1
|
Mita T. Antimalarial drug efficacy and resistance: Insights from northern Uganda. Parasitol Int 2025; 109:103083. [PMID: 40348079 DOI: 10.1016/j.parint.2025.103083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/28/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Artemisinin in combination is the first-line treatment for Plasmodium falciparum malaria in almost all regions. However, by the late 2000s, partial resistance, characterized by delayed parasite clearance after treatment, emerged in the Greater Mekong Subregion and rapidly expanded its range. Since 2013, we have conducted comprehensive malaria drug resistance surveillance in northern Uganda. In 2014, we identified in vitro artemisinin resistance, and by 2017, clinical partial resistance had also been observed. Additionally, we discovered the re-emergence of chloroquine sensitivity in this region as early as 2013, earlier than in the other parts of Uganda. This review aims to summarize our findings from northern Uganda, contextualizing them within similar studies conducted in other regions.
Collapse
Affiliation(s)
- Toshihiro Mita
- Department of Tropical Medicine and Parasitology, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
2
|
Awalt JK, Ooi ZK, Ashton TD, Mansouri M, Calic PPS, Zhou Q, Vasanthan S, Lee S, Loi K, Jarman KE, Penington JS, Qiu D, Zhang X, Lehane AM, Mao EY, Gancheva MR, Wilson DW, Giannangelo C, MacRaild CA, Creek DJ, Yeo T, Sheth T, Fidock DA, Churchyard A, Baum J, Famodimu MT, Delves MJ, Kristan M, Stewart L, Sutherland CJ, Coyle R, Jagoe H, Lee MCS, Chowdury M, de Koning-Ward TF, Baud D, Brand S, Jackson PF, Cowman AF, Dans MG, Sleebs BE. Optimization and Characterization of N-Acetamide Indoles as Antimalarials That Target PfATP4. J Med Chem 2025; 68:8933-8966. [PMID: 40228810 PMCID: PMC12035806 DOI: 10.1021/acs.jmedchem.5c00614] [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: 03/03/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
To discover new antimalarials, a screen of the Janssen Jumpstarter library against Plasmodium falciparum uncovered the N-acetamide indole hit class. The structure-activity relationship of this chemotype was defined and culminated in the optimized frontrunner analog WJM664, which exhibited potent asexual stage activity and high metabolic stability. Resistant selection and whole-genome sequencing revealed mutations in PfATP4, which was validated as the target by showing that analogs exhibited reduced potency against parasites with resistance-conferring mutations in PfATP4, a metabolomic signature similar to that of the PfATP4 inhibitor KAE609, and inhibition of Na+-dependent ATPase activity consistent with on-target inhibition of PfATP4. WJM664 inhibited gamete development and blocked parasite transmission to mosquitoes but exhibited low efficacy in aPlasmodium berghei mouse model, which was attributed to ATP4 species differentiation and its moderate systemic exposure. Optimization of these attributes is required for N-acetamide indoles to be pursued for development as a curative and transmission-blocking therapy.
Collapse
Affiliation(s)
- Jon Kyle Awalt
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Zi Kang Ooi
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Trent D. Ashton
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Mahta Mansouri
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Petar P. S. Calic
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Qingmiao Zhou
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Santhya Vasanthan
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Serena Lee
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Katie Loi
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Kate E. Jarman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Jocelyn S. Penington
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Deyun Qiu
- Research
School of Biology, Australian National University, Canberra 2601, Australia
| | - Xinxin Zhang
- Research
School of Biology, Australian National University, Canberra 2601, Australia
| | - Adele M. Lehane
- Research
School of Biology, Australian National University, Canberra 2601, Australia
| | - Emma Y. Mao
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Maria R. Gancheva
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Danny W. Wilson
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Carlo Giannangelo
- Monash
Institute of Pharmaceutical Sciences, Monash
University, Parkville 3052, Australia
| | | | - Darren J. Creek
- Monash
Institute of Pharmaceutical Sciences, Monash
University, Parkville 3052, Australia
| | - Tomas Yeo
- Department
of Microbiology & Immunology, Columbia
University Irving Medical Center, New York, New York 10032, United States
- Center
for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tanaya Sheth
- Department
of Microbiology & Immunology, Columbia
University Irving Medical Center, New York, New York 10032, United States
- Center
for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David A. Fidock
- Department
of Microbiology & Immunology, Columbia
University Irving Medical Center, New York, New York 10032, United States
- Center
for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division
of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Alisje Churchyard
- Department
of Life Sciences, Imperial College London, South Kensington SW7 2AZ, U.K.
| | - Jake Baum
- Department
of Life Sciences, Imperial College London, South Kensington SW7 2AZ, U.K.
- School
of Biomedical Sciences, University of New
South Wales, Sydney 2031, Australia
| | | | - Michael J. Delves
- London School of
Hygiene and Tropical Medicine, London WC1E 7HT, U.K.
| | - Mojca Kristan
- London School of
Hygiene and Tropical Medicine, London WC1E 7HT, U.K.
| | - Lindsay Stewart
- London School of
Hygiene and Tropical Medicine, London WC1E 7HT, U.K.
| | | | - Rachael Coyle
- London School of
Hygiene and Tropical Medicine, London WC1E 7HT, U.K.
- Wellcome Sanger Institute, Wellcome Genome Campus Hinxton CB10 1SA, U.K.
| | - Hannah Jagoe
- London School of
Hygiene and Tropical Medicine, London WC1E 7HT, U.K.
| | - Marcus C. S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus Hinxton CB10 1SA, U.K.
- Wellcome
Centre for Anti-Infectives Research, Biological Chemistry and Drug
Discovery, University of Dundee, Dundee DD1 5EH, U.K.
| | - Mrittika Chowdury
- School
of Medicine, Deakin University, Waurn Ponds 3216, Australia
- Institute
for Mental and Physical Health and Clinical Translation, Deakin University, Geelong 3216, Australia
| | - Tania F. de Koning-Ward
- School
of Medicine, Deakin University, Waurn Ponds 3216, Australia
- Institute
for Mental and Physical Health and Clinical Translation, Deakin University, Geelong 3216, Australia
| | - Delphine Baud
- Medicines for Malaria Venture, Geneva 1215, Switzerland
| | - Stephen Brand
- Medicines for Malaria Venture, Geneva 1215, Switzerland
| | - Paul F. Jackson
- Emerging
Science & Innovation, Discovery Sciences, Janssen R&D LLC, La Jolla 92121, United States
| | - Alan F. Cowman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Madeline G. Dans
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Brad E. Sleebs
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
3
|
Bremers E, Butler JH, Do Amaral LS, Merino EF, Almolhim H, Zhou B, Baptista RP, Totrov M, Carlier PR, Cassera MB. Stereospecific Resistance to N2-Acyl Tetrahydro-β-carboline Antimalarials Is Mediated by a PfMDR1 Mutation That Confers Collateral Drug Sensitivity. ACS Infect Dis 2025; 11:529-542. [PMID: 39808111 PMCID: PMC11828674 DOI: 10.1021/acsinfecdis.4c01001] [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: 12/12/2024] [Revised: 12/18/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
Half the world's population is at risk of developing a malaria infection, which is caused by parasites of the genus Plasmodium. Currently, resistance has been identified to all clinically available antimalarials, highlighting an urgent need to develop novel compounds and better understand common mechanisms of resistance. We previously identified a novel tetrahydro-β-carboline compound, PRC1590, which potently kills the malaria parasite. To better understand its mechanism of action, we selected for and characterized resistance to PRC1590 in Plasmodium falciparum. Through in vitro selection of resistance to PRC1590, we have identified that a single-nucleotide polymorphism on the parasite's multidrug resistance protein 1 (PfMDR1 G293V) mediates resistance to PRC1590. This mutation results in stereospecific resistance and sensitizes parasites to other antimalarials, such as mefloquine, quinine, and MMV019017. Intraerythrocytic asexual stage specificity assays have revealed that PRC1590 is most potent during the trophozoite stage when the parasite forms a single digestive vacuole (DV) and actively digests hemoglobin. Moreover, fluorescence microscopy revealed that PRC1590 disrupts the function of the DV, indicating a potential molecular target associated with this organelle. Our findings mark a significant step in understanding the mechanism of resistance and the mode of action of this emerging class of antimalarials. In addition, our results suggest a potential link between resistance mediated by PfMDR1 and PRC1590's molecular target. This research underscores the pressing need for future research aimed at investigating the intricate relationship between a compound's chemical scaffold, molecular target, and resistance mutations associated with PfMDR1.
Collapse
Affiliation(s)
- Emily
K. Bremers
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Joshua H. Butler
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Leticia S. Do Amaral
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Emilio F. Merino
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Hanan Almolhim
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Bo Zhou
- Department
of Pharmaceutical Sciences, University of
Illinois Chicago, Chicago, Illinois 60612, United States
| | - Rodrigo P. Baptista
- Department
of Medicine, Houston Methodist Research
Institute, Houston, Texas 77030, United States
| | - Maxim Totrov
- MolSoft
LLC, San Diego, California 92121, United States
| | - Paul R. Carlier
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
- Department
of Pharmaceutical Sciences, University of
Illinois Chicago, Chicago, Illinois 60612, United States
| | - Maria Belen Cassera
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
4
|
Bouzón-Arnáiz I, Rawat M, Coyle R, Feufack-Donfack LB, Ea M, Orban A, Popovici J, Román-Álamo L, Fallica AN, Domínguez-Asenjo B, Moreno J, Arce EM, Mallo-Abreu A, Muñoz-Torrero D, Lee MCS, Fernàndez-Busquets X. YAT2150 is irresistible in Plasmodium falciparum and active against Plasmodium vivax and Leishmania clinical isolates. Sci Rep 2025; 15:2941. [PMID: 39848983 PMCID: PMC11758391 DOI: 10.1038/s41598-025-85346-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: 09/10/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
We recently characterized the potent antiplasmodial activity of the aggregated protein dye YAT2150, whose presumed mode of action is the inhibition of protein aggregation in the malaria parasite. Using single-dose and ramping methods, assays were done to select Plasmodium falciparum parasites resistant to YAT2150 concentrations ranging from 3× to 0.25× the in vitro IC50 of the compound (in the two-digit nM range) and performed a cross-resistance assessment in P. falciparum lines harboring mutations that make them resistant to a variety of antimalarial drugs. Resistant parasites did not emerge in vitro after 60 days of incubation, which postulates YAT2150 as an 'irresistible' antimalarial. The lyophilized compound is stable for at least one year stored at 25 °C. Tests performed in clinical isolates indicated that YAT2150 had also strong activity against Plasmodium vivax (IC50 between 4 and 36 nM) and Leishmania infantum (1.27 and 1.11 µM), placing it as a unique compound with perspectives of becoming the first drug to be used against both malaria and leishmaniasis.
Collapse
Affiliation(s)
- Inés Bouzón-Arnáiz
- Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-University of Barcelona), Rosselló 149-153, Barcelona, 08036, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
| | - Mukul Rawat
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | | | - Malen Ea
- Malaria Research Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, 120210, Phnom Penh, Cambodia
| | - Agnes Orban
- Malaria Research Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, 120210, Phnom Penh, Cambodia
| | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, 120210, Phnom Penh, Cambodia
- Infectious Disease Epidemiology and Analytics, Institut Pasteur, Paris, 75015, France
| | - Lucía Román-Álamo
- Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-University of Barcelona), Rosselló 149-153, Barcelona, 08036, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
| | - Antonino Nicolò Fallica
- Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-University of Barcelona), Rosselló 149-153, Barcelona, 08036, Spain
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain
| | - Bárbara Domínguez-Asenjo
- WHO Collaborating Centre for Leishmaniasis, National Centre for Microbiology Instituto de Salud Carlos III, Majadahonda, 28220, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, National Centre for Microbiology Instituto de Salud Carlos III, Majadahonda, 28220, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, Madrid, Spain
| | - Elsa M Arce
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, Barcelona, 08028, Spain
| | - Ana Mallo-Abreu
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, Barcelona, 08028, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, Barcelona, 08028, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Marcus C S Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Xavier Fernàndez-Busquets
- Barcelona Institute for Global Health (ISGlobal, Hospital Clínic-University of Barcelona), Rosselló 149-153, Barcelona, 08036, Spain.
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona, 08028, Spain.
- Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, Barcelona, 08028, Spain.
| |
Collapse
|
5
|
Kanai M, Mok S, Yeo T, Shears MJ, Ross LS, Jeon JH, Narwal S, Haile MT, Tripathi AK, Mlambo G, Kim J, Gil-Iturbe E, Okombo J, Fairhurst KJ, Bloxham T, Bridgford JL, Sheth T, Ward KE, Park H, Rozenberg FD, Quick M, Mancia F, Lee MC, Small-Saunders JL, Uhlemann AC, Sinnis P, Fidock DA. Identification of the drug/metabolite transporter 1 as a marker of quinine resistance in a NF54×Cam3.II P. falciparum genetic cross. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615529. [PMID: 39386571 PMCID: PMC11463348 DOI: 10.1101/2024.09.27.615529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The genetic basis of Plasmodium falciparum resistance to quinine (QN), a drug used to treat severe malaria, has long been enigmatic. To gain further insight, we used FRG-NOD human liver-chimeric mice to conduct a P. falciparum genetic cross between QN-sensitive and QN-resistant parasites, which also differ in their susceptibility to chloroquine (CQ). By applying different selective conditions to progeny pools prior to cloning, we recovered 120 unique recombinant progeny. These progeny were subjected to drug profiling and QTL analyses with QN, CQ, and monodesethyl-CQ (md-CQ, the active metabolite of CQ), which revealed predominant peaks on chromosomes 7 and 12, consistent with a multifactorial mechanism of resistance. A shared chromosome 12 region mapped to resistance to all three antimalarials and was preferentially co-inherited with pfcrt. We identified an ATP-dependent zinc metalloprotease (FtsH1) as one of the top candidates and observed using CRISPR/Cas9 SNP-edited lines that ftsh1 is a potential mediator of QN resistance and a modulator of md-CQ resistance. As expected, CQ and md-CQ resistance mapped to a chromosome 7 region harboring pfcrt. However, for QN, high-grade resistance mapped to a chromosome 7 peak centered 295kb downstream of pfcrt. We identified the drug/metabolite transporter 1 (DMT1) as the top candidate due to its structural similarity to PfCRT and proximity to the peak. Deleting DMT1 in QN-resistant Cam3.II parasites significantly sensitized the parasite to QN but not to the other drugs tested, suggesting that DMT1 mediates QN response specifically. We localized DMT1 to structures associated with vesicular trafficking, as well as the parasitophorous vacuolar membrane, lipid bodies, and the digestive vacuole. We also observed that mutant DMT1 transports more QN than the wild-type isoform in vitro. Our study demonstrates that DMT1 is a novel marker of QN resistance and a new chromosome 12 locus associates with CQ and QN response, with ftsh1 is a potential candidate, suggesting these genes should be genotyped in surveillance and clinical settings.
Collapse
Affiliation(s)
- Mariko Kanai
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Sachel Mok
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| | - Tomas Yeo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Melanie J. Shears
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Leila S. Ross
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
| | - Jin H. Jeon
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Sunil Narwal
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Meseret T. Haile
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
| | - Abhai K. Tripathi
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, NY, USA
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, NY, USA
| | - John Okombo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Kate J. Fairhurst
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Talia Bloxham
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Jessica L. Bridgford
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Tanaya Sheth
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Kurt E. Ward
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| | - Felix D. Rozenberg
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| | - Matthias Quick
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, NY, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, NY, USA
| | - Marcus C.S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Jennifer L. Small-Saunders
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| | - Anne-Catrin Uhlemann
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, MD, USA
| | - David A. Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, NY, USA
| |
Collapse
|
6
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [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: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
7
|
Platon L, Ménard D. Plasmodium falciparum ring-stage plasticity and drug resistance. Trends Parasitol 2024; 40:118-130. [PMID: 38104024 DOI: 10.1016/j.pt.2023.11.007] [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: 09/26/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
Malaria is a life-threatening tropical disease caused by parasites of the genus Plasmodium, of which Plasmodium falciparum is the most lethal. Malaria parasites have a complex life cycle, with stages occurring in both the Anopheles mosquito vector and human host. Ring stages are the youngest form of the parasite in the intraerythrocytic developmental cycle and are associated with evasion of spleen clearance, temporary growth arrest (TGA), and drug resistance. This formidable ability to survive and develop into mature, sexual, or growth-arrested forms demonstrates the inherent population heterogeneity. Here we highlight the role of the ring stage as a crossroads in parasite development and as a reservoir of surviving cells in the human host via TGA survival mechanisms.
Collapse
Affiliation(s)
- Lucien Platon
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, F-75015 Paris, France; Sorbonne Université, Collège Doctoral ED 515 Complexité du Vivant, F-75015 Paris, France; Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, F-67000 Strasbourg, France.
| | - Didier Ménard
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, F-75015 Paris, France; Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, F-75015 Paris, France; Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, F-67000 Strasbourg, France; CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, F-67000 Strasbourg, France.
| |
Collapse
|
8
|
Morales-Luna L, Vázquez-Bautista M, Martínez-Rosas V, Rojas-Alarcón MA, Ortega-Cuellar D, González-Valdez A, Pérez de la Cruz V, Arreguin-Espinosa R, Rodríguez-Bustamante E, Rodríguez-Flores E, Hernández-Ochoa B, Gómez-Manzo S. Fused Enzyme Glucose-6-Phosphate Dehydrogenase::6-Phosphogluconolactonase (G6PD::6PGL) as a Potential Drug Target in Giardia lamblia, Trichomonas vaginalis, and Plasmodium falciparum. Microorganisms 2024; 12:112. [PMID: 38257939 PMCID: PMC10819308 DOI: 10.3390/microorganisms12010112] [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: 12/04/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Several microaerophilic parasites such as Giardia lamblia, Trichomonas vaginalis, and Plasmodium falciparum are major disease-causing organisms and are responsible for spreading infections worldwide. Despite significant progress made in understanding the metabolism and molecular biology of microaerophilic parasites, chemotherapeutic treatment to control it has seen limited progress. A current proposed strategy for drug discovery against parasitic diseases is the identification of essential key enzymes of metabolic pathways associated with the parasite's survival. In these organisms, glucose-6-phosphate dehydrogenase::6-phosphogluconolactonase (G6PD:: 6PGL), the first enzyme of the pentose phosphate pathway (PPP), is essential for its metabolism. Since G6PD:: 6PGL provides substrates for nucleotides synthesis and NADPH as a source of reducing equivalents, it could be considered an anti-parasite drug target. This review analyzes the anaerobic energy metabolism of G. lamblia, T. vaginalis, and P. falciparum, with a focus on glucose metabolism through the pentose phosphate pathway and the significance of the fused G6PD:: 6PGL enzyme as a therapeutic target in the search for new drugs.
Collapse
Affiliation(s)
- Laura Morales-Luna
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Montserrat Vázquez-Bautista
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Víctor Martínez-Rosas
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Miriam Abigail Rojas-Alarcón
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Abigail González-Valdez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico;
| | - Roberto Arreguin-Espinosa
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
| | - Eduardo Rodríguez-Bustamante
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Eden Rodríguez-Flores
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.A.-E.); (E.R.-B.); (E.R.-F.)
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City 06720, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico; (L.M.-L.); (M.V.-B.); (V.M.-R.); (M.A.R.-A.)
| |
Collapse
|
9
|
Fitri LE, Pawestri AR, Winaris N, Endharti AT, Khotimah ARH, Abidah HY, Huwae JTR. Antimalarial Drug Resistance: A Brief History of Its Spread in Indonesia. Drug Des Devel Ther 2023; 17:1995-2010. [PMID: 37431492 PMCID: PMC10329833 DOI: 10.2147/dddt.s403672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/25/2023] [Indexed: 07/12/2023] Open
Abstract
Malaria remains to be a national and global challenge and priority, as stated in the strategic plan of the Indonesian Ministry of Health and Sustainable Development Goals. In Indonesia, it is targeted that malaria elimination can be achieved by 2030. Unfortunately, the development and spread of antimalarial resistance inflicts a significant risk to the national malaria control programs which can lead to increased malaria morbidity and mortality. In Indonesia, resistance to widely used antimalarial drugs has been reported in two human species, Plasmodium falciparum and Plasmodium vivax. With the exception of artemisinin, resistance has surfaced towards all classes of antimalarial drugs. Initially, chloroquine, sulfadoxine-pyrimethamine, and primaquine were the most widely used antimalarial drugs. Regrettably, improper use has supported the robust spread of their resistance. Chloroquine resistance was first reported in 1974, while sulfadoxine-pyrimethamine emerged in 1979. Twenty years later, most provinces had declared treatment failures of both drugs. Molecular epidemiology suggested that variations in pfmdr1 and pfcrt genes were associated with chloroquine resistance, while dhfr and dhps genes were correlated with sulfadoxine-pyrimethamine resistance. Additionally, G453W, V454C and E455K of pfk13 genes appeared to be early warning sign to artemisinin resistance. Here, we reported mechanisms of antimalarial drugs and their development of resistance. This insight could provide awareness toward designing future treatment guidelines and control programs in Indonesia.
Collapse
Affiliation(s)
- Loeki Enggar Fitri
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Aulia Rahmi Pawestri
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Nuning Winaris
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Agustina Tri Endharti
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Alif Raudhah Husnul Khotimah
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Education, Faculty of Medical and Health Science, Maulana Malik Ibrahim State Islamic University, Malang, Indonesia
| | - Hafshah Yasmina Abidah
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Education, Faculty of Medical and Health Science, Maulana Malik Ibrahim State Islamic University, Malang, Indonesia
| | - John Thomas Rayhan Huwae
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Study Program Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
10
|
Choubey D, Deshmukh B, Rao AG, Kanyal A, Hati AK, Roy S, Karmodiya K. Genomic analysis of Indian isolates of Plasmodium falciparum: Implications for drug resistance and virulence factors. Int J Parasitol Drugs Drug Resist 2023; 22:52-60. [PMID: 37269630 PMCID: PMC10248731 DOI: 10.1016/j.ijpddr.2023.05.003] [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: 02/24/2023] [Revised: 05/04/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023]
Abstract
The emergence of drug resistance to frontline treatments such as Artemisinin-based combination therapy (ACT) is a major obstacle to the control and eradication of malaria. This problem is compounded by the inherent genetic variability of the parasites, as many established markers of resistance do not accurately predict the drug-resistant status. There have been reports of declining effectiveness of ACT in the West Bengal and Northeast regions of India, which have traditionally been areas of drug resistance emergence in the country. Monitoring the genetic makeup of a population can help to identify the potential for drug resistance markers associated with it and evaluate the effectiveness of interventions aimed at reducing the spread of malaria. In this study, we performed whole genome sequencing of 53 isolates of Plasmodium falciparum from West Bengal and compared their genetic makeup to isolates from Southeast Asia (SEA) and Africa. We found that the Indian isolates had a distinct genetic makeup compared to those from SEA and Africa, and were more similar to African isolates, with a high prevalence of mutations associated with antigenic variation genes. The Indian isolates also showed a high prevalence of markers of chloroquine resistance (mutations in Pfcrt) and multidrug resistance (mutations in Pfmdr1), but no known mutations associated with artemisinin resistance in the PfKelch13 gene. Interestingly, we observed a novel L152V mutation in PfKelch13 gene and other novel mutations in genes involved in ubiquitination and vesicular transport that have been reported to support artemisinin resistance in the early stages of ACT resistance in the absence of PfKelch13 polymorphisms. Thus, our study highlights the importance of region-specific genomic surveillance for artemisinin resistance and the need for continued monitoring of resistance to artemisinin and its partner drugs.
Collapse
Affiliation(s)
- Deepak Choubey
- Department of Technology, Savitribai Phule Pune University, Pune, India
| | - Bhagyashree Deshmukh
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Anjani Gopal Rao
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Abhishek Kanyal
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Amiya Kumar Hati
- Department of Medical Entomology, Calcutta School of Tropical Medicine, Kolkata, West Bengal, India
| | - Somenath Roy
- Department of Human Physiology, Vidyasagar University, Paschim Medinipur, West Bengal, India
| | - Krishanpal Karmodiya
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India.
| |
Collapse
|
11
|
Insight into molecular diagnosis for antimalarial drug resistance of Plasmodium falciparum parasites: A review. Acta Trop 2023; 241:106870. [PMID: 36849091 DOI: 10.1016/j.actatropica.2023.106870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Malaria is an infectious disease transmitted by the female Anopheles mosquito and poses a severe threat to human health. At present, antimalarial drugs are the primary treatment for malaria. The widespread use of artemisinin-based combination therapies (ACTs) has dramatically reduced the number of malaria-related deaths; however, the emergence of resistance has the potential to reverse this progress. Accurate and timely diagnosis of drug-resistant strains of Plasmodium parasites via detecting molecular markers (such as Pfnhe1, Pfmrp, Pfcrt, Pfmdr1, Pfdhps, Pfdhfr, and Pfk13) is essential for malaria control and elimination. Here, we review the current techniques which commonly used for molecular diagnosis of antimalarial resistance in P. falciparum and discuss their sensitivities and specificities for different drug resistance-associated molecular markers, with the aim of providing insights into possible directions for future precise point-of-care testing (POCT) of antimalarial drug resistance of malaria parasites.
Collapse
|
12
|
Alves E, Nakaya H, Guimarães E, Garcia CR. Combining IP 3 affinity chromatography and bioinformatics reveals a novel protein-IP 3 binding site on Plasmodium falciparum MDR1 transporter. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 4:100179. [PMID: 36582189 PMCID: PMC9792294 DOI: 10.1016/j.crmicr.2022.100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Intracellular Ca2+ mobilization induced by second messenger IP3 controls many cellular events in most of the eukaryotic groups. Despite the increasing evidence of IP3-induced Ca2+ in apicomplexan parasites like Plasmodium, responsible for malaria infection, no protein with potential function as an IP3-receptor has been identified. The use of bioinformatic analyses based on previously known sequences of IP3-receptor failed to identify potential IP3-receptor candidates in any Apicomplexa. In this work, we combine the biochemical approach of an IP3 affinity chromatography column with bioinformatic meta-analyses to identify potential vital membrane proteins that present binding with IP3 in Plasmodium falciparum. Our analyses reveal that PF3D7_0523000, a gene that codes a transport protein associated with multidrug resistance as a potential target for IP3. This work provides a new insight for probing potential candidates for IP3-receptor in Apicomplexa.
Collapse
Affiliation(s)
- Eduardo Alves
- Life Science Department, Imperial College London, London, United Kingdom
| | - Helder Nakaya
- Department of Clinical and Toxicological Analyses of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil,Computational Systems Biology Laboratory, INOVA, University of Sao Paulo, Sao Paulo, Brazil
| | - Euzébio Guimarães
- Federal University of Rio Grande do Norte, Pharmacy Department, Health Science Center, Natal, Brazil
| | - Célia R.S. Garcia
- Department of Clinical and Toxicological Analyses of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil,Corresponding author.
| |
Collapse
|
13
|
Leski TA, Taitt CR, Colston SM, Bangura U, Holtz A, Yasuda CY, Reynolds ND, Lahai J, Lamin JM, Baio V, Ansumana R, Stenger DA, Vora GJ. Prevalence of malaria resistance-associated mutations in Plasmodium falciparum circulating in 2017–2018, Bo, Sierra Leone. Front Microbiol 2022; 13:1059695. [DOI: 10.3389/fmicb.2022.1059695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
IntroductionIn spite of promising medical, sociological, and engineering strategies and interventions to reduce the burden of disease, malaria remains a source of significant morbidity and mortality, especially among children in sub-Saharan Africa. In particular, progress in the development and administration of chemotherapeutic agents is threatened by evolved resistance to most of the antimalarials currently in use, including artemisinins.MethodsThis study analyzed the prevalence of mutations associated with antimalarial resistance in Plasmodium falciparum from 95 clinical samples collected from individuals with clinically confirmed malaria at a hospital in Bo, Sierra Leone between May 2017 and December 2018. The combination of polymerase chain reaction amplification and subsequent high throughput DNA sequencing was used to determine the presence of resistance-associated mutations in five P. falciparum genes – pfcrt, pfmdr1, pfdhfr, pfdhps and pfkelch13. The geographic origin of parasites was assigned using mitochondrial sequences.ResultsRelevant mutations were detected in the pfcrt (22%), pfmdr1 (>58%), pfdhfr (100%) and pfdhps (>80%) genes while no resistance-associated mutations were found in the pfkelch13 gene. The mitochondrial barcodes were consistent with a West African parasite origin with one exception indicating an isolate imported from East Africa.DiscussionDetection of the pfmdr1 NFSND haplotype in 50% of the samples indicated the increasing prevalence of strains with elevated tolerance to artemeter + lumefantrine (AL) threatening the combination currently used to treat uncomplicated malaria in Sierra Leone. The frequency of mutations linked to resistance to antifolates suggests widespread resistance to the drug combination used for intermittent preventive treatment during pregnancy.
Collapse
|
14
|
Mechanistic basis for multidrug resistance and collateral drug sensitivity conferred to the malaria parasite by polymorphisms in PfMDR1 and PfCRT. PLoS Biol 2022; 20:e3001616. [PMID: 35507548 PMCID: PMC9067703 DOI: 10.1371/journal.pbio.3001616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Polymorphisms in the Plasmodium falciparum multidrug resistance protein 1 (pfmdr1) gene and the Plasmodium falciparum chloroquine resistance transporter (pfcrt) gene alter the malaria parasite’s susceptibility to most of the current antimalarial drugs. However, the precise mechanisms by which PfMDR1 contributes to multidrug resistance have not yet been fully elucidated, nor is it understood why polymorphisms in pfmdr1 and pfcrt that cause chloroquine resistance simultaneously increase the parasite’s susceptibility to lumefantrine and mefloquine—a phenomenon known as collateral drug sensitivity. Here, we present a robust expression system for PfMDR1 in Xenopus oocytes that enables direct and high-resolution biochemical characterizations of the protein. We show that wild-type PfMDR1 transports diverse pharmacons, including lumefantrine, mefloquine, dihydroartemisinin, piperaquine, amodiaquine, methylene blue, and chloroquine (but not the antiviral drug amantadine). Field-derived mutant isoforms of PfMDR1 differ from the wild-type protein, and each other, in their capacities to transport these drugs, indicating that PfMDR1-induced changes in the distribution of drugs between the parasite’s digestive vacuole (DV) and the cytosol are a key driver of both antimalarial resistance and the variability between multidrug resistance phenotypes. Of note, the PfMDR1 isoforms prevalent in chloroquine-resistant isolates exhibit reduced capacities for chloroquine, lumefantrine, and mefloquine transport. We observe the opposite relationship between chloroquine resistance-conferring mutations in PfCRT and drug transport activity. Using our established assays for characterizing PfCRT in the Xenopus oocyte system and in live parasite assays, we demonstrate that these PfCRT isoforms transport all 3 drugs, whereas wild-type PfCRT does not. We present a mechanistic model for collateral drug sensitivity in which mutant isoforms of PfMDR1 and PfCRT cause chloroquine, lumefantrine, and mefloquine to remain in the cytosol instead of sequestering within the DV. This change in drug distribution increases the access of lumefantrine and mefloquine to their primary targets (thought to be located outside of the DV), while simultaneously decreasing chloroquine’s access to its target within the DV. The mechanistic insights presented here provide a basis for developing approaches that extend the useful life span of antimalarials by exploiting the opposing selection forces they exert upon PfCRT and PfMDR1.
Collapse
|
15
|
Wichers JS, Mesén-Ramírez P, Fuchs G, Yu-Strzelczyk J, Stäcker J, von Thien H, Alder A, Henshall I, Liffner B, Nagel G, Löw C, Wilson D, Spielmann T, Gao S, Gilberger TW, Bachmann A, Strauss J. PMRT1, a Plasmodium-Specific Parasite Plasma Membrane Transporter, Is Essential for Asexual and Sexual Blood Stage Development. mBio 2022; 13:e0062322. [PMID: 35404116 PMCID: PMC9040750 DOI: 10.1128/mbio.00623-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Membrane transport proteins perform crucial roles in cell physiology. The obligate intracellular parasite Plasmodium falciparum, an agent of human malaria, relies on membrane transport proteins for the uptake of nutrients from the host, disposal of metabolic waste, exchange of metabolites between organelles, and generation and maintenance of transmembrane electrochemical gradients for its growth and replication within human erythrocytes. Despite their importance for Plasmodium cellular physiology, the functional roles of a number of membrane transport proteins remain unclear, which is particularly true for orphan membrane transporters that have no or limited sequence homology to transporter proteins in other evolutionary lineages. Therefore, in the current study, we applied endogenous tagging, targeted gene disruption, conditional knockdown, and knockout approaches to investigate the subcellular localization and essentiality of six membrane transporters during intraerythrocytic development of P. falciparum parasites. They are localized at different subcellular structures-the food vacuole, the apicoplast, and the parasite plasma membrane-and four out of the six membrane transporters are essential during asexual development. Additionally, the plasma membrane resident transporter 1 (PMRT1; PF3D7_1135300), a unique Plasmodium-specific plasma membrane transporter, was shown to be essential for gametocytogenesis and functionally conserved within the genus Plasmodium. Overall, we reveal the importance of four orphan transporters to blood stage P. falciparum development, which have diverse intracellular localizations and putative functions. IMPORTANCE Plasmodium falciparum-infected erythrocytes possess multiple compartments with designated membranes. Transporter proteins embedded in these membranes not only facilitate movement of nutrients, metabolites, and other molecules between these compartments, but also are common therapeutic targets and can confer antimalarial drug resistance. Orphan membrane transporters in P. falciparum without sequence homology to transporters in other evolutionary lineages and divergent from host transporters may constitute attractive targets for novel intervention approaches. Here, we localized six of these putative transporters at different subcellular compartments and probed their importance during asexual parasite growth by using reverse genetic approaches. In total, only two candidates turned out to be dispensable for the parasite, highlighting four candidates as putative targets for therapeutic interventions. This study reveals the importance of several orphan transporters to blood stage P. falciparum development.
Collapse
Affiliation(s)
- Jan Stephan Wichers
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | | | - Gwendolin Fuchs
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Jing Yu-Strzelczyk
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jan Stäcker
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Heidrun von Thien
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Arne Alder
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Isabelle Henshall
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Benjamin Liffner
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Georg Nagel
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology, Hamburg, Germany
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Danny Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
- Burnet Institute, Melbourne, Victoria, Australia
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Shiqiang Gao
- Institute of Physiology, Department of Neurophysiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Anna Bachmann
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
| | - Jan Strauss
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Biology Department, University of Hamburg, Hamburg, Germany
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| |
Collapse
|
16
|
Identification of polymorphisms in genes associated with drug resistance in Plasmodium falciparum isolates from school-age children in Kinshasa, Democratic Republic of Congo. Parasitol Int 2022; 88:102541. [PMID: 35051550 DOI: 10.1016/j.parint.2022.102541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND The emergence and spread of Plasmodium falciparum parasites resistant to antimalarial drugs constitutes an obstacle to malaria control and elimination. This study aimed to identify the prevalence of polymorphisms in pfk13, pfmdr1, pfdhfr, pfdhps and pfcrt genes in isolates from asymptomatic and symptomatic school-age children in Kinshasa. METHODS Nested-PCR followed by sequencing was performed for the detection of pfk13, pfmdr1, pfdhfr, pfdhps and pfcrt polymorphisms. RESULTS Two mutations in pfk13, C532S and Q613E were identified in the Democratic Republic of Congo for the first time. The prevalence of the drug-resistance associated mutations pfcrt K76T, pfdhps K540E and pfmdr1 N86Y was low, being 27%, 20% and 9%, respectively. CONCLUSION We found a low prevalence of genetic markers associated with chloroquine and sulfadoxine-pyrimethamine resistance in Kinshasa. Furthermore, no mutations previously associated with resistance against artemisinin and is derivatives were observed in the pfK13 gene. These findings support the continued use of ACTs and IPTp-SP. Continuous molecular monitoring of antimalarial resistance markers is recommended.
Collapse
|
17
|
Mesén-Ramírez P, Bergmann B, Elhabiri M, Zhu L, von Thien H, Castro-Peña C, Gilberger TW, Davioud-Charvet E, Bozdech Z, Bachmann A, Spielmann T. The parasitophorous vacuole nutrient channel is critical for drug access in malaria parasites and modulates the artemisinin resistance fitness cost. Cell Host Microbe 2021; 29:1774-1787.e9. [PMID: 34863371 DOI: 10.1016/j.chom.2021.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/14/2021] [Accepted: 11/03/2021] [Indexed: 02/08/2023]
Abstract
Intraerythrocytic malaria parasites proliferate bounded by a parasitophorous vacuolar membrane (PVM). The PVM contains nutrient permeable channels (NPCs) conductive to small molecules, but their relevance for parasite growth for individual metabolites is largely untested. Here we show that growth-relevant levels of major carbon and energy sources pass through the NPCs. Moreover, we find that NPCs are a gate for several antimalarial drugs, highlighting their permeability properties as a critical factor for drug design. Looking into NPC-dependent amino acid transport, we find that amino acid shortage is a reason for the fitness cost in artemisinin-resistant (ARTR) parasites and provide evidence that NPC upregulation to increase amino acids acquisition is a mechanism of ARTR parasites in vitro and in human infections to compensate this fitness cost. Hence, the NPCs are important for nutrient and drug access and reveal amino acid deprivation as a critical constraint in ARTR parasites.
Collapse
Affiliation(s)
- Paolo Mesén-Ramírez
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Bärbel Bergmann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Mourad Elhabiri
- UMR7042 Université de Strasbourg‒CNRS‒UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic and Medicinal Chemistry, European School of Chemistry, Polymers and Materials (ECPM), 25 Rue Becquerel, F-67087 Strasbourg, France
| | - Lei Zhu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Heidrun von Thien
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany; Centre for Structural Systems Biology, Notkestraße 85, Building 15, 22607, University of Hamburg, 20146 Hamburg, Germany
| | - Carolina Castro-Peña
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Tim-Wolf Gilberger
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany; Centre for Structural Systems Biology, Notkestraße 85, Building 15, 22607, University of Hamburg, 20146 Hamburg, Germany
| | - Elisabeth Davioud-Charvet
- UMR7042 Université de Strasbourg‒CNRS‒UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic and Medicinal Chemistry, European School of Chemistry, Polymers and Materials (ECPM), 25 Rue Becquerel, F-67087 Strasbourg, France
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; Honorary Visiting Research Fellow, Nuffield Department of Medicine, University of Oxford, UK
| | - Anna Bachmann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany; Centre for Structural Systems Biology, Notkestraße 85, Building 15, 22607, University of Hamburg, 20146 Hamburg, Germany
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany.
| |
Collapse
|
18
|
Dassonville-Klimpt A, Schneider J, Damiani C, Tisnerat C, Cohen A, Azas N, Marchivie M, Guillon J, Mullié C, Agnamey P, Totet A, Dormoi J, Taudon N, Pradines B, Sonnet P. Design, synthesis, and characterization of novel aminoalcohol quinolines with strong in vitro antimalarial activity. Eur J Med Chem 2021; 228:113981. [PMID: 34782182 DOI: 10.1016/j.ejmech.2021.113981] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/16/2022]
Abstract
Malaria is the fifth most lethal parasitic infections in the world. Herein, five new series of aminoalcohol quinolines including fifty-two compounds were designed, synthesized and evaluated in vitro against Pf3D7 and PfW2 strains. Among them, fourteen displayed IC50 values below or near of 50.0 nM whatever the strain with selectivity index often superior to 100.17b was found as a promising antimalarial candidate with IC50 values of 14.9 nM and 11.0 nM against respectively Pf3D7 and PfW2 and a selectivity index higher than 770 whatever the cell line is. Further experiments were achieved to confirm the safety and to establish the preliminary ADMET profile of compound 17b before the in vivo study performed on a mouse model of P. berghei ANKA infection. The overall data of this study allowed to establish new structure-activity relationships and the development of novel agents with improved pharmacokinetic properties.
Collapse
Affiliation(s)
- A Dassonville-Klimpt
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France.
| | - J Schneider
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - C Damiani
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - C Tisnerat
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - A Cohen
- Université Aix-Marseille, IRD, AP-HM, SSA, VITROME, IHU Méditerranée Infection, Marseille, France
| | - N Azas
- Université Aix-Marseille, IRD, AP-HM, SSA, VITROME, IHU Méditerranée Infection, Marseille, France
| | - M Marchivie
- CNRS, Univ. Bordeaux, Bordeaux INP, ICMCB, UMR 5026, F- 33600 Pessac, France
| | - J Guillon
- Université de Bordeaux, Laboratoire ARNA, UFR des Sciences Pharmaceutiques, Bordeaux, France; INSERM U1212, UMR CNRS 5320, Laboratoire ARNA, Bordeaux, France
| | - C Mullié
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - P Agnamey
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - Anne Totet
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France
| | - J Dormoi
- Unité parasitologie et entomologie, Département de microbiologie et de maladies infectieuses, Institut de recherche biomédicale des armées, Marseille, France; Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, Marseille, France; IHU Méditerranée Infection, Marseille, France
| | - N Taudon
- Unité de Développements Analytiques et Bioanalyse, IRBA, Brétigny-sur-Orge, France
| | - B Pradines
- Unité parasitologie et entomologie, Département de microbiologie et de maladies infectieuses, Institut de recherche biomédicale des armées, Marseille, France; Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, Marseille, France; IHU Méditerranée Infection, Marseille, France; Centre national de référence du paludisme, Marseille, France
| | - P Sonnet
- Université de Picardie Jules Verne, AGIR, UFR de Pharmacie, Amiens, UR, 4294, France.
| |
Collapse
|
19
|
Property activity refinement of 2-anilino 4-amino substituted quinazolines as antimalarials with fast acting asexual parasite activity. Bioorg Chem 2021; 117:105359. [PMID: 34689083 DOI: 10.1016/j.bioorg.2021.105359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/25/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022]
Abstract
Malaria is a devastating disease caused by Plasmodium parasites. Emerging resistance against current antimalarial therapeutics has engendered the need to develop antimalarials with novel structural classes. We recently described the identification and initial optimization of the 2-anilino quinazoline antimalarial class. Here, we refine the physicochemical properties of this antimalarial class with the aim to improve aqueous solubility and metabolism and to reduce adverse promiscuity. We show the physicochemical properties of this class are intricately balanced with asexual parasite activity and human cell cytotoxicity. Structural modifications we have implemented improved LipE, aqueous solubility and in vitro metabolism while preserving fast acting P. falciparum asexual stage activity. The lead compounds demonstrated equipotent activity against P. knowlesi parasites and were not predisposed to resistance mechanisms of clinically used antimalarials. The optimized compounds exhibited modest activity against early-stage gametocytes, but no activity against pre-erythrocytic liver parasites. Confoundingly, the refined physicochemical properties installed in the compounds did not engender improved oral efficacy in a P. berghei mouse model of malaria compared to earlier studies on the 2-anilino quinazoline class. This study provides the framework for further development of this antimalarial class.
Collapse
|
20
|
Erhunse N, Sahal D. Protecting future antimalarials from the trap of resistance: Lessons from artemisinin-based combination therapy (ACT) failures. J Pharm Anal 2021; 11:541-554. [PMID: 34765267 PMCID: PMC8572664 DOI: 10.1016/j.jpha.2020.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 11/01/2022] Open
Abstract
Having faced increased clinical treatment failures with dihydroartemisinin-piperaquine (DHA-PPQ), Cambodia swapped the first line artemisinin-based combination therapy (ACT) from DHA-PPQ to artesunate-mefloquine given that parasites resistant to piperaquine are susceptible to mefloquine. However, triple mutants have now emerged, suggesting that drug rotations may not be adequate to keep resistance at bay. There is, therefore, an urgent need for alternative treatment strategies to tackle resistance and prevent its spread. A proper understanding of all contributors to artemisinin resistance may help us identify novel strategies to keep artemisinins effective until new drugs become available for their replacement. This review highlights the role of the key players in artemisinin resistance, the current strategies to deal with it and suggests ways of protecting future antimalarial drugs from bowing to resistance as their predecessors did.
Collapse
Affiliation(s)
- Nekpen Erhunse
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
- Department of Biochemistry, Faculty of Life Sciences, University of Benin, Benin City, Edo-State, Nigeria
| | - Dinkar Sahal
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| |
Collapse
|
21
|
Bailey BL, Nguyen W, Ngo A, Goodman CD, Gancheva MR, Favuzza P, Sanz LM, Gamo FJ, Lowes KN, McFadden GI, Wilson DW, Laleu B, Brand S, Jackson PF, Cowman AF, Sleebs BE. Optimisation of 2-(N-phenyl carboxamide) triazolopyrimidine antimalarials with moderate to slow acting erythrocytic stage activity. Bioorg Chem 2021; 115:105244. [PMID: 34452759 DOI: 10.1016/j.bioorg.2021.105244] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022]
Abstract
Malaria is a devastating parasitic disease caused by parasites from the genus Plasmodium. Therapeutic resistance has been reported against all clinically available antimalarials, threatening our ability to control the disease and therefore there is an ongoing need for the development of novel antimalarials. Towards this goal, we identified the 2-(N-phenyl carboxamide) triazolopyrimidine class from a high throughput screen of the Janssen Jumpstarter library against the asexual stages of the P. falciparum parasite. Here we describe the structure activity relationship of the identified class and the optimisation of asexual stage activity while maintaining selectivity against the human HepG2 cell line. The most potent analogues from this study were shown to exhibit equipotent activity against P. falciparum multidrug resistant strains and P. knowlesi asexual parasites. Asexual stage phenotyping studies determined the triazolopyrimidine class arrests parasites at the trophozoite stage, but it is likely these parasites are still metabolically active until the second asexual cycle, and thus have a moderate to slow onset of action. Non-NADPH dependent degradation of the central carboxamide and low aqueous solubility was observed in in vitro ADME profiling. A significant challenge remains to correct these liabilities for further advancement of the 2-(N-phenyl carboxamide) triazolopyrimidine scaffold as a potential moderate to slow acting partner in a curative or prophylactic antimalarial treatment.
Collapse
Affiliation(s)
- Brodie L Bailey
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - William Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Anna Ngo
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | | | - Maria R Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Paola Favuzza
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Laura M Sanz
- Global Health Pharma Research Unit, GlaxoSmithKline, Tres Cantos 28760, Spain
| | | | - Kym N Lowes
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Geoffrey I McFadden
- School of Biosciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne 3004, Australia
| | - Benoît Laleu
- Medicines for Malaria Venture, Geneva 1215, Switzerland
| | - Stephen Brand
- Medicines for Malaria Venture, Geneva 1215, Switzerland
| | - Paul F Jackson
- Global Public Health, Janssen Pharmaceuticals, San Diego, CA, United States
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
22
|
Tisnerat C, Dassonville-Klimpt A, Gosselet F, Sonnet P. Antimalarial drug discovery: from quinine to the most recent promising clinical drug candidates. Curr Med Chem 2021; 29:3326-3365. [PMID: 34344287 DOI: 10.2174/0929867328666210803152419] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
Malaria is a tropical threatening disease caused by Plasmodium parasites, resulting in 409,000 deaths in 2019. The delay of mortality and morbidity has been compounded by the widespread of drug resistant parasites from Southeast Asia since two decades. The emergence of artemisinin-resistant Plasmodium in Africa, where most cases are accounted, highlights the urgent need for new medicines. In this effort, the World Health Organization and Medicines for Malaria Venture joined to define clear goals for novel therapies and characterized the target candidate profile. This ongoing search for new treatments is based on imperative labor in medicinal chemistry which is summarized here with particular attention to hit-to-lead optimizations, key properties, and modes of action of these novel antimalarial drugs. This review, after presenting the current antimalarial chemotherapy, from quinine to the latest marketed drugs, focuses in particular on recent advances of the most promising antimalarial candidates in clinical and preclinical phases.
Collapse
Affiliation(s)
- Camille Tisnerat
- AGIR UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens. France
| | | | | | - Pascal Sonnet
- AGIR UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens. France
| |
Collapse
|
23
|
Siddiqui FA, Liang X, Cui L. Plasmodium falciparum resistance to ACTs: Emergence, mechanisms, and outlook. Int J Parasitol Drugs Drug Resist 2021; 16:102-118. [PMID: 34090067 PMCID: PMC8188179 DOI: 10.1016/j.ijpddr.2021.05.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/06/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023]
Abstract
Emergence and spread of resistance in Plasmodium falciparum to the frontline treatment artemisinin-based combination therapies (ACTs) in the epicenter of multidrug resistance of Southeast Asia threaten global malaria control and elimination. Artemisinin (ART) resistance (or tolerance) is defined clinically as delayed parasite clearance after treatment with an ART drug. The resistance phenotype is restricted to the early ring stage and can be measured in vitro using a ring-stage survival assay. ART resistance is associated with mutations in the propeller domain of the Kelch family protein K13. As a pro-drug, ART is activated primarily by heme, which is mainly derived from hemoglobin digestion in the food vacuole. Activated ARTs can react promiscuously with a wide range of cellular targets, disrupting cellular protein homeostasis. Consistent with this mode of action for ARTs, the molecular mechanisms of K13-mediated ART resistance involve reduced hemoglobin uptake/digestion and increased cellular stress response. Mutations in other genes such as AP-2μ (adaptor protein-2 μ subunit), UBP-1 (ubiquitin-binding protein-1), and Falcipain 2a that interfere with hemoglobin uptake and digestion also increase resistance to ARTs. ART resistance has facilitated the development of resistance to the partner drugs, resulting in rapidly declining ACT efficacies. The molecular markers for resistance to the partner drugs are mostly associated with point mutations in the two food vacuole membrane transporters PfCRT and PfMDR1, and amplification of pfmdr1 and the two aspartic protease genes plasmepsin 2 and 3. It has been observed that mutations in these genes can have opposing effects on sensitivities to different partner drugs, which serve as the principle for designing triple ACTs and drug rotation. Although clinical ACT resistance is restricted to Southeast Asia, surveillance for drug resistance using in vivo clinical efficacy, in vitro assays, and molecular approaches is required to prevent or slow down the spread of resistant parasites.
Collapse
Affiliation(s)
- Faiza Amber Siddiqui
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Xiaoying Liang
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Liwang Cui
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
24
|
Abstract
Although the last two decades have seen a substantial decline in malaria incidence and mortality due to the use of insecticide-treated bed nets and artemisinin combination therapy, the threat of drug resistance is a constant obstacle to sustainable malaria control. Given that patients can die quickly from this disease, public health officials and doctors need to understand whether drug resistance exists in the parasite population, as well as how prevalent it is so they can make informed decisions about treatment. As testing for drug efficacy before providing treatment to malaria patients is impractical, researchers need molecular markers of resistance that can be more readily tracked in parasite populations. To this end, much work has been done to unravel the genetic underpinnings of drug resistance in Plasmodium falciparum. The aim of this review is to provide a broad overview of common genomic approaches that have been used to discover the alleles that drive drug response phenotypes in the most lethal human malaria parasite.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
25
|
Plasmodium falciparum Atg18 localizes to the food vacuole via interaction with the multi-drug resistance protein 1 and phosphatidylinositol 3-phosphate. Biochem J 2021; 478:1705-1732. [PMID: 33843972 DOI: 10.1042/bcj20210001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/27/2022]
Abstract
Autophagy, a lysosome-dependent degradative process, does not appear to be a major degradative process in malaria parasites and has a limited repertoire of genes. To better understand the autophagy process, we investigated Plasmodium falciparum Atg18 (PfAtg18), a PROPPIN family protein, whose members like S. cerevisiae Atg18 (ScAtg18) and human WIPI2 bind PI3P and play an essential role in autophagosome formation. Wild type and mutant PfAtg18 were expressed in P. falciparum and assessed for localization, the effect of various inhibitors and antimalarials on PfAtg18 localization, and identification of PfAtg18-interacting proteins. PfAtg18 is expressed in asexual erythrocytic stages and localized to the food vacuole, which was also observed with other Plasmodium Atg18 proteins, indicating that food vacuole localization is likely a shared feature. Interaction of PfAtg18 with the food vacuole-associated PI3P is essential for localization, as PfAtg18 mutants of PI3P-binding motifs neither bound PI3P nor localized to the food vacuole. Interestingly, wild type ScAtg18 interacted with PI3P, but its expression in P. falciparum showed complete cytoplasmic localization, indicating additional requirement for food vacuole localization. The food vacuole multi-drug resistance protein 1 (MDR1) was consistently identified in the immunoprecipitates of PfAtg18 and P. berghei Atg18, and also interacted with PfAtg18. In contrast with PfAtg18, ScAtg18 did not interact with MDR1, which, in addition to PI3P, could play a critical role in localization of PfAtg18. Chloroquine and amodiaquine caused cytoplasmic localization of PfAtg18, suggesting that these target PfAtg18 transport pathway. Thus, PI3P and MDR1 are critical mediators of PfAtg18 localization.
Collapse
|
26
|
Liu S, Huckaby AC, Brown AC, Moore CC, Burbulis I, McConnell MJ, Güler JL. Single-cell sequencing of the small and AT-skewed genome of malaria parasites. Genome Med 2021; 13:75. [PMID: 33947449 PMCID: PMC8094492 DOI: 10.1186/s13073-021-00889-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/17/2021] [Indexed: 12/23/2022] Open
Abstract
Single-cell genomics is a rapidly advancing field; however, most techniques are designed for mammalian cells. We present a single-cell sequencing pipeline for an intracellular parasite, Plasmodium falciparum, with a small genome of extreme base content. Through optimization of a quasi-linear amplification method, we target the parasite genome over contaminants and generate coverage levels allowing detection of minor genetic variants. This work, as well as efforts that build on these findings, will enable detection of parasite heterogeneity contributing to P. falciparum adaptation. Furthermore, this study provides a framework for optimizing single-cell amplification and variant analysis in challenging genomes.
Collapse
Affiliation(s)
- Shiwei Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Adam C Huckaby
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Audrey C Brown
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Christopher C Moore
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Ian Burbulis
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Escuela de Medicina, Universidad San Sebastian, Puerto Montt, Chile
| | - Michael J McConnell
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Current address: Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Jennifer L Güler
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
27
|
McDaniels JM, Huckaby AC, Carter SA, Lingeman S, Francis A, Congdon M, Santos W, Rathod PK, Guler JL. Extrachromosomal DNA amplicons in antimalarial-resistant Plasmodium falciparum. Mol Microbiol 2021; 115:574-590. [PMID: 33053232 PMCID: PMC8246734 DOI: 10.1111/mmi.14624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 12/29/2022]
Abstract
Extrachromosomal (ec) DNAs are genetic elements that exist separately from the genome. Since ecDNA can carry beneficial genes, they are a powerful adaptive mechanism in cancers and many pathogens. For the first time, we report ecDNA contributing to antimalarial resistance in Plasmodium falciparum, the most virulent human malaria parasite. Using pulse field gel electrophoresis combined with PCR-based copy number analysis, we detected two ecDNA elements that differ in migration and structure. Entrapment in the electrophoresis well and low susceptibility to exonucleases revealed that the biologically relevant ecDNA element is large and complex in structure. Using deep sequencing, we show that ecDNA originates from the chromosome and expansion of an ecDNA-specific sequence may improve its segregation or expression. We speculate that ecDNA is maintained using established mechanisms due to shared characteristics with the mitochondrial genome. Implications of ecDNA discovery in this organism are wide-reaching due to the potential for new strategies to target resistance development.
Collapse
Affiliation(s)
| | - Adam C. Huckaby
- Department of BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | | | | | - Audrey Francis
- Department of BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | | | | | | | - Jennifer L. Guler
- Department of BiologyUniversity of VirginiaCharlottesvilleVAUSA
- Division of Infectious Diseases and International HealthDepartment of MedicineUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
28
|
Nguyen W, Dans MG, Ngo A, Gancheva MR, Romeo O, Duffy S, de Koning-Ward TF, Lowes KN, Sabroux HJ, Avery VM, Wilson DW, Gilson PR, Sleebs BE. Structure activity refinement of phenylsulfonyl piperazines as antimalarials that block erythrocytic invasion. Eur J Med Chem 2021; 214:113253. [PMID: 33610028 DOI: 10.1016/j.ejmech.2021.113253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
The emerging resistance to combination therapies comprised of artemisinin derivatives has driven a need to identify new antimalarials with novel mechanisms of action. Central to the survival and proliferation of the malaria parasite is the invasion of red blood cells by Plasmodium merozoites, providing an attractive target for novel therapeutics. A screen of the Medicines for Malaria Venture Pathogen Box employing transgenic P. falciparum parasites expressing the nanoluciferase bioluminescent reporter identified the phenylsulfonyl piperazine class as a specific inhibitor of erythrocyte invasion. Here, we describe the optimization and further characterization of the phenylsulfonyl piperazine class. During the optimization process we defined the functionality required for P. falciparum asexual stage activity and determined the alpha-carbonyl S-methyl isomer was important for antimalarial potency. The optimized compounds also possessed comparable activity against multidrug resistant strains of P. falciparum and displayed weak activity against sexual stage gametocytes. We determined that the optimized compounds blocked erythrocyte invasion consistent with the asexual activity observed and therefore the phenylsulfonyl piperazine analogues described could serve as useful tools for studying Plasmodium erythrocyte invasion.
Collapse
Affiliation(s)
- William Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Madeline G Dans
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia; School of Medicine, Deakin University, Waurn Ponds, 3216, Australia
| | - Anna Ngo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Maria R Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Ornella Romeo
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Sandra Duffy
- Discovery Biology, Griffith University, Nathan, Queensland, 4111, Australia
| | | | - Kym N Lowes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Helene Jousset Sabroux
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith University, Nathan, Queensland, 4111, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia
| | - Paul R Gilson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, 3004, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
29
|
Chemoprotective antimalarials identified through quantitative high-throughput screening of Plasmodium blood and liver stage parasites. Sci Rep 2021; 11:2121. [PMID: 33483532 PMCID: PMC7822874 DOI: 10.1038/s41598-021-81486-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration–response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.
Collapse
|
30
|
Sato S. Plasmodium-a brief introduction to the parasites causing human malaria and their basic biology. J Physiol Anthropol 2021; 40:1. [PMID: 33413683 PMCID: PMC7792015 DOI: 10.1186/s40101-020-00251-9] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Malaria is one of the most devastating infectious diseases of humans. It is problematic clinically and economically as it prevails in poorer countries and regions, strongly hindering socioeconomic development. The causative agents of malaria are unicellular protozoan parasites belonging to the genus Plasmodium. These parasites infect not only humans but also other vertebrates, from reptiles and birds to mammals. To date, over 200 species of Plasmodium have been formally described, and each species infects a certain range of hosts. Plasmodium species that naturally infect humans and cause malaria in large areas of the world are limited to five-P. falciparum, P. vivax, P. malariae, P. ovale and P. knowlesi. The first four are specific for humans, while P. knowlesi is naturally maintained in macaque monkeys and causes zoonotic malaria widely in South East Asia. Transmission of Plasmodium species between vertebrate hosts depends on an insect vector, which is usually the mosquito. The vector is not just a carrier but the definitive host, where sexual reproduction of Plasmodium species occurs, and the parasite's development in the insect is essential for transmission to the next vertebrate host. The range of insect species that can support the critical development of Plasmodium depends on the individual parasite species, but all five Plasmodium species causing malaria in humans are transmitted exclusively by anopheline mosquitoes. Plasmodium species have remarkable genetic flexibility which lets them adapt to alterations in the environment, giving them the potential to quickly develop resistance to therapeutics such as antimalarials and to change host specificity. In this article, selected topics involving the Plasmodium species that cause malaria in humans are reviewed.
Collapse
Affiliation(s)
- Shigeharu Sato
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, 88400 Kota Kinabalu, Sabah, Malaysia.
- Department of Pathobiology and Medical Diagnostics, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Jalan UMS, 88400, Kota Kinabalu, Sabah, Malaysia.
| |
Collapse
|
31
|
Rizwan HM, Abbas H, Sajid MS, Maqbool M, Jones MK, Ullah MI, Ijaz N. Drug Resistance in Protozoal Infections. BIOCHEMISTRY OF DRUG RESISTANCE 2021:95-142. [DOI: 10.1007/978-3-030-76320-6_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
32
|
Transmission of Artemisinin-Resistant Malaria Parasites to Mosquitoes under Antimalarial Drug Pressure. Antimicrob Agents Chemother 2020; 65:AAC.00898-20. [PMID: 33139275 PMCID: PMC7927852 DOI: 10.1128/aac.00898-20] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
Resistance to artemisinin-based combination therapy (ACT) in the Plasmodium falciparum parasite is threatening to reverse recent gains in reducing global deaths from malaria. While resistance manifests as delayed parasite clearance in patients, the phenotype can only spread geographically via the sexual stages and mosquito transmission. In addition to their asexual killing properties, artemisinin and its derivatives sterilize sexual male gametocytes. Whether resistant parasites overcome this sterilizing effect has not, however, been fully tested. Resistance to artemisinin-based combination therapy (ACT) in the Plasmodium falciparum parasite is threatening to reverse recent gains in reducing global deaths from malaria. While resistance manifests as delayed parasite clearance in patients, the phenotype can only spread geographically via the sexual stages and mosquito transmission. In addition to their asexual killing properties, artemisinin and its derivatives sterilize sexual male gametocytes. Whether resistant parasites overcome this sterilizing effect has not, however, been fully tested. Here, we analyzed P. falciparum clinical isolates from the Greater Mekong Subregion, each demonstrating delayed clinical clearance and known resistance-associated polymorphisms in the Kelch13 (PfK13var) gene. As well as demonstrating reduced asexual sensitivity to drug, certain PfK13var isolates demonstrated a marked reduction in sensitivity to artemisinin in an in vitro male gamete formation assay. Importantly, this same reduction in sensitivity was observed when the most resistant isolate was tested directly in mosquito feeds. These results indicate that, under artemisinin drug pressure, while sensitive parasites are blocked, resistant parasites continue transmission. This selective advantage for resistance transmission could favor acquisition of additional host-specificity or polymorphisms affecting partner drug sensitivity in mixed infections. Favored resistance transmission under ACT coverage could have profound implications for the spread of multidrug-resistant malaria beyond Southeast Asia.
Collapse
|
33
|
Zhang X, Deitsch KW, Kirkman LA. The contribution of extrachromosomal DNA to genome plasticity in malaria parasites. Mol Microbiol 2020; 115:503-507. [PMID: 33103309 DOI: 10.1111/mmi.14632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 01/20/2023]
Abstract
Malaria caused by the protozoan parasite Plasmodium falciparum continues to impose significant morbidity and mortality, despite substantial investment into drug and vaccine development and deployment. Underlying the resilience of this parasite is its remarkable ability to undergo genome modifications, thus, providing parasite populations with extensive genetic variability that accelerates selection of drug resistance and limits the efficacy of most vaccines. This genome plasticity is rooted in the mechanisms of DNA repair that parasites employ to maintain genome integrity, a process skewed toward homologous recombination through the evolutionary loss of classical nonhomologous end joining. Repair of DNA double-strand breaks have been shown to enable "shuffling" of antigen-encoding gene sequences to vastly increase antigen diversity and to enable copy number expansion of genes that contribute to drug resistance. The latter phenomenon has been proposed to be a major contributor to the rise of resistance to several classes of antimalarial drugs. In this issue of Molecular Microbiology, McDaniels and colleagues add yet another mechanism that malaria parasites use to reduce drug susceptibility by demonstrating that P. falciparum can maintain expanded arrays of drug resistance cassettes as stably replicating, circular, extrachromosomal DNAs, thus, expanding genome plasticity beyond the parasite's 14 nuclear chromosomes.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Kirk W Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Laura A Kirkman
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA.,Department of Internal Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
34
|
Pimpat Y, Saralamba N, Boonyuen U, Pukrittayakamee S, Nosten F, Smithuis F, Day NPJ, Dondorp AM, Imwong M. Genetic analysis of the orthologous crt and mdr1 genes in Plasmodium malariae from Thailand and Myanmar. Malar J 2020; 19:315. [PMID: 32867773 PMCID: PMC7461347 DOI: 10.1186/s12936-020-03391-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/25/2020] [Indexed: 11/19/2022] Open
Abstract
Background Plasmodium malariae is a widely spread but neglected human malaria parasite, which causes chronic infections. Studies on genetic polymorphisms of anti-malarial drug target genes in P. malariae are limited. Previous reports have shown polymorphisms in the P. malariae dihydrofolate reductase gene associated with pyrimethamine resistance and linked to pyrimethamine drug pressure. This study investigated polymorphisms of the P. malariae homologous genes, chloroquine resistant transporter and multidrug resistant 1, associated with chloroquine and mefloquine resistance in Plasmodium falciparum. Methods The orthologous P. malariae crt and mdr1 genes were studied in 95 patients with P. malariae infection between 2002 and 2016 from Thailand (N = 51) and Myanmar (N = 44). Gene sequences were analysed using BioEdit, MEGA7, and DnaSP programs. Mutations and gene amplifications were compared with P. falciparum and Plasmodium vivax orthologous genes. Protein topology models derived from the observed pmcrt and pmmdr1 haplotypes were constructed and analysed using Phyre2, SWISS MODEL and Discovery Studio Visualization V 17.2. Results Two non-synonymous mutations were observed in exon 2 (H53P, 40%) and exon 8 (E278D, 44%) of pmcrt. The topology model indicated that H53P and E278D were located outside of the transmembrane domain and were unlikely to affect protein function. Pmmdr1 was more diverse than pmcrt, with 10 non-synonymous and 3 synonymous mutations observed. Non-synonymous mutations were located in the parasite cytoplasmic site, transmembrane 11 and nucleotide binding domains 1 and 2. Polymorphisms conferring amino acid changes in the transmembrane and nucleotide binding domains were predicted to have some effect on PmMDR1 conformation, but were unlikely to affect protein function. All P. malariae parasites in this study contained a single copy of the mdr1 gene. Conclusions The observed polymorphisms in pmcrt and pmmdr1 genes are unlikely to affect protein function and unlikely related to chloroquine drug pressure. Similarly, the absence of pmmdr1 copy number variation suggests limited mefloquine drug pressure on the P. malariae parasite population, despite its long time use in Thailand for the treatment of falciparum malaria.
Collapse
Affiliation(s)
- Yupawadee Pimpat
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Naowarat Saralamba
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Usa Boonyuen
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sasithon Pukrittayakamee
- 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
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand.,Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Frank Smithuis
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.,Medical Action Myanmar, Yangon, Myanmar
| | - 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, Churchill Hospital, 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, Churchill Hospital, University of Oxford, Oxford, UK
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
35
|
Noviyanti R, Miotto O, Barry A, Marfurt J, Siegel S, Thuy-Nhien N, Quang HH, Anggraeni ND, Laihad F, Liu Y, Sumiwi ME, Trimarsanto H, Coutrier F, Fadila N, Ghanchi N, Johora FT, Puspitasari AM, Tavul L, Trianty L, Utami RAS, Wang D, Wangchuck K, Price RN, Auburn S. Implementing parasite genotyping into national surveillance frameworks: feedback from control programmes and researchers in the Asia-Pacific region. Malar J 2020; 19:271. [PMID: 32718342 PMCID: PMC7385952 DOI: 10.1186/s12936-020-03330-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/09/2020] [Indexed: 01/13/2023] Open
Abstract
The Asia-Pacific region faces formidable challenges in achieving malaria elimination by the proposed target in 2030. Molecular surveillance of Plasmodium parasites can provide important information on malaria transmission and adaptation, which can inform national malaria control programmes (NMCPs) in decision-making processes. In November 2019 a parasite genotyping workshop was held in Jakarta, Indonesia, to review molecular approaches for parasite surveillance and explore ways in which these tools can be integrated into public health systems and inform policy. The meeting was attended by 70 participants from 8 malaria-endemic countries and partners of the Asia Pacific Malaria Elimination Network. The participants acknowledged the utility of multiple use cases for parasite genotyping including: quantifying the prevalence of drug resistant parasites, predicting risks of treatment failure, identifying major routes and reservoirs of infection, monitoring imported malaria and its contribution to local transmission, characterizing the origins and dynamics of malaria outbreaks, and estimating the frequency of Plasmodium vivax relapses. However, the priority of each use case varies with different endemic settings. Although a one-size-fits-all approach to molecular surveillance is unlikely to be applicable across the Asia-Pacific region, consensus on the spectrum of added-value activities will help support data sharing across national boundaries. Knowledge exchange is needed to establish local expertise in different laboratory-based methodologies and bioinformatics processes. Collaborative research involving local and international teams will help maximize the impact of analytical outputs on the operational needs of NMCPs. Research is also needed to explore the cost-effectiveness of genetic epidemiology for different use cases to help to leverage funding for wide-scale implementation. Engagement between NMCPs and local researchers will be critical throughout this process.
Collapse
Affiliation(s)
| | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- Centre for Genomics and Global Health, Big Data Institute, University of Oxford, Oxford, UK
| | - Alyssa Barry
- School of Medicine, Deakin University, Geelong, VIC, Australia
- Burnet Institute, Melbourne, VIC, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Jutta Marfurt
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Sasha Siegel
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Nguyen Thuy-Nhien
- Centre for Tropical Medicine, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Huynh Hong Quang
- Institute of Malariology, Parasitology and Entomology, Quy Nhon, Vietnam
| | | | | | - 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, Jiangsu Province, China
| | | | | | - Farah Coutrier
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Nadia Fadila
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Najia Ghanchi
- Pathology, Aga Khan University Hospital, Karachi, Pakistan
| | - Fatema Tuj Johora
- Infectious Diseases Division, International Centre for Diarrheal Diseases Research, Bangladesh Mohakhali, Dhaka, Bangladesh
| | | | - Livingstone Tavul
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Leily Trianty
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | - Duoquan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Kesang Wangchuck
- Royal Center for Disease Control, Department of Public Health, Ministry of Health, Thimphu, Bhutan
| | - Ric N Price
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Auburn
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
36
|
Batista FA, Gyau B, Vilacha JF, Bosch SS, Lunev S, Wrenger C, Groves MR. New directions in antimalarial target validation. Expert Opin Drug Discov 2020; 15:189-202. [PMID: 31959021 DOI: 10.1080/17460441.2020.1691996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Malaria is one of the most prevalent human infections worldwide with over 40% of the world's population living in malaria-endemic areas. In the absence of an effective vaccine, emergence of drug-resistant strains requires urgent drug development. Current methods applied to drug target validation, a crucial step in drug discovery, possess limitations in malaria. These constraints require the development of techniques capable of simplifying the validation of Plasmodial targets.Areas covered: The authors review the current state of the art in techniques used to validate drug targets in malaria, including our contribution - the protein interference assay (PIA) - as an additional tool in rapid in vivo target validation.Expert opinion: Each technique in this review has advantages and disadvantages, implying that future validation efforts should not focus on a single approach, but integrate multiple approaches. PIA is a significant addition to the current toolset of antimalarial validation. Validation of aspartate metabolism as a druggable pathway provided proof of concept of how oligomeric interfaces can be exploited to control specific activity in vivo. PIA has the potential to be applied not only to other enzymes/pathways of the malaria parasite but could, in principle, be extrapolated to other infectious diseases.
Collapse
Affiliation(s)
- Fernando A Batista
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands.,Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Benjamin Gyau
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Juliana F Vilacha
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Soraya S Bosch
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands.,Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sergey Lunev
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Matthew R Groves
- Structural Biology Unit, XB20 Drug Design, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Calarco L, Barratt J, Ellis J. Detecting sequence variants in clinically important protozoan parasites. Int J Parasitol 2019; 50:1-18. [PMID: 31857072 DOI: 10.1016/j.ijpara.2019.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 02/06/2023]
Abstract
Second and third generation sequencing methods are crucial for population genetic studies, and variant detection is a popular approach for exploiting this sequence data. While mini- and microsatellites are historically useful markers for studying important Protozoa such as Toxoplasma and Plasmodium spp., detecting non-repetitive variants such as those found in genes can be fundamental to investigating a pathogen's biology. These variants, namely single nucleotide polymorphisms and insertions and deletions, can help elucidate the genetic basis of an organism's pathogenicity, identify selective pressures, and resolve phylogenetic relationships. They also have the added benefit of possessing a comparatively low mutation rate, which contributes to their stability. However, there is a plethora of variant analysis tools with nuanced pipelines and conflicting recommendations for best practise, which can be confounding. This lack of standardisation means that variant analysis requires careful parameter optimisation, an understanding of its limitations, and the availability of high quality data. This review explores the value of variant detection when applied to non-model organisms such as clinically important protozoan pathogens. The limitations of current methods are discussed, including special considerations that require the end-users' attention to ensure that the results generated are reproducible, and the biological conclusions drawn are valid.
Collapse
Affiliation(s)
- Larissa Calarco
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia.
| | - Joel Barratt
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| | - John Ellis
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW 2007, Australia
| |
Collapse
|
38
|
Veale CGL, Jayram J, Naidoo S, Laming D, Swart T, Olivier T, Akerman MP, de Villiers KA, Hoppe HC, Jeena V. Insights into structural and physicochemical properties required for β-hematin inhibition of privileged triarylimidazoles. RSC Med Chem 2019; 11:85-91. [PMID: 33479606 DOI: 10.1039/c9md00468h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 11/21/2022] Open
Abstract
In this study, we investigated a series of triarylimidazoles, in an effort to elucidate critical SAR information pertaining to their anti-plasmodial and β-hematin inhibitory activity. Our results showed that in addition to the positional effects of ring substitution, subtle changes to lipophilicity and imidazole ionisability were important factors in SAR interpretation. Finally, in silico adsorption analysis indicated that these compounds exert their effect by inhibiting β-hematin crystal growth at the fast growing 001 face.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics , Pietermaritzburg Campus , University of KwaZulu-Natal , Private Bag X01 , Scottsville , 3209 , South Africa
| | - Janeeka Jayram
- School of Chemistry and Physics , Pietermaritzburg Campus , University of KwaZulu-Natal , Private Bag X01 , Scottsville , 3209 , South Africa
| | - Shivani Naidoo
- School of Chemistry and Physics , Pietermaritzburg Campus , University of KwaZulu-Natal , Private Bag X01 , Scottsville , 3209 , South Africa
| | - Dustin Laming
- Department of Biochemistry and Microbiology , Rhodes University , Grahamstown , 6140 , South Africa
| | - Tarryn Swart
- Department of Biochemistry and Microbiology , Rhodes University , Grahamstown , 6140 , South Africa
| | - Tania Olivier
- Department of Chemistry and Polymer Science , Stellenbosch University , Private Bag X1 , Matieland , 7602 , South Africa . ;
| | - Matthew P Akerman
- School of Chemistry and Physics , Pietermaritzburg Campus , University of KwaZulu-Natal , Private Bag X01 , Scottsville , 3209 , South Africa
| | - Katherine A de Villiers
- Department of Chemistry and Polymer Science , Stellenbosch University , Private Bag X1 , Matieland , 7602 , South Africa . ;
| | - Heinrich C Hoppe
- Department of Biochemistry and Microbiology , Rhodes University , Grahamstown , 6140 , South Africa
| | - Vineet Jeena
- School of Chemistry and Physics , Pietermaritzburg Campus , University of KwaZulu-Natal , Private Bag X01 , Scottsville , 3209 , South Africa
| |
Collapse
|
39
|
Martin RE. The transportome of the malaria parasite. Biol Rev Camb Philos Soc 2019; 95:305-332. [PMID: 31701663 DOI: 10.1111/brv.12565] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Membrane transport proteins, also known as transporters, control the movement of ions, nutrients, metabolites, and waste products across the membranes of a cell and are central to its biology. Proteins of this type also serve as drug targets and are key players in the phenomenon of drug resistance. The malaria parasite has a relatively reduced transportome, with only approximately 2.5% of its genes encoding transporters. Even so, assigning functions and physiological roles to these proteins, and ascertaining their contributions to drug action and drug resistance, has been very challenging. This review presents a detailed critique and synthesis of the disruption phenotypes, protein subcellular localisations, protein functions (observed or predicted), and links to antimalarial drug resistance for each of the parasite's transporter genes. The breadth and depth of the gene disruption data are particularly impressive, with at least one phenotype determined in the parasite's asexual blood stage for each transporter gene, and multiple phenotypes available for 76% of the genes. Analysis of the curated data set revealed there to be relatively little redundancy in the Plasmodium transportome; almost two-thirds of the parasite's transporter genes are essential or required for normal growth in the asexual blood stage of the parasite, and this proportion increased to 78% when the disruption phenotypes available for the other parasite life stages were included in the analysis. These observations, together with the finding that 22% of the transportome is implicated in the parasite's resistance to existing antimalarials and/or drugs within the development pipeline, indicate that transporters are likely to serve, or are already serving, as drug targets. Integration of the different biological and bioinformatic data sets also enabled the selection of candidates for transport processes known to be essential for parasite survival, but for which the underlying proteins have thus far remained undiscovered. These include potential transporters of pantothenate, isoleucine, or isopentenyl diphosphate, as well as putative anion-selective channels that may serve as the pore component of the parasite's 'new permeation pathways'. Other novel insights into the parasite's biology included the identification of transporters for the potential development of antimalarial treatments, transmission-blocking drugs, prophylactics, and genetically attenuated vaccines. The syntheses presented herein set a foundation for elucidating the functions and physiological roles of key members of the Plasmodium transportome and, ultimately, to explore and realise their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rowena E Martin
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
40
|
Cowell AN, Winzeler EA. The genomic architecture of antimalarial drug resistance. Brief Funct Genomics 2019; 18:314-328. [PMID: 31119263 PMCID: PMC6859814 DOI: 10.1093/bfgp/elz008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/19/2019] [Accepted: 04/09/2019] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum and Plasmodium vivax, the two protozoan parasite species that cause the majority of cases of human malaria, have developed resistance to nearly all known antimalarials. The ability of malaria parasites to develop resistance is primarily due to the high numbers of parasites in the infected person's bloodstream during the asexual blood stage of infection in conjunction with the mutability of their genomes. Identifying the genetic mutations that mediate antimalarial resistance has deepened our understanding of how the parasites evade our treatments and reveals molecular markers that can be used to track the emergence of resistance in clinical samples. In this review, we examine known genetic mutations that lead to resistance to the major classes of antimalarial medications: the 4-aminoquinolines (chloroquine, amodiaquine and piperaquine), antifolate drugs, aryl amino-alcohols (quinine, lumefantrine and mefloquine), artemisinin compounds, antibiotics (clindamycin and doxycycline) and a napthoquinone (atovaquone). We discuss how the evolution of antimalarial resistance informs strategies to design the next generation of antimalarial therapies.
Collapse
Affiliation(s)
- Annie N Cowell
- Division of Infectious Diseases and Global Health, Department of Medicine, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| |
Collapse
|
41
|
Huckaby AC, Granum CS, Carey MA, Szlachta K, Al-Barghouthi B, Wang YH, Guler JL. Complex DNA structures trigger copy number variation across the Plasmodium falciparum genome. Nucleic Acids Res 2019; 47:1615-1627. [PMID: 30576466 PMCID: PMC6393310 DOI: 10.1093/nar/gky1268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Antimalarial resistance is a major obstacle in the eradication of the human malaria parasite, Plasmodium falciparum. Genome amplifications, a type of DNA copy number variation (CNV), facilitate overexpression of drug targets and contribute to parasite survival. Long monomeric A/T tracks are found at the breakpoints of many Plasmodium resistance-conferring CNVs. We hypothesize that other proximal sequence features, such as DNA hairpins, act with A/T tracks to trigger CNV formation. By adapting a sequence analysis pipeline to investigate previously reported CNVs, we identified breakpoints in 35 parasite clones with near single base-pair resolution. Using parental genome sequence, we predicted the formation of stable hairpins within close proximity to all future breakpoint locations. Especially stable hairpins were predicted to form near five shared breakpoints, establishing that the initiating event could have occurred at these sites. Further in-depth analyses defined characteristics of these 'trigger sites' across the genome and detected signatures of error-prone repair pathways at the breakpoints. We propose that these two genomic signals form the initial lesion (hairpins) and facilitate microhomology-mediated repair (A/T tracks) that lead to CNV formation across this highly repetitive genome. Targeting these repair pathways in P. falciparum may be used to block adaptation to antimalarial drugs.
Collapse
Affiliation(s)
- Adam C Huckaby
- Department of Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Claire S Granum
- Department of Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Maureen A Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Karol Szlachta
- Department of Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Basel Al-Barghouthi
- Department of Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA 22908, USA.,Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Yuh-Hwa Wang
- Department of Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Charlottesville, VA 22908, USA.,Division of Infectious Diseases and International Health, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
42
|
Development of artemisinin resistance in malaria therapy. Pharmacol Res 2019; 146:104275. [DOI: 10.1016/j.phrs.2019.104275] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 01/23/2023]
|
43
|
Morita M, Hayashi K, Sato A, Hiramoto A, Kaneko O, Isogawa R, Kurosaki Y, Miyoshi SI, Chang KS, Wataya Y, Kim HS. Genomic and biological features of Plasmodium falciparum resistance against antimalarial endoperoxide N-89. Gene 2019; 716:144016. [PMID: 31377318 DOI: 10.1016/j.gene.2019.144016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 02/05/2023]
Abstract
Drug resistance of malaria parasites remains a problem affecting antimalarial treatment and control of the disease. We previously synthesized an antimalarial endoperoxide, N-89, having high antimalarial effects in vitro and in vivo. In this study we seek to understand the resistant mechanism against N-89 by establishing a highly N-89-resistant clone, named NRC10H, of the Plasmodium falciparum FCR-3 strain. We describe gene mutations in the parent FCR-3 strain and the NRC10H clone using whole-genome sequencing and subsequently by expression profiling using quantitative real-time PCR. Seven genes related to drug resistance, proteolysis, glycophosphatidylinositol anchor biosynthesis, and phosphatidylethanolamine biosynthesis exhibited a single amino acid substitution in the NRC10H clone. Among these seven genes, the multidrug resistance protein 2 (mdr2) variant A532S was found only in NRC10H. The genetic status of the P. falciparum endoplasmic reticulum-resident calcium binding protein (PfERC), a potential target of N-89, was similar between the NRC10H clone and the parent FCR-3 strain. These findings suggest that the genetic alterations of the identified seven genes, in particular mdr2, in NRC10H could give rise to resistance of the antimalarial endoperoxide N-89.
Collapse
Affiliation(s)
- Masayuki Morita
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan; Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Kosuke Hayashi
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan
| | - Akira Sato
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan; Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Akiko Hiramoto
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki 852-8423, Japan
| | - Rena Isogawa
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan
| | - Yuji Kurosaki
- Department of Pharmaceutical Formulation Design, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama, Okayama 700-8530, Japan
| | - Shin-Ichi Miyoshi
- Department of Sanitary Microbiology, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama, Okayama 700-8530, Japan
| | - Kyung-Soo Chang
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yusuke Wataya
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan
| | - Hye-Sook Kim
- Division of International Infectious Diseases Control, Faculty of Pharmaceutical Sciences, Okayama University, Tsushima-Naka, Kita-Ku, Okayama 700-8530, Japan.
| |
Collapse
|
44
|
Efficacy and Safety of Pyronaridine-Artesunate plus Single-Dose Primaquine for Treatment of Uncomplicated Plasmodium falciparum Malaria in Eastern Cambodia. Antimicrob Agents Chemother 2019; 63:AAC.02242-18. [PMID: 30602520 DOI: 10.1128/aac.02242-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/17/2018] [Indexed: 11/20/2022] Open
Abstract
In Cambodia, multidrug-resistant Plasmodium falciparum undermines the treatment of uncomplicated malaria, and new therapeutic options are needed. Pyronaridine-artesunate has not previously been evaluated in eastern Cambodia. We conducted a single-arm, open-label, prospective study between July and December 2017 at the Koh Gnek (Mondulkiri) and Veun Sai (Rattanakiri) health centers in eastern Cambodia. Eligible patients were aged ≥7 years (females, ages 12 to 18 years, were excluded), weighing ≥20 kg, with microscopically confirmed P. falciparum monoinfection and fever. Oral pyronaridine-artesunate was administered once daily for 3 days, dosed according to body weight, plus a single dose of primaquine on day 0. Sixty patients were recruited to Koh Gnek, and 61 patients were recruited to Veun Sai. The primary outcomes, i.e., the day 42 PCR-adjusted adequate clinical and parasitological responses (ACPRs), were 98.3% (95% confidence interval [CI], 88.4 to 99.8) in Koh Gnek and 96.7% (95% CI, 87.3 to 99.2) in Veun Sai (Kaplan-Meier). In a per-protocol analysis, the proportions of patients with day 42 PCR-adjusted ACPRs were 98.3% (57/58; 95% CI, 90.8 to 100.0) at Koh Gnek and 96.7% (58/60; 95% CI, 88.5 to 99.6) at Veun Sai. The Kelch13 (C580Y) mutation was present in 70.0% (77/110) of isolates. The copy numbers were increased in 61.3% (73/119) of isolates for Pfpm2 and in 1.7% (2/119) for Pfmdr1 There was no relationship between outcome and the 50% inhibitory concentration of pyronaridine. Adverse events were consistent with malaria, and there were no serious adverse events. Pyronaridine-artesunate has high efficacy in eastern Cambodia and could be used to increase the diversity of antimalarial therapy in the region. (This study is registered in the Australian New Zealand Clinical Trials Registry [ANZCTR] under no. ACTRN12618001300268.).
Collapse
|
45
|
Evaluation of 4-Amino 2-Anilinoquinazolines against Plasmodium and Other Apicomplexan Parasites In Vitro and in a P. falciparum Humanized NOD- scid IL2Rγ null Mouse Model of Malaria. Antimicrob Agents Chemother 2019; 63:AAC.01804-18. [PMID: 30559138 DOI: 10.1128/aac.01804-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/10/2018] [Indexed: 11/20/2022] Open
Abstract
A series of 4-amino 2-anilinoquinazolines optimized for activity against the most lethal malaria parasite of humans, Plasmodium falciparum, was evaluated for activity against other human Plasmodium parasites and related apicomplexans that infect humans and animals. Four of the most promising compounds from the 4-amino 2-anilinoquinazoline series were equally as effective against the asexual blood stages of the zoonotic P. knowlesi, suggesting that they could also be effective against the closely related P. vivax, another important human pathogen. The 2-anilinoquinazoline compounds were also potent against an array of P. falciparum parasites resistant to clinically available antimalarial compounds, although slightly less so than against the drug-sensitive 3D7 parasite line. The apicomplexan parasites Toxoplasma gondii, Babesia bovis, and Cryptosporidium parvum were less sensitive to the 2-anilinoquinazoline series with a 50% effective concentration generally in the low micromolar range, suggesting that the yet to be discovered target of these compounds is absent or highly divergent in non-Plasmodium parasites. The 2-anilinoquinazoline compounds act as rapidly as chloroquine in vitro and when tested in rodents displayed a half-life that contributed to the compound's capacity to clear P. falciparum blood stages in a humanized mouse model. At a dose of 50 mg/kg of body weight, adverse effects to the humanized mice were noted, and evaluation against a panel of experimental high-risk off targets indicated some potential off-target activity. Further optimization of the 2-anilinoquinazoline antimalarial class will concentrate on improving in vivo efficacy and addressing adverse risk.
Collapse
|
46
|
Resistance to Artemisinin Combination Therapies (ACTs): Do Not Forget the Partner Drug! Trop Med Infect Dis 2019; 4:tropicalmed4010026. [PMID: 30717149 PMCID: PMC6473515 DOI: 10.3390/tropicalmed4010026] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 01/30/2023] Open
Abstract
Artemisinin-based combination therapies (ACTs) have become the mainstay for malaria treatment in almost all malaria endemic settings. Artemisinin derivatives are highly potent and fast acting antimalarials; but they have a short half-life and need to be combined with partner drugs with a longer half-life to clear the remaining parasites after a standard 3-day ACT regimen. When introduced, ACTs were highly efficacious and contributed to the steep decrease of malaria over the last decades. However, parasites with decreased susceptibility to artemisinins have emerged in the Greater Mekong Subregion (GMS), followed by ACTs’ failure, due to both decreased susceptibility to artemisinin and partner drug resistance. Therefore, there is an urgent need to strengthen and expand current resistance surveillance systems beyond the GMS to track the emergence or spread of artemisinin resistance. Great attention has been paid to the spread of artemisinin resistance over the last five years, since molecular markers of decreased susceptibility to artemisinin in the GMS have been discovered. However, resistance to partner drugs is critical, as ACTs can still be effective against parasites with decreased susceptibility to artemisinins, when the latter are combined with a highly efficacious partner drug. This review outlines the different mechanisms of resistance and molecular markers associated with resistance to partner drugs for the currently used ACTs. Strategies to improve surveillance and potential solutions to extend the useful therapeutic lifespan of the currently available malaria medicines are proposed.
Collapse
|
47
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
48
|
Dziekan JM, Yu H, Chen D, Dai L, Wirjanata G, Larsson A, Prabhu N, Sobota RM, Bozdech Z, Nordlund P. Identifying purine nucleoside phosphorylase as the target of quinine using cellular thermal shift assay. Sci Transl Med 2019; 11. [DOI: 10.1126/scitranslmed.aau3174] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
A cellular thermal shift assay (CETSA) protocol identifies and resolves antimalarial drug targets in
P. falciparum
.
Collapse
Affiliation(s)
- Jerzy M. Dziekan
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Dan Chen
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Andreas Larsson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Radoslaw M. Sobota
- Institute of Molecular and Cell Biology, Functional Proteomics Laboratory, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm 17177, Sweden
- Institute of Molecular and Cell Biology, Functional Proteomics Laboratory, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| |
Collapse
|
49
|
Cowell A, Winzeler E. Exploration of the Plasmodium falciparum Resistome and Druggable Genome Reveals New Mechanisms of Drug Resistance and Antimalarial Targets. Microbiol Insights 2018; 11:1178636118808529. [PMID: 30505148 PMCID: PMC6259053 DOI: 10.1177/1178636118808529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 11/29/2022] Open
Abstract
Plasmodium parasites, the causative agent of malaria infections, rapidly evolve drug resistance and escape detection by the human immune response via the incredible mutability of its genome. Understanding the genetic mechanisms by which Plasmodium parasites develop antimalarial resistance is essential to understanding why most drugs fail in the clinic and designing the next generation of therapies. A systematic genomic analysis of 262 Plasmodium falciparum clones with stable in vitro resistance to 37 diverse compounds with potent antimalarial activity was undertaken with the main goal of identifying new drug targets. Despite several challenges inherent to this method of in vitro drug resistance generation followed by whole genome sequencing, the study was able to identify a likely drug target or resistance gene for every compound for which resistant parasites could be generated. Known and novel P falciparum resistance mediators were discovered along with several new promising antimalarial drug targets. Surprisingly, gene amplification events contributed to one-third of the drug resistance acquisition events. The study can serve as a model for drug discovery and resistance analyses in other similar microbial pathogens amenable to in vitro culture.
Collapse
Affiliation(s)
- Annie Cowell
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Elizabeth Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
50
|
Mechanisms of resistance to the partner drugs of artemisinin in the malaria parasite. Curr Opin Pharmacol 2018; 42:71-80. [PMID: 30142480 DOI: 10.1016/j.coph.2018.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/19/2018] [Accepted: 07/26/2018] [Indexed: 01/24/2023]
Abstract
The deployment of artemisinin-based combination therapies (ACTs) has been, and continues to be, integral to reducing the number of malaria cases and deaths. However, their efficacy is being increasingly jeopardized by the emergence and spread of parasites that are resistant (or partially resistant) to the artemisinin derivatives and to their partner drugs, with the efficacy of the latter being especially crucial for treatment success. A detailed understanding of the genetic determinants of resistance to the ACT partner drugs, and the mechanisms by which they mediate resistance, is required for the surveillance of molecular markers and to optimize the efficacy and lifespan of the partner drugs through resistance management strategies. We summarize new insights into the molecular basis of parasite resistance to the ACTs, such as recently-uncovered determinants of parasite susceptibility to the artemisinin derivatives, piperaquine, lumefantrine, and mefloquine, and outline the mechanisms through which polymorphisms in these determinants may be conferring resistance.
Collapse
|