002/jmv.26760
  • Makono, R., & Sibanda, S. (1999). Review of the prevalence of malaria in Zimbabwe with specific reference to parasite drug resistance (1984–1996). Transactions of the Royal Society of Tropical Medicine and Hygiene, 93(5), 449–452. https://doi.org/10.1016/s0035-9203(99)90331-0
  • Manyangadze, T., Chimbari, M. J., Macherera, M., & Mukaratirwa, S. (2017). Micro ‑ spatial distribution of malaria cases and control strategies at ward level in Gwanda district, Matabeleland South, Zimbabwe. Malaria Journal, 16(1), 476. https://doi.org/10.1186/s12936-017-2116-1
  • Masalu, J. P., Finda, M., Killeen, G. F., Ngowo, H. S., Pinda, P. G., & Okumu, F. O. (2020). Creating mosquito-free outdoor spaces using transfluthrin-treated chairs and ribbons. Malaria Journal, 19(1), 109. https://doi.org/10.1186/s12936-020-03180-1
  • Masendu, H. T., Hunt, R. H., Koekemoer, L. L., Brooke, B. D., Govere, J., & Coetzee, M. (2005). Spatial and temporal distributions and insecticide susceptibility of malaria vectors in Zimbabwe. African Entomology, 13, 25–34.
  • Maxmen, A. (2021). Scientists hail historic malaria vaccine approval-but point to challenges ahead. Nature. Available online. https://doi.org/10.1038/d41586-021-02755-5
  • Mbacham, W. F., Ayong, L., Guewo-Fokeng, M., & Makoge, V. (2019). Current situation of malaria in Africa. Methods in Molecular Biology, 2013, 29–44. https://doi.org/10.1007/978-1-4939-9550-9_2
  • Mhamilawa, L. E., Ngasala, B., Morris, U., Kitabi, E. N., Barnes, R., Soe, A. P., Mmbando, B. P., Björkman, A., & Mårtensson, A. (2020). Parasite clearance, cure rate, post‑treatment prophylaxis and safety of standard 3-day versus an extended 6-day treatment of artemether – lumefantrine and a single low-dose primaquine for uncomplicated Plasmodium falciparum malaria in Bagamoyo district, Tanzania : A randomized controlled trial. Malaria Journal, 19(1), 216. https://doi.org/10.1186/s12936-020-03287-5
  • Mharakurwa, S., Matsena-Zingoni, Z., Mudare, N., Matimba, C., Gara, T. X., Makuwaza, A., Maponga, G., Munyati, S., Gwanzura, L., Mutambu, S. L., Mason, P., Kobayashi, T., Midzi, N., Moss, W. J., & Ippolito, M. M. (2021). Steep rebound of chloroquine-sensitive Plasmodium falciparum in Zimbabwe. The Journal of Infectious Diseases, 223(2), 306–309. https://doi.org/10.1093/infdis/jiaa368
  • Mharakurwa, S., & Mugochi, T. (1994). Chloroquine-resistant falciparum malaria in an area of rising endemicity in Zimbabwe. The Journal of Tropical Medicine and Hygiene, 97(1), 39–45.
  • Mharakurwa, S., Mutambu, S. L., Mberikunashe, J., Thuma, P. E., Moss, W. J., & Mason, P. R., Southern Africa ICEMR Team. (2013). Changes in the burden of malaria following scale up of malaria control interventions in Mutasa District, Zimbabwe. Malaria Journal, 12(1), 223. https://doi.org/10.1186/1475-2875-12-223
  • Ministry of Health and Child Welfare. (2007). National malaria control programme strategy. Zimbabwe: Ministry of Health and Child Welfare.
  • Mlambo, G., Sullivan, D., Mutambu, S. L., Soko, W., Mbedzi, J., Chivenga, J., Gemperli, A., & Kumar, N. (2007). High prevalence of molecular markers for resistance to chloroquine and pyrimethamine in Plasmodium falciparum from Zimbabwe. Parasitology Research, 101(4), 1147–1151. https://doi.org/10.1007/s00436-007-0597-5
  • Moon, T. D., Hayes, C. B., Blevins, M., Lopez, M. L., Green, A. F., González-Calvo, L., …& Olupona, O, Ogumaniha-SCIP Zambézia Consortium. (2016). Factors associated with the use of mosquito bed nets: Results from two cross-sectional household surveys in Zambézia Province, Mozambique. Malaria Journal, 15(1), 196. https://doi.org/10.1186/s12936-016-1250-5
  • Mosha, J. F., Sturrock, H. J. W., Greenwood, B., Sutherland, C. J., Gadalla, N. B., Atwal, S., Hemelaar, S., Brown, J. M., Drakeley, C., Kibiki, G., Bousema, T., Chandramohan, D., & Gosling, R. D. (2014). Hot spot or not: A comparison of spatial statistical methods to predict prospective malaria infections. Malaria Journal, 13(1), 53. https://doi.org/10.1186/1475-2875-13-53
  • Mugwagwa, N., Mberikunashe, J., Gombe, N. T., Tshimanga, M., Bangure, D., & Mungati, M. (2015). Honde valley, Mutasa district, Zimbabwe, 2014 : Factors associated with malaria infection in Honde valley, Mutasa district, Zimbabwe, 2014 : A case control study. BMC Research Notes, 8. (1), 829. https://doi.org/10.1186/s13104-015-1831-3
  • Mulaw, T., Wubetu, M., Dessie, B., Demeke, G., & Molla, Y. (2019). Evaluation of antimalarial activity of the 80% methanolic stem bark extract of Combretum molle against Plasmodium berghei in MICE. Journal of Evidence-Based Integrative Medicine, 24, 2515690X19890866. https://doi.org/10.1177/2515690X19890866
  • Mumtaz, H., Nadeem, A., Bilal, W., Ansar, F., Saleem, S., Khan, Q. A., Tango, T., Farkouh, C., Belay, N. F., Verma, R., Farkouh, M., & Saqib, M. (2023). Acceptance, availability, and feasibility of RTS, S/AS01 malaria vaccine: A review. Immunity, Inflammation and Disease, 11(6), e899. https://doi.org/10.1002/iid3.899
  • Mundagowa, P. T., & Chimberengwa, P. T. (2020). Malaria outbreak investigation in a rural area south of Zimbabwe : A case–control study. Malaria Journal, 19(1), 197. 10. https://doi.org/10.1186/s12936-020-03270-0
  • Munhenga, G., Masendu, H. T., Brooke, B. D., Hunt, R. H., & Koekemoer, L. K. (2008). Pyrethroid resistance in the major malaria vector Anopheles arabiensis from Gwave, a malaria-endemic area in Zimbabwe. Malaria Journal, 7(1), 247. https://doi.org/10.1186/1475-2875-7-247
  • Muthaura, C. N., Keriko, J. M., Derese, S., Yenesew, A., & Rukunga, G. M. (2011). Investigation of some medicinal plants traditionally used for treatment of malaria in Kenya as potential sources of antimalarial drugs. Experimental Parasitology, 127(3), 609–626. https://doi.org/10.1016/j.exppara.2010.11.004
  • Mwendera, C., de Jager, C., Longwe, H., Phiri, K., Hongoro, C., & Mutero, C. M. (2016). Malaria research and its influence on anti-malarial drug policy in Malawi: A case study. Health Research Policy and Systems, 14(1), 41. https://doi.org/10.1186/s12961-016-0108-1
  • Nalinya, S., Musoke, D., & Deane, K. (2022). Malaria prevention interventions beyond long-lasting insecticidal nets and indoor residual spraying in low-and middle-income countries: A scoping review. Malaria Journal, 21(1), 31. https://doi.org/10.1186/s12936-022-04052-6
  • Ngarivhume, T.,Van’t Klooster, C. I. E. A.,De Jong, J. T. V. M., &Van Der Westhuizen, J. H. (2015). Medicinal plants used by traditional healers for the treatment of malaria in the Chipinge district in Zimbabwe. Journal of Ethnopharmacology, 159, 224–237. https://doi.org/10.1016/j.jep.2014.11.011 PMID: 25449454
  • Nhama, A., Varo, R., & Bassat, Q. (2020). Highlighting the burden of malarial infection and disease in the neonatal period: Making sense of different concepts. Malaria Journal, 19(1), 311. https://doi.org/10.1186/s12936-020-03394-3
  • Nkya, T. E., Fillinger, U., Sangoro, O. P., Marubu, R., Chanda, E., & Mutero, C. M. (2022). Six decades of malaria vector control in southern Africa: A review of the entomological evidence-base. Malaria Journal, 21(1), 279. https://doi.org/10.1186/s12936-022-04292-6
  • Odufuwa, O. G., Ross, A., Mlacha, Y. P., Juma, O., Mmbaga, S., Msellemu, D., & Moore, S. (2020). Household factors associated with access to insecticide‑treated nets and house modification in Bagamoyo and Ulanga districts, Tanzania. Malaria Journal, 19(1), 220. https://doi.org/10.1186/s12936-020-03303-8
  • Okereke, M., Mashavakure, H. M., & Abdulwasiu, M. A. (2022). An evaluation of local pharmaceutical manufacturing in Zimbabwe: How prepared is Zimbabwe to produce COVID-19 vaccines? Innovations in Pharmacy, 13(1), 10. https://doi.org/10.24926/iip.v13i1.4656
  • Okokon, J. E., Okokon, P. J., & Sahal, D. (2010). In vitro antiplasmodial activity of some medicinal plants from Nigeria. International Journal of Herbal Medicine, 5, 102–109.
  • Oladipo, H. J., Tajudeen, Y. A., Oladunjoye, I. O., Yusuff, S. I., Yusuf, R. O., Oluwaseyi, E. M., AbdulBasit, M. O., Adebisi, Y. A., & El-Sherbini, M. S. (2022). Increasing challenges of malaria control in sub-Saharan Africa: Priorities for public health research and policymakers. Annals of Medicine and Surgery (2012), 81, 104366. https://doi.org/10.1016/j.amsu.2022.104366
  • Olapeju, B., Choiriyyah, I., Lynch, M., Acosta, A., Blaufuss, S., Filemyr, E., Harig, H., Monroe, A., Selby, R. A., Kilian, A., & Koenker, H. (2018). Age and gender trends in insecticide-treated net use in sub-Saharan Africa: A multi-country analysis. Malaria Journal, 17(1), 423. https://doi.org/10.1186/s12936-018-2575-z
  • Park, J., Kang, S., Seok, D., Baek, Y. J., An, S. Y., Lee, J., Jun, A., & Kim, S.-Y. (2023). Barriers against and strategies for malaria control during the COVID-19 pandemic in low-and middle-income countries: A systematic review. Malaria Journal, 22(1), 41. https://doi.org/10.1186/s12936-023-04452-2
  • Pates, H., & Curtis, C. (2005). Mosquito behaviour and vector control. Annual Review of Entomology, 50(1), 53–70. https://doi.org/10.1146/annurev.ento.50.071803.130439
  • Patlolla, A. K., Smith, Z., & Tchounwou, P. (2022). Indirect impacts of COVID-19 on the environment: A Global Review. International Journal of Biomedical and Clinical Analysis, 2(1), 9. https://doi.org/10.61797/ijbca.v2i1.160
  • PMI. (2014). Africa IRS (AIRS). Project indoor residual spraying (IRS 2) task order four. In USAID (Eds.). 2013 Zimbabwe end-of-spray report 2014. PMI|Africa IRS (AIRS) Project Indoor Residual Spraying (IRS 2) Task Order Four, ABT Associates Inc.
  • Pornputtapong, N., Suriyapakorn, B., Satayamapakorn, A., Larpadisorn, K., Janviriyakul, P., & Khemawoot, P. (2020). In silico analysis for factors affecting anti‑malarial penetration into red blood cells. Malaria Journal, 19(1), 215. https://doi.org/10.1186/s12936-020-03280-y
  • Premaratne, R., Wickremasinghe, R., Ranaweera, D., Gunasekera, W. M. K. T. d A. W., Hevawitharana, M., Pieris, L., Fernando, D., & Mendis, K. (2019). Technical and operational underpinnings of malaria elimination from Sri Lanka. Malaria Journal, 18(1), 256. https://doi.org/10.1186/s12936-019-2886-8
  • Pryce, J., Richardson, M., & Lengeler, C, Cochrane Infectious Diseases Group. (2018). Insecticide-treated nets for ­preventing malaria. Cochrane Database of Systematic Reviews, 11, 1–68. https://doi.org/10.1002/14651858.CD000363.pub3
  • Quanquin, N. M., Barres, L. G., Aliyari, S. R., Day, N. T., Gerami, H., Fisher, S. J., Kakuru, A., Kamya, M. R., Havlir, D. V., Feeney, M., Dorsey, G., Cheng, G., & Gaw, S. L. (2020). Gravidity – dependent associations between interferon response and birth weight in placental malaria. Malaria Journal, 19(1), 280. https://doi.org/10.1186/s12936-020-03351-0
  • Rek, J. C., Alegana, V., Arinaitwe, E., Cameron, E., Kamya, M. R., Katureebe, A., Lindsay, S. W., Kilama, M., Staedke, S. G., Todd, J., Dorsey, G., & Tusting, L. S. (2018). Rapid improvements to rural Ugandan housing and their association with malaria from intense to reduced transmission: A cohort study. The Lancet. Planetary Health, 2(2), e83–e94. https://doi.org/10.1016/S2542-5196(18)30010-X
  • Roux, A. T., Maharaj, L., Oyegoke, O., Akoniyon, O. P., Adeleke, M. A., Maharaj, R., & Okpeku, M. (2021). Chloroquine and sulfadoxine–pyrimethamine resistance in Sub-Saharan Africa—a review. Frontiers in Genetics, 12, 668574. https://doi.org/10.3389/fgene.2021.668574
  • RTS. S Clinical Trials Partnership. (2015). Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: Final results of a phase 3, individually randomised, controlled trial. Lancet, 386(9988), 31–45. https://doi.org/10.1016/S0140-6736(15)60721-8
  • RTS. S Clinical Trials Partnership. (2014). Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: A phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLOS Medicine, 11(7), e1001685. https://doi.org/10.1371/journal.pmed.1001685
  • Russell, C. L., Sallau, A., Emukah, E., Graves, P. M., Noland, G. S., Ngondi, J. M., Ozaki, M., Nwankwo, L., Miri, E., McFarland, D. A., Richards, F. O., & Patterson, A. E. (2015). Determinants of bed net use in Southeast Nigeria following mass distribution of LLINs: Implications for social behavior change interventions. PLOS One, 10(10), e0139447. https://doi.org/10.1371/journal.pone.0139447
  • SADC. (2018). Windhoek declaration on eliminating malaria in the SADC region—2018. Windhoek.
  • Sande, S., Zimba, M., Chinwada, P., Masendu, H. T., & Makuwaza, A. (2015). Malaria vector species composition and relative abundance in Mutare and Mutasa Districts, Zimbabwe. Journal of Entomological and Acarological Research, 47(3), 79. 2015https://doi.org/10.4081/jear.2015.4955
  • Sande, S., Zimba, M., Chinwada, P., Masendu, H. T., & Makuwaza, A. (2016). Insights into resting behavior of malaria vector mosquitoes in Mutare and Mutasa Districts of Manicaland Province, Zimbabwe. Journal of Medical Entomology, 53(4), 866–872. https://doi.org/10.1093/jme/tjw044
  • Sande, S., Zimba, M., Mberikunashe, J., Tangwena, A., & Chimusoro, A. (2017). Progress towards malaria elimination in Zimbabwe with special reference to the period 2003–2015. Malaria Journal, 16(1), 295. https://doi.org/10.1186/s12936-017-1939-0
  • Sangaré, L. R., Weiss, N. S., Brentlinger, P. E., Richardson, B. A., Staedke, S. G., Kiwuwa, M. S., & Stergachis, A. (2012). Determinants of use of insecticide treated nets for the prevention of malaria in pregnancy: Jinja, Uganda. PLOS One, 7(6), e39712. https://doi.org/10.1371/journal.pone.0039712
  • SARN. (2010). Malaria elimination technical meeting November 2–4, 2010: Maputo. Southern Africa Roll Back Malaria Network. http://tis.sadc.int/english/sarn/elimination-eight-e8/.
  • Severini, C., & Menegon, M. (2015). Resistance to antimalarial drugs: An endless world war against Plasmodium that we risk losing. Journal of Global Antimicrobial Resistance, 3(2), 58–63. https://doi.org/10.1016/j.jgar.2015.02.002
  • Shah, J. A., Emina, J. B., Eckert, E., & Ye, Y. (2015). Prompt access to effective malaria treatment among children under five in sub-Saharan Africa: A multi-country analysis of national household survey data. Malaria Journal, 14(1), 329. https://doi.org/10.1186/s12936-015-0844-7
  • Shellvarajah, M., Hatz, C., & Schlagenhauf, P. (2017). Malaria prevention recommendations for risk groups visiting sub-Saharan Africa: A survey of European expert opinion and international recommendations. Travel Medicine and Infectious Disease, 19, 49–55. https://doi.org/10.1016/j.tmaid.2017.09.002
  • Singh, M., Brown, G., & Rogerson, S. J. (2013). Ownership and use of insecticide-treated nets during pregnancy in sub-Saharan Africa: A review. Malaria Journal, 12(1), 268. https://doi.org/10.1186/1475-2875-12-268
  • Sinka, M. E., Pironon, S., Massey, N. C., Longbottom, J., Hemingway, J., Moyes, C. L., & Willis, K. J. (2020). A new malaria vector in Africa: predicting the expansion range of Anopheles stephensi and identifying the urban populations at risk. Proceedings of the National Academy of Sciences of the United States of America, 117(40), 24900–24908. https://doi.org/10.1073/pnas.2003976117
  • Slater, H. C., Foy, B. D., Kobylinski, K., Chaccour, C., Watson, O. J., Hellewell, J., Aljayyoussi, G., Bousema, T., Burrows, J., D’Alessandro, U., Alout, H., Ter Kuile, F. O., Walker, P. G. T., Ghani, A. C., & Smit, M. R. (2020). Ivermectin as a novel complementary malaria control tool to reduce incidence and prevalence: A modelling study. The Lancet. Infectious Diseases, 20(4), 498–508. https://doi.org/10.1016/S1473-3099(19)30633-4
  • Snetselaar, J., Njiru, B. N., Gachie, B., Owigo, P., Andriessen, R., Glunt, K., Osinga, A. J., Mutunga, J., Farenhorst, M., & Knols, B. G. J. (2017). Eave tubes for malaria control in Africa: Prototyping and evaluation against Anopheles gambiaess and Anopheles arabiensis under semi-field conditions in western Kenya. Malaria Journal, 16(1), 276. https://doi.org/10.1186/s12936-017-1926-5
  • Somé, F. A., Bazié, T., Ehrlich, H. Y., Goodwin, J., Lehane, A., Neya, C., Zachari, K., Wade, M., Ouattara, J. M., Foy, B. D., Dabiré, R. K., Parikh, S., & Ouédraogo, J. B. (2020). Investigating selected host and parasite factors potentially impacting upon seasonal malaria chemoprevention in Bama, Burkina Faso. Malaria Journal, 19(1), 238. https://doi.org/10.1186/s12936-020-03311-8
  • Sulaiman, S. K., Musa, M. S., Tsiga-Ahmed, F. I., Dayyab, F. M., Sulaiman, A. K., & Bako, A. T. (2022). A systematic review and meta-analysis of the prevalence of caregiver acceptance of malaria vaccine for under-five children in low-income and middle-income countries (LMICs). PLOS One, 17(12), e0278224. https://doi.org/10.1371/journal.pone.0278224
  • Tahghighi, A., Mohamadi-Zarch, S.-M., Rahimi, H., Marashiyan, M., Maleki-Ravasan, N., & Eslamifar, A. (2020). In silico and in vivo anti‑malarial investigation ­methylene) hydrazine derivatives. Malaria Journal, 19(1), 231. https://doi.org/10.1186/s12936-020-03269-7
  • Takarinda, K. P., Nyadundu, S., Govha, E., Gombe, N. T., Chadambuka, A., Juru, T., & Tshimanga, M. (2022). Factors associated with a malaria outbreak at Tongogara refugee camp in Chipinge District, Zimbabwe, 2021: A case–control study. Malaria Journal, 21(1), 94. https://doi.org/10.1186/s12936-022-04106-9
  • Tesfahuneygn, G., & Gebreegziabher, G. (2019). Medicinal plants used in traditional medicine by Ethiopians: A review article. Journal of Respiratory Medicine for Lung Disease, 4(1), 1–3.
  • Teshome, A., Erko, B., Golassa, L., Yohannes, G., Irish, S. R., Zohdy, S., Yoshimizu, M., & Dugassa, S. (2023). Resistance of Anopheles stephensi to selected insecticides used for indoor residual spraying and long-lasting insecticidal nets in Ethiopia. Malaria Journal, 22(1), 218. https://doi.org/10.1186/s12936-023-04649-5
  • Thomas, S., Ravishankaran, S., Justin, J. A., Asokan, A., Mathai, M. T., Valecha, N., Thomas, M. B., & Eapen, A. (2016). Overhead tank is the potential breeding habitat of Anopheles stephensi in an urban transmission setting of Chennai, India. Malaria Journal, 15(1), 274. https://doi.org/10.1186/s12936-016-1321-7
  • Toé, K. H., Jones, C. M., N’Fale, S., Ismail, H. M., Dabiré, R. K., & Ranson, H. (2014). Increased pyrethroid resistance in malaria vectors and decreased bed net effectiveness, Burkina Faso. Emerging Infectious Diseases, 20(10), 1691–1696. https://doi.org/10.3201/eid2010.140619
  • Tumwebaze, P., Tukwasibwe, S., Taylor, A., Conrad, M., Ruhamyankaka, E., Asua, V., Walakira, A., Nankabirwa, J., Yeka, A., Staedke, S. G., Greenhouse, B., Nsobya, S. L., Kamya, M. R., Dorsey, G., & Rosenthal, P. J. (2017). Changing antimalarial drug resistance patterns identified by surveillance at three sites in Uganda. The Journal of Infectious Diseases, 215(4), 631–635. https://doi.org/10.1093/infdis/jiw614
  • Tusting, L. S., Bottomley, C., Gibson, H., Kleinschmidt, I., Tatem, A. J., Lindsay, S. W., & Gething, P. W. (2017). Housing improvements and malaria risk in sub-Saharan Africa: A multi-country analysis of survey data. PLOS Medicine, 14(2), e1002234. https://doi.org/10.1371/journal.pmed.1002234
  • US President’s Malaria Initiative, Zimbabwe. (2020). CDC global health [Internet]. https://www.cdc.gov/globalhealth/countries/zimbabwe/annual-report/pmi.html.
  • US President’s Malaria Initiative. (2018). Zimbabwe malaria operational plan FY 2018.
  • US President’s Malaria Operational Plan FY, (2017). President’ malaria initiative Zimbabwe, malaria operational plan FY 2017.
  • US President’s Malaria Operational Plan FY, (2019). President’ malaria initiative Zimbabwe, malaria operational plan FY 2019.
  • US President’s Malaria Operational Plan FY, (2022). President’ malaria initiative Zimbabwe, malaria operational plan FY 2022.
  • US President’s Initiative. (2016). Fighting malaria and saving lives. Washington DC. Retrieved from https://www.pmi.gov
  • U. S. President’s malaria initiative Zimbabwe. (2018). Malaria operational plan FY 2018. Retrieved from https://www.pmi.gov
  • U. S. President’s malaria initiative Zimbabwe. (2020). Malaria operational plan FY 2020. Retrieved from https://www.pmi.gov
  • Verde, C., Tomé, S., Silva, J. R. D. A., Ramos, A. D. S., Machado, M., De Moura, D. F., Neto, Z., Canto-Cavalheiro, M. M., Figueiredo, P., Rosário, V. E., Amaral, A. C. F., & Lopes, D. (2011). A review of antimalarial plants used in traditional medicine in communities in Portuguese-Speaking countries: Brazil, Mozambique. Memorias do Instituto Oswald Cruz, 106, 142–158.
  • Wesolowski, A., Ippolito, M. M., Gebhardt, M. E., Ferriss, E., Schue, J. L., Kobayashi, T., Chaponda, M., Kabuya, J.-B., Muleba, M., Mburu, M., Matoba, J., Musonda, M., Katowa, B., Lubinda, M., Hamapumbu, H., Simubali, L., Mudenda, T., Shields, T. M., Hackman, A., … Moss, W. J, Southern and Central Africa International Center of Excellence for Malaria Research. (2022). Policy implications of the Southern and Central Africa International Center of Excellence for Malaria Research: Ten years of malaria control impact assessments in hypo-, meso-, and holoendemic transmission zones in Zambia and Zimbabwe. The American Journal of Tropical Medicine and Hygiene, 107(4_Suppl), 68–74. https://doi.org/10.4269/ajtmh.21-1288
  • WHO. (2007). Malaria elimination: A field manual for low and moderate endemic countries. World Health Organization.
  • WHO. (2013). World malaria report 2013. World Health Organization.
  • WHO. (2017). World malaria report 2016. World Health Organization.
  • WHO. (2018). World malaria report 2018. World Health Organization.
  • WHO. (2019). HBHI approach in world malaria report 2019 [Online]. Accessed 5 July 2022. https://www.who.int/publications/i/item/9789240015791.
  • WHO. (2019). World malaria report. World Health Organisation.
  • WHO. (2020). Global trends in the burden of malaria, world malaria report. World Health Organisation.
  • WHO. (2020). Global malaria programme, the potential impact of health service disruptions on the burden of malaria: A modelling analysis for countries in Sub-Saharan Africa. World Health Organisation.
  • World Health Organization. (2020). The E-2020 eliminating 2019 progress report. World Health Organisation.
  • WHO. (2021a). World Malaria Report 2021. World Health Organization. License: CC BY-NC-SA 3.0
  • WHO. (2021b). WHO recommends groundbreaking malaria vaccine for children at risk, historic RTS,S/AS01 recommendation can reinvigorate the fight against malaria. https://www.who.int/news/item/06-10-2021-who-recommends-groundbreaking-malaria-vaccine-for-children-at-risk
  • WHO. (2022a). The transformation agenda of the World Health Organization Secretariat in the African region 2015–2020 [Online]. Accessed 3 June 2022. https://www.afro.who.int/regional-director/transformation-agenda
  • WHO. (2022b). Tackling emerging antimalarial drug resistance in Africa. https://www.who.int/news/item/18-11-2022-tackling-emerging-antimalarial-drug-resistance-in-africa.
  • WHO. (2024). Highlights from the meeting of the strategic advisory group of experts (SAGE) on Immunization 11–13 March 2024. World Health Organization.
  • WHO. (2022c). World malaria report, Progress on malaria control in countries, WHO in an era of transformation, African region. https://www.afro.who.int.
  • Winskill, P., Walker, P. G., Cibulskis, R. E., & Ghani, A. C. (2020). Prioritizing the scale up of interventions for malaria control and elimination. Malaria Journal, 18(1), 122. https://doi.org/10.1186/s12936-019-2755-5
  • Wu, Y., Soe, M. T., Aung, P. L., Zhao, L., Zeng, W., Menezes, L., Yang, Z., Kyaw, M. P., & Cui, L. (2020). Efficacy of artemether ‑ lumefantrine for treating uncomplicated Plasmodium falciparum cases and molecular surveillance of drug resistance genes in Western Myanmar. Malaria Journal, 19(1), 304. https://doi.org/10.1186/s12936-020-03376-5
  • Yukich, J., Digre, P., Scates, S., Boydens, L., Obi, E., Moran, N., Belemvire, A., Chamorro, M., Johns, B., Malm, K. L., Kolyada, L., Williams, I., Asiedu, S., Fomba, S., Mihigo, J., Boko, D., Candrinho, B., Muthoni, R., Opigo, J., … Robertson, M. (2022). Incremental cost and cost-effectiveness of the addition of indoor residual spraying with pirimiphos-methyl in sub-Saharan Africa versus standard malaria control: Results of data collection and analysis in the next generation indoor residual sprays (NgenIRS) project, an economic-evaluation. Malaria Journal, 21(1), 185. https://doi.org/10.1186/s12936-022-04160-3
  • Zimbabwe National Statistical Agency. (2012). Census 2012: Preliminary report. Harare, Zimbabwe: Zimbabwe National Statistics Agency.