1
|
Mita T. Antimalarial drug efficacy and resistance: Insights from northern Uganda. Parasitol Int 2025; 109:103083. [PMID: 40348079 DOI: 10.1016/j.parint.2025.103083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/28/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Artemisinin in combination is the first-line treatment for Plasmodium falciparum malaria in almost all regions. However, by the late 2000s, partial resistance, characterized by delayed parasite clearance after treatment, emerged in the Greater Mekong Subregion and rapidly expanded its range. Since 2013, we have conducted comprehensive malaria drug resistance surveillance in northern Uganda. In 2014, we identified in vitro artemisinin resistance, and by 2017, clinical partial resistance had also been observed. Additionally, we discovered the re-emergence of chloroquine sensitivity in this region as early as 2013, earlier than in the other parts of Uganda. This review aims to summarize our findings from northern Uganda, contextualizing them within similar studies conducted in other regions.
Collapse
Affiliation(s)
- Toshihiro Mita
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
2
|
Kojom Foko LP, Jakhan J, Narang G, Hawadak J, Eboumbou Moukoko CE, Singh V. Novel Plasmodium falciparum Kelch13 polymorphisms in Cameroon with structural and physicochemical impact. Antimicrob Agents Chemother 2025; 69:e0088424. [PMID: 40227034 PMCID: PMC12057353 DOI: 10.1128/aac.00884-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/30/2025] [Indexed: 04/15/2025] Open
Abstract
The recent emergence of Plasmodium falciparum (Pf) parasites resistant to artemisinin-based combination therapies (ACT) in Africa has outlined the need for continuous molecular surveillance of artemisinin partial resistance. Here, the genetic polymorphism in the Kelch 13 gene (pfk13) and its structural impact were analyzed. PfDNA was extracted from dried blood spots of symptomatic and asymptomatic individuals living in different epidemiological facets of Cameroon. The pfk13 gene was amplified by nested polymerase chain reaction, and amplicons were sequenced to detect single nucleotide polymorphisms (SNPs). The evolutionary history and the impact of the polymorphisms on physicochemical properties, structure, and function of the pfK13 protein were appraised using various in silico models. A total of ten SNPs were identified in this study, of which five non-synonymous SNPs have not been previously reported (L647F, D648V, N657S, K658R, and L663P). The genetic diversity of pfk13 sequences was low, and the pfk13 gene evolved under the neutral model. Some mutations, especially L663P, appeared to affect the function and structure of the pfK13 protein. Analysis of the physicochemical properties of the Cameroonian pfK13 protein sequences revealed slight changes in the solvent-accessible surface area, isoelectric point, and hydrophobicity. The results support the ongoing use of ACTs in the study areas, given the absence of validated SNPs associated with artemisinin partial resistance. Computational findings suggest a possible deleterious effect of some novel SNPs on the pfK13 structure and/or function.
Collapse
Affiliation(s)
- Loick P. Kojom Foko
- Parasite & Host Biology, ICMR-National Institute of Malaria Research, New Delhi, Delhi, India
| | - Jahnvi Jakhan
- Parasite & Host Biology, ICMR-National Institute of Malaria Research, New Delhi, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Geetika Narang
- Parasite & Host Biology, ICMR-National Institute of Malaria Research, New Delhi, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Joseph Hawadak
- Parasite & Host Biology, ICMR-National Institute of Malaria Research, New Delhi, Delhi, India
| | - Carole E. Eboumbou Moukoko
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala, Littoral, Cameroon
- Malaria Research Unit, Centre Pasteur Cameroon, Douala, Cameroon
- Laboratory of Parasitology, Mycology and Virology, Postgraduate Training Unit for Health Sciences, Postgraduate School for Pure and Applied Sciences, The University of Douala, Douala, Littoral, Cameroon
| | - Vineeta Singh
- Parasite & Host Biology, ICMR-National Institute of Malaria Research, New Delhi, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
3
|
Meier-Scherling CPG, Watson OJ, Asua V, Ghinai I, Katairo T, Garg S, Conrad MD, Rosenthal PJ, Okell LC, Bailey JA. Selection of Plasmodium falciparum kelch13 mutations in Uganda in comparison with southeast Asia: a modelling study. THE LANCET. MICROBE 2025; 6:101027. [PMID: 40112841 DOI: 10.1016/j.lanmic.2024.101027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/24/2024] [Accepted: 10/16/2024] [Indexed: 03/22/2025]
Abstract
BACKGROUND Artemisinin partial resistance, mediated by mutations in the Plasmodium falciparum kelch13 gene (k13), rapidly spread in southeast Asia, undermining the antimalarial effectiveness of artemisinin-based combination therapies. k13 mutations have also arisen in Africa, but their rates of increase are not well characterised. We aimed to quantify the selection of k13 mutations in Africa and compare the selection with that in southeast Asia. METHODS In this modelling study, we investigated k13 mutation allele frequency at 16 sites in Uganda (2016-22) and five sites in southeast Asia (in Cambodia, Thailand, and Viet Nam; 2003-14). The Ugandan data were obtained from annual clinical surveillance studies and the southeast Asian data were obtained from the MalariaGEN Pf7 dataset. We investigated five validated and candidate k13 mutations: Pro441Leu, Cys469Phe, Cys469Tyr, Arg561His, and Ala675Val. We calculated annual selection coefficients using Bayesian mixed-effect linear models. We then tested whether the k13 mutation allele frequency in southeast Asia could have been forecast accurately using up to the first 5 years of available data and forecast future k13 mutation allele frequency in Uganda. FINDINGS We used data from 7564 samples from Uganda and 6568 samples from southeast Asia. The annual selection coefficient of evaluable k13 mutations (Pro441Leu, Cys469Phe/Tyr, Arg561His, and Ala675Val) across all sites was estimated at 0·381 (95% credible interval 0·298 to 0·472) per year, a 38% increase in relative allele frequency. Selection coefficients across Uganda were 0·494 (-0·462 to 1·410) for Pro441Leu, 0·324 (-0·629 to 1·150) for Cys469Phe, 0·383 (0·207 to 0·591) for Cys469Tyr, and 0·237 (0·087 to 0·403) for Ala675Val. In southeast Asia, the selection coefficients were 0·627 (-0·088 to 1·312) for Cys580Tyr, 0·224 (-0·903 to 1·397) for Arg539Thr, and 0·330 (-0·075 to 0·683) for all validated k13 mutations. Compared with out-of-sample data, the forecasts for southeast Asia underestimated mutation allele frequency and were of variable accuracy. Overall, forecast allele frequencies for Uganda, assuming constant selection, neared fixation (>0·95 allele frequency) within a decade (between 2031 and 2033) for combined k13 mutations. INTERPRETATION k13 mutation selection in Uganda was similar to that observed in southeast Asia, suggesting that frequencies of k13 mutations will continue to increase quickly in Uganda. These commensurate levels of selection indicate a high potential for rapid transmission across other parts of Africa, underscoring the urgent need for treatments and policies to mitigate the spread and impact of k13 mutations. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
| | - Oliver J Watson
- Medical Research Council Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
| | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda; Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Isaac Ghinai
- Big Data Institute, University of Oxford, Oxford, UK
| | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Shreeya Garg
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Melissa D Conrad
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lucy C Okell
- Medical Research Council Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
| | - Jeffrey A Bailey
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
4
|
João MF, Aranda-Díaz A, De Amaral F, Makhanthisa TI, Lauterbach SB, Chisenga M, Mangena B, Maquina P, Routledge I, Sikaala C, Chimumbwa J, Jandondo D, Martins JF, Raman J, Smith JL, Dimbu PR. Geographical Heterogeneity in Antimalarial Resistance Markers Revealed by Genomic Surveillance in Angola, 2023. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.08.25325242. [PMID: 40297444 PMCID: PMC12036384 DOI: 10.1101/2025.04.08.25325242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Plasmodium falciparum malaria remains a leading cause of mortality in Angola, with emerging antimalarial resistance threatening treatment and prevention strategies. Efficacy of artemether-lumefantrine, one of the country's preferred malaria treatments, has been reported below 90% in two provinces, underscoring the need for routine resistance surveillance and efficacy monitoring to guide policy decisions. Between March and July 2023, dried blood spots and demographic data were collected from P. falciparum-positive participants at 16 health facilities across 8 provinces. Multiplexed amplicon deep sequencing was used to characterize single nucleotide polymorphisms in 12 genes linked with resistance, estimate allele frequencies, and detect co-infecting non-falciparum Plasmodium species. Sequence data from 817 samples revealed significant geographic variation in resistance markers. In the southeast, artemisinin partial resistance markers (k13 P574L, P441L), were detected at very low prevalence (<0.1%), while the quintuple dhps/dhfr haplotype, linked to sulfadoxine-pyrimethamine (SP) resistance, was very prevalent (>40% of samples). In the northwest, the sextuple dhps/dhfr haplotype, a marker of higher SP resistance, was most prevalent in Zaire (14.2%). The crt CVIET haplotype, associated with chloroquine resistance, had a national prevalence of 15.9%, detected in over 48% of samples from Zaire and Uíge. The mdr1 N86 genotype, linked to reduced lumefantrine susceptibility, was widespread, detected in 99.3% of samples. Co-infections of P. falciparum and non-falciparum species were rare with no clear geographic distribution. No P. vivax co-infections were detected. These findings highlight the need for continued monitoring to safeguard treatment efficacy, reinforcing the importance of molecular surveillance in malaria control strategies.
Collapse
Affiliation(s)
| | - Andrés Aranda-Díaz
- EPPIcenter Research Program, Department of Medicine, University of California, San Francisco, United States of America
- Malaria Elimination Initiative, Global Health Group, University of California, San Francisco, United States of America
- ISGlobal Barcelona, Spain
| | - Faith De Amaral
- EPPIcenter Research Program, Department of Medicine, University of California, San Francisco, United States of America
| | - Takalani I. Makhanthisa
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research, Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Sonja B. Lauterbach
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research, Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | | | | - Paulo Maquina
- SADC Malaria Elimination Eight Secretariat, Windhoek, Namibia
| | - Isobel Routledge
- Malaria Elimination Initiative, Global Health Group, University of California, San Francisco, United States of America
| | | | - John Chimumbwa
- SADC Malaria Elimination Eight Secretariat, Windhoek, Namibia
| | | | | | - Jaishree Raman
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research, Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
- Wits Research Institute for Malaria, University of Witwatersrand, Johannesburg, South Africa
- University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Jennifer L. Smith
- Malaria Elimination Initiative, Global Health Group, University of California, San Francisco, United States of America
- Department of Epidemiology & Biostatistics, University of California, San Francisco, United States of America
| | | |
Collapse
|
5
|
Sy H, Yadav S, Yangga P, Shaffer A, John A, Rogers W, Westblade L, Marrero Rolón R, Madison-Antenucci S, Thwe PM, Orner E, Murphy K, Maldarelli GA, Kirkman L, Daily JP. Recurrent Plasmodium falciparum Malaria in U.S. Travelers Treated with Artemether-Lumefantrine. Am J Trop Med Hyg 2025; 112:771-774. [PMID: 39773552 PMCID: PMC11965738 DOI: 10.4269/ajtmh.24-0397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/16/2024] [Indexed: 01/11/2025] Open
Abstract
We report two cases of recurrent malaria in U.S. travelers returning from Africa (Ghana and Central African Republic) despite a full course of artemether-lumefantrine (AL). Both patients presented to New York City hospitals, received AL treatment, and clinically improved. Within 2 weeks, they presented with recurrent Plasmodium falciparum malaria. Parasite isolates were sequenced, and P. falciparum kelch 13 propeller domain mutations that are validated or candidate markers of artemisinin partial resistance were not identified. Parasites had mutations within the P. falciparum multidrug resistance protein 1 gene. It is crucial to remain vigilant for recurrent malaria in travelers, even from African regions where partial resistance to artemisinin-based combination therapy has only rarely been reported.
Collapse
Affiliation(s)
- Hendrik Sy
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Sharan Yadav
- Division of Infectious Diseases, Weill Cornell Medicine, New York, New York
| | - Peter Yangga
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Alexander Shaffer
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Amie John
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Wesley Rogers
- Division of Infectious Diseases, Weill Cornell Medicine, New York, New York
| | - Lars Westblade
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Rebecca Marrero Rolón
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Phyu M. Thwe
- Department of Pathology, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Erika Orner
- Department of Pathology, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Kerry Murphy
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | | | - Laura Kirkman
- Division of Infectious Diseases, Weill Cornell Medicine, New York, New York
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York
| | - Johanna P. Daily
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
6
|
Hirai M, Arai M, Hayamichi S, Uchida A, Sudo M, Kubota R, Shinzawa N, Mita T. Deletion of the chloroquine resistance transporter gene confers reduced piperaquine susceptibility to the rodent malaria parasite Plasmodium berghei. Antimicrob Agents Chemother 2025; 69:e0158924. [PMID: 39992104 PMCID: PMC11963562 DOI: 10.1128/aac.01589-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025] Open
Abstract
Malaria parasites acquire drug resistance through genetic changes, the mechanisms of which remain incompletely understood. Understanding the mechanisms of drug resistance is crucial for the development of effective treatments against malaria, and for this purpose, new genetic tools are needed. In a previous study, as a forward genetic tool, we developed the rodent malaria parasite Plasmodium berghei mutator (PbMut) line, which has a greatly increased rate of mutation accumulation and from which we isolated a mutant with reduced susceptibility to piperaquine (PPQ). We identified a mutation in the chloroquine resistance transporter (PbCRT N331I) as responsible for this phenotype. In the current study, we generated a marker-free PbMut to enable further genetic manipulation of the isolated mutants. Here, we screened again for PPQ-resistant mutants in marker-free PbMut and obtained a parasite population with reduced susceptibility to PPQ. Of five isolated clones, none had the mutation PbCRT N331I; rather, they possessed a nonsense mutation at amino acid 119 (PbCRT Y119*), which would truncate the protein before eight of its ten predicted transmembrane domains. The PbCRT orthologue in the human malaria parasite Plasmodium falciparum, PfCRT, is an essential membrane transporter. To address the essentiality of PbCRT, we successfully deleted the full PbCRT gene [PbCRT(-)] from wild-type parasites. PbCRT(-) parasites exhibited reduced susceptibility to PPQ, along with compromised fitness in mice and following transmission to mosquitoes. Taken together, our findings provide the first evidence that P. berghei can acquire reduced PPQ susceptibility through complete loss of PbCRT function.
Collapse
Affiliation(s)
- Makoto Hirai
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Bunkyo-ku Hongo, Tokyo, Japan
| | - Meiji Arai
- Department of International Medical Zoology, School of Medicine, Kagawa University, Kida, Kagawa, Japan
| | - Soki Hayamichi
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Bunkyo-ku Hongo, Tokyo, Japan
| | - Ayako Uchida
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Bunkyo-ku Hongo, Tokyo, Japan
| | - Megumi Sudo
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Bunkyo-ku Hongo, Tokyo, Japan
| | - Rie Kubota
- Department of Parasitology and Tropical Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Bunkyo-ku Yushima, Tokyo, Japan
| | - Naoaki Shinzawa
- Department of Parasitology and Tropical Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Bunkyo-ku Yushima, Tokyo, Japan
| | - Toshihiro Mita
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Bunkyo-ku Hongo, Tokyo, Japan
| |
Collapse
|
7
|
Dame JA, Bockarie YM, Enimil AK. Impact of antimicrobial resistance on infections in children in Africa. Curr Opin Pediatr 2025; 37:145-152. [PMID: 39882690 DOI: 10.1097/mop.0000000000001440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
PURPOSE OF REVIEW Antimicrobial resistance is an escalating public health threat in Africa, and an awareness of the devastating impact on children is growing. This review highlights the prevalence and patterns of antimicrobial resistance among children in Africa, focusing on pathogens responsible for bloodstream infections, community-acquired pneumonia, bacterial meningitis, neonatal infections, diarrhea and malaria. Current strategies to tackle antimicrobial resistance in pediatric populations are discussed. RECENT FINDINGS Bloodstream infections significantly contribute to child mortality, with high resistance observed in pathogens like Salmonella spp., Klebsiella spp., Escherichia coli , and Staphylococcus aureus . Additionally, rising resistance in pathogens causing community-acquired pneumonia, meningitis and bacterial diarrhea challenges the effectiveness of WHO-recommended therapies. Antibiotics used to treat neonatal infections, such as ampicillin, gentamicin and cefotaxime, are threatened by high resistance in Escherichia coli and Klebsiella spp , contributing to adverse neonatal outcomes. PfKelch 13 mutations linked to artemisinin resistance in parts of Africa raise public health concerns, as malaria remains a major cause of illness and death. SUMMARY Stronger collaborative efforts are needed to enhance surveillance, improve diagnostic capabilities and update treatment protocols based on local pathogen sensitivities. More research is required on pediatric antimicrobial resistance in Africa.
Collapse
Affiliation(s)
- Joycelyn Assimeng Dame
- Department of Child Health, University of Ghana Medical School, College of Health Sciences, University of Ghana, Accra
| | | | - Anthony Kwame Enimil
- Department of Child Health, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
8
|
Muriithi B, Chepngetich J, Gachie B, Thiong'o K, Gathirwa J, Kimani F, Mwitari P, Kiboi D. Structural and functional implications of MIT2 and NT2 mutations in amodiaquine and piperaquine resistant Plasmodium berghei parasites. Exp Parasitol 2025; 271:108923. [PMID: 40032183 DOI: 10.1016/j.exppara.2025.108923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/27/2024] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Long-acting drugs, amodiaquine (AQ), lumefantrine (LM), and piperaquine (PQ), are vital components of artemisinin-based combination therapies (ACTs) for malaria treatment. However, the emergence of partial artemisinin-resistant parasites poses significant challenges, particularly in malaria-endemic regions. Despite extensive research, parasite's resistance mechanisms to these drugs still need complete elucidation. This study investigated the genetic basis of resistance to AQ, LM, and PQ using Plasmodium berghei, focusing on selected genes encoding transport proteins in Plasmodium species. In silico bioinformatics tools were used to map genes encoding transport proteins, their ligand-binding sites, and their conservation across different Plasmodium species. PCR amplification and sequence analysis were employed to examine single nucleotide polymorphisms (SNPs) in the genes encoding the selected transporters in AQ, LM, and PQ-resistant P. berghei. The structural impacts of the mutations were evaluated using AlphaFold, ITASSER, UCSF Chimera, and MOTIF Finder. Genes encoding CorA-like Mg2+ transporter protein (MIT2), nucleoside transporter 2 (NT2), ABC Transporter G family member 2 (ABCG2), and novel putative transporter 1 (NPT1) transport proteins with notable conserved motifs and ligand-binding motifs in Plasmodium species were selected and examined. In AQ-resistant (AQR) parasites, a non-synonymous mutation (I433∗) was found in MIT2. PQ-resistant (PQR) parasites possessed a non-synonymous mutation (D511H) in NT2 and a silent mutation in the NPT1 protein. No mutations were observed in the targeted regions of the transporters in LM-resistant (LMR) parasites, nor in the ligand-binding motifs of ABCG2 across all resistant strains. These findings suggest that selection pressure from AQ and PQ leads to mutations in MIT2 and NT2. Further investigation is required to understand how these mutations affect drug susceptibility on a functional level.
Collapse
Affiliation(s)
- Brenda Muriithi
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya
| | - Jean Chepngetich
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya; PanAfrican University Institute for Basic Sciences, Technology and Innovation is 62000, 00200, Nairobi, Kenya
| | - Beatrice Gachie
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; PanAfrican University Institute for Basic Sciences, Technology and Innovation is 62000, 00200, Nairobi, Kenya
| | - Kevin Thiong'o
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya
| | - Jeremiah Gathirwa
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, 54840, 00200, Nairobi, Kenya
| | - Francis Kimani
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya
| | - Peter Mwitari
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, 54840, 00200, Nairobi, Kenya
| | - Daniel Kiboi
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya.
| |
Collapse
|
9
|
Alruwaili M, Elderdery A, Manni E, Mills J. A Narrative Review on the Prevalence of Plasmodium falciparum Resistance Mutations to Antimalarial Drugs in Rwanda. Trop Med Infect Dis 2025; 10:89. [PMID: 40278762 PMCID: PMC12030788 DOI: 10.3390/tropicalmed10040089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/26/2025] Open
Abstract
Malaria has been and remains a significant challenge in Africa and other endemic settings. Roughly, 95% of global morbidity and mortality due to malaria occurs within African populations and affects millions of individuals, especially those living in sub-Saharan countries, predominantly due to disease complications. Cultural factors such as unawareness of and disinterest in using recommended preventive tools and combating the primary host (i.e., the female Anopheles mosquito) play a significant role. This host transmits the malaria-causing Plasmodium parasite by biting an infected individual and spreading it to humans. The current overview focuses on the molecular markers associated with antimalarial drug resistance in Plasmodium falciparum (P. falciparum) in Rwanda, considered an exemplar of sub-Saharan countries where malaria is prevalent and effective policies on the development of malaria treatment, approved recently by WHO in 2025, have been adopted. The prevalence of mutations in key resistance genes, including pfcrt, pfmdr1, and pfdhfr/pfdhps, are linked to resistance against common antimalarial drugs such as chloroquine and sulfadoxine-pyrimethamine (SP). In addition, the Plasmodium falciparum kelch13 (pfk13) gene is linked to resistance against artemisinin, as its mutations can cause delayed parasite clearance and treatment failure. Despite changes in therapeutic use policies owing to high prevalence of variant alleles, which reduce the drug's efficacy resistance to SP, the gene persists in Rwanda. Malaria parasites are becoming more resistant to chloroquine, leading to diminished effectiveness and slower recovery or treatment failure. Surveillance data reported from several studies provide crucial insights into the evolving trends of resistance markers and are vital for guiding treatment protocols and informing therapeutic use policy decisions. It is important that we continue to maintain and develop the effectiveness of malaria prevention strategies and treatments, due to the multiple types of resistance found in the population.
Collapse
Affiliation(s)
- Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Abozer Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Emad Manni
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK;
| |
Collapse
|
10
|
Aranda-Díaz A, Neubauer Vickers E, Murie K, Palmer B, Hathaway N, Gerlovina I, Boene S, García-Ulloa M, Cisteró P, Katairo T, Semakuba FD, Nsengimaana B, Gwarinda H, García-Fernández C, Louie W, Esayas E, Da Silva C, Datta D, Kiyaga S, Wiringilimaana I, Feleke SM, Bennett A, Smith JL, Gadisa E, Parr JB, Conrad MD, Raman J, Tukwasibwe S, Ssewanyana I, Rovira-Vallbona E, Tato CM, Briggs J, Mayor A, Greenhouse B. Sensitive and modular amplicon sequencing of Plasmodium falciparum diversity and resistance for research and public health. Sci Rep 2025; 15:10737. [PMID: 40155691 PMCID: PMC11953298 DOI: 10.1038/s41598-025-94716-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Targeted amplicon sequencing is a powerful and efficient tool for interrogating the Plasmodium falciparum genome, generating actionable data from infections to complement traditional malaria epidemiology. For maximum impact, genomic tools should be multi-purpose, robust, sensitive, and reproducible. We developed, characterized, and implemented MAD4HatTeR, an amplicon sequencing panel based on Multiplex Amplicons for Drug, Diagnostic, Diversity, and Differentiation Haplotypes using Targeted Resequencing, along with a bioinformatic pipeline for data analysis. Additionally, we introduce an analytical approach to detect gene duplications and deletions from amplicon sequencing data. Laboratory control and field samples were used to demonstrate the panel's high sensitivity and robustness. MAD4HatTeR targets 165 highly diverse loci, focusing on multiallelic microhaplotypes, key markers for drug and diagnostic resistance (including duplications and deletions), and CSP and potential vaccine targets. The panel can also detect non-falciparum Plasmodium species. MAD4HatTeR successfully generated data from low-parasite-density dried blood spot and mosquito midgut samples and detected minor alleles at within-sample allele frequencies as low as 1% with high specificity in high-parasite-density dried blood spot samples. Gene deletions and duplications were reliably detected in mono- and polyclonal controls. Data generated by MAD4HatTeR were highly reproducible across multiple laboratories. The successful implementation of MAD4HatTeR in five laboratories, including three in malaria-endemic African countries, showcases its feasibility and reproducibility in diverse settings. MAD4HatTeR is thus a powerful tool for research and a robust resource for malaria public health surveillance and control.
Collapse
Affiliation(s)
- Andrés Aranda-Díaz
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Eric Neubauer Vickers
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Kathryn Murie
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Brian Palmer
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Nicholas Hathaway
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Inna Gerlovina
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Simone Boene
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- ISGlobal, Barcelona, Spain
| | | | | | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Hazel Gwarinda
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research (ARMMOR), Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | | - William Louie
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | | | | | | | - Shahiid Kiyaga
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data Intensive Sciences, Kampala, Uganda
| | | | - Sindew Mekasha Feleke
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
- Department of Environment and Genetics, La Trobe University, Melbourne, Australia
| | | | - Jennifer L Smith
- Global Health Group, Malaria Elimination Initiative, University of California, San Francisco, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, USA
| | | | - Jonathan B Parr
- Division of Infectious Diseases, University of North Carolina, Chapel Hill, NC, USA
| | - Melissa D Conrad
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Jaishree Raman
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research (ARMMOR), Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
- Wits Research Institute for Malaria, University of Witwatersrand, Johannesburg, South Africa
- University of Pretoria Institute for Sustainable Malaria Control (UPISMC), University of Pretoria, Pretoria, South Africa
| | | | | | | | | | - Jessica Briggs
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Alfredo Mayor
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- ISGlobal, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- Department of Physiologic Sciences, Faculty of Medicine, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Bryan Greenhouse
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
11
|
Pernaute-Lau L, Recker M, Tékété M, de Sousa TN, Traore A, Fofana B, Sanogo K, Morris U, Inoue J, Ferreira PE, Diallo N, Burhenne J, Sagara I, Dicko A, Veiga MI, Haefeli W, Björkman A, Djimde AA, Borrmann S, Gil JP. Decreased dihydroartemisinin-piperaquine protection against recurrent malaria associated with Plasmodium falciparum plasmepsin 3 copy number variation in Africa. Nat Commun 2025; 16:2680. [PMID: 40102390 PMCID: PMC11920258 DOI: 10.1038/s41467-025-57726-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Dihydroartemisinin-piperaquine (DHA-PPQ) is being recommended in Africa for the management of uncomplicated Plasmodium falciparum malaria and for chemoprevention strategies, based on the ability of piperaquine to delay re-infections. Although therapeutic resistance to piperaquine has been linked to increased copy number in plasmepsin-coding parasite genes (pfpm), their effect on the duration of the post-treatment prophylactic period remains unclear. Here, we retrospectively analyzed data from a randomized clinical trial, where patients received either DHA-PPQ or artesunate-amodiaquine for recurrent malaria episodes over two years. We observed an increase in the relative risk of re-infection among patients receiving DHA-PPQ compared to artesunate-amodiaquine after the first malaria season. This was driven by shorter average times to reinfection and coincided with an increased frequency of infections comprising pfpm3 multi-copy parasites. The decline in post-treatment protection of DHA-PPQ upon repeated use in a high transmission setting raises concerns for its wider use for chemopreventive strategies in Africa.
Collapse
Affiliation(s)
- Leyre Pernaute-Lau
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- The Art of Discovery, Derio, Basque Country, Spain
| | - Mario Recker
- Centre for Ecology and Conservation, University of Exeter, Penryn Campus, Penryn, UK
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Mamadou Tékété
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
- Dept. of Clinical Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Tais Nóbrega de Sousa
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Molecular Biology and Malaria Immunology Research Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ); Belo, Horizonte, Brasil
| | - Aliou Traore
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Bakary Fofana
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassim Sanogo
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Ulrika Morris
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juliana Inoue
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Pedro E Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B's─PT Government Associate Laboratory; 4806-909 Guimarães, Braga, Portugal
| | - Nouhoum Diallo
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Jürgen Burhenne
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Issaka Sagara
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Maria I Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B's─PT Government Associate Laboratory; 4806-909 Guimarães, Braga, Portugal
| | - Walter Haefeli
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Anders Björkman
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Abdoulaye A Djimde
- Malaria Research and Training Center, Faculty of Pharmacy, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Steffen Borrmann
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - José Pedro Gil
- Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Clinical Tropical Medicine (CTM), Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, Nova University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
12
|
Tebben K, Eng V, Seng D, Tat B, Feufack Donfack LB, Orban A, Yeat R, Salvador J, Sin S, Ko K, Khim N, Flamand C, Sommen C, Lek D, Serre D, Popovici J. Cambodian Plasmodium vivax parasites with reduced hemoglobin digestion display delayed clearance upon artesunate treatment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.09.25323469. [PMID: 40162245 PMCID: PMC11952599 DOI: 10.1101/2025.03.09.25323469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Artemisinin-based combination therapies are the frontline drugs for the treatment of malaria infections but, for Plasmodium falciparum, the efficacy of artemisinin is threatened by the spread of resistance . P. vivax is the second most common cause of human malaria but we have little information on its susceptibility to artemisinin due to the lack of in vitro cultures. Here, we analyze 161 P. vivax infections from Cambodian patients treated with 2 mg/kg/day of artesunate for seven days. All infections were successfully cleared by day 3. However, one third of the infections displayed a slow clearance after treatment, with nine infections (5.7%) with a parasite clearance time greater than 5 hours, meeting the WHO definition of artemisinin resistance. We observed no significant association between slow clearance and either patient- or infection characteristics (including stage composition). We used RNA-seq to characterize the gene expression of parasites from 15 fast- and 16 slow-clearing infections at baseline and 1, 2 and 4 hours after treatment. While fast-clearing parasites showed significant changes in gene expression immediately upon treatment, slow-clearing parasites displayed a significantly delayed gene expression response, with a downregulation of many genes associated with hemoglobin endocytosis and digestion. Overall, our results indicate that some Cambodian P. vivax parasites clear slowly after artesunate treatment, possibly due to a downregulation of hemoglobin metabolism that may reduce the efficiency of the artesunate. Research in context Evidence before this study: The WHO treatment guidelines recommend artemisinin-combination therapy (ACT) for treatment of blood-stage infections caused by Plasmodium vivax in all areas (with chloroquine recommended only in areas where P. vivax are still chloroquine-sensitive). In P. falciparum , partial resistance to artemisinin derivatives is defined in vivo as either detected parasitemia on day 3 post treatment or as a half-life of the parasite clearance slope of ≥ 5 hours. We searched Pubmed for studies containing the terms "vivax" AND "clearance" AND ("artesunate" OR "dihydroartemisinin" OR "artemether" OR "artemisinin") published between 1990 and February 2025, with no language restrictions. Our search retrieved 102 studies for which title and abstracts were screened to identify 21 studies reporting outcomes of P. vivax treatment with an artemisinin derivative. While all these studies concluded that artemisinin derivatives provided rapid clearance of P. vivax parasites, two studies reported a low frequency of day 3 positivity following artesunate-amodiaquine treatment (2.6% in Brazil) or dihydroartemisinin-piperaquine (0.6% in Indonesia). No study reported clearance slope half-life ≥ 5 hours. Added value of this study: This study used a cohort of Cambodian patients infected by P. vivax to rigorously examine the efficacy of artesunate monotherapy at clearing blood stage infections. Our study showed significant variations in clearance rates among infections, with 5.7% of the infections with a clearance slope half-life ≥ 5 hours, meeting the criteria for artemisinin partial resistance used for P. falciparum . Variations in clearance rate upon artesunate treatment were not associated with patient or infection characteristics. Gene expression analyses revealed that the slow-clearing parasites down-regulated upon treatment many genes involved in hemoglobin endocytosis and digestion, possibly resulting in a lesser activation of artesunate. Implications of all the available evidence: Our results confirm that 2 mg/kg of artesunate per day for seven days is effective at clearing P. vivax blood stage infections. However, a subset of the P. vivax parasites displayed a slow clearance following artesunate treatment meeting artemisinin partial resistance definition in P. falciparum . Gene expression analyses suggest that metabolic variations may underlie slow clearance. Increased monitoring of treatment efficacy and drug resistance in P. vivax is therefore recommended.
Collapse
|
13
|
Aranda-Díaz A, Vickers EN, Murie K, Palmer B, Hathaway N, Gerlovina I, Boene S, Garcia-Ulloa M, Cisteró P, Katairo T, Semakuba FD, Nsengimaana B, Gwarinda H, García-Fernández C, Louie W, Esayas E, Da Silva C, Datta D, Kiyaga S, Wiringilimaana I, Fekele SM, Bennett A, Smith JL, Gadisa E, Parr JB, Conrad M, Raman J, Tukwasibwe S, Ssewanyana I, Rovira-Vallbona E, Tato CM, Briggs J, Mayor A, Greenhouse B. Sensitive and modular amplicon sequencing of Plasmodium falciparum diversity and resistance for research and public health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.22.609145. [PMID: 39229023 PMCID: PMC11370457 DOI: 10.1101/2024.08.22.609145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Targeted amplicon sequencing is a powerful and efficient tool for interrogating the Plasmodium falciparum genome, generating actionable data from infections to complement traditional malaria epidemiology. For maximum impact, genomic tools should be multi-purpose, robust, sensitive, and reproducible. Methods We developed, characterized, and implemented MAD4HatTeR, an amplicon sequencing panel based on Multiplex Amplicons for Drug, Diagnostic, Diversity, and Differentiation Haplotypes using Targeted Resequencing, along with a bioinformatic pipeline for data analysis. Additionally, we introduce an analytical approach to detect gene duplications and deletions from amplicon sequencing data. Laboratory control and field samples were used to demonstrate the panel's high sensitivity and robustness. Results MAD4HatTeR targets 165 highly diverse loci, focusing on multiallelic microhaplotypes, key markers for drug and diagnostic resistance (including duplications and deletions), and csp and potential vaccine targets. The panel can also detect non-falciparum Plasmodium species. MAD4HatTeR successfully generated data from low-parasite-density dried blood spot and mosquito midgut samples, and detected minor alleles at within-sample allele frequencies as low as 1% with high specificity in high-parasite-density dried blood spot samples. Gene deletions and duplications were reliably detected in mono- and polyclonal controls. Data generated by MAD4HatTeR were highly reproducible across multiple laboratories. Conclusions The successful implementation of MAD4HatTeR in five laboratories, including three in malaria-endemic African countries, showcases its feasibility and reproducibility in diverse settings. MAD4HatTeR is thus a powerful tool for research and a robust resource for malaria public health surveillance and control.
Collapse
Affiliation(s)
- Andrés Aranda-Díaz
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Eric Neubauer Vickers
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Kathryn Murie
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Brian Palmer
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Nicholas Hathaway
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Inna Gerlovina
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Simone Boene
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | | | | | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Hazel Gwarinda
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research (ARMMOR), Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
| | | | - William Louie
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | | | | | | | - Shahiid Kiyaga
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Sindew Mekasha Fekele
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
- Department of Environment and Genetics, La Trobe University, Melbourne, Australia
| | | | - Jennifer L. Smith
- Malaria Elimination Initiative, Global Health Group, University of California, San Francisco, United States of America
- Department of Epidemiology & Biostatistics, University of California, San Francisco, United States of America
| | | | - Jonathan B. Parr
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Melissa Conrad
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Jaishree Raman
- Laboratory for Antimalarial Resistance Monitoring and Malaria Operational Research (ARMMOR), Centre of Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, Johannesburg, South Africa
- Wits Research Institute for Malaria, University of Witwatersrand, Johannesburg, South Africa
- University of Pretoria Institute for Sustainable Malaria Control (UPISMC), University of Pretoria, Pretoria, South Africa
| | | | | | | | | | - Jessica Briggs
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Alfredo Mayor
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
- ISGlobal, Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- Department of Physiologic Sciences, Faculty of Medicine, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Bryan Greenhouse
- EPPIcenter Research Program, Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
14
|
Srisutham S, Saejeng A, Khantikul N, Sugaram R, Sangsri R, Dondorp AM, Day NPJ, Imwong M. Advancing artemisinin resistance monitoring using a high sensitivity ddPCR assay for Pfkelch13 mutation detection in Asia. Sci Rep 2025; 15:4869. [PMID: 39929914 PMCID: PMC11811204 DOI: 10.1038/s41598-025-86630-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
The spread of Pfkelch13 mutations in Southeast Asia threatens the effectiveness of artemisinin-based combination therapies (ACTs) for malaria. Previous studies revealed a high prevalence of key mutations, including C580Y, P574L, and R561H, emphasizing the need for the surveillance to combat drug resistance. This study, we developed a droplet digital PCR (ddPCR) assay for the rapid screening of common mutations including P441L, Y493H, P527H, G538V, R539T, I543T, R561H, P574L, C580Y, and A675V. The assay was designed to detect minor populations of mutant strain within multiple infection, offering high sensitivity and specificity using artificial mixtures of mutant and wild-type alleles. Field samples collected in Thailand during 2015-2020 and in 2023 (N = 130) were also analyzed to validate the assay in a real-world setting. The ddPCR assay demonstrated exceptional performance, with 100% sensitivity and 90% specificity. The R539T, R561H, and C580Y mutations were detected in clinical samples collected from several study sites in Thailand. Notably, the R561H mutation was detected in 100% of the P. falciparum isolates from Mae Hong Son, Thailand in 2023, underscoring the assay's utility in identifying critical mutations associated with drug resistance. Moreover, ddPCR can detect multiple parasite populations in clinical samples and can be used to analyze the ratios of wild-type and mutant alleles. These results validate the assay's ability to serve as a powerful tool for the early detection of minor allele frequencies, facilitating the timely implementation of interventions to curb the spread of ACT resistance. The ddPCR assays developed in this study provide a sensitive and specific method for detecting Pfkelch13 mutations, allowing the identification of minor parasite populations with artemisinin resistance. These assays enhance our ability to monitor and respond to malaria drug resistance, offering a crucial tool for early detection and contributing to global malaria elimination efforts.
Collapse
Affiliation(s)
- Suttipat Srisutham
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Aungkana Saejeng
- Division of Vector Borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Nardlada Khantikul
- Division of Vector Borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Rungniran Sugaram
- Division of Vector Borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Raweewan Sangsri
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Wernsman Young N, Gashema P, Giesbrecht D, Munyaneza T, Maisha F, Mwebembezi F, Budodo R, Leonetti A, Crudale R, Iradukunda V, Jean Bosco N, Kirby RI, Boyce RM, Mandara CI, Kanyankole GK, Ntaro M, Okell LC, Watson OJ, Mulogo E, Ishengoma DS, Hangi S, Karema C, Mazarati JB, Juliano JJ, Bailey JA. High Frequency of Artemisinin Partial Resistance Mutations in the Great Lakes Region Revealed Through Rapid Pooled Deep Sequencing. J Infect Dis 2025; 231:269-280. [PMID: 39367758 PMCID: PMC11793061 DOI: 10.1093/infdis/jiae475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/13/2024] [Accepted: 10/03/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND In Africa, the first Plasmodium falciparum artemisinin partial resistance mutation was Kelch13 (K13) 561H, detected and validated at appreciable frequency in Rwanda in 2014. Surveillance to better define the extent of the emergence in Rwanda and neighboring countries is critical. METHODS We used novel liquid blood drop preservation with pooled sequencing to provide cost-effective rapid assessment of resistance mutation frequencies at multiple collection sites across Rwanda and neighboring regions in Uganda, Tanzania, and the Democratic Republic of the Congo. Malaria-positive samples (N = 5465) from 39 health facilities collected between May 2022 and March 2023 were sequenced in 199 pools. RESULTS In Rwanda, K13 561H and 675V were detected in 90% and 65% of sites, with an average frequency of 19.0% (range, 0%-54.5%) and 5.0% (0%-35.5%), respectively. In Tanzania, 561H had high frequency in multiple sites. 561H appeared at 1.6% in Uganda. 561H was absent from the Democratic Republic of the Congo, although 675V was seen at low frequency. Concerningly, candidate mutations were observed: 441L, 449A, and 469F co-occurred with validated mutations, suggesting that they are arising under the same pressures. Other markers for decreased susceptibility to artemether-lumefantrine are common: P falciparum multidrug resistance protein 1 N86 at 98.0% (range, 63.3%-100%) and 184F at 47.0% (0%-94.3%) and P falciparum chloroquine resistance transporter 76T at 14.7% (0%-58.6%). Additionally, sulfadoxine-pyrimethamine-associated mutations show high frequencies. CONCLUSIONS K13 mutations are rapidly expanding in the region, further endangering control efforts with the potential of engendering partner drug resistance.
Collapse
Affiliation(s)
- Neeva Wernsman Young
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
- Center for Computational and Molecular Biology, Brown University, Providence, Rhode Island
| | - Pierre Gashema
- Center for Genomic Biology, Institut d’Enseignement Supérieur de Ruhengeri, Ruhengeri, Rwanda
| | - David Giesbrecht
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | | | - Felicien Maisha
- Department of Pediatrics, HEAL Africa Hospital, Goma, Democratic Republic of the Congo
| | - Fred Mwebembezi
- Department of Community Health, Mbarara University of Science and Technology, Uganda
| | - Rule Budodo
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Alec Leonetti
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Rebecca Crudale
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Vincent Iradukunda
- Center for Genomic Biology, Institut d’Enseignement Supérieur de Ruhengeri, Ruhengeri, Rwanda
| | - Ntwari Jean Bosco
- Center for Genomic Biology, Institut d’Enseignement Supérieur de Ruhengeri, Ruhengeri, Rwanda
| | - Rebecca I Kirby
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Ross M Boyce
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill
| | - Celine I Mandara
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | | | - Moses Ntaro
- Department of Community Health, Mbarara University of Science and Technology, Uganda
| | - Lucy C Okell
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, United Kingdom
| | - Oliver J Watson
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, United Kingdom
| | - Edgar Mulogo
- Department of Community Health, Mbarara University of Science and Technology, Uganda
| | - Deus S Ishengoma
- National Institute for Medical Research, Dar es Salaam, Tanzania
- Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam
| | - Stan Hangi
- Department of Pediatrics, HEAL Africa Hospital, Goma, Democratic Republic of the Congo
| | | | - Jean-Baptiste Mazarati
- Center for Genomic Biology, Institut d’Enseignement Supérieur de Ruhengeri, Ruhengeri, Rwanda
| | - Jonathan J Juliano
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill
| | - Jeffrey A Bailey
- Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
- Center for Computational and Molecular Biology, Brown University, Providence, Rhode Island
| |
Collapse
|
16
|
Trickey ML, Chowdury M, Bramwell G, Counihan NA, de Koning-Ward TF. Utilisation of an in vivo malaria model to provide functional proof for RhopH1/CLAG essentiality and conserved orthology with P. falciparum. J Biomed Sci 2025; 32:13. [PMID: 39894870 PMCID: PMC11789411 DOI: 10.1186/s12929-024-01105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Malaria parasites establish new permeation pathways (NPPs) at the red blood cell membrane to facilitate the transport of essential nutrients from the blood plasma into the infected host cell. The NPPs are critical to parasite survival and, therefore, in the pursuit of novel therapeutics are an attractive drug target. The NPPs of the human parasite, P. falciparum, have been linked to the RhopH complex, with the monoallelic paralogues clag3.1 and clag3.2 encoding the protein RhopH1/CLAG3 that likely forms the NPP channel-forming component. Yet curiously, the combined knockout of both clag3 genes does not completely eliminate NPP function. The essentiality of the clag3 genes is, however, complicated by three additional clag paralogs (clag2, clag8 and clag9) in P. falciparum that could also be contributing to NPP formation. METHODS Here, the rodent malaria species, P. berghei, was utilised to investigate clag essentiality since it contains only two clag genes, clagX and clag9. Allelic replacement of the regions encompassing the functional components of P. berghei clagX with either P. berghei clag9 or P. falciparum clag3.1 examined the relationship between the two P. berghei clag genes as well as functional orthology across the two species. An inducible P. berghei clagX knockout was created to examine the essentiality of the clag3 ortholog to both survival and NPP functionality. RESULTS It was revealed P. berghei CLAGX and CLAG9, which belong to two distinct phylogenetic clades, have separate non-complementary functions, and that clagX is the functional orthologue of P. falciparum clag3. The inducible clagX knockout in conjunction with a guanidinium chloride induced-haemolysis assay to assess NPP function provided the first evidence of CLAG essentiality to Plasmodium survival and NPP function in an in vivo model of infection. CONCLUSIONS This work provides valuable insight regarding the essentiality of the RhopH1 clag genes to the NPPs functionality and validates the continued investigation of the RhopH complex as a therapeutic target to treat malaria infections.
Collapse
Affiliation(s)
- Mitchell L Trickey
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Mrittika Chowdury
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Georgina Bramwell
- School of Life and Environmental Sciences, Deakin University, Geelong, Australia
| | - Natalie A Counihan
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Geelong, Australia.
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia.
| |
Collapse
|
17
|
Dereje N, Fallah MP, Shaweno T, Duga A, Mazaba ML, Raji T, Folayan MO, Ngongo N, Ndembi N, Kaseya J. Resurgence of malaria and artemisinin resistance in Africa requires a concerted response. Nat Med 2025; 31:362-363. [PMID: 39753972 DOI: 10.1038/s41591-024-03439-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Affiliation(s)
- Nebiyu Dereje
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia.
| | - Mosoka Papa Fallah
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Tamrat Shaweno
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Alemayehu Duga
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Mazyanga Lucy Mazaba
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Tajudeen Raji
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Morenike O Folayan
- Department of Child Dental Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Ngashi Ngongo
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Nicaise Ndembi
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| | - Jean Kaseya
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa, Ethiopia
| |
Collapse
|
18
|
Gare CL, Palombi IR, White AM, Chavchich M, Edstein MD, Lock A, Avery VM, Craik DJ, McMorran BJ, Lawrence N, Malins LR. Exploring the Utility of Cell-Penetrating Peptides as Vehicles for the Delivery of Distinct Antimalarial Drug Cargoes. ChemMedChem 2025; 20:e202400637. [PMID: 39379289 DOI: 10.1002/cmdc.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
The devastating impact of malaria includes significant mortality and illness worldwide. Increasing resistance of the causative parasite, Plasmodium, to existing antimalarial drugs underscores a need for additional compounds with distinct modes of action in the therapeutic development pipeline. Here we showcase peptide-drug conjugates (PDCs) as an attractive compound class, in which therapeutic or lead antimalarials are chemically conjugated to cell-penetrating peptides. This approach aims to enhance selective uptake into Plasmodium-infected red blood cells and impart additional cytotoxic actions on the intraerythrocytic parasite, thereby enabling targeted drug delivery and dual modes of action. We describe the development of PDCs featuring four compounds with antimalarial activity-primaquine, artesunate, tafenoquine and methotrexate-conjugated to three cell-penetrating peptide scaffolds with varied antiplasmodial activity, including active and inactive analogues of platelet factor 4 derived internalization peptide (PDIP), and a cyclic polyarginine peptide. Development of this diverse set of PDCs featured distinct and adaptable conjugation strategies, to produce conjugates with in vitro antiplasmodial activities ranging from low nanomolar to low micromolar potencies according to the drug cargo and bioactivity of the partner peptide. Overall, this study establishes a strategic and methodological framework for the further development of dual mode of action peptide-drug antimalarial therapeutics.
Collapse
Affiliation(s)
- Caitlin L Gare
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Isabella R Palombi
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Andrew M White
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Marina Chavchich
- Department of Drug Evaluation, Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, QLD, 4051, Australia
| | - Michael D Edstein
- Department of Drug Evaluation, Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, QLD, 4051, Australia
| | - Aaron Lock
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Brendan J McMorran
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nicole Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lara R Malins
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
19
|
Funwei RI, Olaleye A, Uyaiabasi GN, Hammed W, Obadimeji MM, Elikwu CJ, Adepoju A, Okangba C, Akinyede A, Ojurongbe O, Falade C, Walker O. Genetic profiling of Plasmodium falciparum antigenic biomarkers among asymptomatic pregnant women on intermittent preventive treatment with sulfadoxine-pyrimethamine from southwest Nigeria. Placenta 2025; 159:161-169. [PMID: 39731848 DOI: 10.1016/j.placenta.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024]
Abstract
INTRODUCTION The genetic complexity of Plasmodium falciparum is contributory to the emergence of drug resistant-parasites. Intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP) in malaria endemic settings is recommended by WHO. This study evaluated the prevalence of Plasmodium falciparum multidrug resistance-1 gene (Pfmdr-1), genetic diversity of merozoite surface proteins (msp-1, msp-2) and glutamate-rich protein (glurp) among pregnant women with sub-patent parasitaemia from southwest Nigeria. METHODS One hundred PCR-confirmed Plasmodium falciparum isolates, collected at first visit-V-1 (n = 52), delivery (n = 31) and cord blood (n = 17), were selected for analysis. The Pfmdr-1 alleles was evaluated using restriction fragment length polymorphism (RLFP), while msp-1, msp-2 and glurp genes were genotyped. Allelic frequency distribution and multiplicity of infection were calculated at p-value ≤0.05. RESULTS The Pfmdr-1 (N86/N86Y) combination was detected in 11.8 %, 61.3 % and 58.8 % (p ≤ 0.05) in V-1, Delivery and Cord isolates respectively. The N86Y haplotype was detected only in cord (5.9 %). The allelic frequency distribution for msp-1 was 244 (K1 = 81, MAD20 = 84 and RO33 = 79), and msp-2; 110 alleles, representing 43.6 % (FC27) and 56.4 % (3D7). While glurp expressed 25 alleles, 84 % (V-1), 12 % (delivery) and 4 % (cord), respectively (p ≤ 0.05). The msp-1 and msp-2 recorded higher MOIs than glurp. DISCUSSION Genetically diverse P. falciparum strains with Pfmdr-1 mutant alleles were detected in pregnant women with sub-patent parasitaemia in southwest Nigeria, which may reduce IPTp-SP effectiveness. Thus, continuous molecular surveillance of resistant-parasites to sulphadoxine-pyrimethamine and ACTs is essential.
Collapse
Affiliation(s)
- R I Funwei
- Department of Pharmacology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria.
| | - A Olaleye
- Two Hills Medical Clinic, Two Hills, Alberta, Canada; Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria
| | - G N Uyaiabasi
- Department of Pharmacology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria
| | - W Hammed
- Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Department of Pharmacology, Therapeutics and Toxicology, University of Lagos, Idiaraba, Nigeria
| | - M M Obadimeji
- Research Innovation and International Cooperation (RIIC) Unit, Babcock University, Ogun State, Nigeria
| | - C J Elikwu
- Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Department of Medical Microbiology, Babcock University, Ilishan-Remo, Ogun, Nigeria
| | - A Adepoju
- Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Centre for Research, Innovation and Development, Babcock University, Ilishan-Remo, Ogun, Nigeria
| | - C Okangba
- Department of Medical Microbiology, Babcock University, Ilishan-Remo, Ogun, Nigeria
| | - A Akinyede
- Department of Pharmacology, Therapeutics and Toxicology, University of Lagos, Idiaraba, Nigeria
| | - O Ojurongbe
- Department of Medical Microbiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - C Falade
- Institute for Advanced Medical Research and Training, University of Ibadan, Ibadan, Oyo, Nigeria
| | - O Walker
- Department of Pharmacology, Babcock University, Ilishan-Remo, Ogun, Nigeria; Centre for Advanced Medical Research and Biotechnology, Babcock University, Ilishan-Remo, Ogun, Nigeria
| |
Collapse
|
20
|
Chaves CRS, da Silva C, Salamandane A, Nogueira F. Mapping Antimalarial Drug Resistance in Mozambique: A Systematic Review of Plasmodium falciparum Genetic Markers Post-ACT Implementation. Int J Mol Sci 2024; 25:13645. [PMID: 39769406 PMCID: PMC11728251 DOI: 10.3390/ijms252413645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Malaria continues to be a significant public health burden in many tropical and subtropical regions. Mozambique ranks among the top countries affected by malaria, where it is a leading cause of morbidity and mortality, accounting for 29% of all hospital deaths in the general population and 42% of deaths amongst children under five. This review presents a comparative analysis of data on five critical genes associated with antimalarial drug resistance: pfmdr1, pfcrt, pfk13, pfdhfr, and pfdhps, along with the copy number variation (CNV) in genes pfmdr1 and pfpm2/3. These are genes associated with parasite response to antimalarials currently used to treat uncomplicated P. falciparum malaria in Mozambique. The review synthesizes data collected from published studies conducted in Mozambique after the introduction of artemisinin-based combination therapies (ACTs) (2006) up to June 2024, highlighting the presence or absence of mutations in these genes across Mozambique. We aimed at mapping the prevalence and distribution of these molecular markers across the country in order to contribute to the development of targeted interventions to sustain the efficacy of malaria treatments in Mozambique. Four databases were used to access the articles: PubMed, Science Direct, Scopus, and Google scholar. The search strategy identified 132 studies addressing malaria and antimalarial resistance. Of these, 112 were excluded for various reasons, leaving 20 studies to be included in this review. Children and pregnant women represent the majority of target groups in studies on all types of antimalarials. Most studies (87.5%) were conducted in the provinces of Maputo and Gaza. The primary alleles reported were pfcrt CVMNK, and in the most recent data, its wild-type form was found in the majority of patients. A low prevalence of mutations in the pfk13 gene was identified reflecting the effectiveness of ACTs. In pfk13, only mutation A578S was reported in Niassa and Tete. CNVs were observed in studies carried out in the south of Mozambique, with a frequency of 1.1-5.1% for pfmdr1 and a frequency of 1.1-3.4% for pfpm2. This review indicates that molecular markers linked to malaria resistance show considerable variation across provinces in Mozambique, with most up-to-date data accessible for Maputo and Gaza. In contrast, provinces such as Zambezia and Inhambane have limited data on several genes, while Nampula lacks data on all drug resistance markers.
Collapse
Affiliation(s)
- Celso Raul Silambo Chaves
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal; (C.R.S.C.); (C.d.S.)
| | - Clemente da Silva
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal; (C.R.S.C.); (C.d.S.)
| | - Acácio Salamandane
- Faculdade de Ciências de Saúde, Universidade Lúrio, Campus Universitário de Marrere, Nampula 4250, Mozambique;
| | - Fatima Nogueira
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal; (C.R.S.C.); (C.d.S.)
| |
Collapse
|
21
|
White NJ, Chotivanich K. Artemisinin-resistant malaria. Clin Microbiol Rev 2024; 37:e0010924. [PMID: 39404268 PMCID: PMC11629630 DOI: 10.1128/cmr.00109-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024] Open
Abstract
SUMMARYThe artemisinin antimalarials are the cornerstone of current malaria treatment. The development of artemisinin resistance in Plasmodium falciparum poses a major threat to malaria control and elimination. Recognized first in the Greater Mekong subregion of Southeast Asia nearly 20 years ago, artemisinin resistance has now been documented in Guyana, South America, in Papua New Guinea, and most recently, it has emerged de novo in East Africa (Rwanda, Uganda, South Sudan, Tanzania, Ethiopia, Eritrea, and eastern DRC) where it has now become firmly established. Artemisinin resistance is associated with mutations in the propeller region of the PfKelch gene, which play a causal role, although the parasites' genetic background also makes an important contribution to the phenotype. Clinically, artemisinin resistance manifests as reduced parasiticidal activity and slower parasite clearance and thus an increased risk of treatment failure following artemisinin-based combination therapy (ACT). This results from the loss of artemisinin activity against the younger circulating ring stage parasites. This loss of activity is likely to diminish the life-saving advantage of artesunate in the treatment of severe falciparum malaria. Gametocytocidal and thus transmission blocking activities are also reduced. At current levels of resistance, artemisinin-resistant parasites still remain susceptible at the trophozoite stage of asexual development, and so, artemisinin still contributes to the therapeutic response. As ACTs are the most widely used antimalarial drugs in the world, it is essential from a malaria control perspective that ACT cure rates remain high. Better methods of identifying uncomplicated hyperparasitemia, the main cause of ACT treatment failure, are required so that longer courses of treatment can be given to these high-risk patients. Reducing the use of artemisinin monotherapies will reduce the continued selection pressure which could lead potentially to higher levels of artemisinin resistance. Triple artemisinin combination therapies should be deployed as soon as possible to protect the ACT partner drugs and thereby delay the emergence of higher levels of resistance. As new affordable antimalarial drugs are still several years away, the control of artemisinin resistance must depend on the better use of available tools.
Collapse
Affiliation(s)
- N. J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - K. Chotivanich
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
22
|
Nayak S, Peto TJ, Kucharski M, Tripura R, Callery JJ, Quang Huy DT, Gendrot M, Lek D, Nghia HDT, van der Pluijm RW, Dong N, Long LT, Vongpromek R, Rekol H, Hoang Chau N, Miotto O, Mukaka M, Dhorda M, von Seidlein L, Imwong M, Roca X, Day NPJ, White NJ, Dondorp AM, Bozdech Z. Population genomics and transcriptomics of Plasmodium falciparum in Cambodia and Vietnam uncover key components of the artemisinin resistance genetic background. Nat Commun 2024; 15:10625. [PMID: 39639029 PMCID: PMC11621345 DOI: 10.1038/s41467-024-54915-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
The emergence of Plasmodium falciparum parasites resistant to artemisinins compromises the efficacy of Artemisinin Combination Therapies (ACTs), the global first-line malaria treatment. Artemisinin resistance is a complex genetic trait in which nonsynonymous SNPs in PfK13 cooperate with other genetic variations. Here, we present population genomic/transcriptomic analyses of P. falciparum collected from patients with uncomplicated malaria in Cambodia and Vietnam between 2018 and 2020. Besides the PfK13 SNPs, several polymorphisms, including nonsynonymous SNPs (N1131I and N821K) in PfRad5 and an intronic SNP in PfWD11 (WD40 repeat-containing protein on chromosome 11), appear to be associated with artemisinin resistance, possibly as new markers. There is also a defined set of genes whose steady-state levels of mRNA and/or splice variants or antisense transcripts correlate with artemisinin resistance at the base level. In vivo transcriptional responses to artemisinins indicate the resistant parasite's capacity to decelerate its intraerythrocytic developmental cycle (IDC), which can contribute to the resistant phenotype. During this response, PfRAD5 and PfWD11 upregulate their respective alternatively/aberrantly spliced isoforms, suggesting their contribution to the protective response to artemisinins. PfRAD5 and PfWD11 appear under selective pressure in the Greater Mekong Sub-region over the last decade, suggesting their role in the genetic background of the artemisinin resistance.
Collapse
Affiliation(s)
- Sourav Nayak
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Thomas J Peto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Michal Kucharski
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Amsterdam UMC, University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Rupam Tripura
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - James J Callery
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Duong Tien Quang Huy
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Mathieu Gendrot
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Dysoley Lek
- Centre for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
- National Institute for Public Health, Phnom Penh, Cambodia
| | - Ho Dang Trung Nghia
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
- Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Rob W van der Pluijm
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institut Pasteur, Université Paris Cité, G5 Infectious Disease Epidemiology and Analytics, Paris, France
| | - Nguyen Dong
- Khanh Hoa Hospital for Tropical diseases, Ho Chi Minh City, Khanh Hoa province, Vietnam
| | - Le Thanh Long
- Phuoc Long Hospital, Ho Chi Minh City, Binh Phuoc province, Vietnam
| | - Ranitha Vongpromek
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- WorldWide Antimalarial Resistance Network - Asia-Pacific Regional Centre, Bangkok, Thailand
| | - Huy Rekol
- Amsterdam UMC, University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | | | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mavuto Mukaka
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mehul Dhorda
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- WorldWide Antimalarial Resistance Network - Asia-Pacific Regional Centre, Bangkok, Thailand
| | - Lorenz von Seidlein
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Mallika Imwong
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam.
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
23
|
Rosenthal MR, Vijayrajratnam S, Firestone TM, Ng CL. Enhanced cell stress response and protein degradation capacity underlie artemisinin resistance in Plasmodium falciparum. mSphere 2024; 9:e0037124. [PMID: 39436072 PMCID: PMC11580438 DOI: 10.1128/msphere.00371-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/23/2024] Open
Abstract
Malaria remains a global health burden, killing over half a million people each year. Decreased therapeutic efficacy to artemisinin, the most efficacious antimalarial, has been detected in sub-Saharan Africa, a worrying fact given that over 90% of deaths occur on this continent. Mutations in Kelch13 are the most well-established molecular marker for artemisinin resistance, but these do not explain all artemisinin-resistant isolates. Understanding the biological underpinnings of drug resistance is key to curbing the emergence and spread of artemisinin resistance. Artemisinin-mediated non-specific alkylation leads to the accumulation of misfolded and damaged proteins and activation of the parasite unfolded protein response (UPR). In addition, the parasite proteasome is vital to artemisinin resistance, as we have previously shown that chemical inhibition of the proteasome or mutations in the β2 proteasome subunit increase parasite susceptibility to dihydroartemisinin (DHA), the active metabolite of artemisinins. Here, we investigate parasites with mutations at the Kelch13 and/or 19S and 20S proteasome subunits with regard to UPR regulation and proteasome activity in the context of artemisinin resistance. Our data show that perturbing parasite proteostasis kills parasites, early parasite UPR signaling dictates DHA survival outcomes, and DHA susceptibility correlates with impairment of proteasome-mediated protein degradation. Importantly, we show that functional proteasomes are required for artemisinin resistance in a Kelch13-independent manner, and compound-selective proteasome inhibition demonstrates why artemisinin-resistant Kelch13 mutants remain susceptible to the related antimalarial peroxide OZ439. These data provide further evidence for targeting the parasite proteasome and UPR to overcome existing artemisinin resistance.IMPORTANCEDecreased therapeutic efficacy represents a major barrier to malaria treatment control strategies. The malaria proteasome and accompanying unfolded protein response are crucial to artemisinin resistance, revealing novel antimalarial therapeutic strategies.
Collapse
Affiliation(s)
- Melissa R. Rosenthal
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sukhithasri Vijayrajratnam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tessa M. Firestone
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Caroline L. Ng
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biology, University of Omaha, Omaha, Nebraska, USA
| |
Collapse
|
24
|
Kojom Foko LP, Hawadak J, Eboumbou Moukoko CE, Das A, Singh V. Genetic analysis of the circumsporozoite gene in Plasmodium falciparum isolates from Cameroon: Implications for efficacy and deployment of RTS,S/AS01 vaccine. Gene 2024; 927:148744. [PMID: 38964492 DOI: 10.1016/j.gene.2024.148744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Current understanding of genetic polymorphisms and natural selection in Plasmodium falciparum circumsporozoite (PfCSP), the leading malaria vaccine, is crucial for the development of next-generation vaccines, and such data is lacking in Africa. Blood samples were collected among Plasmodium-infected individuals living in four Cameroonian areas (Douala, Maroua, Mayo-Oulo, Pette). DNA samples were amplified using nested PCR protocols, sequenced, and BLASTed. Single nucleotide polymorphisms (SNPs) were analysed in each PfCSP region, and their impact on PfCSP function/structure was predicted in silico. The N-terminal region showed a limited polymorphism with four haplotypes, and three novel SNPs (N68Y, R87W, K93E) were found. Thirty-five haplotypes were identified in the central region, with several variants (e.g., NVNP and KANP). The C-terminal region was also highly diverse, with 25 haplotypes and eight novel SNPs (N290D, N308I, S312G, K317A, V344I, D356E, E357L, D359Y). Most polymorphic codon sites were mainly observed in the Th2R subregion in isolates from Douala and Pette. The codon site 321 was under episodic positive selection. One novel (E357L) and three known (K322I, G349D, D359Y) SNPs show an impact on function/structure. This study showed extensive genetic diversity with geographical patterns and evidence of the selection of Cameroonian PfCSP central and C-terminal regions.
Collapse
Affiliation(s)
| | - Joseph Hawadak
- Parasite & Host Biology Group, National Institute of Malaria Research, New-Delhi, India
| | - Carole Else Eboumbou Moukoko
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Cameroon; Malaria Research Unit, Centre Pasteur Cameroon, Yaoundé, Cameroon; Laboratory of Parasitology, Mycology and Virology, Postgraduate Training Unit for Health Sciences, Postgraduate School for Pure and Applied Sciences, The University of Douala, Cameroon
| | - Aparup Das
- Division of Vector Borne Diseases, National Institute of Research in Tribal Health, Madhya Pradesh, India
| | - Vineeta Singh
- Parasite & Host Biology Group, National Institute of Malaria Research, New-Delhi, India; Academy of Scientific and Innovative Research, Ghaziabad, India.
| |
Collapse
|
25
|
Rosenthal PJ. Artemisinin Partial Resistance and the Treatment of Severe Malaria. JAMA 2024:2826318. [PMID: 39540803 DOI: 10.1001/jama.2024.23100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
|
26
|
Baina MT, Djontu JC, Mbama Ntabi JD, Mfoutou Mapanguy CC, Lissom A, Vouvoungui CJ, Boumpoutou RK, Mouanga AM, Nguimbi E, Ntoumi F. Polymorphisms in the Pfcrt, Pfmdr1, and Pfk13 genes of Plasmodium falciparum isolates from southern Brazzaville, Republic of Congo. Sci Rep 2024; 14:27988. [PMID: 39543235 PMCID: PMC11564878 DOI: 10.1038/s41598-024-78670-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
This study aimed to analyze polymorphisms in Pfcrt, Pfmdr1, and Pfk13 genes' markers of resistance to Artemisinin-based combination therapy (ACT), in Plasmodium falciparum isolates from southern Brazzaville, 15 years after the adoption of ACT in the Republic of Congo. A total of 369 microscopy-confirmed malaria-infected individuals were enrolled from March to October 2021 in the community and in health facilities during a cross-sectional study. The K76T mutation in the Pfcrt gene, N86Y and Y184F mutations in the Pfmdr1 gene were investigated using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) while the codons region (1005-1300) of the Pfmdr1gene, and Pfk13 gene were sequenced. The prevalences of K76T, N86Y, Y184F mutations were 26.0%, 6.8%, and 27.7%, respectively. However, no mutations were detected in codons 1034, 1042, and 1246 of the Pfmdr1 gene. None of the mutations previously associated with artemisinin-based resistance were detected in the Pfk13 gene. The results reveal a significant decrease in the prevalence of K76T, N86Y, Y184F mutations, in Plasmodium falciparum isolates following the change of therapeutic policy. As artemisinin resistance is emerging throughout Africa, continued surveillance for early detection of these mutations and relevant partner markers of drug resistance are recommended in the Republic of Congo.
Collapse
Affiliation(s)
- Marcel Tapsou Baina
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | - Jean Claude Djontu
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo.
- Biotechnology Center, University of Yaounde I, Yaounde, Cameroon.
| | - Jacques Dollon Mbama Ntabi
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | - Claujens Chastel Mfoutou Mapanguy
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | - Abel Lissom
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Department of Zoology, Faculty of Science, University of Bamenda, Bamenda, Cameroon
| | - Christevy Jeannhey Vouvoungui
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | | | - Alain Maxime Mouanga
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo
- Faculté des Sciences de la santé, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | - Etienne Nguimbi
- Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Republic of Congo
| | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo.
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
27
|
Delandre O, Pradines B, Javelle E. Dihydroartemisinin-Piperaquine Combination in the Treatment of Uncomplicated Plasmodium falciparum Malaria: Update on Clinical Failures in Africa and Tools for Surveillance. J Clin Med 2024; 13:6828. [PMID: 39597971 PMCID: PMC11594973 DOI: 10.3390/jcm13226828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Dihydroartemisinin (or artenimol)-piperaquine is one of the six artemisinin-based combination therapies recommended in uncomplicated malaria treatment. However, artemisinin partial resistance has been reported in Cambodia, Laos, Vietnam, India, and, recently, in Africa. Polymorphisms in the Pfk13 gene have been described as molecular markers of artemisinin resistance and the amplification of the plasmepsine II/III (Pfpmp2/Pfpmp3) gene has been associated with piperaquine resistance. However, some therapeutic failures with this combination remain unexplained by strains' characterization. We provide an overview on the use of dihydroartemisinin-piperaquine in malaria treatment and discuss tools available to monitor its efficacy.
Collapse
Affiliation(s)
- Océane Delandre
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (B.P.); (E.J.)
- Aix Marseille Univ, SSA, AP-HM, RITMES, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (B.P.); (E.J.)
- Aix Marseille Univ, SSA, AP-HM, RITMES, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Emilie Javelle
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 13005 Marseille, France; (B.P.); (E.J.)
- Aix Marseille Univ, SSA, AP-HM, RITMES, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France
- Centre National de Référence du Paludisme, 13005 Marseille, France
| |
Collapse
|
28
|
Stopper D, de Carvalho LP, de Souza ML, Kponomaizoun CE, Winzeler EA, Held J, Hansen FK. Development of peptoid-based heteroaryl-decorated histone deacetylase (HDAC) inhibitors with dual-stage antiplasmodial activity. Eur J Med Chem 2024; 277:116782. [PMID: 39208744 DOI: 10.1016/j.ejmech.2024.116782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Dynamics of epigenetic modifications such as acetylation and deacetylation of histone proteins have been shown to be crucial for the life cycle development and survival of Plasmodium falciparum, the deadliest malaria parasite. In this study, we present a novel series of peptoid-based histone deacetylase (HDAC) inhibitors incorporating nitrogen-containing bicyclic heteroaryl residues as a new generation of antiplasmodial peptoid-based HDAC inhibitors. We synthesized the HDAC inhibitors by an efficient multicomponent protocol based on the Ugi four-component reaction. The subsequent screening of 16 compounds from our mini-library identified 6i as the most promising candidate, demonstrating potent activity against asexual blood-stage parasites (IC50Pf3D7 = 30 nM; IC50PfDd2 = 98 nM), low submicromolar activity against liver-stage parasites (IC50PbEEF = 0.25 μM), excellent microsomal stability (t1/2 > 60 min), and low cytotoxicity to HEK293 cells (IC50 = 136 μM).
Collapse
Affiliation(s)
- Daniel Stopper
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | | | - Mariana Laureano de Souza
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Cindy-Esther Kponomaizoun
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, 72074, Tübingen, Germany; German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Finn K Hansen
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany.
| |
Collapse
|
29
|
Ishengoma DS, Mandara CI, Bakari C, Fola AA, Madebe RA, Seth MD, Francis F, Buguzi CC, Moshi R, Garimo I, Lazaro S, Lusasi A, Aaron S, Chacky F, Mohamed A, Njau RJA, Kitau J, Rasmussen C, Bailey JA, Juliano JJ, Warsame M. Evidence of artemisinin partial resistance in northwestern Tanzania: clinical and molecular markers of resistance. THE LANCET. INFECTIOUS DISEASES 2024; 24:1225-1233. [PMID: 39159633 PMCID: PMC11511676 DOI: 10.1016/s1473-3099(24)00362-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND In 2021, nationwide malaria molecular surveillance revealed a high prevalence of a validated artemisinin resistance marker, the kelch13 (k13) Arg561His mutation, in the Kagera region of northwestern Tanzania. We aimed to investigate the efficacy of artemether-lumefantrine and artesunate-amodiaquine and to confirm the presence of artemisinin partial resistance (ART-R) in the Karagwe district of this region. METHODS This single-arm, therapeutic efficacy study was carried out at the Bukangara dispensary in the Karagwe district of the Kagera region in northwestern Tanzania. Eligible participants were aged between 6 months and 120 months, had confirmed Plasmodium falciparum asexual parasitaemia, and met other inclusion criteria according to WHO's standard protocol. Participants were enrolled, treated sequentially with either artemether-lumefantrine or artesunate-amodiaquine, and assessed clinically and parasitologically for 28 days as per WHO protocol. Parasitaemia was measured every 8 h until day 3, on day 7, and then during weekly follow-up visits until day 28. Mutations in the k13 gene and extended haplotypes with the mutations were analysed, and comparisons were made with previous samples collected in the same region of Kagera and in Rwanda and southeast Asia. The primary endpoint was PCR-corrected cure rate. FINDINGS Between April 29 and Sept 1, 2022, 343 patients were screened, of whom 176 were enrolled: 88 in each treatment group. The PCR-corrected cure rate was 98% (95% CI 91-100) in the artemether-lumefantrine group and 100% (96-100) in the artesunate-amodiaquine group. Persistent parasitaemia on day 3 occurred in 11 (13%) of 88 patients in the artemether-lumefantrine group and 17 (19%) of 88 patients in the artesunate-amodiaquine group. Arg561His mutations on day 0 and parasitaemia on day 3 were reported in eight (9%) of 87 patients in the artemether-lumefantrine group and ten (12%) of 86 patients in the artesunate-amodiaquine group. The median parasite clearance half-life in patients harbouring parasites with Arg561His mutation was 6·1 h in the artemether-lumefantrine group and 6·0 h in the artesunate-amodiaquine group. Parasites with the Arg561His mutation were not similar to those from southeast Asia and Rwanda but had similar haplotypes to parasites reported in the same Tanzanian region of Kagera in 2021. INTERPRETATION This study confirms the presence of ART-R in Tanzania, although artemether-lumefantrine and artesunate-amodiaquine showed high efficacy. A context-specific response strategy and vigilance to detect the reduced efficacy of current antimalarial treatments and ART-R in other parts of the country are urgently needed. FUNDING The Bill & Melinda Gates Foundation and the US National Institutes of Health.
Collapse
Affiliation(s)
- Deus S Ishengoma
- National Institute for Medical Research, Dar es Salaam, Tanzania; Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam, Tanzania.
| | - Celine I Mandara
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Catherine Bakari
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Abebe A Fola
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA; Centre for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Rashid A Madebe
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Misago D Seth
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Filbert Francis
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Creyton C Buguzi
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Ramadhan Moshi
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Issa Garimo
- National Malaria Control Program, Dodoma, Tanzania
| | | | | | | | - Frank Chacky
- National Malaria Control Program, Dodoma, Tanzania
| | - Ally Mohamed
- National Malaria Control Program, Dodoma, Tanzania
| | - Ritha J A Njau
- Department of Parasitology and Medical Entomology, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Jovin Kitau
- World Health Organization Country Office, Dar es Salaam, Tanzania
| | | | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA; Centre for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Jonathan J Juliano
- Division of Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marian Warsame
- Department of Public Health and Community Medicine, Gothenburg University, Gothenburg, Sweden; Research Unit in Rector's Office, Benadir University, Mogadishu, Somalia
| |
Collapse
|
30
|
Petro DA, Shaban N, Aaron S, Chacky F, Lazaro S, Boni MF, Ishengoma DS. Geospatial Analysis of Malaria Burden in Kagera Region, Northwestern Tanzania Using Health Facility and Community Survey Data. Open Forum Infect Dis 2024; 11:ofae609. [PMID: 39507880 PMCID: PMC11540141 DOI: 10.1093/ofid/ofae609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background Malaria transmission in Tanzania has declined significantly over the last 2 decades due to scaled-up control interventions. However, recent confirmation of artemisinin partial resistance (ART-R) in Kagera region in northwest Tanzania threatens the ongoing efforts to eliminate malaria in the country. This study was conducted according to the World Health Organization recommendation to generate evidence of malaria burden in areas with confirmed ART-R as the first step before developing a response strategy to the resistance. Methods We assessed the local burden of malaria in Kagera region by geospatial analysis, using data collected retrospectively from health facilities and community surveys from 2015 to 2023 to identify malaria hot spots. Results From 2017 to 2023, a total of 8 124 363 suspected malaria cases were reported by health facilities, and 2 983 717 (36.7% [95% range across wards, 22.7%-50.7%]) tested positive by rapid diagnostic tests. Test positivity rates were similar among patients aged <5 years (33.1% [95% range, 19.7%-46.5%]) and those aged ≥5 years (33.7% [21.0%-46.5%]). The malaria prevalence was 10.0% (95% range across wards, 5.1%-14.9% [n = 84 999 of 853 761]) in pregnant women and 26.1% (11.7%-40.6% [n = 3409 of 13 065]) in schoolchildren. Despite high temporal variations, we identified hot spots and cold spots, including persistently high burden in 69 of 192 wards (35.9%). Conclusions The malaria burden in Kagera exhibited high temporal and spatial heterogeneity, with schoolchildren showing the highest prevalence. This demographic pattern underlines the need for targeted interventions and provides evidence for developing an ART-R response for the region.
Collapse
Affiliation(s)
- Daniel A Petro
- Department of Mathematics, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Nyimvua Shaban
- Department of Mathematics, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Sijenunu Aaron
- Office of the Chief Medical Officer, Ministry of Health, National Malaria Control Programme, Dodoma, Tanzania
| | - Frank Chacky
- Office of the Chief Medical Officer, Ministry of Health, National Malaria Control Programme, Dodoma, Tanzania
| | - Samuel Lazaro
- Office of the Chief Medical Officer, Ministry of Health, National Malaria Control Programme, Dodoma, Tanzania
| | - Maciej F Boni
- Department of Biology, Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Deus S Ishengoma
- Genomics Laboratory, National Institute for Medical Research, Dar es Salaam, Tanzania
- Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam, Tanzania
| |
Collapse
|
31
|
Schnoz A, Beuret C, Concu M, Hosch S, Rutaihwa LK, Golumbeanu M, Nsanzabana C. Genotyping methods to distinguish Plasmodium falciparum recrudescence from new infection for the assessment of antimalarial drug efficacy: an observational, single-centre, comparison study. THE LANCET. MICROBE 2024; 5:100914. [PMID: 39426395 DOI: 10.1016/s2666-5247(24)00153-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 03/24/2024] [Accepted: 06/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Distinguishing Plasmodium falciparum recrudescence from new infections is crucial for the assessment of antimalarial drug efficacy against P falciparum. We aimed to compare the efficacy of different genotyping methods to assess their effect on drug efficacy estimates, particularly in patients from high-transmission settings with polyclonal infections. METHODS In this head-to-head comparison study, we compared five different genotyping methods currently used: fast capillary electrophoresis (F-CE) using msp1, msp2, and glurp; high-resolution capillary electrophoresis (H-CE) using msp1, msp2, and glurp; H-CE using microsatellites; targeted amplicon deep sequencing (TADS) using single nucleotide polymorphism (SNP)-rich markers; and high-resolution melting (HRM) analysis using msp1 and msp2. We assessed their sensitivity in detecting minority clones in polyclonal infections, their reproducibility, and the genetic diversity of the markers used. Our study used four well characterised P falciparum laboratory strains mixed in varying ratios, and 20 paired samples collected from an in-vivo clinical trial. The experiments were performed at the Swiss Tropical and Public Health Institute in Basel, Switzerland between May 5, 2020, and Aug 23, 2021. FINDINGS H-CE using msp1 and msp2 and TADS revealed the highest sensitivity in detecting minority clones (up to ratios of 1:100 for H-CE and 50:1:1:1 for TADS in the FCB1:HB3 and 3D7:K1:HB3:FCB1 laboratory strain mixtures, respectively), highest reproducibility (intra-assay: 99% and 91% for H-CE and TADS, respectively; inter-assay: 98% and 92% for H-CE and TADS, respectively), and highest genetic diversity in the used markers (up to 36 and 32 unique genotypes in 20 paired samples for H-CE using msp2 and TADS using cpmp, respectively). Microsatellites assessed by H-CE had a lower genetic diversity compared with msp1, msp2, and glurp assessed by H-CE and the SNP-rich markers assessed by TADS, with a maximum of 13 unique genotypes, and some genotypes having allelic frequencies larger than 30%. Markers used by TADS gave the most consistent results in distinguishing recrudescence from new infection across all methods (in 18 of 20 pairs of samples vs 15 of 20 pairs for H-CE). INTERPRETATION WHO currently recommends replacing glurp with microsatellites. However, in this study, the replacement of glurp with microsatellites did not change the genotyping outcome, probably due to the lower genetic diversity of microsatellites. More studies with large sample sizes are required to identify the most suitable microsatellites that could replace glurp. Our study indicates that TADS should be considered the gold standard for genotyping to distinguish recrudescence from new infection, and that it should be used to validate other methods. FUNDING Swiss Tropical and Public Health Institute.
Collapse
Affiliation(s)
- Annina Schnoz
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Carla Beuret
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Maura Concu
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Salome Hosch
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Liliana K Rutaihwa
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Monica Golumbeanu
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Christian Nsanzabana
- Department of Medicine, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
32
|
Kucharski M, Nayak S, Gendrot M, Dondorp AM, Bozdech Z. Peeling the onion: how complex is the artemisinin resistance genetic trait of malaria parasites? Trends Parasitol 2024; 40:970-986. [PMID: 39358163 DOI: 10.1016/j.pt.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The genetics of Plasmodium as an intracellular, mostly haploid, sexually reproducing, eukaryotic organism with a complex life cycle, presents unprecedented challenges in studying drug resistance. This article summarizes current knowledge on the genetic basis of artemisinin resistance (AR) - a main component of current drug therapies for falciparum malaria. Although centered on nonsynonymous single-nucleotide polymorphisms (nsSNPs), we describe multifaceted resistance mechanisms as part of a complex, cumulative genetic trait that involves regulation of expression by a wide array of polymorphisms in noncoding regions. These genetic variations alter transcriptome profiles linked to Plasmodium's development and population dynamics, ultimately influencing the emergence and spread of the resistance.
Collapse
Affiliation(s)
- Michal Kucharski
- School of Biological Sciences, Nanyang Technological University, Singapore; Amsterdam UMC, University of Amsterdam, Department of Global Health, Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Sourav Nayak
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Mathieu Gendrot
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Arjen M Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore; Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Makau M, Kanoi BN, Mgawe C, Maina M, Bitshi M, Too EK, Naruse TK, Abkallo HM, Waweru H, Adung'o F, Kaneko O, Gitaka J. Presence of Plasmodium falciparum strains with artemisinin-resistant K13 mutation C469Y in Busia County, Western Kenya. Trop Med Health 2024; 52:72. [PMID: 39425178 PMCID: PMC11488207 DOI: 10.1186/s41182-024-00640-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/29/2024] [Indexed: 10/21/2024] Open
Abstract
Malaria remains a key health and economic problem, particularly in sub-Saharan Africa. The emergence of artemisinin drug resistance (ART-R) parasite strains poses a serious threat to the control and elimination of this scourge. This is because artemisinin-based combination therapies (ACTs) remain the first-line treatment in the majority of malaria-endemic regions in Sub-Saharan Africa. Certain single-nucleotide polymorphisms in the propeller domains of Plasmodium falciparum Kelch 13 protein (K13) have been associated with delayed parasite clearance in vivo and in vitro. These mutations serve as vital molecular markers for tracking the emergence and dispersion of resistance. Recently, there have been increasing reports of the emergence and spread of P. falciparum ART-R parasites in the Eastern Africa region. This necessitates continued surveillance to best inform mitigation efforts. This study investigated the presence of all reported mutations of K13 propeller domains in the parasite population in Busia County, Kenya, a known malaria-endemic region. Two hundred twenty-six participants with microscopically confirmed uncomplicated malaria were recruited for this study. They were treated with artemether-lumefantrine under observation for the first dose, and microscopic examination was repeated 1 day later after ensuring the participants had taken the second and third doses. P. falciparum DNA from all samples underwent targeted amplification of the K13 gene using a semi-nested PCR approach, followed by Sanger sequencing. The recently validated ART-R K13 mutation C469Y was identified in three samples. These three samples were among 63 samples with a low reduction in parasitemia on day 1, suggesting day 1 parasitemia reduction rate is a useful parameter to enrich the ART-R parasites for further analysis. Our findings highlight the need for continuous surveillance of ART-R in western Kenya and the region to determine the spread of ART-R and inform containment.
Collapse
Affiliation(s)
- Mark Makau
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Bernard N Kanoi
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Calvin Mgawe
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
| | - Michael Maina
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
| | - Mimie Bitshi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Edwin K Too
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Taeko K Naruse
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Hussein M Abkallo
- Animal and Human Health Program, International Livestock Research Institute (ILRI), Nairobi, Kenya
| | - Harrison Waweru
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya
| | - Ferdinand Adung'o
- Centre for Infectious and Parasitic Diseases Control Research, Kenya Medical Research Institute, Busia, Kenya
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - Jesse Gitaka
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika, Kenya.
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya.
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
34
|
Juliano JJ, Giesbrecht DJ, Simkin A, Fola AA, Lyimo BM, Pereus D, Bakari C, Madebe RA, Seth MD, Mandara CI, Popkin-Hall ZR, Moshi R, Mbwambo RB, Niaré K, MacInnis B, Francis F, Mbwambo D, Garimo I, Chacky F, Aaron S, Lusasi A, Molteni F, Njau RJA, Nhiga SL, Mohamed A, Bailey JA, Ishengoma DS. Prevalence of mutations associated with artemisinin partial resistance and sulfadoxine-pyrimethamine resistance in 13 regions in Tanzania in 2021: a cross-sectional survey. THE LANCET. MICROBE 2024; 5:100920. [PMID: 39159629 PMCID: PMC11464622 DOI: 10.1016/s2666-5247(24)00160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND The emergence of the artemisinin partial resistance (ART-R) mutation in the Plasmodium falciparum kelch13 gene (k13), Arg561His, in Rwanda and the regional presence of polymorphisms affecting sulfadoxine-pyrimethamine have raised concern in neighbouring Tanzania. The goal of this study was to assess the status of antimalarial resistance in Tanzania, with a focus on the border with Rwanda, to understand the distribution of the Arg561His mutation, partner drug resistance, and resistance to chemoprevention drugs. METHODS In this cross-sectional survey, capillary dried blood spots were collected from malaria positive asymptomatic individuals in the community and symptomatic individuals in health facilities aged 6 months and older, in 13 regions of mainland Tanzania from Jan 31 to June 26, 2021. Exclusion criteria included residence of the areas other than the target sites, presenting to the health facility for care and treatment of conditions other than malaria, and not providing informed consent. Samples were assessed for antimalarial resistance polymorphisms and genetic relatedness using molecular inversion probes targeting P falciparum and short-read whole-genome sequencing. The primary outcome was the prevalence of molecular markers of antimalarial resistance at the region level, as well as at the district level in Kagera, a region in the northwest of the country at the border with Rwanda. FINDINGS 6855 (88·1%) of 7782 capillary dried blood spot samples collected were successfully genotyped. The overall prevalence of k13 Arg561His in Kagera was 7·7% (90% CI 6·0-9·4; 50 of 649), with the highest prevalence in the districts near the Rwandan border (22·8% [31 of 136] in Karagwe, 14·4% [17 of 118]) in Kyerwa, and 1·4% [two of 144] in Ngara). k13 Arg561His was uncommon in the other regions. Haplotype analysis suggested that some of these parasites are related to isolates collected in Rwanda in 2015, supporting regional spread of Arg561His. However, a novel k13 Arg561His haplotype was observed, potentially indicating a second origin in the region. Other validated k13 resistance polymorphisms (one Arg622Ile and two Ala675Val isolates) were also identified. A region of prevalent dihydrofolate reductase Ile164Leu mutation, associated with sulfadoxine-pyrimethamine resistance, was also identified in Kagera (15·2% [12·6-17·8%]; 80 of 526). The mutant crt Lys76Thr mutation, associated with chloroquine and amodiaquine resistance, was uncommon, occurring only in 75 of 2861 genotyped isolates, whereases the wild-type mdr1 Asn86Tyr allele, associated with reduced sensitivity to lumefantrine, was found in 99·7% (3819 of 3830) of samples countrywide. INTERPRETATION These findings show that the k13 Arg561His mutation is common in northwest Tanzania and that multiple emergences of ART-R, similar as to what was seen in southeast Asia, have occurred. Mutations associated with high levels of sulfadoxine-pyrimethamine resistance are common. These results raise concerns about the long-term efficacy of artemisinin and chemoprevention antimalarials in the region. Understanding how multiple emergences interact with drivers of regional spread is essential for combating ART-R in Africa. FUNDING This study was funded by the Bill & Melinda Gates Foundation and the National Institutes of Health.
Collapse
Affiliation(s)
- Jonathan J Juliano
- Division of Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum of Genetics and Molecular Biology, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Institute for Global Health and Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David J Giesbrecht
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Alfred Simkin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Abebe A Fola
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Beatus M Lyimo
- National Institute for Medical Research, Dar es Salaam, Tanzania; School of Life Sciences and Bioengineering, Nelson Mandela African Institute of Science and Technology, Arusha, Tanzania
| | - Dativa Pereus
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Catherine Bakari
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Rashid A Madebe
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Misago D Seth
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Celine I Mandara
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Zachary R Popkin-Hall
- Institute for Global Health and Infectious Diseases, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ramadhan Moshi
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Ruth B Mbwambo
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Karamoko Niaré
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Bronwyn MacInnis
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA; Infectious Disease and Microbiome Program, Broad Institute, Boston, MA, USA
| | - Filbert Francis
- National Institute for Medical Research, Dar es Salaam, Tanzania
| | | | - Issa Garimo
- National Malaria Control Programme, Dodoma, Tanzania
| | - Frank Chacky
- National Malaria Control Programme, Dodoma, Tanzania
| | | | | | | | - Ritha J A Njau
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Ally Mohamed
- National Malaria Control Programme, Dodoma, Tanzania
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Deus S Ishengoma
- National Institute for Medical Research, Dar es Salaam, Tanzania; Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA; Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam, Tanzania.
| |
Collapse
|
35
|
Attaway C, Mathison BA, Misra A. No longer stuck in the past: new advances in artificial intelligence and molecular assays for parasitology screening and diagnosis. Curr Opin Infect Dis 2024; 37:357-366. [PMID: 39133581 DOI: 10.1097/qco.0000000000001041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW Emerging technologies are revolutionizing parasitology diagnostics and challenging traditional methods reliant on microscopic analysis or serological confirmation, which are known for their limitations in sensitivity and specificity. This article sheds light on the transformative potential of artificial intelligence and molecular assays in the field, promising more accurate and efficient detection methods. RECENT FINDINGS Artificial intelligence has emerged as a promising tool for blood and stool parasite review, when paired with comprehensive databases and expert oversight result in heightened specificity and sensitivity of diagnoses while also increasing efficiency. Significant strides have been made in nucleic acid testing for multiplex panels for enteric pathogen. Both multiplex and single target panels for Plasmodium , Babesia , filaria, and kinetoplastids have been developed and garnered regulatory approval, notably for blood donor screening in the United States. Additional technologies such as MALDI-TOF, metagenomics, flow cytometry, and CRISPR-Cas are under investigation for their diagnostic utility and are currently in the preliminary stages of research and feasibility assessment. SUMMARY Recent implementation of artificial intelligence and digital microscopy has enabled swift smear screening and diagnosis, although widespread implementation remains limited. Simultaneously, molecular assays - both targeted and multiplex panels are promising and have demonstrated promise in numerous studies with some assays securing regulatory approval recently. Additional technologies are under investigation for their diagnostic utility and are compelling avenues for future proof-of-concept diagnostics.
Collapse
Affiliation(s)
| | - Blaine A Mathison
- Scientist III, Institute for Clinical and Experimental Pathology, ARUP Laboratories, Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
36
|
Chahine Z, Abel S, Hollin T, Barnes GL, Chung JH, Daub ME, Renard I, Choi JY, Vydyam P, Pal A, Alba-Argomaniz M, Banks CAS, Kirkwood J, Saraf A, Camino I, Castaneda P, Cuevas MC, De Mercado-Arnanz J, Fernandez-Alvaro E, Garcia-Perez A, Ibarz N, Viera-Morilla S, Prudhomme J, Joyner CJ, Bei AK, Florens L, Ben Mamoun C, Vanderwal CD, Le Roch KG. A kalihinol analog disrupts apicoplast function and vesicular trafficking in P. falciparum malaria. Science 2024; 385:eadm7966. [PMID: 39325875 PMCID: PMC11793105 DOI: 10.1126/science.adm7966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/27/2024] [Accepted: 07/09/2024] [Indexed: 09/28/2024]
Abstract
We report the discovery of MED6-189, an analog of the kalihinol family of isocyanoterpene natural products that is effective against drug-sensitive and drug-resistant Plasmodium falciparum strains, blocking both asexual replication and sexual differentiation. In vivo studies using a humanized mouse model of malaria confirm strong efficacy of the compound in animals with no apparent hemolytic activity or toxicity. Complementary chemical, molecular, and genomics analyses revealed that MED6-189 targets the parasite apicoplast and acts by inhibiting lipid biogenesis and cellular trafficking. Genetic analyses revealed that a mutation in PfSec13, which encodes a component of the parasite secretory machinery, reduced susceptibility to the drug. Its high potency, excellent therapeutic profile, and distinctive mode of action make MED6-189 an excellent addition to the antimalarial drug pipeline.
Collapse
Affiliation(s)
- Z. Chahine
- Department of Chemistry, University of California, Irvine, CA, USA
| | - S. Abel
- Department of Chemistry, University of California, Irvine, CA, USA
| | - T. Hollin
- Department of Chemistry, University of California, Irvine, CA, USA
| | - G. L. Barnes
- Department of Chemistry, University of California, Irvine, CA, USA
| | - J. H. Chung
- Department of Chemistry, University of California, Irvine, CA, USA
| | - M. E. Daub
- Department of Chemistry, University of California, Irvine, CA, USA
| | - I. Renard
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - J. Y. Choi
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - P. Vydyam
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - A. Pal
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - M. Alba-Argomaniz
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - C. A. S. Banks
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - J. Kirkwood
- Metabolomics Core Facility, University of California, Riverside, CA, USA
| | - A. Saraf
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | | | | | | | | | | | - N. Ibarz
- GSK, Tres Cantos (Madrid), Spain
| | | | - J. Prudhomme
- Department of Chemistry, University of California, Irvine, CA, USA
| | - C. J. Joyner
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - A. K. Bei
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - L. Florens
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - C. Ben Mamoun
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - C. D. Vanderwal
- Department of Chemistry, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - K. G. Le Roch
- Department of Chemistry, University of California, Irvine, CA, USA
| |
Collapse
|
37
|
Zheng D, Liu T, Yu S, Liu Z, Wang J, Wang Y. Antimalarial Mechanisms and Resistance Status of Artemisinin and Its Derivatives. Trop Med Infect Dis 2024; 9:223. [PMID: 39330912 PMCID: PMC11435542 DOI: 10.3390/tropicalmed9090223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Artemisinin is an endoperoxide sesquiterpene lactone isolated from Artemisia annua and is often used to treat malaria. Artemisinin's peroxide bridge is the key structure behind its antimalarial action. Scientists have created dihydroartemisinin, artemether, artesunate, and other derivatives preserving artemisinin's peroxide bridge to increase its clinical utility value. Artemisinin compounds exhibit excellent efficacy, quick action, and minimal toxicity in malaria treatment and have greatly contributed to malaria control. With the wide and unreasonable application of artemisinin-based medicines, malaria parasites have developed artemisinin resistance, making malaria prevention and control increasingly challenging. Artemisinin-resistant Plasmodium strains have been found in many countries and regions. The mechanisms of antimalarials and artemisinin resistance are not well understood, making malaria prevention and control a serious challenge. Understanding the antimalarial and resistance mechanisms of artemisinin drugs helps develop novel antimalarials and guides the rational application of antimalarials to avoid the spread of resistance, which is conducive to malaria control and elimination efforts. This review will discuss the antimalarial mechanisms and resistance status of artemisinin and its derivatives, which will provide a reference for avoiding drug resistance and the research and development of new antimalarial drugs.
Collapse
Affiliation(s)
- Dan Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Zhilong Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
38
|
Daka D, Woldeyes D, Golassa L, Alemayehu GS, Zewde Z, Tamiru G, Misganaw T, Massebo F, Wondale B. Therapeutic efficacy of artemether-lumefantrine in the treatment of uncomplicated Plasmodium falciparum malaria in Arba Minch Zuria District, Gamo Zone, Southwest Ethiopia. Malar J 2024; 23:282. [PMID: 39289715 PMCID: PMC11406784 DOI: 10.1186/s12936-024-05087-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Artemether-lumefantrine (AL) has been the primary anti-malarial drug used to treat uncomplicated Plasmodium falciparum malaria in Ethiopia since 2004. However, there have been recent reports of AL resistance mutations in different African countries, including Ethiopia. This is concerning and requires periodic monitoring of anti-malarial drug resistance. Therefore, the current study aimed to evaluate the therapeutic efficacy of AL in treating uncomplicated P. falciparum malaria in the Arba Minch Zuria District, Gamo Zone, Southwest Ethiopia. METHODS A single-arm prospective study with a 28-day follow-up period was conducted from July to October 2022. Capillary blood samples were collected for RDT and microscopic examination. The study enrolled monoinfected P. falciparum patients aged ≥ 18 years at Ganta Sira Health Post. Sociodemographic and clinical data were recorded, and a dried blood spot (DBS) was prepared for each participant. Nested polymerase chain reaction (nPCR) genotyping of the msp-1 and msp-2 genes was only performed for recurrent cases to distinguish between recurrence and reinfection. Data entry and analysis were performed using the WHO Excel spreadsheet and SPSS version 26. RESULTS A total of 89 patients were enrolled, and 67 adequately completed the 28-day follow-up period. AL showed a 100% clearance rate for fever on day 2 and asexual parasites on day 3. Gametocytes were detected in 13.5% (12/89) of the participants. The gametocyte clearance rate was 58.3% (7/12) until day 7 and 100% (12/12) until day 14. Five participants developed recurrent malaria, three of whom experienced relapse and two of whom experienced reinfection. Based on the Kaplan-Meier survival analysis, the PCR-uncorrected and PCR-corrected cumulative incidence of success were 93.7% (95% CI 85.5-97.3) and 96.2% (95% CI 85.5-98.7), respectively. CONCLUSION AL was efficacious in treating uncomplicated P. falciparum malaria in the study area. However, the detection of recurrent patients highlights the need for continuous efficacy studies in this area.
Collapse
Affiliation(s)
- Demeke Daka
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia
- Department of Biology, Madda Walabu University, Bale Robe, Ethiopia
| | - Daniel Woldeyes
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia
| | - Lemu Golassa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Zerihun Zewde
- Arba Minch Public Health Laboratory, South Ethiopia Region Public Health Institute, Arba Minch, Ethiopia
| | - Girum Tamiru
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia
| | - Tadesse Misganaw
- Department of Medical Laboratory Science, Woldia University, Woldia, Ethiopia
| | - Fekadu Massebo
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia
| | - Biniam Wondale
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia.
| |
Collapse
|
39
|
Ogwang R, Osoti V, Wamae K, Ndwiga L, Muteru K, Ningwa A, Tuju J, Kinyanjui S, Osier F, Marsh K, Bejon P, Idro R, Ochola-Oyier LI. A retrospective analysis of P. falciparum drug resistance markers detects an early (2016/17) high prevalence of the k13 C469Y mutation in asymptomatic infections in Northern Uganda. Antimicrob Agents Chemother 2024; 68:e0157623. [PMID: 39136465 PMCID: PMC11382623 DOI: 10.1128/aac.01576-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 07/20/2024] [Indexed: 09/05/2024] Open
Abstract
The emergence of drug-resistant Plasmodium falciparum parasites in sub-Saharan Africa will substantially challenge malaria control. Here, we evaluated the frequency of common drug resistance markers among adolescents from Northern Uganda with asymptomatic infections. We used an established amplicon deep sequencing strategy to screen dried blood spot samples collected from 2016 to 2017 during a reported malaria epidemic within the districts of Kitgum and Pader in Northern Uganda. We screened single-nucleotide polymorphisms within: kelch13 (Pfk13), dihydropteroate synthase (Pfdhps), multidrug resistance-1 (Pfmdr1), dihydrofolate reductase (Pfdhfr), and apical membrane antigen (Pfama1) genes. Within the study population, the median age was 15 years (14.3-15.0, 95% CI), and 54.9% (78/142) were Plasmodium positive by 18S rRNA qPCR, which were subsequently targeted for sequencing analysis. We observed a high frequency of resistance markers particularly for sulfadoxine-pyrimethamine (SP), with no wild-type-only parasites observed for Pfdhfr (N51I, C59R, and S108N) and Pfdhps (A437G and K540E) mutations. Within Pfmdr1, mixed infections were common for NF/NY (98.5%). While for artemisinin resistance, in kelch13, there was a high frequency of C469Y (34%). Using the pattern for Pfama1, we found a high level of polygenomic infections with all individuals presenting with complexity of infection greater than 2 with a median of 6.9. The high frequency of the quintuple SP drug-resistant parasites and the C469Y artemisinin resistance-associated mutation in asymptomatic individuals suggests an earlier high prevalence than previously reported from symptomatic malaria surveillance studies (in 2016/2017). Our data demonstrate the urgency for routine genomic surveillance programs throughout Africa and the value of deep sequencing.
Collapse
Affiliation(s)
- Rodney Ogwang
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Makerere University College of Health Sciences, Kampala, Uganda
- Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda
| | - Victor Osoti
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kevin Wamae
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Leonard Ndwiga
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kelvin Muteru
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Albert Ningwa
- Makerere University College of Health Sciences, Kampala, Uganda
- Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Sam Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Faith Osier
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Kevin Marsh
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Philip Bejon
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Richard Idro
- Makerere University College of Health Sciences, Kampala, Uganda
- Centre of Tropical Neuroscience (CTN), Kitgum Site, Uganda
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lynette Isabella Ochola-Oyier
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| |
Collapse
|
40
|
Kreutzfeld O, Orena S, Okitwi M, Tumwebaze PK, Byaruhanga O, Katairo T, Conrad MD, Legac J, Garg S, Crudale R, Aydemir O, Giesbrecht D, Nsobya SL, Blasco B, Duffey M, Rouillier M, Bailey JA, Cooper RA, Rosenthal PJ. Ex vivo susceptibilities to ganaplacide and diversity in potential resistance mediators in Ugandan Plasmodium falciparum isolates. Antimicrob Agents Chemother 2024; 68:e0046624. [PMID: 39136468 PMCID: PMC11373204 DOI: 10.1128/aac.00466-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/20/2024] [Indexed: 09/05/2024] Open
Abstract
Novel antimalarials are urgently needed to combat rising resistance to available drugs. The imidazolopiperazine ganaplacide is a promising drug candidate, but decreased susceptibility of laboratory strains has been linked to polymorphisms in the Plasmodium falciparum cyclic amine resistance locus (PfCARL), acetyl-CoA transporter (PfACT), and UDP-galactose transporter (PfUGT). To characterize parasites causing disease in Africa, we assessed ex vivo drug susceptibilities to ganaplacide in 750 P. falciparum isolates collected in Uganda from 2017 to 2023. Drug susceptibilities were assessed using a 72-hour SYBR Green growth inhibition assay. The median IC50 for ganaplacide was 13.8 nM, but some isolates had up to 31-fold higher IC50s (31/750 with IC50 > 100 nM). To assess genotype-phenotype associations, we sequenced genes potentially mediating altered ganaplacide susceptibility in the isolates using molecular inversion probe and dideoxy sequencing methods. PfCARL was highly polymorphic, with eight mutations present in >5% of isolates. None of these eight mutations had previously been selected in laboratory strains with in vitro drug pressure and none were found to be significantly associated with decreased ganaplacide susceptibility. Mutations in PfACT and PfUGT were found in ≤5% of isolates, except for two frequent (>20%) mutations in PfACT; one mutation in PfACT (I140V) was associated with a modest decrease in susceptibility. Overall, Ugandan P. falciparum isolates were mostly highly susceptible to ganaplacide. Known resistance mediators were polymorphic, but mutations previously selected with in vitro drug pressure were not seen, and mutations identified in the Ugandan isolates were generally not associated with decreased ganaplacide susceptibility.
Collapse
Affiliation(s)
| | - Stephen Orena
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Martin Okitwi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Jennifer Legac
- University of California, San Francisco, California, USA
| | - Shreeya Garg
- University of California, San Francisco, California, USA
| | | | - Ozkan Aydemir
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ouédraogo A, Pouplin JNN, Mukaka M, Kaendiao T, Ruecker A, Millet P, Vallet T, Ruiz F, Sirima SB, Taylor WR. Anti-infectivity efficacy and pharmacokinetics of WHO recommended single low-dose primaquine in children with acute Plasmodium falciparum in Burkina Faso: study protocol. Trials 2024; 25:583. [PMID: 39227956 PMCID: PMC11373093 DOI: 10.1186/s13063-024-08428-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Primaquine (PQ) has activity against mature P. falciparum gametocytes and proven transmission blocking efficacy (TBE) between humans and mosquitoes. WHO formerly recommended a single transmission blocking dose of 0.75 mg/kg but this was little used. Then in 2012, faced with the emergence of artemisinin-resistant P. falciparum (ARPf) in SE Asia, the WHO recommended a lower dose of 0.25 mg/kg to be added to artemisinin-based combination therapy in falciparum-infected patients in low transmission areas. This dose was considered safe in glucose-6-phosphate dehydrogenase deficiency (G6PDd) and not requiring G6PD testing. Subsequent single low-dose primaquine (SLDPQ) studies have demonstrated safety in different G6PD variants. Dosing remains challenging in children under the age of 5 because of the paucity of PQ pharmacokinetic (PK) data. We plan to assess the anti-infectivity efficacy of SLDPQ using an allometrically scaled, weight-based regimen, with a target dose of 0.25 mg/kg, in children with acute uncomplicated falciparum malaria. METHODS This study is an open label, randomised 1:1, phase IIb study to assess TBE, tolerability, pharmacokinetics and acceptability of artesunate pyronaridine (ASPYR) administered alone or combined with SLDPQ in 56 Burkinabe children aged ≥ 6 months- < 5 years, with uncomplicated P. falciparum and a haemoglobin (Hb) concentration of ≥ 5 g/dL. We will assess TBE, using direct membrane feeding assays (DMFA), and further investigate PQ pharmacokinetics, adverse events, Hb dynamics, G6PD, sickle cells, thalassaemia and cytochrome 2D6 (CYP2D6) status, acceptability of flavoured PQ [CAST-ClinSearch Acceptability Score Test®], and the population's knowledge, attitude and practices on malaria. EXPECTED RESULTS AND DISCUSSION We expect children to accept tablets, confirm the TBE and gametocytocidal effects of SLDPQ and then construct a PK infectivity model (including age, sex, baseline Hb, G6PD and CYP2D6 status) to define the dose response TBE relationship that may lead to fine tuning our SLDPQ regimen. Our study will complement others that have examined factors associated with Hb dynamics and PQ PK. It will provide much needed, high-quality evidence of SLDPQ in sick African children and provide reassurance that SLDPQ should be used as a strategy against emerging ARPf in Africa. TRIAL REGISTRATION ISRCTN16297951. Registered on September 26, 2021.
Collapse
Affiliation(s)
- Alphonse Ouédraogo
- Groupe de Recherche Action en Santé (GRAS), 06 BP 10248, Ouagadougou 06, Burkina Faso.
| | | | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thoopmanee Kaendiao
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
| | - Andrea Ruecker
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pascal Millet
- ReMeD, 21bis Avenue du Commandant de L'Herminier, Saint-Nazaire, 44 600, France
| | - Thibaut Vallet
- ClinSearch, 110 Avenue Pierre Brossolette, Malakoff, 92240, France
| | - Fabrice Ruiz
- ClinSearch, 110 Avenue Pierre Brossolette, Malakoff, 92240, France
| | - Sodiomon B Sirima
- Groupe de Recherche Action en Santé (GRAS), 06 BP 10248, Ouagadougou 06, Burkina Faso
| | - Walter R Taylor
- Mahidol Oxford Tropical Medicine Research Unit (MORU), 420/6 Rajvithi Road, Rajthevee, Bangkok, 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Rosenthal PJ, Asua V, Bailey JA, Conrad MD, Ishengoma DS, Kamya MR, Rasmussen C, Tadesse FG, Uwimana A, Fidock DA. The emergence of artemisinin partial resistance in Africa: how do we respond? THE LANCET. INFECTIOUS DISEASES 2024; 24:e591-e600. [PMID: 38552654 DOI: 10.1016/s1473-3099(24)00141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 04/21/2024]
Abstract
Malaria remains one of the most important infectious diseases in the world, with the greatest burden in sub-Saharan Africa, primarily from Plasmodium falciparum infection. The treatment and control of malaria is challenged by resistance to most available drugs, but partial resistance to artemisinins (ART-R), the most important class for the treatment of malaria, was until recently confined to southeast Asia. This situation has changed, with the emergence of ART-R in multiple countries in eastern Africa. ART-R is mediated primarily by single point mutations in the P falciparum kelch13 protein, with several mutations present in African parasites that are now validated resistance mediators based on clinical and laboratory criteria. Major priorities at present are the expansion of genomic surveillance for ART-R mutations across the continent, more frequent testing of the efficacies of artemisinin-based regimens against uncomplicated and severe malaria in trials, more regular assessment of ex-vivo antimalarial drug susceptibilities, consideration of changes in treatment policy to deter the spread of ART-R, and accelerated development of new antimalarial regimens to overcome the impacts of ART-R. The emergence of ART-R in Africa is an urgent concern, and it is essential that we increase efforts to characterise its spread and mitigate its impact.
Collapse
Affiliation(s)
- Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, USA.
| | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda; University of Tübingen, Tübingen, Germany
| | - Jeffrey A Bailey
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA; Departments of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Melissa D Conrad
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Deus S Ishengoma
- National Institute for Medical Research, Dar es Salaam, Tanzania; Department of Biochemistry, Kampala International University in Tanzania, Dar es Salaam, Tanzania; School of Public Health, Harvard University, Boston, MA, USA
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda; Department of Medicine, Makerere University, Kampala, Uganda
| | | | - Fitsum G Tadesse
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia; London School of Hygiene and Tropical Medicine, London, UK
| | - Aline Uwimana
- Rwanda Biomedical Center, Kigali, Rwanda; Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Woldesenbet D, Birhanie M, Abere A, Zeleke AJ, Bezabih MK, Semaw M, Wubetie M, Abebe W, Tamene E, Tegegne Y. Therapeutic efficacy and safety of artemether-lumefantrine combination therapy for the treatment of uncomplicated Plasmodium falciparum malaria at Teda Health Centre, Northwest Ethiopia, 2022/23. Malar J 2024; 23:266. [PMID: 39215366 PMCID: PMC11363363 DOI: 10.1186/s12936-024-05082-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The emergence of Plasmodium falciparum drug resistance against artemisinin-based combination therapy has threatened malaria control efforts. Since malaria control and elimination plans are dependent on these drugs, they must remain efficacious. However, resistance to these drugs was detected in low-transmission settings and is predicted to emerge in high-transmission settings, including in unspecified areas of Ethiopia. Therefore, this study aimed to assess the therapeutic efficacy and safety of artemether-lumefantrine for the treatment of uncomplicated P. falciparum malaria. METHODS A single-arm prospective observational study was conducted at Teda Health Centre, Northwest Ethiopia, by following the 2009 World Health Organization efficacy study guidelines from September 2022 to February 2023. Patients with uncomplicated falciparum malaria were conveniently selected and treated with a standard dose of artemether-lumefantrine, along with a single low dose of primaquine. Then clinical and parasitological responses and haemoglobin levels were assessed during the 28-day scheduled follow-up. Blood films were examined and asexual parasites were quantified; axillary temperature was measured; and drug adverse events were assessed throughout the follow-up. Finally, the drug efficacy (adequate clinical and parasitological response) was determined by Kaplan-Meier and per-protocol analyses. The data were analysed using the WHO Excel spreadsheet and SPSS version 25 software. RESULTS The success rates of PCR uncorrected and corrected Kaplan-Meier analysis on day 28 were 95.8% (95% CI 87.5-98.6) and 97.3% (95% CI 89.4-99.3), respectively. The per-protocol PCR uncorrected and corrected adequate clinical and parasitological responses were 95.5% (95% CI 87.5-99.1) and 97% (95% CI 89.5-99.6), respectively. On day-3, 97% of study participants were free of asexual parasitaemia, and all of them were fever-free on day-2. All of the gametocyte-positive patients at baseline were found to be negative for gametocytes on day-2. Moreover, the baseline mean hemoglobin of 13.10 g/dl increased slightly on day-14 to 13.27 g/dl but significantly on day-28 to 13.69 g/dl in a paired sample t test. All adverse events reported were mild. CONCLUSION Artemether-lumefantrine continued to be an efficacious and safe drug for the treatment of uncomplicated Plasmodium falciparum malaria at the Teda Health Centre. TRIAL REGISTRATION unique ID# PACTR202309773069812 at https://pactr.samrc.ac.za on September 1, 2023.
Collapse
Affiliation(s)
- Dagmawi Woldesenbet
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Wachemo University, Hossana, Ethiopia.
| | - Meseret Birhanie
- Department of Medical Parasitology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Aberham Abere
- Department of Medical Parasitology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Ayalew Jejaw Zeleke
- Department of Medical Parasitology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Migbaru Keffale Bezabih
- Malaria and Neglected Tropical Diseases Directorate, Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Muluken Semaw
- Medical Laboratory Unit, Sanja General Hospital, Amhara National Regional State Health Bureau, Sanja, Ethiopia
| | - Menberu Wubetie
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Dire Dawa University, Dire Dawa, Ethiopia
| | - Wagaw Abebe
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Elias Tamene
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Wachemo University, Hossana, Ethiopia
| | - Yalewayker Tegegne
- Department of Medical Parasitology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
44
|
Tripathi J, Stoklasa M, Nayak S, En Low K, Qian Hui Lee E, Duong Tien QH, Rénia L, Malleret B, Bozdech Z. The artemisinin-induced dormant stages of Plasmodium falciparum exhibit hallmarks of cellular quiescence/senescence and drug resilience. Nat Commun 2024; 15:7485. [PMID: 39209862 PMCID: PMC11362153 DOI: 10.1038/s41467-024-51846-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Recrudescent infections with the human malaria parasite, Plasmodium falciparum, presented traditionally the major setback of artemisinin-based monotherapies. Although the introduction of artemisinin combination therapies (ACT) largely solved the problem, the ability of artemisinin to induce dormant parasites still poses an obstacle for current as well as future malaria chemotherapeutics. Here, we use a laboratory model for induction of dormant P. falciparum parasites and characterize their transcriptome, drug sensitivity profile, and cellular ultrastructure. We show that P. falciparum dormancy requires a ~ 5-day maturation process during which the genome-wide gene expression pattern gradually transitions from the ring-like state to a unique form. The transcriptome of the mature dormant stage carries hallmarks of both cellular quiescence and senescence, with downregulation of most cellular functions associated with growth and development and upregulation of selected metabolic functions and DNA repair. Moreover, the P. falciparum dormant stage is considerably more resistant to antimalaria drugs compared to the fast-growing asexual stages. Finally, the irregular cellular ultrastructure further suggests unique properties of this developmental stage of the P. falciparum life cycle that should be taken into consideration by malaria control strategies.
Collapse
Affiliation(s)
- Jaishree Tripathi
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore.
| | - Michal Stoklasa
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore
| | - Sourav Nayak
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore
| | - Kay En Low
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Erica Qian Hui Lee
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Quang Huy Duong Tien
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore
| | - Laurent Rénia
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University (NTU), Singapore, 636921, Singapore
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Benoit Malleret
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore, 637551, Singapore.
| |
Collapse
|
45
|
Galick DS, Donfack OT, Mifumu TAO, Onvogo CNO, Dougan TB, Mikue MIAA, Nguema GE, Eribo CO, Euka MMB, Marone Martin KP, Phiri WP, Guerra CA, García GA. Adapting malaria indicator surveys to investigate treatment adherence: a pilot study on Bioko Island, Equatorial Guinea. Malar J 2024; 23:244. [PMID: 39138464 PMCID: PMC11323597 DOI: 10.1186/s12936-024-05057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Adherence to anti-malarial treatment regimens is an important aspect of understanding and improving the impact of malaria case management. However, both adherence to artemisinin-based combination therapy (ACT) and the factors driving it vary widely. While many other evaluation activities have been conducted on Bioko Island, until now adherence to anti-malarial treatments, and in particular ACT has not been evaluated. METHODS The implementation of a malaria indicator survey (MIS) conducted on Bioko in 2023 was leveraged to evaluate adherence to ACT provided to individuals testing positive following the survey. A follow-up team visited the targeted households, physically observed treatment blisters where possible, and provided messaging to household members on the importance of adhering to the treatment guidelines to household members. The team used survey data from the targeted households to make messaging as relevant to the household's particular context as possible. RESULTS Overall ACT adherence on Bioko Island was low, around 50%, and this varied demographically and geographically. Some of the highest transmission areas had exceptionally low adherence, but no systematic relationship between proper adherence and Plasmodium falciparum prevalence was detected. Estimates of adherence from follow-up visits were much lower than survey-based estimates in the same households (52.5% versus 87.1%), suggesting that lack of proper adherence may be a much larger issue on Bioko Island than previously thought. CONCLUSION Representative surveys can be easily adapted to provide empirical estimates of adherence to anti-malarial treatments, complementary to survey-based and health facility-based estimates. The large discrepancy between adherence as measured in this study and survey-based estimates on Bioko Island suggests a health facility-based study to quantify adherence among the population receiving treatment for symptomatic malaria may be necessary.
Collapse
|
46
|
Fola AA, Ciubotariu II, Dorman J, Mwenda MC, Mambwe B, Mulube C, Kasaro R, Hawela MB, Hamainza B, Miller JM, Bailey JA, Moss WJ, Bridges DJ, Carpi G. National genomic profiling of Plasmodium falciparum antimalarial resistance in Zambian children participating in the 2018 Malaria Indicator Survey. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.05.24311512. [PMID: 39148823 PMCID: PMC11326323 DOI: 10.1101/2024.08.05.24311512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The emergence of antimalarial drug resistance is a major threat to malaria control and elimination. Using whole genome sequencing of 282 P. falciparum samples collected during the 2018 Zambia National Malaria Indicator Survey, we determined the prevalence and spatial distribution of known and candidate antimalarial drug resistance mutations. High levels of genotypic resistance were found across Zambia to pyrimethamine, with over 94% (n=266) of samples having the Pfdhfr triple mutant (N51I, C59R, and S108N), and sulfadoxine, with over 84% (n=238) having the Pfdhps double mutant (A437G and K540E). In northern Zambia, 5.3% (n=15) of samples also harbored the Pfdhps A581G mutation. Although 29 mutations were identified in Pfkelch13, these mutations were present at low frequency (<2.5%), and only three were WHO-validated artemisinin partial resistance mutations: P441L (n=1, 0.35%), V568M (n=2, 0.7%) and R622T (n=1, 0.35%). Notably, 91 (32%) of samples carried the E431K mutation in the Pfatpase6 gene, which is associated with artemisinin resistance. No specimens carried any known mutations associated with chloroquine resistance in the Pfcrt gene (codons 72-76). P. falciparum strains circulating in Zambia were highly resistant to sulfadoxine and pyrimethamine but remained susceptible to chloroquine and artemisinin. Despite this encouraging finding, early genetic signs of developing artemisinin resistance highlight the urgent need for continued vigilance and expanded routine genomic surveillance to monitor these changes.
Collapse
Affiliation(s)
- Abebe A. Fola
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Ilinca I. Ciubotariu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jack Dorman
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Mulenga C. Mwenda
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Brenda Mambwe
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Conceptor Mulube
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Rachael Kasaro
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Moonga B. Hawela
- National Malaria Elimination Centre, Zambia Ministry of Health, Chainama Hospital Grounds, Lusaka, Zambia
| | - Busiku Hamainza
- National Malaria Elimination Centre, Zambia Ministry of Health, Chainama Hospital Grounds, Lusaka, Zambia
| | - John M. Miller
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Jeffrey A. Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - William J. Moss
- The Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daniel J. Bridges
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Giovanna Carpi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- The Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
47
|
Konyanee A, Chaniad P, Chukaew A, Payaka A, Septama AW, Phuwajaroanpong A, Plirat W, Punsawad C. Antiplasmodial potential of isolated xanthones from Mesua ferrea Linn. roots: an in vitro and in silico molecular docking and pharmacokinetics study. BMC Complement Med Ther 2024; 24:282. [PMID: 39054443 PMCID: PMC11270968 DOI: 10.1186/s12906-024-04580-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Malaria is a major global health concern, particularly in tropical and subtropical countries. With growing resistance to first-line treatment with artemisinin, there is an urgent need to discover novel antimalarial drugs. Mesua ferrea Linn., a plant used in traditional medicine for various purposes, has previously been investigated by our research group for its cytotoxic properties. The objective of this study was to explore the compounds isolated from M. ferrea with regards to their potential antiplasmodial activity, their interaction with Plasmodium falciparum lactate dehydrogenase (PfLDH), a crucial enzyme for parasite survival, and their pharmacokinetic and toxicity profiles. METHODS The isolated compounds were assessed for in vitro antiplasmodial activity against a multidrug-resistant strain of P. falciparum K1 using a parasite lactate dehydrogenase (pLDH) assay. In vitro cytotoxicity against Vero cells was determined using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. The interactions between the isolated compounds and the target enzyme PfLDH were investigated using molecular docking. Additionally, pharmacokinetic and toxicity properties were estimated using online web tools SwissADME and ProTox-II, respectively. RESULTS Among the seven compounds isolated from M. ferrea roots, rheediachromenoxanthone (5), which belongs to the pyranoxanthone class, demonstrated good in vitro antiplasmodial activity, with the IC50 being 19.93 µM. Additionally, there was no toxicity towards Vero cells (CC50 = 112.34 µM) and a selectivity index (SI) of 5.64. Molecular docking analysis revealed that compound (5) exhibited a strong binding affinity of - 8.6 kcal/mol towards PfLDH and was stabilized by forming hydrogen bonds with key amino acid residues, including ASP53, TYR85, and GLU122. Pharmacokinetic predictions indicated that compound (5) possessed favorable drug-like properties and desired pharmacokinetic characteristics. These include high absorption in the gastrointestinal tract, classification as a non-substrate of permeability glycoprotein (P-gp), non-inhibition of CYP2C19, ease of synthesis, a high predicted LD50 value of 4,000 mg/kg, and importantly, non-hepatotoxic, non-carcinogenic, and non-cytotoxic effects. CONCLUSIONS This study demonstrated that compounds isolated from M. ferrea exhibit activity against P. falciparum. Rheediachromenoxanthone has significant potential as a scaffold for the development of potent antimalarial drugs.
Collapse
Affiliation(s)
- Atthaphon Konyanee
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Prapaporn Chaniad
- School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Arnon Chukaew
- Chemistry Department, Faculty of Science and Technology, Suratthani Rajabhat University, Surat Thani, 84100, Thailand
| | - Apirak Payaka
- School of Science, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Abdi Wira Septama
- Research Center for Pharmaceutical Ingredient and Traditional Medicine, Cibinong Science Center, National Research and Innovation Agency (BRIN), West Java, 16915, Indonesia
| | - Arisara Phuwajaroanpong
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Walaiporn Plirat
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| |
Collapse
|
48
|
Dhorda M, Kaneko A, Komatsu R, Kc A, Mshamu S, Gesase S, Kapologwe N, Assefa A, Opigo J, Adoke Y, Ebong C, Karema C, Uwimana A, Mangara JLN, Amaratunga C, Peto TJ, Tripura R, Callery JJ, Adhikari B, Mukaka M, Cheah PY, Mutesa L, Day NPJ, Barnes KI, Dondorp A, Rosenthal PJ, White NJ, von Seidlein L. Artemisinin-resistant malaria in Africa demands urgent action. Science 2024; 385:252-254. [PMID: 39024426 DOI: 10.1126/science.adp5137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Investment in community health workers is essential.
Collapse
Affiliation(s)
- Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Akira Kaneko
- Department of Microbiology, Tumor and Cell Biology (C1), Karolinska Institutet, Solna, Sweden
| | | | - Achyut Kc
- Department of Microbiology, Tumor and Cell Biology (C1), Karolinska Institutet, Solna, Sweden
| | | | - Samwel Gesase
- National Institute of Malaria Research, Tanga Medical Research Centre, Tanga, Tanzania
| | - Ntuli Kapologwe
- Department of Preventive Health Services, Ministry of Health, Dodoma, Tanzania
| | - Ashenafi Assefa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, NC, USA
| | | | - Yeka Adoke
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Chris Ebong
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | | | - Chanaki Amaratunga
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thomas J Peto
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rupam Tripura
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James J Callery
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bipin Adhikari
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Phaik Yeong Cheah
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Leon Mutesa
- Centre for Human Genetics, College of Medicine and Health Sciences University of Rwanda, Kigali, Rwanda
| | - Nicholas P J Day
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Karen I Barnes
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Arjen Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
49
|
Milong Melong CS, Peloewetse E, Russo G, Tamgue O, Tchoumbougnang F, Paganotti GM. An overview of artemisinin-resistant malaria and associated Pfk13 gene mutations in Central Africa. Parasitol Res 2024; 123:277. [PMID: 39023630 DOI: 10.1007/s00436-024-08301-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Malaria caused by Plasmodium falciparum is one of the deadliest and most common tropical infectious diseases. However, the emergence of artemisinin drug resistance associated with the parasite's Pfk13 gene, threatens the public health of individual countries as well as current efforts to reduce malaria burdens globally. It is of concern that artemisinin-resistant parasites may be selected or have already emerged in Africa. This narrative review aims to evaluate the published evidence concerning validated, candidate, and novel Pfk13 polymorphisms in ten Central African countries. Results show that four validated non-synonymous polymorphisms (M476I, R539T, P553L, and P574L), directly associated with a delayed therapy response, have been reported in the region. Also, two Pfk13 polymorphisms associated to artemisinin resistance but not validated (C469F and P527H) have been reported. Furthermore, several non-validated mutations have been observed in Central Africa, and one allele A578S, is commonly found in different countries, although additional molecular and biochemical studies are needed to investigate whether those mutations alter artemisinin effects. This information is discussed in the context of biochemical and genetic aspects of Pfk13, and related to the regional malaria epidemiology of Central African countries.
Collapse
Affiliation(s)
- Charlotte Sabine Milong Melong
- Department of Biochemistry, Faculty of Sciences, University of Douala, P.O. Box 24157, Douala, Cameroon
- Botswana-University of Pennsylvania Partnership, P.O. Box 45498, Gaborone, Riverwalk, Botswana
| | - Elias Peloewetse
- Department of Biological Sciences, Faculty of Sciences, University of Botswana, Private Bag, 0022, Gaborone, UB, Botswana
| | - Gianluca Russo
- Department of Public Health and Infectious Diseases, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.Le Aldo Moro 5, 00185, Rome, Italy
| | - Ousman Tamgue
- Department of Biochemistry, Faculty of Sciences, University of Douala, P.O. Box 24157, Douala, Cameroon
| | - Francois Tchoumbougnang
- Department of Processing and Quality Control of Aquatic Products, Institute of Fisheries and Aquatic Sciences, University of Douala, P.O. Box 7236, Douala, Cameroon
| | - Giacomo Maria Paganotti
- Botswana-University of Pennsylvania Partnership, P.O. Box 45498, Gaborone, Riverwalk, Botswana.
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
50
|
Yoshida N, Kikuchi H, Hirai M, Balikagala B, Anywar DA, Taka H, Kaga N, Miura Y, Fukuda N, Odongo-Aginya EI, Kubohara Y, Mita T. A longer-chain acylated derivative of Dictyostelium differentiation-inducing factor-1 enhances the antimalarial activity against Plasmodium parasites. Biochem Pharmacol 2024; 225:116243. [PMID: 38697310 DOI: 10.1016/j.bcp.2024.116243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024]
Abstract
The spread of malarial parasites resistant to first-line treatments such as artemisinin combination therapies is a global health concern. Differentiation-inducing factor 1 (DIF-1) is a chlorinated alkylphenone (1-(3,5-dichloro-2,6-dihydroxy-4-methoxyphenyl) hexan-1-one) originally found in the cellular slime mould Dictyostelium discoideum. We previously showed that some derivatives of DIF-1, particularly DIF-1(+2) (1-(3,5-dichloro-2,6-dihydroxy-4-methoxyphenyl) octan-1-one), exert potent antimalarial activities. In this study, we synthesised DIF-1(+3) (1-(3,5-dichloro-2,6-dihydroxy-4-methoxyphenyl) nonan-1-one). We then evaluated the effects of DIF-1(+3) in vitro on Plasmodium falciparum and in vivo over 7 days (50-100 mg/kg/day) in a mouse model of Plasmodium berghei. DIF-1(+3) exhibited a half-maximal inhibitory concentration of approximately 20-30 % of DIF-1(+2) in three laboratory strains with a selectivity index > 263, including in strains resistant to chloroquine and artemisinin. Parasite growth and multiplication were almost completely suppressed by treatment with 100 mg/kg DIF-1(+3). The survival time of infected mice was significantly increased (P = 0.006) with no apparent adverse effects. In summary, addition of an acyl group to DIF-1(+2) to prepare DIF-1(+3) substantially enhanced antimalarial activity, even in drug-resistant malaria, indicating the potential of applying DIF-1(+3) for malaria treatment.
Collapse
Affiliation(s)
- Naoko Yoshida
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Haruhisa Kikuchi
- Division of Natural Medicines, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Makoto Hirai
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Betty Balikagala
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Denis A Anywar
- Faculty of Medicine, Gulu University, P.O. Box 166, Gulu, Uganda
| | - Hikari Taka
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Naoko Kaga
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Naoyuki Fukuda
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | | | - Yuzuru Kubohara
- Laboratory of Health and Life Science, Graduate School of Health and Sports Science, Juntendo University, Inzai, Chiba 270-1695, Japan.
| | - Toshihiro Mita
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|