1
|
Erhunse N, Kumari S, Anmol, Singh P, Omoregie ES, Singh AP, Sharma U, Sahal D. Annickia affinis (Exell) Versteegh & Sosef methanol stem bark extract, potent fractions and isolated Berberine alkaloid target both blood and liver stages of malaria parasites. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117269. [PMID: 37813288 DOI: 10.1016/j.jep.2023.117269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Having identified Annickia affinis as the most potent antiplasmodial plant constituent in a hepta-herbal Agbo-iba (HHA) formula commonly used to manage malaria in Benin city, Nigeria, we have in this study attempted to identify the specialized metabolites responsible for antiplasmodial activity of A. affinis through anti-blood stage malaria parasite activity guided isolation of potent molecules from its stem bark methanol extract. After that, phenotypic effects, including stage-specific kill kinetics, were investigated. Further, the crude extract, its potent fractions, and specialized metabolites were also tested against the liver-stage malaria parasite. MATERIALS AND METHODS A. affinis was subjected to molecular PCR-based analysis to confirm its identity. Thereafter, extraction of its stem bark with methanol was carried out. Alkaloid enriched fractions from this stem bark extract were obtained using the acid-base-solvent extraction method. These alkaloid-enriched fractions were subjected to various chromatographic techniques that led to the isolation of two protoberberine alkaloids identified as berberine and palmatine based on NMR and mass spectrometry analysis. The efficacy of crude extract, fractions and purified alkaloids was tested against the malaria parasite's blood and liver stages, respectively. RESULTS AND DISCUSSION Annickia affinis methanol extract, fractions, and the isolated protoberberine alkaloids showed excellent antiplasmodial activity with good selectivity against blood-stage malaria parasite. Thus, their IC50 against various strains of the parasite ranged from 0.95 to 18.65 μg/ml, while CC50 against Human embryonic kidney (HEK) and the human hepatoma (HUH-7) cell lines ranged between 10 and > 100 μg/ml. Interestingly, the crude extract and the alkaloid enriched fractions showed promising activity against the liver-stage malaria parasite. Between berberine and palmatine isolated from the potent fractions, only the former showed ∼100% and 90% inhibitions of liver stage parasite at 5 μg/ml and 1 μg/ml, respectively, while the latter showed no inhibition even at 20 μg/ml. CONCLUSION This study reports that the ethnomedicinal use of HHA to manage malaria can be attributed to the presence of promising antiplasmodial protoberberine alkaloids together with synergistic effects via either enhancement of bioavailability or improved pharmacokinetics by other phytoconstituent(s) coming from other HHA constituent plants. The protoberberine alkaloids isolated have been identified as fast-acting antiplasmodial agents, with activity against all erythrocytic stages of the malaria parasite. Further, A. affinis methanol stembark extract and the protoberberine alkaloid berberine isolated from it also displayed excellent activity (>90% inhibition at 1 μg/ml) against the liver-stage malaria parasite. A. affinis and HHA can thus be useful as both liver-stage prophylactic and blood-stage curative agents.
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, Nigeria
| | - Surekha Kumari
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anmol
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pooja Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhiz, 110067, India
| | | | - Agam Prasad Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhiz, 110067, India
| | - Upendra Sharma
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Dinkar Sahal
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.
| |
Collapse
|
2
|
Demarta-Gatsi C, Andenmatten N, Jiménez-Díaz MB, Gobeau N, Cherkaoui-Rabti MH, Fuchs A, Díaz P, Berja S, Sánchez R, Gómez H, Ruiz E, Sainz P, Salazar E, Gil-Merino R, Mendoza LM, Eguizabal C, Leroy D, Moehrle JJ, Tornesi B, Angulo-Barturen I. Predicting Optimal Antimalarial Drug Combinations from a Standardized Plasmodium falciparum Humanized Mouse Model. Antimicrob Agents Chemother 2023; 67:e0157422. [PMID: 37133382 PMCID: PMC10269072 DOI: 10.1128/aac.01574-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/29/2023] [Indexed: 05/04/2023] Open
Abstract
The development of new combinations of antimalarial drugs is urgently needed to prevent the spread of parasites resistant to drugs in clinical use and contribute to the control and eradication of malaria. In this work, we evaluated a standardized humanized mouse model of erythrocyte asexual stages of Plasmodium falciparum (PfalcHuMouse) for the selection of optimal drug combinations. First, we showed that the replication of P. falciparum was robust and highly reproducible in the PfalcHuMouse model by retrospective analysis of historical data. Second, we compared the relative value of parasite clearance from blood, parasite regrowth after suboptimal treatment (recrudescence), and cure as variables of therapeutic response to measure the contributions of partner drugs to combinations in vivo. To address the comparison, we first formalized and validated the day of recrudescence (DoR) as a new variable and found that there was a log-linear relationship with the number of viable parasites per mouse. Then, using historical data on monotherapy and two small cohorts of PfalcHuMice evaluated with ferroquine plus artefenomel or piperaquine plus artefenomel, we found that only measurements of parasite killing (i.e., cure of mice) as a function of drug exposure in blood allowed direct estimation of the individual drug contribution to efficacy by using multivariate statistical modeling and intuitive graphic displays. Overall, the analysis of parasite killing in the PfalcHuMouse model is a unique and robust experimental in vivo tool to inform the selection of optimal combinations by pharmacometric pharmacokinetic and pharmacodynamic (PK/PD) modeling.
Collapse
Affiliation(s)
| | | | | | | | | | - Aline Fuchs
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Pablo Díaz
- The Art of Discovery, Derio, Basque Country, Spain
| | - Sandra Berja
- The Art of Discovery, Derio, Basque Country, Spain
| | | | - Hazel Gómez
- The Art of Discovery, Derio, Basque Country, Spain
| | | | - Paula Sainz
- The Art of Discovery, Derio, Basque Country, Spain
| | | | | | | | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
- Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Bizkaia, Spain
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | | | | | | |
Collapse
|
3
|
Umumararungu T, Nkuranga JB, Habarurema G, Nyandwi JB, Mukazayire MJ, Mukiza J, Muganga R, Hahirwa I, Mpenda M, Katembezi AN, Olawode EO, Kayitare E, Kayumba PC. Recent developments in antimalarial drug discovery. Bioorg Med Chem 2023; 88-89:117339. [PMID: 37236020 DOI: 10.1016/j.bmc.2023.117339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Although malaria remains a big burden to many countries that it threatens their socio-economic stability, particularly in the countries where malaria is endemic, there have been great efforts to eradicate this disease with both successes and failures. For example, there has been a great improvement in malaria prevention and treatment methods with a net reduction in infection and mortality rates. However, the disease remains a global threat in terms of the number of people affected because it is one of the infectious diseases that has the highest prevalence rate, especially in Africa where the deadly Plasmodium falciparum is still widely spread. Methods to fight malaria are being diversified, including the use of mosquito nets, the target candidate profiles (TCPs) and target product profiles (TPPs) of medicine for malarial venture (MMV) strategy, the search for newer and potent drugs that could reverse chloroquine resistance, and the use of adjuvants such as rosiglitazone and sevuparin. Although these adjuvants have no antiplasmodial activity, they can help to alleviate the effects which result from plasmodium invasion such as cytoadherence. The list of new antimalarial drugs under development is long, including the out of ordinary new drugs MMV048, CDRI-97/78 and INE963 from South Africa, India and Novartis, respectively.
Collapse
Affiliation(s)
- Théoneste Umumararungu
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda.
| | - Jean Bosco Nkuranga
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Gratien Habarurema
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Jean Baptiste Nyandwi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Marie Jeanne Mukazayire
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Janvier Mukiza
- Department of Mathematical Science and Physical Education, School of Education, College of Education, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Raymond Muganga
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Innocent Hahirwa
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Matabishi Mpenda
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Alain Nyirimigabo Katembezi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Emmanuel Oladayo Olawode
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N Miami Ave #1, Miami, FL 33169, USA
| | - Egide Kayitare
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Pierre Claver Kayumba
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| |
Collapse
|
4
|
Azmi WA, Rizki AFM, Djuardi Y, Artika IM, Siregar JE. Molecular insights into artemisinin resistance in Plasmodium falciparum: An updated review. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105460. [PMID: 37269964 DOI: 10.1016/j.meegid.2023.105460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Malaria still poses a major burden on human health around the world, especially in endemic areas. Plasmodium resistance to several antimalarial drugs has been one of the major hindrances in control of malaria. Thus, the World Health Organization recommended artemisinin-based combination therapy (ACT) as a front-line treatment for malaria. The emergence of parasites resistant to artemisinin, along with resistant to ACT partner drugs, has led to ACT treatment failure. The artemisinin resistance is mostly related to the mutations in the propeller domain of the kelch13 (k13) gene that encodes protein Kelch13 (K13). The K13 protein has an important role in parasite reaction to oxidative stress. The most widely spread mutation in K13, with the highest degree of resistance, is a C580Y mutation. Other mutations, which are already identified as markers of artemisinin resistance, are R539T, I543T, and Y493H. The objective of this review is to provide current molecular insights into artemisinin resistance in Plasmodium falciparum. The trending use of artemisinin beyond its antimalarial effect is described. Immediate challenges and future research directions are discussed. Better understanding of the molecular mechanisms underlying artemisinin resistance will accelerate implementation of scientific findings to solve problems with malarial infection.
Collapse
Affiliation(s)
- Wihda Aisarul Azmi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Andita Fitri Mutiara Rizki
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - I Made Artika
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Bogor 16680, Indonesia
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia.
| |
Collapse
|
5
|
Muema JM, Mutunga JM, Obonyo MA, Getahun MN, Mwakubambanya RS, Akala HM, Cheruiyot AC, Yeda RA, Juma DW, Andagalu B, Johnson JL, Roth AL, Bargul JL. Isoliensinine from Cissampelos pariera rhizomes exhibits potential gametocytocidal and anti-malarial activities against Plasmodium falciparum clinical isolates. Malar J 2023; 22:161. [PMID: 37208735 DOI: 10.1186/s12936-023-04590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND The unmet demand for effective malaria transmission-blocking agents targeting the transmissible stages of Plasmodium necessitates intensive discovery efforts. In this study, a bioactive bisbenzylisoquinoline (BBIQ), isoliensinine, from Cissampelos pariera (Menispermaceae) rhizomes was identified and characterized for its anti-malarial activity. METHODS Malaria SYBR Green I fluorescence assay was performed to evaluate the in vitro antimalarial activity against D6, Dd2, and F32-ART5 clones, and immediate ex vivo (IEV) susceptibility for 10 freshly collected P. falciparum isolates. To determine the speed- and stage-of-action of isoliensinine, an IC50 speed assay and morphological analyses were performed using synchronized Dd2 asexuals. Gametocytocidal activity against two culture-adapted gametocyte-producing clinical isolates was determined using microscopy readouts, with possible molecular targets and their binding affinities deduced in silico. RESULTS Isoliensinine displayed a potent in vitro gametocytocidal activity at mean IC50gam values ranging between 0.41 and 0.69 µM for Plasmodium falciparum clinical isolates. The BBIQ compound also inhibited asexual replication at mean IC50Asexual of 2.17 µM, 2.22 µM, and 2.39 µM for D6, Dd2 and F32-ART5 respectively, targeting the late-trophozoite to schizont transition. Further characterization demonstrated a considerable immediate ex vivo potency against human clinical isolates at a geometric mean IC50IEV = 1.433 µM (95% CI 0.917-2.242). In silico analyses postulated a probable anti-malarial mechanism of action by high binding affinities for four mitotic division protein kinases; Pfnek1, Pfmap2, Pfclk1, and Pfclk4. Additionally, isoliensinine was predicted to possess an optimal pharmacokinetics profile and drug-likeness properties. CONCLUSION These findings highlight considerable grounds for further exploration of isoliensinine as an amenable scaffold for malaria transmission-blocking chemistry and target validation.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya.
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya.
| | - James M Mutunga
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
- Department of Biological Sciences, School of Pure and Applied Sciences, Mount Kenya University, Thika, Kenya
- School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, Egerton, Kenya
| | - Merid N Getahun
- International Centre of Insect Physiology and Ecology (Icipe), Nairobi, Kenya
| | | | - Hoseah M Akala
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Agnes C Cheruiyot
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Redemptah A Yeda
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Dennis W Juma
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Ben Andagalu
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Jaree L Johnson
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Amanda L Roth
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya.
- International Centre of Insect Physiology and Ecology (Icipe), Nairobi, Kenya.
| |
Collapse
|
6
|
Recent approaches in the drug research and development of novel antimalarial drugs with new targets. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:1-27. [PMID: 36692468 DOI: 10.2478/acph-2023-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 01/25/2023]
Abstract
Malaria is a serious worldwide medical issue that results in substantial annual death and morbidity. The availability of treatment alternatives is limited, and the rise of resistant parasite types has posed a significant challenge to malaria treatment. To prevent a public health disaster, novel antimalarial agents with single-dosage therapies, extensive curative capability, and new mechanisms are urgently needed. There are several approaches to developing antimalarial drugs, ranging from alterations of current drugs to the creation of new compounds with specific targeting abilities. The availability of multiple genomic techniques, as well as recent advancements in parasite biology, provides a varied collection of possible targets for the development of novel treatments. A number of promising pharmacological interference targets have been uncovered in modern times. As a result, our review concentrates on the most current scientific and technical progress in the innovation of new antimalarial medications. The protein kinases, choline transport inhibitors, dihydroorotate dehydrogenase inhibitors, isoprenoid biosynthesis inhibitors, and enzymes involved in the metabolism of lipids and replication of deoxyribonucleic acid, are among the most fascinating antimalarial target proteins presently being investigated. The new cellular targets and drugs which can inhibit malaria and their development techniques are summarised in this study.
Collapse
|
7
|
Rathod GK, Jain M, Sharma KK, Das S, Basak A, Jain R. New structural classes of antimalarials. Eur J Med Chem 2022; 242:114653. [PMID: 35985254 DOI: 10.1016/j.ejmech.2022.114653] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/19/2022]
Abstract
Malaria remains a major vector borne disease claiming millions of lives worldwide due to infections caused by Plasmodium sp. Discovery and development of antimalarial drugs have previously been dominated majorly by single drug therapy. The malaria parasite has developed resistance against first line and second line antimalarial drugs used in the single drug therapy. This has drawn attention to find ways to alleviate the disease burden supplanted by combination therapy with multiple drugs to overcome drug resistance. Emergence of resistant strains even against the combination therapy has now mandated the revision of the current antimalarial pharmacotherapy. Research efforts of the past decade led to the discovery and identification of several new structural classes of antimalarial agents with improved biological attributes over the older ones. The following is a comprehensive review, addressed to the new structural classes of heterocyclic and natural compounds that have been identified during the last decade as antimalarial agents. Some of the classes included herein contain one or more pharmacophores amalgamated into a single bioactive scaffold as antimalarial agents, which act upon the conventional and novel targets.
Collapse
Affiliation(s)
- Gajanan K Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Meenakshi Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Krishna K Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Samarpita Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Ahana Basak
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India.
| |
Collapse
|
8
|
Gupta Y, Sharma N, Singh S, Romero JG, Rajendran V, Mogire RM, Kashif M, Beach J, Jeske W, Poonam, Ogutu BR, Kanzok SM, Akala HM, Legac J, Rosenthal PJ, Rademacher DJ, Durvasula R, Singh AP, Rathi B, Kempaiah P. The Multistage Antimalarial Compound Calxinin Perturbates P. falciparum Ca 2+ Homeostasis by Targeting a Unique Ion Channel. Pharmaceutics 2022; 14:1371. [PMID: 35890267 PMCID: PMC9319510 DOI: 10.3390/pharmaceutics14071371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 12/22/2022] Open
Abstract
Malaria elimination urgently needs novel antimalarial therapies that transcend resistance, toxicity, and high costs. Our multicentric international collaborative team focuses on developing multistage antimalarials that exhibit novel mechanisms of action. Here, we describe the design, synthesis, and evaluation of a novel multistage antimalarial compound, 'Calxinin'. A compound that consists of hydroxyethylamine (HEA) and trifluoromethyl-benzyl-piperazine. Calxinin exhibits potent inhibitory activity in the nanomolar range against the asexual blood stages of drug-sensitive (3D7), multidrug-resistant (Dd2), artemisinin-resistant (IPC4912), and fresh Kenyan field isolated Plasmodium falciparum strains. Calxinin treatment resulted in diminished maturation of parasite sexual precursor cells (gametocytes) accompanied by distorted parasite morphology. Further, in vitro liver-stage testing with a mouse model showed reduced parasite load at an IC50 of 79 nM. A single dose (10 mg/kg) of Calxinin resulted in a 30% reduction in parasitemia in mice infected with a chloroquine-resistant strain of the rodent parasite P. berghei. The ex vivo ookinete inhibitory concentration within mosquito gut IC50 was 150 nM. Cellular in vitro toxicity assays in the primary and immortalized human cell lines did not show cytotoxicity. A computational protein target identification pipeline identified a putative P. falciparum membrane protein (Pf3D7_1313500) involved in parasite calcium (Ca2+) homeostasis as a potential Calxinin target. This highly conserved protein is related to the family of transient receptor potential cation channels (TRP-ML). Target validation experiments showed that exposure of parasitized RBCs (pRBCs) to Calxinin induces a rapid release of intracellular Ca2+ from pRBCs; leaving de-calcinated parasites trapped in RBCs. Overall, we demonstrated that Calxinin is a promising antimalarial lead compound with a novel mechanism of action and with potential therapeutic, prophylactic, and transmission-blocking properties against parasites resistant to current antimalarials.
Collapse
Affiliation(s)
- Yash Gupta
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| | - Neha Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
| | - Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
| | - Jesus G. Romero
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
- School of Biology, Institute of Experimental Biology, Central University of Venezuela, Caracas 1040, Venezuela
| | - Vinoth Rajendran
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | - Reagan M. Mogire
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Mohammad Kashif
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi 110067, India; (M.K.); (A.P.S.)
| | - Jordan Beach
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
| | - Walter Jeske
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi, New Delhi 110021, India;
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi 110007, India
| | - Bernhards R. Ogutu
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Stefan M. Kanzok
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA;
| | - Hoseah M. Akala
- Centre Clinical Research, Kenya Medical Research Institute, Nairobi P.O. Box 54840-00200, Kenya; (R.M.M.); (B.R.O.); (H.M.A.)
| | - Jennifer Legac
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (J.L.); (P.J.R.)
| | - Philip J. Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (J.L.); (P.J.R.)
| | - David J. Rademacher
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60660, USA; (J.G.R.); (J.B.); (W.J.); (D.J.R.)
- Core Imaging Facility and Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Ravi Durvasula
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi 110067, India; (M.K.); (A.P.S.)
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, New Delhi 110021, India; (N.S.); (S.S.)
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi 110007, India
| | - Prakasha Kempaiah
- Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (Y.G.); (R.D.)
| |
Collapse
|
9
|
Bernard MM, Mohanty A, Rajendran V. Title: A Comprehensive Review on Classifying Fast-acting and Slow-acting Antimalarial Agents Based on Time of Action and Target Organelle of Plasmodium sp. Pathog Dis 2022; 80:6589403. [PMID: 35588061 DOI: 10.1093/femspd/ftac015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/20/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical resistance towards malarial parasites has rendered many antimalarials ineffective, likely due to a lack of understanding of time of action and stage specificity of all life stages. Therefore, to tackle this problem a more incisive comprehensive analysis of the fast and slow-acting profile of antimalarial agents relating to parasite time-kill kinetics and the target organelle on the progression of blood-stage parasites was carried out. It is evident from numerous findings that drugs targeting food vacuole, nuclear components, and endoplasmic reticulum mainly exhibit a fast-killing phenotype within 24h affecting first-cycle activity. Whereas drugs targeting mitochondria, apicoplast, microtubules, parasite invasion and egress exhibit a largely slow-killing phenotype within 96-120h, affecting second-cycle activity with few exemptions as moderately fast-killing. It is essential to understand the susceptibility of drugs on rings, trophozoites, schizonts, merozoites, and the appearance of organelle at each stage of 48h intraerythrocytic parasite cycle. Therefore, these parameters may facilitate the paradigm for understanding the timing of antimalarials action in deciphering its precise mechanism linked with time. Thus, classifying drugs based on the time of killing may promote designing new combination regimens against varied strains of P. falciparum and evaluating potential clinical resistance.
Collapse
Affiliation(s)
- Monika Marie Bernard
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Abhinab Mohanty
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Vinoth Rajendran
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| |
Collapse
|
10
|
Marfurt J, Wirjanata G, Prayoga P, Chalfein F, Leonardo L, Sebayang BF, Apriyanti D, Sihombing MAEM, Trianty L, Suwanarusk R, Brockman A, Piera KA, Luo I, Rumaseb A, MacHunter B, Auburn S, Anstey NM, Kenangalem E, Noviyanti R, Russell B, Poespoprodjo JR, Price RN. Longitudinal ex vivo and molecular trends of chloroquine and piperaquine activity against Plasmodium falciparum and P. vivax before and after introduction of artemisinin-based combination therapy in Papua, Indonesia. Int J Parasitol Drugs Drug Resist 2021; 17:46-56. [PMID: 34193398 PMCID: PMC8358472 DOI: 10.1016/j.ijpddr.2021.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 01/13/2023]
Abstract
Drug resistant Plasmodium parasites are a major threat to malaria control and elimination. After reports of high levels of multidrug resistant P. falciparum and P. vivax in Indonesia, in 2005, the national first-line treatment policy for uncomplicated malaria was changed in March 2006, to dihydroartemisinin-piperaquine against all species. This study assessed the temporal trends in ex vivo drug susceptibility to chloroquine (CQ) and piperaquine (PIP) for both P. falciparum and P. vivax clinical isolates collected between 2004 and 2018, by using schizont maturation assays, and genotyped a subset of isolates for known and putative molecular markers of CQ and PIP resistance by using Sanger and next generation whole genome sequencing. The median CQ IC50 values varied significantly between years in both Plasmodium species, but there was no significant trend over time. In contrast, there was a significant trend for increasing PIP IC50s in both Plasmodium species from 2010 onwards. Whereas the South American CQ resistant 7G8 pfcrt SVMNT isoform has been fixed since 2005 in the study area, the pfmdr1 86Y allele frequencies decreased and became fixed at the wild-type allele in 2015. In P. vivax isolates, putative markers of CQ resistance (no pvcrt-o AAG (K10) insertion and pvmdr1 Y967F and F1076L) were fixed at the mutant alleles since 2005. None of the putative PIP resistance markers were detected in P. falciparum. The ex vivo drug susceptibility and molecular analysis of CQ and PIP efficacy for P. falciparum and P. vivax after 12 years of intense drug pressure with DHP suggests that whilst the degree of CQ resistance appears to have been sustained, there has been a slight decline in PIP susceptibility, although this does not appear to have reached clinically significant levels. The observed decreasing trend in ex vivo PIP susceptibility highlights the importance of ongoing surveillance.
Collapse
Affiliation(s)
- Jutta Marfurt
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia.
| | - Grennady Wirjanata
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Pak Prayoga
- Papuan Health and Community Development Foundation (PHCDF), Jl. Caritas No. 1, 99961, Timika, Papua, Indonesia
| | - Ferryanto Chalfein
- Papuan Health and Community Development Foundation (PHCDF), Jl. Caritas No. 1, 99961, Timika, Papua, Indonesia
| | - Leo Leonardo
- Papuan Health and Community Development Foundation (PHCDF), Jl. Caritas No. 1, 99961, Timika, Papua, Indonesia
| | - Boni F Sebayang
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, 10430, Jakarta, Indonesia
| | - Dwi Apriyanti
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, 10430, Jakarta, Indonesia
| | - Maic A E M Sihombing
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, 10430, Jakarta, Indonesia
| | - Leily Trianty
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, 10430, Jakarta, Indonesia
| | - Rossarin Suwanarusk
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Alan Brockman
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Kim A Piera
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Irene Luo
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Angela Rumaseb
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Barbara MacHunter
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Sarah Auburn
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Enny Kenangalem
- Papuan Health and Community Development Foundation (PHCDF), Jl. Caritas No. 1, 99961, Timika, Papua, Indonesia; District Health Authority, Timika, Papua, Indonesia
| | - Rintis Noviyanti
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, 10430, Jakarta, Indonesia
| | - Bruce Russell
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia
| | - Jeanne R Poespoprodjo
- Papuan Health and Community Development Foundation (PHCDF), Jl. Caritas No. 1, 99961, Timika, Papua, Indonesia; Paediatric Research Office, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina, NT, 0811, Darwin, Australia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| |
Collapse
|
11
|
Fernandes VDS, da Rosa R, Zimmermann LA, Rogério KR, Kümmerle AE, Bernardes LSC, Graebin CS. Antiprotozoal agents: How have they changed over a decade? Arch Pharm (Weinheim) 2021; 355:e2100338. [PMID: 34661935 DOI: 10.1002/ardp.202100338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/21/2022]
Abstract
Neglected tropical diseases are a diverse group of communicable diseases that are endemic in low- or low-to-middle-income countries located in tropical and subtropical zones. The number and availability of drugs for treating these diseases are low, the administration route is inconvenient in some cases, and most of them have safety, efficacy, or adverse/toxic reaction issues. The need for developing new drugs to deal with these issues is clear, but one of the most drastic consequences of this negligence is the lack of interest in the research and development of new therapeutic options among major pharmaceutical companies. Positive changes have been achieved over the last few years, although the overall situation remains alarming. After more than one decade since the original work reviewing antiprotozoal agents came to light, now it is time to question ourselves: How has the scenario for the treatment of protozoal diseases such as malaria, leishmaniasis, human African trypanosomiasis, and American trypanosomiasis changed? This review covers the last decade in terms of the drugs currently available for the treatment of these diseases as well as the clinical candidates being currently investigated.
Collapse
Affiliation(s)
- Vitória de Souza Fernandes
- Department of Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry Laboratory, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Rafael da Rosa
- Department of Organic Chemistry, Medicinal Chemistry and Molecular Diversity Laboratory, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Lara A Zimmermann
- Department of Organic Chemistry, Medicinal Chemistry and Molecular Diversity Laboratory, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Kamilla R Rogério
- Department of Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry Laboratory, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Arthur E Kümmerle
- Department of Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry Laboratory, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Lilian S C Bernardes
- Department of Organic Chemistry, Medicinal Chemistry and Molecular Diversity Laboratory, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Cedric S Graebin
- Department of Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry Laboratory, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
12
|
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: 9] [Impact Index Per Article: 3.0] [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
|
13
|
Madhav H, Hoda N. An insight into the recent development of the clinical candidates for the treatment of malaria and their target proteins. Eur J Med Chem 2020; 210:112955. [PMID: 33131885 DOI: 10.1016/j.ejmech.2020.112955] [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: 07/03/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 01/18/2023]
Abstract
Malaria is an endemic disease, prevalent in tropical and subtropical regions which cost half of million deaths annually. The eradication of malaria is one of the global health priority nevertheless, current therapeutic efforts seem to be insufficient due to the emergence of drug resistance towards most of the available drugs, even first-line treatment ACT, unavailability of the vaccine, and lack of drugs with a new mechanism of action. Intensification of antimalarial research in recent years has resulted into the development of single dose multistage therapeutic agents which has advantage of overcoming the antimalarial drug resistance. The present review explored the current progress in the development of new promising antimalarials against prominent target proteins that have the potential to be a clinical candidate. Here, we also reviewed different aspects of drug resistance and highlighted new drug candidates that are currently in a clinical trial or clinical development, along with a few other molecules with excellent antimalarial activity overs ACTs. The summarized scientific value of previous approaches and structural features of antimalarials related to the activity are highlighted that will be helpful for the development of next-generation antimalarials.
Collapse
Affiliation(s)
- Hari Madhav
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| |
Collapse
|
14
|
Burgert L, Rottmann M, Wittlin S, Gobeau N, Krause A, Dingemanse J, Möhrle JJ, Penny MA. Ensemble modeling highlights importance of understanding parasite-host behavior in preclinical antimalarial drug development. Sci Rep 2020; 10:4410. [PMID: 32157151 PMCID: PMC7064600 DOI: 10.1038/s41598-020-61304-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/20/2020] [Indexed: 11/23/2022] Open
Abstract
Emerging drug resistance and high-attrition rates in early and late stage drug development necessitate accelerated development of antimalarial compounds. However, systematic and meaningful translation of drug efficacy and host-parasite dynamics between preclinical testing stages is missing. We developed an ensemble of mathematical within-host parasite growth and antimalarial action models, fitted to extensive data from four antimalarials with different modes of action, to assess host-parasite interactions in two preclinical drug testing systems of murine parasite P. berghei in mice, and human parasite P. falciparum in immune-deficient mice. We find properties of the host-parasite system, namely resource availability, parasite maturation and virulence, drive P. berghei dynamics and drug efficacy, whereas experimental constraints primarily influence P. falciparum infection and drug efficacy. Furthermore, uninvestigated parasite behavior such as dormancy influences parasite recrudescence following non-curative treatment and requires further investigation. Taken together, host-parasite interactions should be considered for meaningful translation of pharmacodynamic properties between murine systems and for predicting human efficacious treatment.
Collapse
Affiliation(s)
- Lydia Burgert
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Andreas Krause
- Idorsia Pharmaceuticals Ltd, Clinical Pharmacology, Allschwil, Switzerland
| | - Jasper Dingemanse
- Idorsia Pharmaceuticals Ltd, Clinical Pharmacology, Allschwil, Switzerland
| | - Jörg J Möhrle
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Medicines for Malaria Venture, Geneva, Switzerland
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Abstract
The scientific community worldwide has realized that malaria elimination will not be possible without development of safe and effective transmission-blocking interventions. Primaquine, the only WHO recommended transmission-blocking drug, is not extensively utilized because of the toxicity issues in G6PD deficient individuals. Therefore, there is an urgent need to develop novel therapeutic interventions that can target malaria parasites and effectively block transmission. But at first, it is imperative to unravel the existing portfolio of transmission-blocking drugs. This review highlights transmission-blocking potential of current antimalarial drugs and drugs that are in various stages of clinical development. The collective analysis of the relationships between the structure and the activity of transmission-blocking drugs is expected to help in the design of new transmission-blocking antimalarials.
Collapse
|
16
|
Marin-Mogollon C, Salman AM, Koolen KMJ, Bolscher JM, van Pul FJA, Miyazaki S, Imai T, Othman AS, Ramesar J, van Gemert GJ, Kroeze H, Chevalley-Maurel S, Franke-Fayard B, Sauerwein RW, Hill AVS, Dechering KJ, Janse CJ, Khan SM. A P. falciparum NF54 Reporter Line Expressing mCherry-Luciferase in Gametocytes, Sporozoites, and Liver-Stages. Front Cell Infect Microbiol 2019; 9:96. [PMID: 31058097 PMCID: PMC6477837 DOI: 10.3389/fcimb.2019.00096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/22/2019] [Indexed: 12/16/2022] Open
Abstract
Transgenic malaria parasites expressing fluorescent and bioluminescent proteins are valuable tools to interrogate malaria-parasite biology and to evaluate drugs and vaccines. Using CRISPR/Cas9 methodology a transgenic Plasmodium falciparum (Pf) NF54 line was generated that expresses a fusion of mCherry and luciferase genes under the control of the Pf etramp10.3 gene promoter (line mCherry-luc@etramp10.3). Pf etramp10.3 is related to rodent Plasmodium uis4 and the uis4 promoter has been used to drive high transgene expression in rodent parasite sporozoites and liver-stages. We examined transgene expression throughout the complete life cycle and compared this expression to transgenic lines expressing mCherry-luciferase and GFP-luciferase under control of the constitutive gapdh and eef1a promoters. The mCherry-luc@etramp10.3 parasites express mCherry in gametocytes, sporozoites, and liver-stages. While no mCherry signal was detected in asexual blood-stage parasites above background levels, luciferase expression was detected in asexual blood-stages, as well as in gametocytes, sporozoites and liver-stages, with the highest levels of reporter expression detected in stage III-V gametocytes and in sporozoites. The expression of mCherry and luciferase in gametocytes and sporozoites makes this transgenic parasite line suitable to use in in vitro assays that examine the effect of transmission blocking inhibitors and to analyse gametocyte and sporozoite biology.
Collapse
Affiliation(s)
| | - Ahmed M Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Fiona J A van Pul
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ahmad Syibli Othman
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Robert W Sauerwein
- TropIQ Health Sciences, Nijmegen, Netherlands.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Tse EG, Korsik M, Todd MH. The past, present and future of anti-malarial medicines. Malar J 2019; 18:93. [PMID: 30902052 PMCID: PMC6431062 DOI: 10.1186/s12936-019-2724-z] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/12/2019] [Indexed: 11/10/2022] Open
Abstract
Great progress has been made in recent years to reduce the high level of suffering caused by malaria worldwide. Notably, the use of insecticide-treated mosquito nets for malaria prevention and the use of artemisinin-based combination therapy (ACT) for malaria treatment have made a significant impact. Nevertheless, the development of resistance to the past and present anti-malarial drugs highlights the need for continued research to stay one step ahead. New drugs are needed, particularly those with new mechanisms of action. Here the range of anti-malarial medicines developed over the years are reviewed, beginning with the discovery of quinine in the early 1800s, through to modern day ACT and the recently-approved tafenoquine. A number of new potential anti-malarial drugs currently in development are outlined, along with a description of the hit to lead campaign from which it originated. Finally, promising novel mechanisms of action for these and future anti-malarial medicines are outlined.
Collapse
Affiliation(s)
- Edwin G Tse
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marat Korsik
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Matthew H Todd
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia. .,School of Pharmacy, University College London, London, WC1N 1AX, United Kingdom.
| |
Collapse
|
18
|
Hooft van Huijsduijnen R, Wells T, Tanner M, Wittlin S. Two successful decades of Swiss collaborations to develop new anti-malarials. Malar J 2019; 18:94. [PMID: 30902051 PMCID: PMC6431002 DOI: 10.1186/s12936-019-2728-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/14/2019] [Indexed: 04/24/2023] Open
Abstract
Over the last two decades there has been a renaissance in the pipeline of new drugs targeting malaria, with the launch of new products that help save the lives of children throughout the world. In addition, there is a wealth of new molecules both entering and progressing through clinical development. These bring hope for a new generation of simpler and more effective cures that could overcome the emerging threat of drug resistance. In addition, there is hope that some of these medicines will have prophylactic activity and can be used to protect vulnerable populations, given the absence of a highly effective vaccine. Switzerland has played a key role in the development of these medicines. First, the country has a long history of understanding the biology of parasites and the pharmacology of drug responses through the leadership of the Swiss Tropical and Public Health Institute in Basel. Second, the highly successful Swiss pharmaceutical industry brings, beyond excellence, a strong interest in neglected diseases, building on work at Hoffmann-La Roche in the last century and with more recent products from Novartis and other Swiss companies. Third, the emergence of product-development-partnerships, in this case led by the Medicines for Malaria Venture, based in Geneva, has helped to catalyze the development of new medicines and bring the community together within Switzerland and beyond. Finally, this progress would not have been possible without the engagement of the Swiss people and the support of the federal government through the Swiss Agency for Development and Cooperation (SDC), the State Secretariat of Education, Research and Innovation (SERI) and the Swiss Republic and Canton of Geneva.
Collapse
Affiliation(s)
| | - Timothy Wells
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva, Switzerland.
| | - Marcel Tanner
- Swiss Tropical & Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Sergio Wittlin
- Swiss Tropical & Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Singh S, Rajendran V, He J, Singh AK, Achieng AO, Vandana, Pant A, Nasamu AS, Pandit M, Singh J, Quadiri A, Gupta N, Poonam, Ghosh PC, Singh BK, Narayanan L, Kempaiah P, Chandra R, Dunn BM, Pandey KC, Goldberg DE, Singh AP, Rathi B. Fast-Acting Small Molecules Targeting Malarial Aspartyl Proteases, Plasmepsins, Inhibit Malaria Infection at Multiple Life Stages. ACS Infect Dis 2019; 5:184-198. [PMID: 30554511 DOI: 10.1021/acsinfecdis.8b00197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The eradication of malaria remains challenging due to the complex life cycle of Plasmodium and the rapid emergence of drug-resistant forms of Plasmodium falciparum and Plasmodium vivax. New, effective, and inexpensive antimalarials against multiple life stages of the parasite are urgently needed to combat the spread of malaria. Here, we synthesized a set of novel hydroxyethylamines and investigated their activities in vitro and in vivo. All of the compounds tested had an inhibitory effect on the blood stage of P. falciparum at submicromolar concentrations, with the best showing 50% inhibitory concentrations (IC50) of around 500 nM against drug-resistant P. falciparum parasites. These compounds showed inhibitory actions against plasmepsins, a family of malarial aspartyl proteases, and exhibited a marked killing effect on blood stage Plasmodium. In chloroquine-resistant Plasmodium berghei and P. berghei ANKA infected mouse models, treating mice with both compounds led to a significant decrease in blood parasite load. Importantly, two of the compounds displayed an inhibitory effect on the gametocyte stages (III-V) of P. falciparum in culture and the liver-stage infection of P. berghei both in in vitro and in vivo. Altogether, our findings suggest that fast-acting hydroxyethylamine-phthalimide analogs targeting multiple life stages of the parasite could be a valuable chemical lead for the development of novel antimalarial drugs.
Collapse
Affiliation(s)
- Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | - Jiang He
- Institute for Medical Engineering and Science, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Amit K. Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Angela O. Achieng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
| | - Vandana
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Akansha Pant
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Mansi Pandit
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Jyoti Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Afshana Quadiri
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Nikesh Gupta
- Special Centre for Nanosciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi North Campus, Delhi 110007, India
| | - Prahlad C. Ghosh
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | | | - Latha Narayanan
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Prakasha Kempaiah
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
- Department of Medicine, Loyola University Stritch School of Medicine, 2160 South First Avenue, Chicago, Illinois 60153, United States
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Ben M. Dunn
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, Florida 32610, United States
| | - Kailash C. Pandey
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
- Department of Biochemistry, National Institute for Research in Environmental Health, ICMR, Bhopal 462001, India
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
20
|
Kumari A, Karnatak M, Singh D, Shankar R, Jat JL, Sharma S, Yadav D, Shrivastava R, Verma VP. Current scenario of artemisinin and its analogues for antimalarial activity. Eur J Med Chem 2018; 163:804-829. [PMID: 30579122 DOI: 10.1016/j.ejmech.2018.12.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 11/17/2022]
Abstract
Human malaria, one of the most striking, reemerging infectious diseases, is caused by several types of Plasmodium parasites. Whilst advances have been made in lowering the numbers of cases and deaths, it is clear that a strategy based solely on disease control year on year, without reducing transmission and ultimately eradicating the parasite, is unsustainable. Natural products have served as a template for the design and development of antimalarial drugs currently in the clinic or in the development phase. Artemisinin combine potent, rapid antimalarial activity with a wide therapeutic index and an absence of clinically important resistance. The alkylating ability of artemisinin and its semi-synthetic analogues toward heme related to their antimalarial efficacy are underlined. Although impressive results have already been achieved in malaria research, more systematization and concentration of efforts are required if real breakthroughs are to be made. This review will concisely cover the clinical, preclinical antimalarial and current updates in artemisinin based antimalarial drugs. Diverse classes of semi-synthetic analogs of artemisinin reported in the last decade have also been extensively studied. The experience gained in this respect is discussed.
Collapse
Affiliation(s)
- Akriti Kumari
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India
| | - Manvika Karnatak
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India
| | - Davinder Singh
- Bio-Organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, Jammu and Kashmir, India
| | - Ravi Shankar
- Bio-Organic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, Jammu and Kashmir, India
| | - Jawahar L Jat
- Department of Applied Chemistry, BabaSaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar Raebareli Road, Lucknow, 226025, India
| | - Siddharth Sharma
- Department of Chemistry, Mohanlal Sukhadia University, Udaipur, 313001, India
| | - Dinesh Yadav
- Department of Chemistry, Mohanlal Sukhadia University, Udaipur, 313001, India
| | - Rahul Shrivastava
- Department of Chemistry, Manipal University Jaipur, Jaipur, 303007, India
| | - Ved Prakash Verma
- Department of Chemistry, Banasthali University, Banasthali Newai, 304022, Rajasthan, India.
| |
Collapse
|
21
|
Investigating Sulfoxide-to-Sulfone Conversion as a Prodrug Strategy for a Phosphatidylinositol 4-Kinase Inhibitor in a Humanized Mouse Model of Malaria. Antimicrob Agents Chemother 2018; 62:AAC.00261-18. [PMID: 30249687 DOI: 10.1128/aac.00261-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/07/2018] [Indexed: 11/20/2022] Open
Abstract
The in vivo antimalarial efficacies of two phosphatidylinositol 4-kinase (PI4K) inhibitors, a 3,5-diaryl-2-aminopyrazine sulfoxide and its corresponding sulfone metabolite, were evaluated in the NOD-scid IL2Rγnull (NSG) murine malaria disease model of Plasmodium falciparum infection. We hypothesized that the sulfoxide would serve as a more soluble prodrug for the sulfone, which would lead to improved drug exposure with oral dosing. Both compounds had similar efficacy (90% effective dose [ED90], 0.1 mg kg-1 of body weight) across a quadruple-dose regimen. Pharmacokinetic profiling revealed rapid sulfoxide clearance via conversion to sulfone, with sulfone identified as the major active metabolite. When the sulfoxide was dosed, the exposure of the sulfone achieved was as much as 2.9-fold higher than when the sulfone was directly dosed, thereby demonstrating that the sulfoxide served as an effective prodrug for the treatment of malaria.
Collapse
|
22
|
Meier A, Erler H, Beitz E. Targeting Channels and Transporters in Protozoan Parasite Infections. Front Chem 2018; 6:88. [PMID: 29637069 PMCID: PMC5881087 DOI: 10.3389/fchem.2018.00088] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Infectious diseases caused by pathogenic protozoa are among the most significant causes of death in humans. Therapeutic options are scarce and massively challenged by the emergence of resistant parasite strains. Many of the current anti-parasite drugs target soluble enzymes, generate unspecific oxidative stress, or act by an unresolved mechanism within the parasite. In recent years, collections of drug-like compounds derived from large-scale phenotypic screenings, such as the malaria or pathogen box, have been made available to researchers free of charge boosting the identification of novel promising targets. Remarkably, several of the compound hits have been found to inhibit membrane proteins at the periphery of the parasites, i.e., channels and transporters for ions and metabolites. In this review, we will focus on the progress made on targeting channels and transporters at different levels and the potential for use against infections with apicomplexan parasites mainly Plasmodium spp. (malaria) and Toxoplasma gondii (toxoplasmosis), with kinetoplastids Trypanosoma brucei (sleeping sickness), Trypanosoma cruzi (Chagas disease), and Leishmania ssp. (leishmaniasis), and the amoeba Entamoeba histolytica (amoebiasis).
Collapse
Affiliation(s)
- Anna Meier
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Holger Erler
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Eric Beitz
- Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
23
|
Poonam, Gupta Y, Gupta N, Singh S, Wu L, Chhikara BS, Rawat M, Rathi B. Multistage inhibitors of the malaria parasite: Emerging hope for chemoprotection and malaria eradication. Med Res Rev 2018; 38:1511-1535. [DOI: 10.1002/med.21486] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/09/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Poonam
- Department of Chemistry; Miranda House, University of Delhi; India
| | - Yash Gupta
- National Institute of Malaria Research (ICMR); New Delhi India
| | - Nikesh Gupta
- Special Centre for Nanosciences; Jawaharlal Nehru University; New Delhi India
| | - Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry; Hansraj College University Enclave, University of Delhi; Delhi India
| | - Lidong Wu
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA USA
- Key Laboratory of Control of Quality and Safety for Aquatic Products; Ministry of Agriculture, Chinese Academy of Fishery Sciences; Beijing China
| | | | - Manmeet Rawat
- Department of Internal Medicine; University of New Mexico School of Medicine; Albuquerque NM USA
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry; Hansraj College University Enclave, University of Delhi; Delhi India
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA USA
| |
Collapse
|
24
|
Abstract
A marked decrease in malaria-related deaths worldwide has been attributed to the administration of effective antimalarials against Plasmodium falciparum, in particular, artemisinin-based combination therapies (ACTs). Increasingly, ACTs are also used to treat Plasmodium vivax, the second major human malaria parasite. However, resistance to frontline artemisinins and partner drugs is now causing the failure of P. falciparum ACTs in southeast Asia. In this Review, we discuss our current knowledge of markers and mechanisms of resistance to artemisinins and ACTs. In particular, we describe the identification of mutations in the propeller domains of Kelch 13 as the primary marker for artemisinin resistance in P. falciparum and explore two major mechanisms of resistance that have been independently proposed: the activation of the unfolded protein response and proteostatic dysregulation of parasite phosphatidylinositol 3- kinase. We emphasize the continuing challenges and the imminent need to understand mechanisms of resistance to improve parasite detection strategies, develop new combinations to eliminate resistant parasites and prevent their global spread.
Collapse
|
25
|
Moreno-Sabater A, Pérignon JL, Mazier D, Lavazec C, Soulard V. Humanized mouse models infected with human Plasmodium species for antimalarial drug discovery. Expert Opin Drug Discov 2017; 13:131-140. [DOI: 10.1080/17460441.2018.1410136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Alicia Moreno-Sabater
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
- Assistance Publique - Hopitaux de Paris - Hôpitaux Universitaires Paris-Est - Site Saint-Antoine, Paris, Île-de-France France
| | | | - Dominique Mazier
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| | - Catherine Lavazec
- Institut Cochin – INSERM U1016, Paris, Île-de-France France
- CNRS - UMR8104, Paris, France
- Universite Paris Descartes, Paris, Île-de-France France
| | - Valerie Soulard
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| |
Collapse
|
26
|
Dingemanse J, Krause A. Impact of pharmacokinetic-pharmacodynamic modelling in early clinical drug development. Eur J Pharm Sci 2017; 109S:S53-S58. [PMID: 28535992 DOI: 10.1016/j.ejps.2017.05.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/27/2022]
Abstract
Early clinical pharmacology studies in healthy subjects are often dissociated from patient studies. In this review we encourage the use of modelling and simulation techniques to generate valuable information at an early stage of clinical development. We illustrate these principles by presenting 5 different case studies from a spectrum of therapeutic drug classes. Their application leads to a better understanding of drug characteristics early on, thereby facilitating the design of dose-finding studies in the target patient population and saving resources.
Collapse
Affiliation(s)
- Jasper Dingemanse
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland.
| | - Andreas Krause
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland
| |
Collapse
|
27
|
Chiu JE, Thekkiniath J, Choi JY, Perrin BA, Lawres L, Plummer M, Virji AZ, Abraham A, Toh JY, Zandt MV, Aly ASI, Voelker DR, Mamoun CB. The antimalarial activity of the pantothenamide α-PanAm is via inhibition of pantothenate phosphorylation. Sci Rep 2017; 7:14234. [PMID: 29079738 PMCID: PMC5660193 DOI: 10.1038/s41598-017-14074-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022] Open
Abstract
The biosynthesis of the major acyl carrier Coenzyme A from pantothenic acid (PA) is critical for survival of Plasmodium falciparum within human erythrocytes. Accordingly, a PA analog α-PanAm showed potent activity against blood stage parasites in vitro; however, its efficacy in vivo and its mode of action remain unknown. We developed a new synthesis route for α-PanAm and showed that the compound is highly effective against blood stages of drug-sensitive and -resistant P. falciparum strains, inhibits development of P. berghei in hepatocytes, and at doses up to 100 mg/kg also inhibits blood stage development of P. chabaudi in mice. We used yeast and its pantothenate kinase Cab1 as models to characterize mode of action of α-PanAm and found that α-PanAm inhibits yeast growth in a PA-dependent manner, and its potency increases dramatically in a yeast mutant with defective pantothenate kinase activity. Biochemical analyses using 14C-PA as a substrate demonstrated that α-PanAm is a competitive inhibitor of Cab1. Interestingly, biochemical and mass spectrometry analyses also showed that the compound is phosphorylated by Cab1. Together, these data suggest that α-PanAm exerts its antimicrobial activity by direct competition with the natural substrate PA for phosphorylation by the pantothenate kinase.
Collapse
Affiliation(s)
- Joy E Chiu
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jose Thekkiniath
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jae-Yeon Choi
- Basic Science Section, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, Colorado, 80206, USA
| | - Benjamin A Perrin
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lauren Lawres
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mark Plummer
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Azan Z Virji
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Amanah Abraham
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112, USA
| | - Justin Y Toh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Ahmed S I Aly
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70112, USA
| | - Dennis R Voelker
- Basic Science Section, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, Colorado, 80206, USA
| | - Choukri Ben Mamoun
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
28
|
Probst D, Heitz M, Poirier M, Gan BH, Delalande C, Reymond JL. Frontiers in Medicinal Chemistry 2017 in Bern, Switzerland. ChemMedChem 2017; 12:1645-1651. [PMID: 28941184 DOI: 10.1002/cmdc.201700306] [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: 05/19/2017] [Indexed: 11/06/2022]
Abstract
Sharing capital ideas: The 2017 Frontiers in Medicinal Chemistry (FiMC) conference, organized jointly by the German Chemical Society, the German Pharmaceutical Society, and the Swiss Chemical Society, was held at the Department of Chemistry and Biochemistry of the University of Bern in February 2017. Herein we summarize the many conference highlights, and look forward to the next FiMC meeting, to be held in Jena (Germany) in March 2018.
Collapse
Affiliation(s)
- Daniel Probst
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| | - Marc Heitz
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| | - Marion Poirier
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| | - Bee Ha Gan
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| | - Clémence Delalande
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| | - Jean-Louis Reymond
- Universität Bern, Departement für Chemie und Biochemie, Freiestrasse 3, Bern, 3012, Switzerland
| |
Collapse
|
29
|
Andrews KA, Wesche D, McCarthy J, Möhrle JJ, Tarning J, Phillips L, Kern S, Grasela T. Model-Informed Drug Development for Malaria Therapeutics. Annu Rev Pharmacol Toxicol 2017; 58:567-582. [PMID: 28992431 PMCID: PMC7198115 DOI: 10.1146/annurev-pharmtox-010715-103429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Malaria is a critical public health problem resulting in substantial morbidity and
mortality, particularly in developing countries. Owing to the development of resistance
toward current therapies, novel approaches to accelerate the development efforts of new
malaria therapeutics are urgently needed. There have been significant advancements in the
development of in vitro and in vivo experiments that generate data used to inform
decisions about the potential merit of new compounds. A comprehensive disease-drug model
capable of integrating discrete data from different preclinical and clinical components
would be a valuable tool across all stages of drug development. This could have an
enormous impact on the otherwise slow and resource-intensive process of traditional
clinical drug development.
Collapse
Affiliation(s)
- Kayla Ann Andrews
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , , .,Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York 14214, USA
| | - David Wesche
- Bill and Melinda Gates Foundation, Seattle, Washington 98109, USA; ,
| | - James McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Medicine, University of Queensland, Brisbane, Australia;
| | - Jörg J Möhrle
- Medicines for Malaria Venture, Geneva 1215, Switzerland;
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; .,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Luann Phillips
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , ,
| | - Steven Kern
- Bill and Melinda Gates Foundation, Seattle, Washington 98109, USA; ,
| | - Thaddeus Grasela
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , ,
| |
Collapse
|
30
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
31
|
Hundreds of dual-stage antimalarial molecules discovered by a functional gametocyte screen. Nat Commun 2017; 8:15160. [PMID: 28513586 PMCID: PMC5442318 DOI: 10.1038/ncomms15160] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/03/2017] [Indexed: 02/07/2023] Open
Abstract
Plasmodium falciparum stage V gametocytes are responsible for parasite transmission, and drugs targeting this stage are needed to support malaria elimination. We here screen the Tres Cantos Antimalarial Set (TCAMS) using the previously developed P. falciparum female gametocyte activation assay (Pf FGAA), which assesses stage V female gametocyte viability and functionality using Pfs25 expression. We identify over 400 compounds with activities <2 μM, chemically classified into 57 clusters and 33 singletons. Up to 68% of the hits are chemotypes described for the first time as late-stage gametocyte-targeting molecules. In addition, the biological profile of 90 compounds representing the chemical diversity is assessed. We confirm in vitro transmission-blocking activity of four of the six selected molecules belonging to three distinct scaffold clusters. Overall, this TCAMS gametocyte screen provides 276 promising antimalarial molecules with dual asexual/sexual activity, representing starting points for target identification and candidate selection. There is a need for Plasmodium transmission blocking drugs for malaria elimination. Here, Miguel-Blanco et al. screen >10,000 compounds against stage V female gametocytes, identify active compounds belonging to 57 chemotypes and confirm transmission blocking activity of four selected compounds in vitro.
Collapse
|