1
|
Wagner A, Trombley R, Podgurski M, Ruberto AA, Cui M, Cooper CA, Long WE, Nguyen GB, Marin AA, Mai SL, Lombardo F, Maher SP, Kyle DE, Manetsch R. Discovery and optimization of a novel carboxamide scaffold with selective antimalarial activity. Eur J Med Chem 2025; 291:117572. [PMID: 40199028 DOI: 10.1016/j.ejmech.2025.117572] [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: 02/21/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Artemisinin combination therapies (ACTs) are critical components of malaria control worldwide. Alarmingly, ACTs have begun to fail, owing to the rise in artemisinin resistance. Thus, there is an urgent need for an expanded set of novel antimalarials to generate new combination therapies. Herein, we have identified a 1,2,4-triazole-containing carboxamide scaffold that, through scaffold hopping efforts, resulted in a nanomolar potent deuterated picolinamide (110). The lead compound of this class (110) displays moderate aqueous solubility (13.4 μM) and metabolic stability (CLintapp HLM 17.3 μL/min/mg) in vitro, as well as moderate oral bioavailability (%F 16.2) in invivo pharmacokinetic studies. Compound 110 also displayed activity against various P. falciparum isolates with different genetic backgrounds and a slow-to-moderate rate of killing (average parasite reduction ratio 2.4), making the series appealing for further development.
Collapse
Affiliation(s)
- Alicia Wagner
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, 02115, Massachusetts, USA
| | - Roger Trombley
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, 02115, Massachusetts, USA
| | - Maris Podgurski
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, 02115, Massachusetts, USA
| | - Anthony A Ruberto
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University, Boston, 02115, Massachusetts, USA
| | - Caitlin A Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - William E Long
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Gia-Bao Nguyen
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Adriana A Marin
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Sarah Lee Mai
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Franco Lombardo
- CmaxDMPK LLC, P.O. Box 505549, Chelsea, 02150, Massachusetts, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr Ste 370, Athens, 30602, Georgia, USA
| | - Roman Manetsch
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, 02115, Massachusetts, USA; Department of Pharmaceutical Sciences, Northeastern University, Boston, 02115, Massachusetts, USA; Center for Drug Discovery and Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, 02115, Massachusetts, USA.
| |
Collapse
|
2
|
Wasakul V, Verschuuren TD, Thuy-Nhien N, Booth E, Quang HH, Thang ND, Chindavongsa K, Sovannaroth S, Banouvong V, Sengsavath V, Mayxay M, Tuyen NTK, Phuong VNL, Duc Trung P, Gonçalves S, Chen S, Phalivong S, Xayvanghang S, Mahaphontrakoon S, Pearson RD, Newton PN, Maude RJ, Ashley EA, Ariani CV, Simpson VJ, Day NP, Dondorp AM, Miotto O. Genetic surveillance of Plasmodium falciparum populations following treatment policy revisions in the Greater Mekong Subregion. Nat Commun 2025; 16:4689. [PMID: 40394107 DOI: 10.1038/s41467-025-59946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Genetic surveillance of Plasmodium falciparum (Pf) can track antimalarial-resistant strains, to inform decision-making by National Malaria Control Programmes (NMCPs). The GenRe-Mekong project prospectively collected 5982 samples in the Greater Mekong Subregion (GMS) between 2017 and 2022, genotyping drug resistance markers, and barcodes that recapitulate genetic variation. Genotypes were analyzed with the grcMalaria R package, first described in this paper, to translate genetic epidemiology data into actionable visual information. Since 2020, Pf incidences decreased rapidly, accompanied by a decline of dihydroartemisinin-piperaquine (DHA-PPQ) resistant lineages, previously dominant in the eastern GMS. The frequency of plasmepsin2/3 amplifications, conferring piperaquine resistance, dropped from 62% in 2017-2019 to 2% in 2022, coinciding with a switch in frontline therapy in Cambodia, Thailand, and Vietnam. While regional artemisinin resistance levels remained high, no evidence of emerging mefloquine resistance was found. Routine genetic surveillance proved valuable in monitoring rapid parasite population changes in response to public health interventions, providing actionable information for NMCPs.
Collapse
Affiliation(s)
- Varanya Wasakul
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tess D Verschuuren
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Ethan Booth
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Huynh Hong Quang
- Institute of Malariology, Parasitology and Entomology, Quy Nhon, Vietnam
| | - Ngo Duc Thang
- National Institute of Malariology, Parasitology and Entomology, Hanoi, Vietnam
| | | | - Siv Sovannaroth
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Virasak Banouvong
- Centre for Malariology, Parasitology and Entomology, Vientiane, Lao PDR
| | | | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Institute of Research and Education Development, University of Health Sciences, Ministry of Health, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | | | | | - Pham Duc Trung
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | | | - Soun Chen
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Sonexay Phalivong
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Saiamphone Xayvanghang
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Supaporn Mahaphontrakoon
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Paul N Newton
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Richard J Maude
- 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
- The Open University, Milton Keynes, UK
| | - Elizabeth A Ashley
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Nicholas P 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
| | - 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
| | - 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, UK.
| |
Collapse
|
3
|
Dondorp AM. MMV533, a promising new antimalarial on the horizon. THE LANCET. INFECTIOUS DISEASES 2025; 25:473-474. [PMID: 39708823 DOI: 10.1016/s1473-3099(24)00730-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Fogang B, Guillochon E, Kamaliddin C, Agbota G, Ezinmegnon S, Alao MJ, Deloron P, Bertin G, Claessens A. Detection of novel Plasmodium falciparum haplotypes under treatment pressure in paediatric severe malaria. Microb Genom 2025; 11:001386. [PMID: 40340804 PMCID: PMC12064853 DOI: 10.1099/mgen.0.001386] [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: 08/16/2024] [Accepted: 02/26/2025] [Indexed: 05/10/2025] Open
Abstract
Background. In Africa, the clearance time for Plasmodium falciparum severe malaria varies significantly, likely due to the complexity of P. falciparum infections and the sequestration phenomenon exhibited by this parasite. This study aims to evaluate different methods to study the intra-host dynamics of polyclonal infections during parasite clearance under antimalarial treatment. Additionally, it seeks to determine the association between parasite clearance rate following artesunate or quinine treatment and the genetic complexity of P. falciparum in Beninese children with severe malaria.Methods. Sixty-five P. falciparum severe malaria individuals diagnosed by microscopy and treated with artesunate or quinine were sampled every 8 h for 24 h. Using whole-genome sequencing (WGS) data, we estimated the multiplicity of infection (MOI) with three algorithms (Fws, THE REAL McCOIL and RoH). We then characterized the P. falciparum genetic complexity in WGS-identified polyclonal infections using amplicon sequencing (AmpSeq) on DNA extracted from plasma and the red blood cell pellet.Results. AmpSeq demonstrated greater sensitivity in detecting multiple genomes within isolates compared to WGS methods. The MOI from AmpSeq was significantly higher in red blood cell pellets compared to plasma (2.4 vs. 1.8 distinct microhaplotypes per isolate). However, at parasitaemia over 1,000 parasites per microlitre, the same MOI was detected in both plasma and pellet samples in 85.4% of the isolates. We observed a high variability in parasite clearance rate among participants, but it was not associated with parasite MOI at diagnosis. Interestingly, in 60.9% of participants, previously undetected microhaplotypes appeared in circulation 16 h after treatment initiation.Conclusion. These findings demonstrate that combining different haplotyping techniques effectively determines parasite genetic complexity. Additionally, plasma can be effectively used for parasite genotyping at sufficient parasitaemia levels. The parasite clearance rate of severe malaria is independent of parasite MOI. However, genotyping a single blood sample upon hospital admission does not capture the full spectrum of parasite genotypes present in the infection.
Collapse
Affiliation(s)
- Balotin Fogang
- LPHI, CNRS, INSERM, University of Montpellier, Montpellier, France
| | | | | | - Gino Agbota
- Université Paris Cité, MERIT, IRD, Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Abomey-Calavi, Benin
| | - Sem Ezinmegnon
- Université Paris Cité, MERIT, IRD, Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Abomey-Calavi, Benin
| | - Maroufou Jules Alao
- Paediatric Department, Mother and Child University and Hospital Center (CHU-MEL), Cotonou, Benin
| | | | | | | |
Collapse
|
5
|
Abebe W, Ashagre A, Misganaw T, Dejazmach Z, Kumie G, Nigatie M, Jemal A, Asmare Z, Kassahun W, Gedfie S, Getachew E, Gashaw M, Ayana S, Gashaw Y, Sisay A, Tadesse S, Eshetu T, Awoke M, Kassanew B, Kidie AA, Abate BB, Reta MA. Prevalence of antimalaria drug resistance-conferring mutations associated with sulphadoxine-pyrimethamineine-resistant Plasmodium falciparum in East Africa: a systematic review and meta-analysis. Ann Clin Microbiol Antimicrob 2025; 24:25. [PMID: 40241183 PMCID: PMC12004884 DOI: 10.1186/s12941-025-00795-7] [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: 08/30/2024] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The emergence and spread of drug resistance to antimalarial drugs pose a severe threat to effective malaria control and treatment. Although sulfadoxine-pyrimethamine resistance is well-documented, it is still the drug of choice for treating intermittent resistance. Molecular markers play a crucial role in tracking and understanding the prevalence of antimalarial drug resistance. Currently, there is insufficient information on the prevalence of molecular markers associated with sulfadoxine-pyrimethamine resistance in P. falciparum. OBJECTIVE This systematic review and meta-analysis aimed to determine the pooled prevalence of antimalaria drug resistance-conferring markers associated with sulphadoxine-pyrimethamineine in Plasmodium falciparum in East Africa. METHODS Systematic searche was performed to retrieve articles from PubMed, Scopus, Science Direct databases, and Google Scholar search engine. Sixteen potential studies that provided important data on markers for sulphadoxine-pyrimethamineine resistance in Plasmodium falciparum were systematically reviewed and analyzed. Nine antimalarial drug resistance markers responsible for sulphadoxine-pyrimethamineine resistance in Plasmodium falciparum were extracted separately into Microsoft Excel and analyzed using STATA 17.0. The inverse of variance was done to evaluate heterogeneity across studies. A funnel plot was used to determine the presence of publication bias. A trim-and-fill-meta-analysis was carried out to generate a bias-adjusted effect estimate. A random effect model was used to determine the pooled prevalence of markers responsible for sulphadoxine-pyrimethamineine resistance. Subgroup analysis was performed based on country and year of publication. RESULTS A total of 16 studies were included for this systematic review and meta-analysis.The molecular markers like dhfr (N51I, C59R, S108N, 108N, 59R, and I164L), and dhps (A437G, K540E, & 540E) were selected for meta-analysis. From this meta-analysis, the pooled prevalence of dhfr N51I, dhfr C59R, dhfr S108N, dhfr 108N, dhfr 59R, and dhfr I164L was 88.6%, 85.3%, 89.6%, 92.2%, 71.5%, and 3.9%, respectively. Likewise, the aggregated prevalence of dhps A437G, dhps K540E, and dhps 540E was 90.2%, 80.9%, and 91.5%, respectively. The subgroup analysis based on year of publication showed that the pooled prevalence of dhfr N51I, dhfr C59R, dhfr S108N, dhps A437G, and dhps K540E, in studies conducted 2014-2018 was 97.11%, 90.57%, 96.45%, 90.89%, and 89.45%, respectively, while it was 82.03%, 81.78%, 85.12%, 89.24%, and 73.98%, respectively, in studies conducted 2019-2023. On the other hand, country-based analysis showed that the pooled prevalence of dhfr N51I, dhfr C59R, dhfr S108N, dhps A437G, and dhps K540E, in Kenya was 85.88%, 84.02%, 86.56%, 90.7%, and 77.55%, respectively. CONCLUSIONS This systematic review and meta-analysis reveal a high prevalence of drug resistance markers associated with sulphadoxine-pyrimethamine resistance in Plasmodium falciparum across the East African region. This underscores the significant challenges in managing malaria infections caused by Plasmodium falciparum in the region. Therefore, regular monitoring, identification, and limiting of drug-resistance markers and drug-resistant P. falciparum strains must be sustained to ensure the effectiveness of malaria treatment.
Collapse
Affiliation(s)
- Wagaw Abebe
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia.
| | - Agenagnew Ashagre
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Tadesse Misganaw
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Zelalem Dejazmach
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Getinet Kumie
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Marye Nigatie
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Abdu Jemal
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Zelalem Asmare
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Woldeteklehaymanot Kassahun
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Solomon Gedfie
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Ermias Getachew
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Muluken Gashaw
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Sisay Ayana
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Yalewayker Gashaw
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Assefa Sisay
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Selamyhun Tadesse
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Tegegne Eshetu
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulat Awoke
- Department of Nursing, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Birhanu Kassanew
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
| | - Atitegeb Abera Kidie
- Department of Public Health, College of Health Sciences, Woldia University, PO Box 400, Woldia, Ethiopia
| | - Biruk Beletew Abate
- Department of Nursing, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Melese Abate Reta
- Department of Medical Laboratory Science, College of Health Sciences, Woldia University, P.O. Box 400, Woldia, Ethiopia
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Prinshof, Pretoria, 0084, South Africa
| |
Collapse
|
6
|
Wada A, Asahi H. Identification of a Class of Iron-Grabbing Compounds with Antiplasmodial Activity: Impact of Coordination Structures and Electronic Regularity on the Intraerythrocytic Growth Cycle of Plasmodium falciparum. ACS Infect Dis 2025; 11:799-804. [PMID: 40162597 DOI: 10.1021/acsinfecdis.4c00846] [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] [Indexed: 04/02/2025]
Abstract
Innovative antimalarials are required to combat malaria, a global infectious disease caused by Plasmodium falciparum. To explore the untapped antiplasmodial compounds that can target the iron source vital at the blood stages of P. falciparum, we investigated the antiplasmodial activities of natural siderophores and synthetic compounds with metal-binding affinity. The assessment of their IC50 values and spectroscopic analytical data revealed that terpyridyl compounds specifically bound to target Fe(II) ions and strongly induced the growth inhibition of intraerythrocytic parasites. Furthermore, the IC50 values of the 4,4',4''-substituted terpyridines were linearly correlated with the sum of the para Hammett constants of their substitutions, suggesting that their growth inhibitory effects depended on the electronic states of the coordinating nitrogen atoms. Considering the specific developmental blockage at the trophozoite stage and selective antiplasmodial activities of the iron-grabbing compounds, these findings provide insights into the development of antimalarials that can disrupt iron homeostasis.
Collapse
Affiliation(s)
- Akira Wada
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroko Asahi
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
7
|
Daily JP, Parikh S. Malaria. N Engl J Med 2025; 392:1320-1333. [PMID: 40174226 DOI: 10.1056/nejmra2405313] [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: 04/04/2025]
Affiliation(s)
- Johanna P Daily
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York
| | - Sunil Parikh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
8
|
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
|
9
|
Hadjimichael E, Deitsch KW. Variable surface antigen expression, virulence, and persistent infection by Plasmodium falciparum malaria parasites. Microbiol Mol Biol Rev 2025; 89:e0011423. [PMID: 39807932 PMCID: PMC11948492 DOI: 10.1128/mmbr.00114-23] [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] [Indexed: 01/16/2025] Open
Abstract
SUMMARYThe human malaria parasite Plasmodium falciparum is known for its ability to maintain lengthy infections that can extend for over a year. This property is derived from the parasite's capacity to continuously alter the antigens expressed on the surface of the infected red blood cell, thereby avoiding antibody recognition and immune destruction. The primary target of the immune system is an antigen called PfEMP1 that serves as a cell surface receptor and enables infected cells to adhere to the vascular endothelium and thus avoid filtration by the spleen. The parasite's genome encodes approximately 60 antigenically distinct forms of PfEMP1, each encoded by individual members of the multicopy var gene family. This provides the parasite with a repertoire of antigenic types that it systematically cycles through over the course of an infection, thereby maintaining an infection until the repertoire is exhausted. While this model of antigenic variation based on var gene switching explains the dynamics of acute infections in individuals with limited anti-malarial immunity, it fails to explain reports of chronic, asymptomatic infections that can last over a decade. Recent field studies have led to a re-evaluation of previous conclusions regarding the prevalence of chronic infections, and the application of new technologies has provided insights into the molecular mechanisms that enable chronic infections and how these processes evolved.
Collapse
Affiliation(s)
- Evi Hadjimichael
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
10
|
Haines LR, Trett A, Rose C, García N, Sterkel M, McGuinness D, Regnault C, Barrett MP, Leroy D, Burrows JN, Biagini G, Ranganath LR, Aljayyoussi G, Acosta-Serrano Á. Anopheles mosquito survival and pharmacokinetic modeling show the mosquitocidal activity of nitisinone. Sci Transl Med 2025; 17:eadr4827. [PMID: 40138457 DOI: 10.1126/scitranslmed.adr4827] [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: 07/02/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
One approach to interrupting the transmission of insect-borne diseases that is successfully used in veterinary medicine is exploiting the ability of antiparasitic drugs to make vertebrate blood toxic for blood-feeding insects. Recent studies have identified 4-hydroxyphenylpyruvate dioxygenase (HPPD), an enzyme of the tyrosine detoxification pathway, as essential for hematophagous arthropods to digest their blood meals. Such blood-feeding insects include anopheline mosquitoes, which transmit malaria-causing Plasmodium parasites. A US Food and Drug Administration-approved HPPD enzyme inhibitor called nitisinone is a drug used to treat rare human-inherited disorders of the tyrosine pathway. Here, we demonstrate that feeding human blood containing nitisinone to insectary-reared female Anopheles gambiae mosquitoes was mosquitocidal to both young and old mosquitoes as well as insecticide-resistant Anopheles strains. Pharmacokinetic-pharmacodynamic (PK/PD) modeling of nitisinone's dose-response relationship (when administered at the highest recommended doses for adults and children) demonstrated improved efficacy against mosquitoes compared with the gold standard endectocidal drug, ivermectin. Furthermore, blood samples from individuals with alkaptonuria (a rare genetic metabolic disorder in the tyrosine degradation pathway), who were taking a daily low dose of 2 milligrams of nitisinone, were shown to be lethal to mosquitoes. Thus, inhibiting the Anopheles HPPD enzyme with nitisinone warrants further investigation as a complementary intervention for vector control and the prevention of malaria transmission.
Collapse
Affiliation(s)
- Lee R Haines
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Anna Trett
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Clair Rose
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Natalia García
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Marcos Sterkel
- Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, CENEXA, CONICET, La Plata, Argentina
| | - Dagmara McGuinness
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Clément Regnault
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Glasgow Polyomics, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Michael P Barrett
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Glasgow Polyomics, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Jeremy N Burrows
- Medicines for Malaria Venture, Geneva, Switzerland
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Giancarlo Biagini
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Lakshminarayan R Ranganath
- National Alkaptonuria Centre, Department of Clinical Biochemistry and Metabolism, Royal Liverpool University Hospital, Liverpool, UK
| | - Ghaith Aljayyoussi
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Álvaro Acosta-Serrano
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
11
|
Okombo J, Fidock DA. Towards next-generation treatment options to combat Plasmodium falciparum malaria. Nat Rev Microbiol 2025; 23:178-191. [PMID: 39367132 PMCID: PMC11832322 DOI: 10.1038/s41579-024-01099-x] [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] [Accepted: 08/23/2024] [Indexed: 10/06/2024]
Abstract
Malaria, which is caused by infection of red blood cells with Plasmodium parasites, can be fatal in non-immune individuals if left untreated. The recent approval of the pre-erythrocytic vaccines RTS, S/AS01 and R21/Matrix-M has ushered in hope of substantial reductions in mortality rates, especially when combined with other existing interventions. However, the efficacy of these vaccines is partial, and chemotherapy remains central to malaria treatment and control. For many antimalarial drugs, clinical efficacy has been compromised by the emergence of drug-resistant Plasmodium falciparum strains. Therefore, there is an urgent need for new antimalarial medicines to complement the existing first-line artemisinin-based combination therapies. In this Review, we discuss various opportunities to expand the present malaria treatment space, appraise the current antimalarial drug development pipeline and highlight examples of promising targets. We also discuss other approaches to circumvent antimalarial resistance and how potency against drug-resistant parasites could be retained.
Collapse
Affiliation(s)
- John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
12
|
Mahamar A, Vanheer LN, Smit MJ, Sanogo K, Sinaba Y, Niambele SM, Diallo M, Dicko OM, Diarra RS, Maguiraga SO, Youssouf A, Sacko A, Keita S, Samake S, Dembele A, Teelen K, Dicko Y, Traore SF, Dondorp A, Drakeley C, Stone W, Dicko A. Artemether-lumefantrine-amodiaquine or artesunate-amodiaquine combined with single low-dose primaquine to reduce Plasmodium falciparum malaria transmission in Ouélessébougou, Mali: a five-arm, phase 2, single-blind, randomised controlled trial. THE LANCET. MICROBE 2025; 6:100966. [PMID: 39701119 PMCID: PMC11798902 DOI: 10.1016/j.lanmic.2024.100966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/21/2024] [Accepted: 07/26/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Triple artemisinin-based combination therapies (TACTs) can delay the spread of antimalarial drug resistance. Artesunate-amodiaquine is widely used for uncomplicated Plasmodium falciparum malaria. We therefore aimed to determine the safety and efficacy of artemether-lumefantrine-amodiaquine and artesunate-amodiaquine with and without single low-dose primaquine for reducing gametocyte carriage and transmission to mosquitoes. METHODS We did a five-arm, single-blind, phase 2 randomised controlled trial at the Ouélessébougou Clinical Research Unit of the Malaria Research and Training Centre of the University of Sciences, Techniques and Technologies of Bamako in Mali. Eligible participants were aged 10-50 years, with asymptomatic P falciparum microscopy-detected gametocyte carriage. Eligible participants were randomly allocated (1:1:1:1:1) to receive either artemether-lumefantrine, artemether-lumefantrine-amodiaquine, artemether-lumefantrine-amodiaquine plus primaquine, artesunate-amodiaquine, or artesunate-amodiaquine plus primaquine. Treatment regimens were administered on days 0, 1, and 2; primaquine was given as a single dose on day 0. All staff except the trial pharmacist and participants were masked to the treatment allocation. The primary outcome was the median percentage change in mosquito infection rate between pretreatment and 2 days after treatment initiation, assessed by direct membrane feeding assay. Data were analysed using a per-protocol analysis. This study is registered with ClinicalTrials.gov, NCT05550909. FINDINGS Between Oct 16, 2022, and Dec 28, 2022, a total of 1249 individuals were screened; of whom, 100 were enrolled and randomly assigned to one of the five treatment groups (20 per group). Before treatment, 61 (61%) of 100 participants were infectious to mosquitoes, with a median of 7·3% (IQR 3·2 to 23·5) of mosquitoes becoming infected. Among infectious participants, the median percentage reduction in mosquito infection rate between pretreatment and 2 days after treatment was 100% (IQR 100 to 100) in the artemether-lumefantrine (p=0·0018), artemether-lumefantrine-amodiaquine (p=0·0018), and artemether-lumefantrine-amodiaquine plus primaquine (p=0·0009) treatment groups. In the artesunate-amodiaquine group the median percentage reduction in mosquito infection rate was only 32% (IQR -10·9 to 79·4; p=0·19), whereas a 100% median reduction was seen in the artesunate-amodiaquine plus primaquine group (IQR 100 to 100; p=0·0009). At day 2, two (10%) of 20 participants in the artemether-lumefantrine group, two (11%) of 19 in the artemether-lumefantrine-amodiaquine group, and 15 (75%) of 20 in the artesunate-amodiaquine group infected any number of mosquitoes whereas no infected mosquitoes were observed at this timepoint in the groups with primaquine. 85 (85%) of 100 participants had a total of 262 adverse events during follow-up; of which, 181 (69%) were categorised as mild and 81 (31%) as moderate. No serious adverse events were reported. INTERPRETATION Our findings support the effectiveness of artemether-lumefantrine alone or as part of TACT for preventing nearly all human-mosquito malaria parasite transmission within 48 h. By contrast, substantial transmission was observed following treatment with artesunate-amodiaquine. The addition of a single low dose of primaquine blocks transmission to mosquitoes rapidly regardless of schizonticide. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Almahamoudou Mahamar
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Leen N Vanheer
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Merel J Smit
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Koualy Sanogo
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Youssouf Sinaba
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sidi M Niambele
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Makonon Diallo
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Oumar M Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Richard S Diarra
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Seydina O Maguiraga
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Ahamadou Youssouf
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Adama Sacko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sekouba Keita
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Siaka Samake
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Adama Dembele
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Karina Teelen
- Department of Medical Microbiology and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yahia Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sekou F Traore
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Arjen 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 Clinical Medicine, University of Oxford, Oxford, UK
| | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - William Stone
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| |
Collapse
|
13
|
Shi Q, Wang C, Yang W, Ma X, Tang J, Zhang J, Zhu G, Wang Y, Liu Y, He X. Plasmodium falciparum transcription factor AP2-06B is mutated at high frequency in Southeast Asia but does not associate with drug resistance. Front Cell Infect Microbiol 2025; 14:1521152. [PMID: 39835275 PMCID: PMC11744005 DOI: 10.3389/fcimb.2024.1521152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction A continuing challenge for malaria control is the ability of Plasmodium falciparum to develop resistance to antimalarial drugs. Members within the Plasmodium transcription factor family AP2 regulate the growth and development of the parasite, and are also thought to be involved in unclear aspects of drug resistance. Here we screened for single nucleotide polymorphisms (SNPs) within the AP2 family and identified 6 non-synonymous mutations within AP2-06B (PF3D7_0613800), with allele frequencies greater than 0.05. One mutation, K3124R, was located in a PfAP2-06B AP2 domain. Methods To investigate transcriptional regulation by PfAP2-06B, ChIP-seq assays were performed on 3D7/PfAP2-06B-GFP schizonts using antibodies against GFP. The DNA sequences of the artemisinin-resistant CWX and the quinoline-resistant strains PfDd2 and Pf7G8 were analyzed for the genetic diversity of AP2-06B, compared with the Pf3D7 strain as a reference sequence. To determine whether AP2-06B can alter the expression of pfk13 and pfcrt, as well as cause artemisinin and quinoline resistance in Plasmodium, we generated both a K3124R mutation and conditional knockdown of AP2-06B in Pf3D7 using CRISPR/Cas9-mediated genome editing. Results ChIP-Seq analysis showed that AP2-06B can bind to the loci of the Plasmodium genes pfk13 and pfcrt. The AP2-06B K3124R mutation was also found in the artemisinin-resistant parasite strain CWX and the chloroquine-resistant strains Dd2 and 7G8. Contrary to expectation, Pf3D7 Plasmodium lines modified by either K3124R mutation of AP2-06B or conditional knockdown of AP2-06B did not have altered sensitivity to artemisinin or quinolines by modulating pfk13 or pfcrt expression. Discussion AP2-06B was predicted to be associated with artemisinin and quinoline resistance, but no change in resistance was observed after mutation or conditional knockdown. Given the multigenic nature of resistance, it might be difficult to recreate a resistance phenotype. In conclusion, whether AP2-06B regulates the development of artemisinin or quinoline resistance remains to be studied.
Collapse
Affiliation(s)
- Qiyang Shi
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Changhong Wang
- Laboratory of Molecular Parasitology, State Key Laboratory of Cardiology and Research Center for Translational Medicine, Shanghai East Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, Shanghai, China
| | - Wenluan Yang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Xiaoqin Ma
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Jianxia Tang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Jiayao Zhang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Guoding Zhu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Yinlong Wang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Yaobao Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Xiaoqin He
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| |
Collapse
|
14
|
Mohd Azam NNSFN, Othman S, Choo YM. Antimalarial Drug Discovery from Natural and Synthetic Sources. Curr Med Chem 2025; 32:87-110. [PMID: 38818916 DOI: 10.2174/0109298673312727240527064833] [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: 03/21/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Malaria remains a significant global health threat despite extensive efforts aimed at its eradication. Numerous challenges persist in eliminating the disease, chief among them being the parasite's ability to mutate, resulting in drug resistance. The discovery of antimalarial drugs has relied on both phenotypic and target-based approaches. While phenotypic screening has identified promising candidates, target-based methods offer a more precise approach by leveraging chemically validated targets and computational tools. Analysis of Plasmodium spp . protein structures reveal druggable targets, offering opportunities for in silico screening. Combining compounds from natural and synthetic sources in a target-based approach accelerates the discovery of new antimalarial agents. This review explores previous breakthroughs in antimalarial drug discovery from natural products and synthetic origins, emphasizing their specific target proteins within Plasmodium species.
Collapse
Affiliation(s)
| | - Shatrah Othman
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Yeun-Mun Choo
- Department of Chemistry, University of Malaya, Kuala Lumpur, 50603, Malaysia
| |
Collapse
|
15
|
Saeung M, Jupatanakul N, Hii J, Thanispong K, Chareonviriyaphap T, Manguin S. Overview of national and local efforts to eliminate malaria in Thailand. Trends Parasitol 2025; 41:52-65. [PMID: 39694742 DOI: 10.1016/j.pt.2024.11.006] [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: 10/22/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
Success in the national control of malaria during the past decades has led to the reorientation of Thailand's program toward the elimination of this disease. The country established and implemented a National Malaria Elimination Strategy, resulting in a substantial decline in cases. Although the reduction varied, Sisaket Province stands out as a success. In accordance with the national strategy, the province adopted a 1-3-7 surveillance strategy and engaged in multisectoral collaboration, positioning itself to achieve malaria-free status and advancing to the prevention of re-establishment (PoR) phase. Scaling up this approach and applying the lessons learned from the success in Sisaket Province, as a solid foundation, could prove beneficial at both the national and international levels.
Collapse
Affiliation(s)
- Manop Saeung
- Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand; HSM, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Natapong Jupatanakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, Thailand
| | - Jeffrey Hii
- Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand; College of Public Health, Medical, and Veterinary Sciences, James Cook University, North Queensland, Australia
| | - Kanutcharee Thanispong
- Division of Vector Borne Diseases, Department of Disease Control, Ministry of Public Health, Nonthaburi, Thailand
| | - Theeraphap Chareonviriyaphap
- Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand; Research and Lifelong Learning Center for Urban and Environmental Entomology, Kasetsart University Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Sylvie Manguin
- Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand; HSM, University of Montpellier, CNRS, IRD, Montpellier, France.
| |
Collapse
|
16
|
Hagenah LM, Yeo T, Schindler KA, Jeon JH, Bloxham TS, Small-Saunders JL, Mok S, Fidock DA. Plasmodium falciparum African PfCRT Mutant Isoforms Conducive to Piperaquine Resistance are Infrequent and Impart a Major Fitness Cost. J Infect Dis 2024:jiae617. [PMID: 39661643 DOI: 10.1093/infdis/jiae617] [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: 08/26/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Piperaquine, used in combination with dihydroartemisinin, has been identified as a promising partner drug for uncomplicated treatment and chemoprevention of Plasmodium falciparum malaria in Africa. In light of the earlier spread of piperaquine resistance in Southeast Asia, mediated primarily by mutations in the drug efflux transporter PfCRT, we have explored whether PfCRT mutations would represent a probable path to piperaquine resistance becoming established in Africa. METHODS We edited PfCRT mutations known to mediate piperaquine resistance in Southeast Asia into P. falciparum asexual blood stage parasites expressing three prevalent African mutant PfCRT haplotypes. Gene-edited clones were profiled in antimalarial concentration-response and competitive fitness assays. RESULTS pfcrt-edited parasites expressing the contemporary Southeast Asian T93S or I218F mutations added to the GB4 and Cam783 haplotypes common in Africa did not mediate piperaquine resistance, with partial survival only at low drug concentrations. In contrast, parasites expressing these mutations on the rare PfCRT FCB haplotype, observed mostly in North-East Africa, acquired a moderate level of piperaquine resistance. Dd2GB4, Dd2Cam783, and Dd2FCB lines edited to express the T93S or I218F mutations showed increased susceptibility to chloroquine. Piperaquine-resistant African PfCRT isoforms conferred a substantial fitness cost, manifesting as reduced asexual blood stage parasite growth rates. CONCLUSIONS These findings suggest that piperaquine-resistant PfCRT mutations that emerged in Southeast Asia mediate resistance only in a limited subset of African PfCRT haplotypes, with fitness costs that we suspect would likely preclude dissemination in high-transmission malaria-endemic African regions.
Collapse
Affiliation(s)
- Laura M Hagenah
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Kyra A Schindler
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Jin H Jeon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Talia S Bloxham
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jennifer L Small-Saunders
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sachel Mok
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Kattenberg JH, Mutsaers M, Nguyen VH, Nguyen THN, Umugwaneza A, Lara-Escandell M, Nguyen XX, Nguyen THB, Rosanas-Urgell A. Genetic surveillance shows spread of ACT resistance during period of malaria decline in Vietnam (2018-2020). Front Genet 2024; 15:1478706. [PMID: 39687741 PMCID: PMC11646998 DOI: 10.3389/fgene.2024.1478706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Vietnam's goal to eliminate malaria by 2030 is challenged by the further spread of drug-resistant Plasmodium falciparum malaria to key antimalarials, particularly dihydroartemisinin-piperaquine (DHA-PPQ). Methods The custom targeted NGS amplicon sequencing assay, AmpliSeq Pf Vietnam v2, targeting drug resistance, population genetic- and other markers, was applied to detect genetic diversity and resistance profiles in samples from 8 provinces in Vietnam (n = 354), in a period of steep decline of incidence (2018-2020). Variants in 14 putative resistance genes, including P. falciparum Kelch 13 (PfK13) and P. falciparum chloroquine resistance transporter (Pfcrt), were analyzed and within-country parasite diversity was evaluated. Other targets included KEL1-lineage markers and diagnostic markers of Pfhrp2/3. Results A concerning level of DHA-PPQ resistance was detected. The C580Y mutation in PfK13 was found in nearly 80% of recent samples, a significant rise from previous data. Vietnam has experienced a significant challenge with the spread of DHA-PPQ resistant malaria parasites, particularly in the provinces of Binh Phuoc and Gia Lai. Resistance spread to high levels in Binh Thuan prior to the country-wide treatment policy change from DHA-PPQ to pyronadine-artesunate (PA). A complex picture of PPQ-resistance dynamics was observed, with an increase of PPQ-resistance associated Pfcrt mutations, indicating an evolutionary response to antimalarial pressure. Additionally, the compensatory mutation C258W in Pfcrt, which increases chloroquine (CQ) resistance while reversing PPQ resistance, is emerging in Gia Lai following the adoption of PA as the first-line treatment. This study found high levels of multidrug resistance, with over 70% of parasites in 6 out of 8 provinces showing significant sulfadoxine-pyrimethamine (SP) resistance and widespread chloroquine-resistant Pfcrt haplotypes. We also report an absence of P. falciparum histidine rich protein 2 and 3 (Pfhrp2/3) gene deletions, ensuring the continued reliability of HRP2/3-based rapid diagnostic tests. P. falciparum populations in Vietnam are becoming more isolated, with clonal populations showing high geographical clustering by province. The central highlands, particularly Gia Lai province, have the highest residual malaria burden but exhibit low diversity and clonal populations, likely due to the pressures from the antimalarial drugs and targeted national malaria control program (NMCP) efforts. Discussion In conclusion, examining a broad panel of full-length resistance genes and SNPs provided high-resolution insights into genetic diversity and resistance evolution in Vietnam, offering valuable information to inform local treatment and intervention strategies.
Collapse
Affiliation(s)
| | - Mathijs Mutsaers
- Biomedical Sciences Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Van Hong Nguyen
- Department of Clinical Research, National Institute of Malariology, Parasitology and Entomology, Hanoi, Vietnam
| | - Thi Hong Ngoc Nguyen
- Department of Molecular Biology, National Institute of Malariology, Parasitology and Entomology, Hanoi, Vietnam
| | - Arlette Umugwaneza
- Biomedical Sciences Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Maria Lara-Escandell
- Biomedical Sciences Department, Institute of Tropical Medicine, Antwerp, Belgium
| | - Xuan Xa Nguyen
- Regional Artemisinin Initiative, RAI project, National Institute of Malariology, Parasitology and Entomology, Hanoi, Vietnam
| | - Thi Huong Binh Nguyen
- Department of Clinical Research, National Institute of Malariology, Parasitology and Entomology, Hanoi, Vietnam
| | - Anna Rosanas-Urgell
- Biomedical Sciences Department, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
20
|
Xie SC, Tai CW, Morton CJ, Ma L, Huang SC, Wittlin S, Du Y, Hu Y, Dogovski C, Salimimarand M, Griffin R, England D, de la Cruz E, Deni I, Yeo T, Burkhard AY, Striepen J, Schindler KA, Crespo B, Gamo FJ, Khandokar Y, Hutton CA, Rabie T, Birkholtz LM, Famodimu MT, Delves MJ, Bolsher J, Koolen KMJ, van der Laak R, Aguiar ACC, Pereira DB, Guido RVC, Creek DJ, Fidock DA, Dick LR, Brand SL, Gould AE, Langston S, Griffin MDW, Tilley L. A potent and selective reaction hijacking inhibitor of Plasmodium falciparum tyrosine tRNA synthetase exhibits single dose oral efficacy in vivo. PLoS Pathog 2024; 20:e1012429. [PMID: 39652589 DOI: 10.1371/journal.ppat.1012429] [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: 07/22/2024] [Revised: 12/26/2024] [Accepted: 10/31/2024] [Indexed: 12/28/2024] Open
Abstract
The Plasmodium falciparum cytoplasmic tyrosine tRNA synthetase (PfTyrRS) is an attractive drug target that is susceptible to reaction-hijacking by AMP-mimicking nucleoside sulfamates. We previously identified an exemplar pyrazolopyrimidine ribose sulfamate, ML901, as a potent reaction hijacking inhibitor of PfTyrRS. Here we examined the stage specificity of action of ML901, showing very good activity against the schizont stage, but lower trophozoite stage activity. We explored a series of ML901 analogues and identified ML471, which exhibits improved potency against trophozoites and enhanced selectivity against a human cell line. Additionally, it has no inhibitory activity against human ubiquitin-activating enzyme (UAE) in vitro. ML471 exhibits low nanomolar activity against asexual blood stage P. falciparum and potent activity against liver stage parasites, gametocytes and transmissible gametes. It is fast-acting and exhibits a long in vivo half-life. ML471 is well-tolerated and shows single dose oral efficacy in the SCID mouse model of P. falciparum malaria. We confirm that ML471 is a reaction hijacking inhibitor that is converted into a tight binding Tyr-ML471 conjugate by the PfTyrRS enzyme. A crystal structure of the PfTyrRS/ Tyr-ML471 complex offers insights into improved potency, while molecular docking into UAE provides a rationale for improved selectivity.
Collapse
Affiliation(s)
- Stanley C Xie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Chia-Wei Tai
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Craig J Morton
- Biomedical Manufacturing Program, CSIRO, Clayton South, Victoria, Australia
| | - Liting Ma
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Shih-Chung Huang
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Yawei Du
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Yongbo Hu
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Con Dogovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mina Salimimarand
- School of Chemistry, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert Griffin
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Dylan England
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Elisa de la Cruz
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Ioanna Deni
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Tomas Yeo
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Anna Y Burkhard
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Josefine Striepen
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Kyra A Schindler
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Benigno Crespo
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | | | - Craig A Hutton
- School of Chemistry, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tayla Rabie
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, South Africa
| | - Mufuliat T Famodimu
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael J Delves
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | | | - Anna C C Aguiar
- Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Dhelio B Pereira
- Research Center for Tropical Medicine of Rondonia, Porto Velho, Brazil
| | - Rafael V C Guido
- Sao Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Darren J Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - David A Fidock
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, United States of America
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Lawrence R Dick
- Seofon Consulting, Natick, Massachusetts, United States of America
| | | | - Alexandra E Gould
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Steven Langston
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Leann Tilley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Kumar A, Chithanna S, Li Y, Zhang X, Dodean RA, Caridha D, Madejczyk MS, Lee PJ, Jin X, Chetree R, Blount C, Dennis WE, DeLuca J, Vuong C, Pannone K, Dinh HT, Leed S, Roth A, Reynolds KA, Kelly JX, Kancharla P. Optimization of B-Ring-Functionalized Antimalarial Tambjamines and Prodiginines. J Med Chem 2024; 67:19755-19776. [PMID: 39425665 PMCID: PMC11563898 DOI: 10.1021/acs.jmedchem.4c02093] [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] [Indexed: 10/21/2024]
Abstract
Malaria has been a deadly enemy of mankind throughout history, affecting over 200 million people annually, along with approximately half a million deaths. Resistance to current therapies is of great concern, and there is a dire need for novel and well-tolerated antimalarials that operate by clinically unexploited mechanisms. We have previously reported that both tambjamines and prodiginines are highly potent novel antiplasmodial agents, but they required rigor optimizations to enhance the oral efficacy, safety, and physicochemical properties. Here, we launched a comprehensive structure-activity relationship study for B-ring-functionalized tambjamines and prodiginines with 54 novel analogues systematically designed and synthesized. A number of compounds exhibited remarkable antiplasmodial activities against asexual erythrocytic Plasmodium parasites, with improved safety and metabolic profiles. Notably, several prodiginines cured erythrocytic Plasmodium yoelii infections after oral 25 mg/kg × 4 days in a murine model and provided partial protection against liver stage Plasmodium berghei sporozoite-induced infection in mice.
Collapse
Affiliation(s)
- Amrendra Kumar
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Sivanna Chithanna
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Yuexin Li
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Xiaowei Zhang
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Rozalia A. Dodean
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Diana Caridha
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Michael S. Madejczyk
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Patricia J. Lee
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Xiannu Jin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Ravi Chetree
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Cameron Blount
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - William E. Dennis
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jesse DeLuca
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chau Vuong
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kristina Pannone
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Hieu T. Dinh
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan Leed
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Alison Roth
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kevin A. Reynolds
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Jane X. Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| |
Collapse
|
22
|
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
|
23
|
Fola AA, Kobayashi T, Hamapumbu H, Musonda M, Katowa B, Matoba J, Stevenson JC, Norris DE, Thuma PE, Wesolowski A, Moss WJ, Juliano JJ, Bailey JA. Temporal genomics in Southern Zambia shows rising prevalence of Plasmodium falciparum mutations linked to delayed clearance after artemisinin-lumefantrine treatment. Sci Rep 2024; 14:26789. [PMID: 39500918 PMCID: PMC11538544 DOI: 10.1038/s41598-024-76442-6] [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: 06/18/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
The emergence of antimalarial drug resistance is an impediment to malaria control and elimination in Africa. Analysis of temporal trends in molecular markers of resistance is critical to inform policy makers and guide malaria treatment guidelines. In a low and seasonal transmission region of southern Zambia, we successfully genotyped 85.5% (389/455) of Plasmodium falciparum samples collected between 2013 and 2018 from 8 spatially clustered health centres using molecular inversion probes (MIPs) targeting key drug resistance genes. Aside from one sample from 2016 carrying K13 622I, no other World Health Organization-validated or candidate artemisinin partial resistance (ART-R) mutations were observed. However, in the more recent years (2016-2017) five novel K13-propeller-domain mutations, C532S, A578S, Q613E, D680N and G718S were identified at low prevalence. Moreover, 13% (CI, 9.6-17.2) of isolates had the AP2MU 160N mutation, which has been associated with delayed clearance following artemisinin combination therapy in Africa. This mutation increased in prevalence between 2015 and 2018 and bears a genomic signature of selection. During this time period, there was an increase in the MDR1 NFD haplotype that is associated with reduced susceptibility to lumefantrine. Sulfadoxine-pyrimethamine polymorphisms were near fixation. While validated ART-R mutations are rare, a mutation associated with slow parasite clearance in Africa appears to be under selection in southern Zambia.
Collapse
Affiliation(s)
- Abebe A Fola
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02906, USA
| | - Tamaki Kobayashi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | | | | | | | | | - Douglas E Norris
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | - Amy Wesolowski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - William J Moss
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Jonathan J Juliano
- Institute for Global Health and Infectious Diseases, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Infectious Diseases, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02906, USA.
| |
Collapse
|
24
|
Barzkar N, Sukhikh S, Babich O. A comprehensive review of marine sponge metabolites, with emphasis on Neopetrosia sp. Int J Biol Macromol 2024; 280:135823. [PMID: 39313052 DOI: 10.1016/j.ijbiomac.2024.135823] [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: 07/13/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
The secondary metabolites that marine sponges create are essential to the advancement of contemporary medicine and are often employed in clinical settings. Over the past five years, microbes associated with sponges have yielded the identification of 140 novel chemicals. Statistics show that most are derived from actinomycetes (bacteria) and ascomycotes (fungi). The aim of this study was to investigate the biological activity of metabolites from marine sponges. Chlocarbazomycins A-D, which are a group of novel chlorinated carbazole alkaloids isolated from the sponge Neopetrosia fennelliae KUFA 0811, exhibit antimicrobial, cytotoxic, and enzyme inhibitory activities. Recently, marine sponges of the genus Neopetrosia have attracted attention due to the unique chemical composition of the compounds they produce, including alkaloids of potential importance in drug discovery. Fridamycin H and fridamycin I are two novel type II polyketides synthesized by sponge-associated bacteria exhibit antitrypanosomal activity. Fintiamin, composed of amino acids and terpenoid moieties, shows affinity for the cannabinoid receptor CB 1. It was found that out of 27 species of Neopetrosia sponges, the chemical composition of only 9 species has been studied. These species mainly produce bioactive substances such as alkaloids, quinones, sterols, and terpenoids. The presence of motuporamines is a marker of the species Neopetrosia exigua. Terpenoids are specific markers of Neopetrosia vanilla species. Although recently discovered, secondary metabolites from marine sponges have been shown to have diverse biological activities, antimicrobial, antiviral, antibacterial, antimicrobial, antioxidant, antimalarial, and anticancer properties, providing many lead compounds for drug development. The data presented in this review on known and future natural products derived from sponges will further clarify the role and importance of microbes in marine sponges and trace the prospects of their applications, especially in medicine, cosmeceuticals, environmental protection, and manufacturing industries.
Collapse
Affiliation(s)
- Noora Barzkar
- Higher Institution Center of Excellence, Borneo Marine Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia.
| | - Stanislav Sukhikh
- SEC "Applied Biotechnologies", Immanuel Kant Baltic Federal University, A. Nevskogo Street 14, Kaliningrad, 236016, Russia
| | - Olga Babich
- SEC "Applied Biotechnologies", Immanuel Kant Baltic Federal University, A. Nevskogo Street 14, Kaliningrad, 236016, Russia
| |
Collapse
|
25
|
Ferreira LT, Cassiano GC, Alvarez LCS, Okombo J, Calit J, Fontinha D, Gil-Iturbe E, Coyle R, Andrade CH, Sunnerhagen P, Bargieri DY, Prudêncio M, Quick M, Cravo PV, Lee MCS, Fidock DA, Costa FTM. A novel 4-aminoquinoline chemotype with multistage antimalarial activity and lack of cross-resistance with PfCRT and PfMDR1 mutants. PLoS Pathog 2024; 20:e1012627. [PMID: 39471233 PMCID: PMC11521309 DOI: 10.1371/journal.ppat.1012627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 11/01/2024] Open
Abstract
Artemisinin-based combination therapy (ACT) is the mainstay of effective treatment of Plasmodium falciparum malaria. However, the long-term utility of ACTs is imperiled by widespread partial artemisinin resistance in Southeast Asia and its recent emergence in parts of East Africa. This underscores the need to identify chemotypes with new modes of action (MoAs) to circumvent resistance to ACTs. In this study, we characterized the asexual blood stage antiplasmodial activity and resistance mechanisms of LDT-623, a 4-aminoquinoline (4-AQ). We also detected LDT-623 activity against multiple stages (liver schizonts, stage IV-V gametocytes, and ookinetes) of Plasmodium's life cycle, a feature unlike other 4-AQs such as chloroquine (CQ) and piperaquine (PPQ). Using heme fractionation profiling and drug uptake studies in PfCRT-containing proteoliposomes, we observed inhibition of hemozoin formation and PfCRT-mediated transport, which constitute characteristic features of 4-AQs' MoA. We also found minimal cross-resistance to LDT-623 in a panel of mutant pfcrt or pfmdr1 lines, but not the PfCRT F145I mutant that is highly resistant to PPQ resistance yet is very unfit. No P. falciparum parasites were recovered in an in vitro resistance selection study, suggesting a high barrier for resistance to emerge. Finally, a competitive growth assay comprising >50 barcoded parasite lines with mutated resistance mediators or major drug targets found no evidence of cross-resistance. Our findings support further exploration of this promising 4-AQ.
Collapse
Affiliation(s)
- Letícia Tiburcio Ferreira
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Gustavo Capatti Cassiano
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Luis Carlos Salazar Alvarez
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Juliana Calit
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
- Center for the Research and Advancement in Fragments and molecular Targets, School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Center for Excellence in Artificial Intelligence, Institute of Informatics, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Youssef Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Physiology & Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States of America
- New York State Psychiatric Institute, Area Neuroscience – Molecular Therapeutics, New York, New York, United States of America
| | - Pedro V. Cravo
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Marcus C. S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
26
|
Manh ND, Thanh NV, Quang HH, Van NTT, San NN, Phong NC, Birrell GW, Edgel KA, Martin NJ, Edstein MD, Chavchich M. Therapeutic efficacy of pyronaridine-artesunate (Pyramax) in treating Plasmodium vivax malaria in the central highlands of Vietnam. Antimicrob Agents Chemother 2024; 68:e0004424. [PMID: 39046237 PMCID: PMC11373200 DOI: 10.1128/aac.00044-24] [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: 01/09/2024] [Accepted: 05/19/2024] [Indexed: 07/25/2024] Open
Abstract
The emergence and spread of chloroquine-resistant Plasmodium vivax have necessitated the assessment of alternative blood schizonticidal drugs. In Vietnam, chloroquine-resistant P. vivax malaria has been reported. In an open-label, single-arm trial, the safety, tolerability, and efficacy of pyronaridine-artesunate (Pyramax, PA) was evaluated in Dak Nong province, Vietnam. A 3-day course of PA was administered to adults and children (≥20 kg) infected with P. vivax. Patients also received primaquine (0.25 mg/kg daily for 14 days). PA was well tolerated with transient asymptomatic increases in liver transaminases. The per-protocol proportion of patients with day 42 PCR-unadjusted adequate clinical and parasitological response was 96.0% (95% CI, 84.9%-99.0%, n = 48/50). The median parasite clearance time was 12 h (range, 12-36 h), with a median fever clearance time of 24 h (range, 12-60 h). Single nucleotide polymorphisms (SNPs) as potential genetic markers of reduced drug susceptibility were analyzed in three putative drug resistance markers, Pvcrt-o, Pvmdr1, and PvK12. Insertion at position K10 of the Pvcrt-o gene was found in 74.6% (44/59) of isolates. Pvmdr1 SNPs at Y976F and F1076L were present in 61% (36/59) and 78% (46/59), respectively. Amplification of Pvmdr1 gene (two copies) was found in 5.1% (3/59) of parasite samples. Only 5.1% (3/59) of isolates had mutation 552I of the PvK12 gene. Overall, PA rapidly cleared P. vivax blood asexual stages and was highly efficacious in treating vivax malaria, with no evidence of artemisinin resistance found. PA provides an alternative to chloroquine treatment for vivax malaria in Vietnam. CLINICAL TRIALS This study is registered with the Australian New Zealand Clinical Trials Registry as ACTRN12618001429246.
Collapse
Affiliation(s)
- Nguyen Duc Manh
- Vietnam People’s Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Nguyen Van Thanh
- Vietnam People’s Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Huynh Hong Quang
- Vietnam Ministry of Health Institute of Malariology, Parasitology and Entomology, Qui Nhon, Vietnam
| | | | - Nguyen Ngoc San
- Vietnam People’s Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Nguen Chinh Phong
- Vietnam People’s Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Geoffrey W. Birrell
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | | | | | - Michael D. Edstein
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Marina Chavchich
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| |
Collapse
|
27
|
Maiga M, Dembele L, Courlet P, Khandelwal A, Dara A, Sogore F, Diakité O, Maiga FO, Dao F, Sissoko S, Barre Y, Goita S, Diakite M, Diakite SAS, Djimde AA, Oeuvray C, Spangenberg T, Wicha SG, Demarta-Gatsi C. Towards clinically relevant dose ratios for Cabamiquine and Pyronaridine combination using P. falciparum field isolate data. Nat Commun 2024; 15:7659. [PMID: 39227370 PMCID: PMC11372057 DOI: 10.1038/s41467-024-51994-3] [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: 02/15/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
The selection and combination of dose regimens for antimalarials involve complex considerations including pharmacokinetic and pharmacodynamic interactions. In this study, we use immediate ex vivo P. falciparum field isolates to evaluate the effect of cabamiquine and pyronaridine as standalone treatments and in combination therapy. We feed the data into a pharmacometrics model to generate an interaction map and simulate meaningful clinical dose ratios. We demonstrate that the pharmacometrics model of parasite growth and killing provides a detailed description of parasite kinetics against cabamiquine-susceptible and resistant parasites. Pyronaridine monotherapy provides suboptimal killing rates at doses as high as 720 mg. In contrast, the combination of a single dose of 330 mg cabamiquine and 360 mg pyronaridine provides over 90% parasite killing in most of the simulated patients. The described methodology that combines a rapid, 3R-compliant in vitro method and modelling to set meaningful doses for new antimalarials could contribute to clinical drug development.
Collapse
Affiliation(s)
- Mohamed Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Laurent Dembele
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali.
| | - Perrine Courlet
- Merck Institute of Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | - Akash Khandelwal
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
- UCB Biosciences GmbH, Rolf-Schwarz-Schütte-Platz 1, Monheim am Rhein, Germany
| | - Antoine Dara
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fanta Sogore
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Ousmaila Diakité
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fatoumata O Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - François Dao
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Sekou Sissoko
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Yacouba Barre
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Siaka Goita
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Mahamadou Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Seidina A S Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Abdoulaye A Djimde
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali
| | - Claude Oeuvray
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Thomas Spangenberg
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstr. 45, Hamburg, Germany.
| | - Claudia Demarta-Gatsi
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali.
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Watson OJ, Muchiri S, Ward A, Meier-Sherling C, Asua V, Katairo T, Brewer T, Cuomo-Dannenburg G, Winskill P, Bailey JA, Okell L, Scudu G, Woolsey AM. Risk of selection and timelines for the continued spread of artemisinin and partner drug resistance in Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.28.24312699. [PMID: 39252921 PMCID: PMC11383480 DOI: 10.1101/2024.08.28.24312699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The introduction of artemisinin combination therapies (ACTs) has significantly reduced the burden of Plasmodium falciparum malaria, yet the emergence of artemisinin partial resistance (ART-R) as well as partner drug resistance threatens these gains. Recent confirmations of prevalent de novo ART-R mutations in Africa, in particular in Rwanda, Uganda and Ethiopia, underscore the urgency of addressing this issue in Africa. Our objective is to characterise this evolving resistance landscape in Africa and understand the speed with which ART-R will continue to spread. We produce estimates of both ART-R and partner drug resistance by bringing together WHO, WWARN and MalariaGen Pf7k data on antimalarial resistance in combination with a literature review. We integrate these estimates within a mathematical modelling approach, aincorporating to estimate parameters known to impact the selection of ART-R for each malaria-endemic country and explore scenarios of ART-R spread and establishment. We identify 16 malaria-endemic countries in Africa to prioritise for surveillance and future deployment of alternative antimalarial strategies, based on ART-R reaching greater than 10% prevalence by 2040 under current malaria burden and effective-treatment coverage. If resistance continues to spread at current rates with no change in drug policy, we predict that partner drug resistance will emerge and the mean percentage of treatment failure across Africa will reach 30.74% by 2060 (parameter uncertainty range: 24.98% - 34.54%). This translates to an alarming number of treatment failures, with 52,980,600 absolute cases of treatment failure predicted in 2060 in Africa (parameter uncertainty range: 26,374,200 - 93,672,400) based on current effective treatment coverage. Our results provide a refined and updated prediction model for the emergence of ART-R to help guide antimalarial policy and prioritise future surveillance efforts and innovation in Africa. These results put into stark context the speed with which antimalarial resistance may spread in Africa if left unchecked, confirming the need for swift and decisive action in formulating antimalarial treatment policies focused on furthering malaria control and containing antimalarial resistance in Africa. The rise of artemisinin partial resistance (ART-R) and increasing partner drug tolerance by Plasmodium falciparum malaria in Africa threatens to undo malaria control efforts. Recent confirmations of de novo ART-R markers in Rwanda, Uganda, and Ethiopia highlight the urgent need to address this threat in Africa, where the vast majority of cases and deaths occur. This study characterises the resistance landscape and predicts the spread of antimalarial resistance across Africa. We estimate and map the current levels of resistance markers related to artemisinin and its partner drugs using WHO, WWARN, and MalariaGen Pf7k data. We combine these estimates with current malaria transmission and treatment data and use an established individual-based model of malaria resistance to simulate future resistance spread. We identify 16 African countries at highest risk of ART-R for prioritisation of enhanced surveillance and alternative antimalarial strategies. We project that, without policy changes, ART-R will exceed 10% in these regions by 2040. By 2060, if resistance spreads unchecked, we predict mean treatment failure rates will reach 30.74% (parameter uncertainty range: 24.98% - 34.54%) across Africa. This alarming spread of resistance is predicted to cause 52.98 million treatment failures (uncertainty range: 26.37 million - 93.67 million) in 2060. The impact of antimalarial resistance in Africa, if left unchecked, would hugely damage efforts to reduce malaria burden. Our results underscore the critical need for swift policy action to contain resistance and guide future surveillance and intervention efforts.
Collapse
Affiliation(s)
- Oliver J Watson
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, London, UK
| | - Salome Muchiri
- Clinton Health Access Initiative, Boston, MA, 21127, USA
| | - Abby Ward
- Clinton Health Access Initiative, Boston, MA, 21127, USA
| | | | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Tom Brewer
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, London, UK
| | - Gina Cuomo-Dannenburg
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, London, UK
| | - Peter Winskill
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, 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
| | - Lucy Okell
- MRC Centre for Global Infectious Disease Analysis, Faculty of Medicine, Imperial College London, London, UK
| | | | | |
Collapse
|
31
|
Tam LT, Thinkhamrop K, Suttiprapa S, Clements ACA, Wangdi K, Suwannatrai AT. Bayesian spatio-temporal modelling of environmental, climatic, and socio-economic influences on malaria in Central Vietnam. Malar J 2024; 23:258. [PMID: 39182127 PMCID: PMC11344946 DOI: 10.1186/s12936-024-05074-y] [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] [Accepted: 08/09/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Despite the successful efforts in controlling malaria in Vietnam, the disease remains a significant health concern, particularly in Central Vietnam. This study aimed to assess correlations between environmental, climatic, and socio-economic factors in the district with malaria cases. METHODS The study was conducted in 15 provinces in Central Vietnam from January 2018 to December 2022. Monthly malaria cases were obtained from the Institute of Malariology, Parasitology, and Entomology Quy Nhon, Vietnam. Environmental, climatic, and socio-economic data were retrieved using a Google Earth Engine script. A multivariable Zero-inflated Poisson regression was undertaken using a Bayesian framework with spatial and spatiotemporal random effects with a conditional autoregressive prior structure. The posterior random effects were estimated using Bayesian Markov Chain Monte Carlo simulation with Gibbs sampling. RESULTS There was a total of 5,985 Plasmodium falciparum and 2,623 Plasmodium vivax cases during the study period. Plasmodium falciparum risk increased by five times (95% credible interval [CrI] 4.37, 6.74) for each 1-unit increase of normalized difference vegetation index (NDVI) without lag and by 8% (95% CrI 7%, 9%) for every 1ºC increase in maximum temperature (TMAX) at a 6-month lag. While a decrease in risk of 1% (95% CrI 0%, 1%) for a 1 mm increase in precipitation with a 6-month lag was observed. A 1-unit increase in NDVI at a 1-month lag was associated with a four-fold increase (95% CrI 2.95, 4.90) in risk of P. vivax. In addition, the risk increased by 6% (95% CrI 5%, 7%) and 3% (95% CrI 1%, 5%) for each 1ºC increase in land surface temperature during daytime with a 6-month lag and TMAX at a 4-month lag, respectively. Spatial analysis showed a higher mean malaria risk of both species in the Central Highlands and southeast parts of Central Vietnam and a lower risk in the northern and north-western areas. CONCLUSION Identification of environmental, climatic, and socio-economic risk factors and spatial malaria clusters are crucial for designing adaptive strategies to maximize the impact of limited public health resources toward eliminating malaria in Vietnam.
Collapse
Affiliation(s)
- Le Thanh Tam
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Epidemiology, Institute of Malariology, Parasitology, and Entomology Quy Nhon, Quy Nhon, Binh Dinh, Vietnam
| | - Kavin Thinkhamrop
- Health and Epidemiology Geoinformatics Research (HEGER), Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand
| | - Sutas Suttiprapa
- Department of Tropical Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Kinley Wangdi
- HEAL Global Research Centre, Health Research Institute, University of Canberra, Canberra, ACT 2617, Australia
- National Centre for Epidemiology and Population Health, College of Health and Medicine, Australian National University, Canberra, Australia
| | - Apiporn T Suwannatrai
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
32
|
Fowkes FJ, Robinson LJ, Simpson JA, Thriemer K, Laman M, Oo WH. The challenges of malaria elimination in the Asia-Pacific region. Med J Aust 2024; 221:182-185. [PMID: 39104144 DOI: 10.5694/mja2.52398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Affiliation(s)
- Freya Ji Fowkes
- University of Melbourne, Melbourne, VIC
- Burnet Institute, Melbourne, VIC
- Monash University, Melbourne, VIC
| | | | | | | | - Moses Laman
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Win Han Oo
- University of Melbourne, Melbourne, VIC
- Burnet Institute, Melbourne, VIC
| |
Collapse
|
33
|
Gawriljuk VO, Godoy AS, Oerlemans R, Welker LAT, Hirsch AKH, Groves MR. Cryo-EM structure of 1-deoxy-D-xylulose 5-phosphate synthase DXPS from Plasmodium falciparum reveals a distinct N-terminal domain. Nat Commun 2024; 15:6642. [PMID: 39103329 PMCID: PMC11300867 DOI: 10.1038/s41467-024-50671-9] [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: 12/18/2023] [Accepted: 07/17/2024] [Indexed: 08/07/2024] Open
Abstract
Plasmodium falciparum is the main causative agent of malaria, a deadly disease that mainly affects children under five years old. Artemisinin-based combination therapies have been pivotal in controlling the disease, but resistance has arisen in various regions, increasing the risk of treatment failure. The non-mevalonate pathway is essential for the isoprenoid synthesis in Plasmodium and provides several under-explored targets to be used in the discovery of new antimalarials. 1-deoxy-D-xylulose-5-phosphate synthase (DXPS) is the first and rate-limiting enzyme of the pathway. Despite its importance, there are no structures available for any Plasmodium spp., due to the complex sequence which contains large regions of high disorder, making crystallisation a difficult task. In this manuscript, we use cryo-electron microscopy to solve the P. falciparum DXPS structure at a final resolution of 2.42 Å. Overall, the structure resembles other DXPS enzymes but includes a distinct N-terminal domain exclusive to the Plasmodium genus. Mutational studies show that destabilization of the cap domain interface negatively impacts protein stability and activity. Additionally, a density for the co-factor thiamine diphosphate is found in the active site. Our work highlights the potential of cryo-EM to obtain structures of P. falciparum proteins that are unfeasible by means of crystallography.
Collapse
Affiliation(s)
- Victor O Gawriljuk
- Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Andre S Godoy
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 - Jardim Santa Angelina, Sao Carlos, 13563-120, Brazil
| | - Rick Oerlemans
- Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Luise A T Welker
- Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
- Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Matthew R Groves
- Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
34
|
Kane J, Li X, Kumar S, Button-Simons KA, Vendrely Brenneman KM, Dahlhoff H, Sievert MAC, Checkley LA, Shoue DA, Singh PP, Abatiyow BA, Haile MT, Nair S, Reyes A, Tripura R, Peto TJ, Lek D, Mukherjee A, Kappe SHI, Dhorda M, Nkhoma SC, Cheeseman IH, Vaughan AM, Anderson TJC, Ferdig MT. A Plasmodium falciparum genetic cross reveals the contributions of pfcrt and plasmepsin II/III to piperaquine drug resistance. mBio 2024; 15:e0080524. [PMID: 38912775 PMCID: PMC11253641 DOI: 10.1128/mbio.00805-24] [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: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Piperaquine (PPQ) is widely used in combination with dihydroartemisinin as a first-line treatment against malaria. Multiple genetic drivers of PPQ resistance have been reported, including mutations in the Plasmodium falciparum chloroquine resistance transporter (pfcrt) and increased copies of plasmepsin II/III (pm2/3). We generated a cross between a Cambodia-derived multidrug-resistant KEL1/PLA1 lineage isolate (KH004) and a drug-susceptible Malawian parasite (Mal31). Mal31 harbors a wild-type (3D7-like) pfcrt allele and a single copy of pm2/3, while KH004 has a chloroquine-resistant (Dd2-like) pfcrt allele with an additional G367C substitution and multiple copies of pm2/3. We recovered 104 unique recombinant parasites and examined a targeted set of progeny representing all possible combinations of variants at pfcrt and pm2/3. We performed a detailed analysis of competitive fitness and a range of PPQ susceptibility phenotypes with these progenies, including PPQ survival assay, area under the dose response curve, and a limited point IC50. We find that inheritance of the KH004 pfcrt allele is required for reduced PPQ sensitivity, whereas copy number variation in pm2/3 further decreases susceptibility but does not confer resistance in the absence of additional mutations in pfcrt. A deep investigation of genotype-phenotype relationships demonstrates that progeny clones from experimental crosses can be used to understand the relative contributions of pfcrt, pm2/3, and parasite genetic background to a range of PPQ-related traits. Additionally, we find that the resistance phenotype associated with parasites inheriting the G367C substitution in pfcrt is consistent with previously validated PPQ resistance mutations in this transporter.IMPORTANCEResistance to piperaquine, used in combination with dihydroartemisinin, has emerged in Cambodia and threatens to spread to other malaria-endemic regions. Understanding the causal mutations of drug resistance and their impact on parasite fitness is critical for surveillance and intervention and can also reveal new avenues to limiting the evolution and spread of drug resistance. An experimental genetic cross is a powerful tool for pinpointing the genetic determinants of key drug resistance and fitness phenotypes and has the distinct advantage of quantifying the effects of naturally evolved genetic variation. Our study was strengthened since the full range of copies of KH004 pm2/3 was inherited among the progeny clones, allowing us to directly test the role of the pm2/3 copy number on resistance-related phenotypes in the context of a unique pfcrt allele. Our multigene model suggests an important role for both loci in the evolution of this multidrug-resistant parasite lineage.
Collapse
Affiliation(s)
- John Kane
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Xue Li
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Katrina A. Button-Simons
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Katelyn M. Vendrely Brenneman
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Haley Dahlhoff
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Mackenzie A. C. Sievert
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Lisa A. Checkley
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Douglas A. Shoue
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Puspendra P. Singh
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Biley A. Abatiyow
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Meseret T. Haile
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Shalini Nair
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ann Reyes
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - 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 Research Building, University of Oxford Old Road Campus, Oxford, United Kingdom
| | - 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 Research Building, University of Oxford Old Road Campus, Oxford, United Kingdom
| | - Dysoley Lek
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
- School of Public Health, National Institute of Public Health, Phnom Penh, Cambodia
| | - Angana Mukherjee
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
- Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana, USA
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - 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 Research Building, University of Oxford Old Road Campus, Oxford, United Kingdom
| | - Standwell C. Nkhoma
- BEI Resources, American Type Culture Collection (ATCC), Manassas, Virginia, USA
| | - Ian H. Cheeseman
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Timothy J. C. Anderson
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Michael T. Ferdig
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
35
|
Giannangelo C, Challis MP, Siddiqui G, Edgar R, Malcolm TR, Webb CT, Drinkwater N, Vinh N, Macraild C, Counihan N, Duffy S, Wittlin S, Devine SM, Avery VM, De Koning-Ward T, Scammells P, McGowan S, Creek DJ. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. eLife 2024; 13:RP92990. [PMID: 38976500 PMCID: PMC11230628 DOI: 10.7554/elife.92990] [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: 07/10/2024] Open
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum (PfA-M1) and Plasmodium vivax (PvA-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets PfA-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on PfA-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of PfA-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
Affiliation(s)
- Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Matthew P Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Rebecca Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Tess R Malcolm
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Chaille T Webb
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Nyssa Drinkwater
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Natalie Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Christopher Macraild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Natalie Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
| | - Sergio Wittlin
- Swiss Tropical and Public Health InstituteAllschwilSwitzerland
- University of BaselBaselSwitzerland
| | - Shane M Devine
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, The University of MelbourneParkvilleAustralia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
- School of Environment and Science, Griffith UniversityNathanAustralia
| | - Tania De Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Peter Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| |
Collapse
|
36
|
Tran THT, Hien BTT, Dung NTL, Huong NT, Binh TT, Van Long N, Ton ND. Evaluation of Dihydroartemisinin-Piperaquine Efficacy and Molecular Markers in Uncomplicated Falciparum Patients: A Study across Binh Phuoc and Dak Nong, Vietnam. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1013. [PMID: 38929629 PMCID: PMC11205605 DOI: 10.3390/medicina60061013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Malaria continues to be a significant global health challenge. The efficacy of artemisinin-based combination therapies (ACTs) has declined in many parts of the Greater Mekong Subregion, including Vietnam, due to the spread of resistant malaria strains. This study was conducted to assess the efficacy of the Dihydroartemisinin (DHA)-Piperaquine (PPQ) regimen in treating uncomplicated falciparum malaria and to conduct molecular surveillance of antimalarial drug resistance in Binh Phuoc and Dak Nong provinces. Materials and Methods: The study included 63 uncomplicated malaria falciparum patients from therapeutic efficacy studies (TES) treated following the WHO treatment guidelines (2009). Molecular marker analysis was performed on all 63 patients. Methods encompassed Sanger sequencing for pfK13 mutations and quantitative real-time PCR for the pfpm2 gene. Results: This study found a marked decrease in the efficacy of the DHA-PPQ regimen, with an increased rate of treatment failures at two study sites. Genetic analysis revealed a significant presence of pfK13 mutations and pfpm2 amplifications, indicating emerging resistance to artemisinin and its partner drug. Conclusions: The effectiveness of the standard DHA-PPQ regimen has sharply declined, with rising treatment failure rates. This decline necessitates a review and possible revision of national malaria treatment guidelines. Importantly, molecular monitoring and clinical efficacy assessments together provide a robust framework for understanding and addressing detection drug resistance in malaria.
Collapse
Affiliation(s)
- Thu Huyen Thi Tran
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi 100000, Vietnam; (T.H.T.T.); (B.T.T.H.); (N.T.L.D.); (N.V.L.)
- Department of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| | - Bui Thi Thu Hien
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi 100000, Vietnam; (T.H.T.T.); (B.T.T.H.); (N.T.L.D.); (N.V.L.)
| | - Nguyen Thi Lan Dung
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi 100000, Vietnam; (T.H.T.T.); (B.T.T.H.); (N.T.L.D.); (N.V.L.)
| | - Nguyen Thi Huong
- National Burn Hospital, Vietnam Military Medical University, Hanoi 100000, Vietnam
| | - Tran Thanh Binh
- 103 Hospital, Vietnam Military Medical University, Hanoi 100000, Vietnam;
| | - Nguyen Van Long
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi 100000, Vietnam; (T.H.T.T.); (B.T.T.H.); (N.T.L.D.); (N.V.L.)
| | - Nguyen Dang Ton
- Department of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| |
Collapse
|
37
|
Fola AA, Kobayashi T, Shields T, Hamapumbu H, Musonda M, Katowa B, Matoba J, Stevenson JC, Norris DE, Thuma PE, Wesolowski A, Moss WJ, Juliano JJ, Bailey JA. Temporal genomic analysis of Plasmodium falciparum reveals increased prevalence of mutations associated with delayed clearance following treatment with artemisinin-lumefantrine in Choma District, Southern Province, Zambia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.05.24308497. [PMID: 38883763 PMCID: PMC11178023 DOI: 10.1101/2024.06.05.24308497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The emergence of antimalarial drug resistance is an impediment to malaria control and elimination in Africa. Analysis of temporal trends in molecular markers of resistance is critical to inform policy makers and guide malaria treatment guidelines. In a low and seasonal transmission region of southern Zambia, we successfully genotyped 85.5% (389/455) of Plasmodium falciparum samples collected between 2013-2018 from 8 spatially clustered health centres using molecular inversion probes (MIPs) targeting key drug resistance genes. Aside from one sample carrying K13 R622I, none of the isolates carried other World Health Organization-validated or candidate artemisinin partial resistance (ART-R) mutations in K13. However, 13% (CI, 9.6-17.2) of isolates had the AP2MU S160N mutation, which has been associated with delayed clearance following artemisinin combination therapy in Africa. This mutation increased in prevalence between 2015-2018 and bears a genomic signature of selection. During this time period, there was an increase in the MDR1 NFD haplotype that is associated with reduced susceptibility to lumefantrine. Sulfadoxine-pyrimethamine polymorphisms were near fixation. While validated ART-R mutations are rare, a mutation associated with slow parasite clearance in Africa appears to be under selection in southern Zambia.
Collapse
Affiliation(s)
- Abebe A. Fola
- Department of Pathology and Laboratory Medicine, Brown University, RI, USA, 02906
| | - Tamaki Kobayashi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
| | - Timothy Shields
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
| | | | | | - Ben Katowa
- Macha Research Trust, Choma District, Zambia
| | | | | | - Douglas E. Norris
- Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
| | | | - Amy Wesolowski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
| | - William J. Moss
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
- Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 21205
| | - Jonathan J. Juliano
- Institute for Global Health and Infectious Diseases, University of North Carolina Chapel Hill, NC, USA, 27599
- Division of Infectious Diseases, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina Chapel Hill, Chapel Hill, NC, USA, 27599
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599
| | - Jeffrey A. Bailey
- Department of Pathology and Laboratory Medicine, Brown University, RI, USA, 02906
| |
Collapse
|
38
|
Achan J, Barry A, Leroy D, Kamara G, Duparc S, Kaszubska W, Gandhi P, Buffet B, Tshilab P, Ogutu B, Taylor T, Krishna S, Richardson N, Ramachandruni H, Rietveld H. Defining the next generation of severe malaria treatment: a target product profile. Malar J 2024; 23:174. [PMID: 38835069 DOI: 10.1186/s12936-024-04986-z] [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: 02/09/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Severe malaria is a life-threatening infection, particularly affecting children under the age of 5 years in Africa. Current treatment with parenteral artemisinin derivatives is highly efficacious. However, artemisinin partial resistance is widespread in Southeast Asia, resulting in delayed parasite clearance after therapy, and has emerged independently in South America, Oceania, and Africa. Hence, new treatments for severe malaria are needed, and it is prudent to define their characteristics now. This manuscript focuses on the target product profile (TPP) for new treatments for severe malaria. It also highlights preparedness when considering ways of protecting the utility of artemisinin-based therapies. TARGET PRODUCT PROFILE Severe malaria treatments must be highly potent, with rapid onset of antiparasitic activity to clear the infection as quickly as possible to prevent complications. They should also have a low potential for drug resistance selection, given the high parasite burden in patients with severe malaria. Combination therapies are needed to deter resistance selection and dissemination. Partner drugs which are approved for uncomplicated malaria treatment would provide the most rapid development pathway for combinations, though new candidate molecules should be considered. Artemisinin combination approaches to severe malaria would extend the lifespan of current therapy, but ideally, completely novel, non-artemisinin-based combination therapies for severe malaria should be developed. These should be advanced to at least phase 2 clinical trials, enabling rapid progression to patient use should current treatment fail clinically. New drug combinations for severe malaria should be available as injectable formulations for rapid and effective treatment, or as rectal formulations for pre-referral intervention in resource-limited settings. CONCLUSION Defining the TPP is a key step to align responses across the community to proactively address the potential for clinical failure of artesunate in severe malaria. In the shorter term, artemisinin-based combination therapies should be developed using approved or novel drugs. In the longer term, novel combination treatments should be pursued. Thus, this TPP aims to direct efforts to preserve the efficacy of existing treatments while improving care and outcomes for individuals affected by this life-threatening disease.
Collapse
Affiliation(s)
| | - Aïssata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Didier Leroy
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | - George Kamara
- Médecins Sans Frontières, Magburaka District Hospital, Freetown, Sierra Leone
| | - Stephan Duparc
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | - Wiweka Kaszubska
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | | | - Bénédicte Buffet
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | | | - Bernhards Ogutu
- Centre for Clinical Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Terrie Taylor
- Queen Elizabeth Central Hospital and Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Sanjeev Krishna
- Institut Für Tropenmedizin, Eberhard Karls Universität Tübingen, and German Center for Infection Research (Dzif), Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
- Clinical Academic Group, Institute for Infection and Immunity, St. George's University of London, London, UK
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Hanu Ramachandruni
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland.
| | - Hans Rietveld
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland.
| |
Collapse
|
39
|
Fukuda N, Yoshida N, Balikagala B, Tsuru I, Ikeda M, Hirai M, Anywar DA, Odongo-Aginya EI, Mita T. Detection of drug-resistant malaria in resource-limited settings: efficient and high-throughput surveillance of artemisinin and partner drug resistance. J Antimicrob Chemother 2024; 79:1418-1422. [PMID: 38661223 DOI: 10.1093/jac/dkae120] [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: 01/08/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024] Open
Abstract
OBJECTIVES Artemisinin-resistant Plasmodium falciparum malaria is currently spreading globally, including in Africa. Artemisinin resistance also leads to resistance to partner drugs used in artemisinin-based combination therapies. Sequencing of kelch13, which is associated with artemisinin resistance, culture-based partner drug susceptibility tests, and ELISA-based growth measurement are conventionally used to monitor resistance; however, their application is challenging in resource-limited settings. METHODS An experimental package for field studies with minimum human/material requirements was developed. RESULTS First, qPCR-based SNP assay was applied in artemisinin resistance screening, which can detect mutations within 1 h and facilitate sample selection for subsequent processes. It had 100% sensitivity and specificity compared with DNA sequencing in the detection of the two common artemisinin resistance mutations in Uganda, C469Y and A675V. Moreover, in the partner drug susceptibility test, the cultured samples were dry-preserved on a 96-well filter paper plate and shipped to the central laboratory. Parasite growth was measured by ELISA using redissolved samples. It well reproduced the results of direct ELISA, reducing significant workload in the field (Pearson correlation coefficient: 0.984; 95% CI: 0.975-0.990). CONCLUSIONS Large-scale and sustainable monitoring is required urgently to track rapidly spreading drug-resistant malaria. In malaria-endemic areas, where research resources are often limited, simplicity and feasibility of the procedure is especially important. Our approach combines a qPCR-based rapid test, which is also applicable to point-of-care diagnosis of artemisinin resistance and centralized analysis of ex vivo culture. The approach could improve efficiency of field experiments and accelerate global drug resistance surveillance.
Collapse
Affiliation(s)
- Naoyuki Fukuda
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Naoko Yoshida
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Betty Balikagala
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Ibuki Tsuru
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Mie Ikeda
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Makoto Hirai
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | | | | | - Toshihiro Mita
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
40
|
Duffey M, Shafer RW, Timm J, Burrows JN, Fotouhi N, Cockett M, Leroy D. Combating antimicrobial resistance in malaria, HIV and tuberculosis. Nat Rev Drug Discov 2024; 23:461-479. [PMID: 38750260 DOI: 10.1038/s41573-024-00933-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 06/07/2024]
Abstract
Antimicrobial resistance poses a significant threat to the sustainability of effective treatments against the three most prevalent infectious diseases: malaria, human immunodeficiency virus (HIV) infection and tuberculosis. Therefore, there is an urgent need to develop novel drugs and treatment protocols capable of reducing the emergence of resistance and combating it when it does occur. In this Review, we present an overview of the status and underlying molecular mechanisms of drug resistance in these three diseases. We also discuss current strategies to address resistance during the research and development of next-generation therapies. These strategies vary depending on the infectious agent and the array of resistance mechanisms involved. Furthermore, we explore the potential for cross-fertilization of knowledge and technology among these diseases to create innovative approaches for minimizing drug resistance and advancing the discovery and development of new anti-infective treatments. In conclusion, we advocate for the implementation of well-defined strategies to effectively mitigate and manage resistance in all interventions against infectious diseases.
Collapse
Affiliation(s)
- Maëlle Duffey
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland
- The Global Antibiotic Research & Development Partnership, Geneva, Switzerland
| | - Robert W Shafer
- Department of Medicine/Infectious Diseases, Stanford University, Palo Alto, CA, USA
| | | | - Jeremy N Burrows
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland
| | | | | | - Didier Leroy
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland.
| |
Collapse
|
41
|
Rosenthal PJ, Asua V, Conrad MD. Emergence, transmission dynamics and mechanisms of artemisinin partial resistance in malaria parasites in Africa. Nat Rev Microbiol 2024; 22:373-384. [PMID: 38321292 DOI: 10.1038/s41579-024-01008-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
Malaria, mostly due to Plasmodium falciparum infection in Africa, remains one of the most important infectious diseases in the world. Standard treatment for uncomplicated P. falciparum malaria is artemisinin-based combination therapy (ACT), which includes a rapid-acting artemisinin derivative plus a longer-acting partner drug, and standard therapy for severe P. falciparum malaria is intravenous artesunate. The efficacy of artemisinins and ACT has been threatened by the emergence of artemisinin partial resistance in Southeast Asia, mediated principally by mutations in the P. falciparum Kelch 13 (K13) protein. High ACT treatment failure rates have occurred when resistance to partner drugs is also seen. Recently, artemisinin partial resistance has emerged in Rwanda, Uganda and the Horn of Africa, with independent emergences of different K13 mutants in each region. In this Review, we summarize our current knowledge of artemisinin partial resistance and focus on the emergence of resistance in Africa, including its epidemiology, transmission dynamics and mechanisms. At present, the clinical impact of emerging resistance in Africa is unclear and most available evidence suggests that the efficacies of leading ACTs remain excellent, but there is an urgent need to better appreciate the extent of the problem and its consequences for the treatment and control of malaria.
Collapse
Affiliation(s)
| | - Victor Asua
- Infectious Diseases Research Collaboration, Kampala, Uganda
- University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
42
|
Creek D, Giannangelo C, Challis M, Siddiqui G, Edgar R, Malcolm T, Webb C, Drinkwater N, Vinh N, MacRaild C, Counihan N, Duffy S, Wittlin S, Devine S, Avery V, de Koning-Ward T, Scammells P, McGowan S. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. RESEARCH SQUARE 2024:rs.3.rs-3251230. [PMID: 38746424 PMCID: PMC11092810 DOI: 10.21203/rs.3.rs-3251230/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum ( Pf A-M1) and Plasmodium vivax ( Pv A-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets Pf A-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on Pf A-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of Pf A-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
|
43
|
Ajibaye O, Olukosi YA, Oriero EC, Oboh MA, Iwalokun B, Nwankwo IC, Nnam CF, Adaramoye OV, Chukwemeka S, Okanazu J, Gabriel E, Balogun EO, Amambua-Ngwa A. Detection of novel Plasmodium falciparum coronin gene mutations in a recrudescent ACT-treated patient in South-Western Nigeria. Front Cell Infect Microbiol 2024; 14:1366563. [PMID: 38716192 PMCID: PMC11074373 DOI: 10.3389/fcimb.2024.1366563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/28/2024] [Indexed: 05/12/2024] Open
Abstract
Background Routine surveillance for antimalarial drug resistance is critical to sustaining the efficacy of artemisinin-based Combination Therapies (ACTs). Plasmodium falciparum kelch-13 (Pfkelch-13) and non-Pfkelch-13 artemisinin (ART) resistance-associated mutations are uncommon in Africa. We investigated polymorphisms in Plasmodium falciparum actin-binding protein (Pfcoronin) associated with in vivo reduced sensitivity to ART in Nigeria. Methods Fifty-two P. falciparum malaria subjects who met the inclusion criteria were followed up in a 28-day therapeutic efficacy study of artemether-lumefantrine in Lagos, Nigeria. Parasite detection was done by microscopy and molecular diagnostic approaches involving PCR amplification of genes for Pf18S rRNA, varATS, telomere-associated repetitive elements-2 (TARE-2). Pfcoronin and Pfkelch-13 genes were sequenced bi-directionally while clonality of infections was determined using 12 neutral P. falciparum microsatellite loci and msp2 analyses. Antimalarial drugs (sulfadoxine-pyrimethamine, amodiaquine, chloroquine and some quinolones) resistance variants (DHFR_51, DHFR_59, DHFR_108, DHFR_164, MDR1_86, MDR1_184, DHPS_581 and DHPS_613) were genotyped by high-resolution melting (HRM) analysis. Results A total of 7 (26.92%) cases were identified either as early treatment failure, late parasitological failure or late clinical failure. Of the four post-treatment infections identified as recrudescence by msp2 genotypes, only one was classified as recrudescence by multilocus microsatellites genotyping. Microsatellite analysis revealed no significant difference in the mean allelic diversity, He, (P = 0.19, Mann-Whitney test). Allele sizes and frequency per locus implicated one isolate. Genetic analysis of this isolate identified two new Pfcoronin SNVs (I68G and L173F) in addition to the P76S earlier reported. Linkage-Disequilibrium as a standardized association index, IAS, between multiple P. falciparum loci revealed significant LD (IAS = 0.2865, P=0.02, Monte-Carlo simulation) around the neutral microsatellite loci. The pfdhfr/pfdhps/pfmdr1 drug resistance-associated haplotypes combinations, (108T/N/51I/164L/59R/581G/86Y/184F), were observed in two samples. Conclusion Pfcoronin mutations identified in this study, with potential to impact parasite clearance, may guide investigations on emerging ART tolerance in Nigeria, and West African endemic countries.
Collapse
Affiliation(s)
- Olusola Ajibaye
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
- Medical Research Council Unit, the Gambia – The London School of Hygiene and Tropical Medicine, Fajara, Banjul, Gambia
| | - Yetunde Adeola Olukosi
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Eniyou C. Oriero
- Medical Research Council Unit, the Gambia – The London School of Hygiene and Tropical Medicine, Fajara, Banjul, Gambia
| | - Mary Aigbiremo Oboh
- Medical Research Council Unit, the Gambia – The London School of Hygiene and Tropical Medicine, Fajara, Banjul, Gambia
| | - Bamidele Iwalokun
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Ikechukwu Chidiebere Nwankwo
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Chinaza Favour Nnam
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Olawunmi Victoria Adaramoye
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Idi-araba, Surulere, Lagos, Nigeria
| | - Somadina Chukwemeka
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Judith Okanazu
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Eniafe Gabriel
- Malaria Genomics Research and Training Centre, Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Emmanuel Oluwadare Balogun
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, Gilman Drive, La Jolla, CA, United States
| | - Alfred Amambua-Ngwa
- Medical Research Council Unit, the Gambia – The London School of Hygiene and Tropical Medicine, Fajara, Banjul, Gambia
| |
Collapse
|
44
|
Lucky AB, Wang C, Li X, Liang X, Muneer A, Miao J. Transforming the CRISPR/dCas9-based gene regulation technique into a forward screening tool in Plasmodium falciparum. iScience 2024; 27:109602. [PMID: 38617559 PMCID: PMC11015506 DOI: 10.1016/j.isci.2024.109602] [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: 08/09/2023] [Revised: 02/11/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024] Open
Abstract
It is a significant challenge to assess the functions of many uncharacterized genes in human malaria parasites. Here, we present a genetic screening tool to assess the contribution of essential genes from Plasmodium falciparum by the conditional CRISPR-/deadCas9-based interference and activation (i/a) systems. We screened both CRISPRi and CRISPRa sets, consisting of nine parasite lines per set targeting nine genes via their respective gRNAs. By conducting amplicon sequencing of gRNA loci, we identified the contribution of each targeted gene to parasite fitness upon drug (artemisinin, chloroquine) and stress (starvation, heat shock) treatment. The screening was highly reproducible, and the screening libraries were easily generated by transfection of mixed plasmids expressing different gRNAs. We demonstrated that this screening is straightforward, robust, and can provide a fast and efficient tool to study essential genes that have long presented a bottleneck in assessing their functions using existing genetic tools.
Collapse
Affiliation(s)
- Amuza Byaruhanga Lucky
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Xiaolian Li
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Xiaoying Liang
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Azhar Muneer
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| |
Collapse
|
45
|
Wirjanata G, Lin J, Dziekan JM, El Sahili A, Chung Z, Tjia S, Binte Zulkifli NE, Boentoro J, Tham R, Jia LS, Go KD, Yu H, Partridge A, Olsen D, Prabhu N, Sobota RM, Nordlund P, Lescar J, Bozdech Z. Identification of an inhibitory pocket in falcilysin provides a new avenue for malaria drug development. Cell Chem Biol 2024; 31:743-759.e8. [PMID: 38593807 DOI: 10.1016/j.chembiol.2024.03.002] [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: 03/29/2023] [Revised: 09/02/2023] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
Identification of new druggable protein targets remains the key challenge in the current antimalarial development efforts. Here we used mass-spectrometry-based cellular thermal shift assay (MS-CETSA) to identify potential targets of several antimalarials and drug candidates. We found that falcilysin (FLN) is a common binding partner for several drug candidates such as MK-4815, MMV000848, and MMV665806 but also interacts with quinoline drugs such as chloroquine and mefloquine. Enzymatic assays showed that these compounds can inhibit FLN proteolytic activity. Their interaction with FLN was explored systematically by isothermal titration calorimetry and X-ray crystallography, revealing a shared hydrophobic pocket in the catalytic chamber of the enzyme. Characterization of transgenic cell lines with lowered FLN expression demonstrated statistically significant increases in susceptibility toward MK-4815, MMV000848, and several quinolines. Importantly, the hydrophobic pocket of FLN appears amenable to inhibition and the structures reported here can guide the development of novel drugs against malaria.
Collapse
Affiliation(s)
- Grennady Wirjanata
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Jianqing Lin
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore; Infectious Diseases Labs & Singapore Immunology Network, Agency for Science, Technology and Research, 138648 Singapore, Singapore
| | - Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Abbas El Sahili
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore
| | - Zara Chung
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Seth Tjia
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | | | - Josephine Boentoro
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Roy Tham
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Lai Si Jia
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Ka Diam Go
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | | | - David Olsen
- Merck & Co., Inc., West Point, PA 19486, USA
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), Singapore 138673, Singapore; Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), Singapore 138673, Singapore; Department of Oncology and Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore; NTU Institute of Structural Biology, Nanyang Technology University, Singapore 637551, Singapore; Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 637551, Singapore.
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technology University, Singapore 637551, Singapore.
| |
Collapse
|
46
|
van Loon W, Bisimwa BC, Byela V, Kirby R, Bugeme PM, Balagizi A, Lupande D, Malembaka EB, Mockenhaupt FP, Bahizire E. Detection of Artemisinin Resistance Marker Kelch-13 469Y in Plasmodium falciparum, South Kivu, Democratic Republic of the Congo, 2022. Am J Trop Med Hyg 2024; 110:653-655. [PMID: 38377612 PMCID: PMC10993838 DOI: 10.4269/ajtmh.23-0740] [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: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 02/22/2024] Open
Abstract
Partial artemisinin resistance has emerged in East Africa, posing a threat to malaria control across the continent. The Democratic Republic of the Congo carries one of the heaviest malaria burdens globally, and the South Kivu province directly borders current artemisinin resistance hot spots, but indications of such resistance have not been observed so far. We assessed molecular markers of antimalarial drug resistance in 256 Plasmodium falciparum isolates collected in 2022 in South Kivu, Democratic Republic of the Congo. One isolate carried the P. falciparum Kelch-13 469Y variant, a marker associated with partial artemisinin resistance and decreased lumefantrine susceptibility in Uganda. In addition, the multidrug resistance-1 mutation pattern suggested increased lumefantrine tolerance.
Collapse
Affiliation(s)
- Welmoed van Loon
- Institute of International Health, Center for Global Health, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Bertin C. Bisimwa
- Center for Tropical Diseases & Global Health, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Institut Supérieur des Techniques Médicales de Bukavu, Bukavu, Democratic Republic of the Congo
| | - Valéry Byela
- Institut Supérieur des Techniques Médicales de Bukavu, Bukavu, Democratic Republic of the Congo
| | - Rebecca Kirby
- University of California, San Diego School of Medicine, San Diego, California
| | - Patrick M. Bugeme
- Center for Tropical Diseases & Global Health, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
| | - Aimé Balagizi
- Hôpital Général de Référence de Nyantende, Nyantende, Democratic Republic of the Congo
| | - David Lupande
- Center for Tropical Diseases & Global Health, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Hôpital Provincial Général de Référence de Bukavu, Bukavu, Democratic Republic of the Congo
| | - Espoir B. Malembaka
- Center for Tropical Diseases & Global Health, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
| | - Frank P. Mockenhaupt
- Institute of International Health, Center for Global Health, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Esto Bahizire
- Center for Tropical Diseases & Global Health, Université Catholique de Bukavu, Bukavu, Democratic Republic of the Congo
- Centre de Recherche en Sciences Naturelles de Lwiro, Bukavu, Democratic Republic of the Congo
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
47
|
Dorkenoo AM, Warsame M, Ataba E, Hemou M, Yakpa K, Sossou E, Mitigmsagou M, Teou CD, Caspar E, Ma L, Djadou KE, Atcha-Oubou T, Rasmussen C, Menard D. Efficacy of artemether-lumefantrine and dihydroartemisinin-piperaquine and prevalence of molecular markers of anti-malarial drug resistance in children in Togo in 2021. Malar J 2024; 23:92. [PMID: 38570791 PMCID: PMC10988893 DOI: 10.1186/s12936-024-04922-1] [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: 10/27/2023] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Artemether-lumefantrine (AL) and dihydroartemisinin-piperaquine (DP) are the currently recommended first- and second-line therapies for uncomplicated Plasmodium falciparum infections in Togo. This study assessed the efficacy of these combinations, the proportion of Day3-positive patients (D3 +), the proportion of molecular markers associated with P. falciparum resistance to anti-malarial drugs, and the variable performance of HRP2-based malaria rapid diagnostic tests (RDTs). METHODS A single arm prospective study evaluating the efficacy of AL and DP was conducted at two sites (Kouvé and Anié) from September 2021 to January 2022. Eligible children were enrolled, randomly assigned to treatment at each site and followed up for 42 days after treatment initiation. The primary endpoint was polymerase chain reaction (PCR) adjusted adequate clinical and parasitological response (ACPR). At day 0, samples were analysed for mutations in the Pfkelch13, Pfcrt, Pfmdr-1, dhfr, dhps, and deletions in the hrp2/hrp3 genes. RESULTS A total of 179 and 178 children were included in the AL and DP groups, respectively. After PCR correction, cure rates of patients treated with AL were 97.5% (91.4-99.7) at day 28 in Kouvé and 98.6% (92.4-100) in Anié, whereas 96.4% (CI 95%: 89.1-98.8) and 97.3% (CI 95%: 89.5-99.3) were observed at day 42 in Kouvé and Anié, respectively. The cure rates of patients treated with DP at day 42 were 98.9% (CI 95%: 92.1-99.8) in Kouvé and 100% in Anié. The proportion of patients with parasites on day 3 (D3 +) was 8.5% in AL and 2.6% in DP groups in Anié and 4.3% in AL and 2.1% DP groups in Kouvé. Of the 357 day 0 samples, 99.2% carried the Pfkelch13 wild-type allele. Two isolates carried nonsynonymous mutations not known to be associated with artemisinin partial resistance (ART-R) (A578S and A557S). Most samples carried the Pfcrt wild-type allele (97.2%). The most common Pfmdr-1 allele was the single mutant 184F (75.6%). Among dhfr/dhps mutations, the quintuple mutant haplotype N51I/C59R/S108N + 437G/540E, which is responsible for SP treatment failure in adults and children, was not detected. Single deletions in hrp2 and hrp3 genes were detected in 1/357 (0.3%) and 1/357 (0.3%), respectively. Dual hrp2/hrp3 deletions, which could affect the performances of HRP2-based RDTs, were not observed. CONCLUSION The results of this study confirm that the AL and DP treatments are highly effective. The absence of the validated Pfkelch13 mutants in the study areas suggests the absence of ART -R, although a significant proportion of D3 + cases were found. The absence of dhfr/dhps quintuple or sextuple mutants (quintuple + 581G) supports the continued use of SP for IPTp during pregnancy and in combination with amodiaquine for seasonal malaria chemoprevention. TRIAL REGISTRATION ACTRN12623000344695.
Collapse
Affiliation(s)
| | - Marian Warsame
- School of Public Health and Community Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Essoham Ataba
- Programme National de Lutte Contre le Paludisme, Lomé, Togo
| | - Manani Hemou
- Service de Pédiatrie, Centre Hospitalier Universitaire Campus, Lomé, Togo
| | - Kossi Yakpa
- Programme National de Lutte Contre le Paludisme, Lomé, Togo
| | - Efoe Sossou
- Service des Laboratoires, Centre Hospitalier Universitaire Sylvanus Olympio Lomé, Lomé, Togo
| | | | | | - Emmanuelle Caspar
- Institute of Parasitology and Tropical Diseases, Université de Strasbourg, UR7292 Dynamics of Host-Pathogen Interactions, 67000, Strasbourg, France
| | - Laurence Ma
- Biomics Platform, C2RT, Institut Pasteur, 75015, Paris, France
| | | | | | | | - Didier Menard
- Institute of Parasitology and Tropical Diseases, Université de Strasbourg, UR7292 Dynamics of Host-Pathogen Interactions, 67000, Strasbourg, France
- Malaria Genetics and Resistance Unit, Institut Pasteur, Université Paris Cité, INSERM U1201, 75015, Paris, France
- Malaria Parasite Biology and Vaccines, Institut Pasteur, Université Paris Cité, 75015, Paris, France
- Laboratory of Parasitology and Medical Mycology, CHU Strasbourg, 67000, Strasbourg, France
| |
Collapse
|
48
|
Wamae K, Ndwiga L, Kharabora O, Kimenyi K, Osoti V, de Laurent Z, Wambua J, Musyoki J, Ngetsa C, Kalume P, Mwambingu G, Hamaluba M, van der Pluijm R, Dondorp AM, Bailey J, Juliano J, Bejon P, Ochola-Oyier L. Targeted amplicon deep sequencing of ama1 and mdr1 to track within-host P. falciparum diversity throughout treatment in a clinical drug trial. Wellcome Open Res 2024; 7:95. [PMID: 37456906 PMCID: PMC10349275 DOI: 10.12688/wellcomeopenres.17736.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 07/20/2023] Open
Abstract
Introduction Antimalarial therapeutic efficacy studies are routinely conducted in malaria-endemic countries to assess the effectiveness of antimalarial treatment strategies. Targeted amplicon sequencing (AmpSeq) uniquely identifies and quantifies genetically distinct parasites within an infection. In this study, AmpSeq of Plasmodium falciparum apical membrane antigen 1 ( ama1), and multidrug resistance gene 1 ( mdr1), were used to characterise the complexity of infection (COI) and drug-resistance genotypes, respectively. Methods P. falciparum-positive samples were obtained from a triple artemisinin combination therapy clinical trial conducted in 30 children under 13 years of age between 2018 and 2019 in Kilifi, Kenya. Nine of the 30 participants presented with recurrent parasitemia from day 26 (624h) onwards. The ama1 and mdr1 genes were amplified and sequenced, while msp1, msp2 and glurp data were obtained from the original clinical study. Results The COI was comparable between ama1 and msp1, msp2 and glurp; overall, ama1 detected more microhaplotypes. Based on ama1, a stable number of microhaplotypes were detected throughout treatment until day 3. Additionally, a recrudescent infection was identified with an ama1 microhaplotype initially observed at 30h and later in an unscheduled follow-up visit. Using the relative frequencies of ama1 microhaplotypes and parasitemia, we identified a fast (<1h) and slow (>5h) clearing microhaplotype. As expected, only two mdr1 microhaplotypes (NF and NY) were identified based on the combination of amino acid polymorphisms at codons 86 and 184. Conclusions This study highlights AmpSeq as a tool for highly-resolution tracking of parasite microhaplotypes throughout treatment and can detect variation in microhaplotype clearance estimates. AmpSeq can also identify slow-clearing microhaplotypes, a potential early sign of selection during treatment. Consequently, AmpSeq has the capability of improving the discriminatory power to distinguish recrudescences from reinfections accurately.
Collapse
Affiliation(s)
- Kevin Wamae
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Leonard Ndwiga
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Oksana Kharabora
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Kelvin Kimenyi
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Victor Osoti
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Juliana Wambua
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Jennifer Musyoki
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Caroline Ngetsa
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Peter Kalume
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Mainga Hamaluba
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rob van der Pluijm
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M. Dondorp
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jeffrey Bailey
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Jonathan Juliano
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27516, USA
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philip Bejon
- Bioscience, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | |
Collapse
|
49
|
Zupko RJ, Servadio JL, Nguyen TD, Tran TNA, Tran KT, Somé AF, Boni MF. Role of seasonal importation and genetic drift on selection for drug-resistant genotypes of Plasmodium falciparum in high-transmission settings. J R Soc Interface 2024; 21:20230619. [PMID: 38442861 PMCID: PMC10914515 DOI: 10.1098/rsif.2023.0619] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Historically Plasmodium falciparum has followed a pattern of drug resistance first appearing in low-transmission settings before spreading to high-transmission settings. Several features of low-transmission regions are hypothesized as explanations: higher chance of symptoms and treatment seeking, better treatment access, less within-host competition among clones and lower rates of recombination. Here, we test whether importation of drug-resistant parasites is more likely to lead to successful emergence and establishment in low-transmission or high-transmission periods of the same epidemiological setting, using a spatial, individual-based stochastic model of malaria and drug-resistance evolution calibrated for Burkina Faso. Upon controlling for the timing of importation of drug-resistant genotypes and examination of key model variables, we found that drug-resistant genotypes imported during the low-transmission season were (i) more susceptible to stochastic extinction due to the action of genetic drift, and (ii) more likely to lead to establishment of drug resistance when parasites are able to survive early stochastic loss due to drift. This implies that rare importation events are more likely to lead to establishment if they occur during a high-transmission season, but that constant importation (e.g. neighbouring countries with high levels of resistance) may produce a greater risk during low-transmission periods.
Collapse
Affiliation(s)
- Robert J. Zupko
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Joseph L. Servadio
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Tran Dang Nguyen
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Thu Nguyen-Anh Tran
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Kien Trung Tran
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Anyirékun Fabrice Somé
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l'Ouest, Bobo Dioulasso, Burkina Faso
| | - Maciej F. Boni
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Li EZ, Nguyen TD, Tran TNA, Zupko RJ, Boni MF. Assessing emergence risk of double-resistant and triple-resistant genotypes of Plasmodium falciparum. Nat Commun 2024; 15:1390. [PMID: 38360803 PMCID: PMC10869733 DOI: 10.1038/s41467-024-45547-x] [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: 03/09/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
Delaying and slowing antimalarial drug resistance evolution is a priority for malaria-endemic countries. Until novel therapies become available, the mainstay of antimalarial treatment will continue to be artemisinin-based combination therapy (ACT). Deployment of different ACTs can be optimized to minimize evolutionary pressure for drug resistance by deploying them as a set of co-equal multiple first-line therapies (MFT) rather than rotating therapies in and out of use. Here, we consider one potential detriment of MFT policies, namely, that the simultaneous deployment of multiple ACTs could drive the evolution of different resistance alleles concurrently and that these resistance alleles could then be brought together by recombination into double-resistant or triple-resistant parasites. Using an individual-based model, we compare MFT and cycling policies in malaria transmission settings ranging from 0.1% to 50% prevalence. We define a total risk measure for multi-drug resistance (MDR) by summing the area under the genotype-frequency curves (AUC) of double- and triple-resistant genotypes. When prevalence ≥ 1%, total MDR risk ranges from statistically similar to 80% lower under MFT policies than under cycling policies, irrespective of whether resistance is imported or emerges de novo. At 0.1% prevalence, there is little statistical difference in MDR risk between MFT and cycling.
Collapse
Affiliation(s)
- Eric Zhewen Li
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Tran Dang Nguyen
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Thu Nguyen-Anh Tran
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Robert J Zupko
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Maciej F Boni
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, PA, USA.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|