1
|
Adebayo G, Ayanda OI, Rottmann M, Ajibaye OS, Oduselu G, Mulindwa J, Ajani OO, Aina O, Mäser P, Adebiyi E. The Importance of Murine Models in Determining In Vivo Pharmacokinetics, Safety, and Efficacy in Antimalarial Drug Discovery. Pharmaceuticals (Basel) 2025; 18:424. [PMID: 40143200 PMCID: PMC11944934 DOI: 10.3390/ph18030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
New chemical entities are constantly being investigated towards antimalarial drug discovery, and they require animal models for toxicity and efficacy testing. Murine models show physiological similarities to humans and are therefore indispensable in the search for novel antimalarial drugs. They provide a preclinical basis (following in vitro assessments of newly identified lead compounds) for further assessment in the drug development pipeline. Specific mouse strains, non-humanized and humanized, have successfully been infected with rodent Plasmodium species and the human Plasmodium species, respectively. Infected mice provide a platform for the assessment of treatment options being sought. In vivo pharmacokinetic evaluations are necessary when determining the fate of potential antimalarials in addition to the efficacy assessment of these chemical entities. This review describes the role of murine models in the drug development pipeline. It also explains some in vivo pharmacokinetic, safety, and efficacy parameters necessary for making appropriate choices of lead compounds in antimalarial drug discovery. Despite the advantages of murine models in antimalarial drug discovery, certain limitations are also highlighted.
Collapse
Affiliation(s)
- Glory Adebayo
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Opeyemi I. Ayanda
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Olusola S. Ajibaye
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Gbolahan Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Julius Mulindwa
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, Kampala P.O. Box 7062, Uganda;
| | - Olayinka O. Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Oluwagbemiga Aina
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- African Centre of Excellence in Bioinformatics & Data Intensive Science (ACE), Kampala P.O. Box 7062, Uganda
- Infectious Diseases Institute, Makerere University, Kampala P.O. Box 22418, Uganda
| |
Collapse
|
2
|
Andrade Meirelles M, Almeida VM, Sullivan JR, de Toledo I, Dos Reis CV, Cunha MR, Zigweid R, Shim A, Sankaran B, Woodward EL, Seibold S, Liu L, Mian MR, Battaile KP, Riley J, Duncan C, Simeons FRC, Ferguson L, Joji H, Read KD, Lovell S, Staker BL, Behr MA, Pilli RA, Couñago RM. Rational Exploration of 2,4-Diaminopyrimidines as DHFR Inhibitors Active against Mycobacterium abscessus and Mycobacterium avium, Two Emerging Human Pathogens. J Med Chem 2024; 67:19143-19164. [PMID: 39468773 DOI: 10.1021/acs.jmedchem.4c01594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Nontuberculous mycobacteria (NTM) are emerging human pathogens linked to severe pulmonary diseases. Current treatments involve the prolonged use of multiple drugs and are often ineffective. Bacterial dihydrofolate reductase (DHFR) is a key enzyme targeted by antibiotics in Gram-negative bacterial infections. However, existing DHFR inhibitors designed for Gram-negative bacteria often fail against mycobacterial DHFRs. Here, we detail the rational design of NTM DHFR inhibitors based on P218, a malarial DHFR inhibitor. We identified compound 8, a 2,4-diaminopyrimidine exhibiting improved pharmacological properties and activity against purified DHFR, and whole cell cultures of two predominant NTM species: Mycobacterium avium and Mycobacterium abscessus. This study underscores the potential of compound 8 as a promising candidate for the in vivo validation of DHFR as an effective treatment against NTM infections.
Collapse
Affiliation(s)
- Matheus Andrade Meirelles
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Vitor M Almeida
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Jaryd R Sullivan
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
| | - Ian de Toledo
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Caio Vinicius Dos Reis
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Micael Rodrigues Cunha
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
| | - Rachel Zigweid
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Abraham Shim
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Elijah L Woodward
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Steve Seibold
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Lijun Liu
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mohammad Rasel Mian
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Kevin P Battaile
- New York Structural Biology Center, Upton, New York 11973, United States
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Christina Duncan
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Frederick R C Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Liam Ferguson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Halimatu Joji
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry, University of Dundee, Dundee DD1 5EH, U.K
| | - Scott Lovell
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
- Protein Structure and X-ray Crystallography Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Bart L Staker
- Center for Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, United States
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington 98109, United States
| | - Marcel A Behr
- Department of Microbiology & Immunology, McGill University, Montréal H3A 2B4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal H4A 3J1, Canada
- McGill International TB Centre, Montréal H4A 3S5, Canada
- Department of Medicine, McGill University Health Centre, Montréal H4A 3J1, Canada
| | - Ronaldo A Pilli
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, UNICAMP, 13083-970-Campinas, SP, Brazil
| | - Rafael M Couñago
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, 13083-886-Campinas, SP, Brazil
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
3
|
Lin M, Cai J, Wei Y, Peng X, Luo Q, Li B, Chen Y, Wang L. MalariaFlow: A comprehensive deep learning platform for multistage phenotypic antimalarial drug discovery. Eur J Med Chem 2024; 277:116776. [PMID: 39173285 DOI: 10.1016/j.ejmech.2024.116776] [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: 05/11/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024]
Abstract
Malaria remains a significant global health challenge due to the growing drug resistance of Plasmodium parasites and the failure to block transmission within human host. While machine learning (ML) and deep learning (DL) methods have shown promise in accelerating antimalarial drug discovery, the performance of deep learning models based on molecular graph and other co-representation approaches warrants further exploration. Current research has overlooked mutant strains of the malaria parasite with varying degrees of sensitivity or resistance, and has not covered the prediction of inhibitory activities across the three major life cycle stages (liver, asexual blood, and gametocyte) within the human host, which is crucial for both treatment and transmission blocking. In this study, we manually curated a benchmark antimalarial activity dataset comprising 407,404 unique compounds and 410,654 bioactivity data points across ten Plasmodium phenotypes and three stages. The performance was systematically compared among two fingerprint-based ML models (RF::Morgan and XGBoost:Morgan), four graph-based DL models (GCN, GAT, MPNN, and Attentive FP), and three co-representations DL models (FP-GNN, HiGNN, and FG-BERT), which reveal that: 1) The FP-GNN model achieved the best predictive performance, outperforming the other methods in distinguishing active and inactive compounds across balanced, more positive, and more negative datasets, with an overall AUROC of 0.900; 2) Fingerprint-based ML models outperformed graph-based DL models on large datasets (>1000 compounds), but the three co-representations DL models were able to incorporate domain-specific chemical knowledge to bridge this gap, achieving better predictive performance. These findings provide valuable guidance for selecting appropriate ML and DL methods for antimalarial activity prediction tasks. The interpretability analysis of the FP-GNN model revealed its ability to accurately capture the key structural features responsible for the liver- and blood-stage activities of the known antimalarial drug atovaquone. Finally, we developed a web server, MalariaFlow, incorporating these high-quality models for antimalarial activity prediction, virtual screening, and similarity search, successfully predicting novel triple-stage antimalarial hits validated through experimental testing, demonstrating its effectiveness and value in discovering potential multistage antimalarial drug candidates.
Collapse
Affiliation(s)
- Mujie Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Junxi Cai
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, 510006, China
| | - Yuancheng Wei
- School of Software Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Xinru Peng
- School of Software Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Qianhui Luo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Biaoshun Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yihao Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Ling Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [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: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
5
|
Decharuangsilp S, Arwon U, Sooksai N, Rattanajak R, Saeyang T, Vitsupakorn D, Vanichtanankul J, Yuthavong Y, Kamchonwongpaisan S, Hoarau M. Novel flexible biphenyl PfDHFR inhibitors with improved antimalarial activity. RSC Med Chem 2024; 15:2496-2507. [PMID: 39026651 PMCID: PMC11253869 DOI: 10.1039/d4md00197d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/09/2024] [Indexed: 07/20/2024] Open
Abstract
As pregnant women and young children remain the first victims of malaria worldwide, the search for new antimalarials has been focusing on compounds with a high safety profile and extended efficacy. In a previous study, a rigid biphenyl PfDHFR inhibitor was developed by fragment-based screening, displaying sub nM enzyme inhibition but poor antiparasitic activity, presumably due to its low flexibility. Here, we report a new series of compounds that combines the biphenyl fragment with a flexible linker. Interestingly, their mode of binding differs from previously reported compounds, taking advantage of strong hydrophobic interaction. The new flexible biphenyl compounds show overall improved antiparasitic activity compared to rigid ones, with the best compound displaying a 2 nM antiplasmodial IC50 and suitable drug-like properties. This confirms the importance of compound flexibility for antimalarial activity and opens the way to new opportunities for antimalarial drug design.
Collapse
Affiliation(s)
- Sasithorn Decharuangsilp
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Uthai Arwon
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Nawarat Sooksai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Roonglawan Rattanajak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Thanaya Saeyang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Danoo Vitsupakorn
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Jarunee Vanichtanankul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Marie Hoarau
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| |
Collapse
|
6
|
Olotu A, Möhrle JJ. Moving seasonal malaria chemoprevention out of its geographical isolation. THE LANCET. INFECTIOUS DISEASES 2023; 23:1102-1103. [PMID: 37414067 DOI: 10.1016/s1473-3099(23)00268-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 07/08/2023]
Affiliation(s)
- Ally Olotu
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Jörg J Möhrle
- Medicines for Malaria Venture, 1215 Geneva 15, Switzerland.
| |
Collapse
|
7
|
van der Plas JL, Kuiper VP, Bagchus WM, Bödding M, Yalkinoglu Ö, Tappert A, Seitzinger A, Spangenberg T, Bezuidenhout D, Wilkins J, Oeuvray C, Dhingra SK, Thathy V, Fidock DA, Smidt LCA, Roozen GVT, Koopman JPR, Lamers OAC, Sijtsma J, van Schuijlenburg R, Wessels E, Meij P, Kamerling IMC, Roestenberg M, Khandelwal A. Causal chemoprophylactic activity of cabamiquine against Plasmodium falciparum in a controlled human malaria infection: a randomised, double-blind, placebo-controlled study in the Netherlands. THE LANCET. INFECTIOUS DISEASES 2023; 23:1164-1174. [PMID: 37414066 DOI: 10.1016/s1473-3099(23)00212-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Cabamiquine is a novel antimalarial that inhibits Plasmodium falciparum translation elongation factor 2. We investigated the causal chemoprophylactic activity and dose-exposure-response relationship of single oral doses of cabamiquine following the direct venous inoculation (DVI) of P falciparum sporozoites in malaria-naive, healthy volunteers. METHODS This was a phase 1b, randomised, double-blind, placebo-controlled, adaptive, dose-finding, single-centre study performed in Leiden, Netherlands. Malaria-naive, healthy adults aged 18-45 years were divided into five cohorts and randomly assigned (3:1) to receive cabamiquine or placebo. Randomisation was done by an independent statistician using codes in a permuted block schedule with a block size of four. Participants, investigators, and study personnel were masked to treatment allocation. A single, oral dose regimen of cabamiquine (200, 100, 80, 60, or 30 mg) or matching placebo was administered either at 2 h (early liver-stage) or 96 h (late liver-stage) after DVI. The primary endpoints based on a per-protocol analysis set were the number of participants who developed parasitaemia within 28 days of DVI, time to parasitaemia, number of participants with documented parasite blood-stage growth, clinical symptoms of malaria, and exposure-efficacy modelling. The impact of cabamiquine on liver stages was evaluated indirectly by the appearance of parasitaemia in the blood. The Clopper-Pearson CI (nominal 95%) was used to express the protection rate. The secondary outcomes were safety and tolerability, assessed in those who had received DVI and were administered one dose of the study intervention. The trial was prospectively registered on ClinicalTrials.gov (NCT04250363). FINDINGS Between Feb 17, 2020 and April 29, 2021, 39 healthy participants were enrolled (early liver-stage: 30 mg [n=3], 60 mg [n=6], 80 mg [n=6], 100 mg [n=3], 200 mg [n=3], pooled placebo [n=6]; late liver-stage: 60 mg [n=3], 100 mg [n=3], 200 mg [n=3], pooled placebo [n=3]). A dose-dependent causal chemoprophylactic effect was observed, with four (67%) of six participants in the 60 mg, five (83%) of six participants in the 80 mg, and all three participants in the 100 and 200 mg cabamiquine dose groups protected from parasitaemia up to study day 28, whereas all participants in the pooled placebo and 30 mg cabamiquine dose group developed parasitaemia. A single, oral dose of 100 mg cabamiquine or higher provided 100% protection against parasitaemia when administered during early or late liver-stage malaria. The median time to parasitaemia in those with early liver-stage malaria was prolonged to 15, 22, and 24 days for the 30, 60, and 80 mg dose of cabamiquine, respectively, compared with 10 days for the pooled placebo. All participants with positive parasitaemia showed documented blood-stage parasite growth, apart from one participant in the pooled placebo group and one participant in the 30 mg cabamiquine group. Most participants did not exhibit any malaria symptoms in both the early and late liver-stage groups, and those reported were mild in severity. A positive dose-exposure-efficacy relationship was established across exposure metrics. The median maximum concentration time was 1-6 h, with a secondary peak observed between 6 h and 12 h in all cabamiquine dose groups (early liver-stage). All cabamiquine doses were safe and well tolerated. Overall, 26 (96%) of 27 participants in the early liver-stage group and ten (83·3%) of 12 participants in the late liver-stage group reported at least one treatment-emergent adverse event (TEAE) with cabamiquine or placebo. Most TEAEs were of mild severity, transient, and resolved without sequelae. The most frequently reported cabamiquine-related TEAE was headache. No dose-related trends were observed in the incidence, severity, or causality of TEAEs. INTERPRETATION The results from this study show that cabamiquine has a dose-dependent causal chemoprophylactic activity. Together with previously demonstrated activity against the blood stages combined with a half-life of more than 150 h, these results indicate that cabamiquine could be developed as a single-dose monthly regimen for malaria prevention. FUNDING The healthcare business of Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
- Johan L van der Plas
- Centre for Human Drug Research, Leiden, Netherlands; Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent P Kuiper
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Wilhelmina M Bagchus
- Merck Institute for Pharmacometrics, Merck Serono (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | | | | | - Aliona Tappert
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Thomas Spangenberg
- Global Health Institute of Merck, Ares Trading (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Deon Bezuidenhout
- Merck (Pty) (an affiliate of Merck KGaA, Darmstadt, Germany), Modderfontein, South Africa
| | | | - Claude Oeuvray
- Global Health Institute of Merck, Ares Trading (a subsidiary of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | | | - Vandana Thathy
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Geert V T Roozen
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jan Pieter R Koopman
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Olivia A C Lamers
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen Sijtsma
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Roos van Schuijlenburg
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Els Wessels
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Pauline Meij
- Center for Cell and Gene Therapy, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Ingrid M C Kamerling
- Centre for Human Drug Research, Leiden, Netherlands; Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Department of Infectious Diseases and Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| | | |
Collapse
|
8
|
Siqueira-Neto JL, Wicht KJ, Chibale K, Burrows JN, Fidock DA, Winzeler EA. Antimalarial drug discovery: progress and approaches. Nat Rev Drug Discov 2023; 22:807-826. [PMID: 37652975 PMCID: PMC10543600 DOI: 10.1038/s41573-023-00772-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Recent antimalarial drug discovery has been a race to produce new medicines that overcome emerging drug resistance, whilst considering safety and improving dosing convenience. Discovery efforts have yielded a variety of new molecules, many with novel modes of action, and the most advanced are in late-stage clinical development. These discoveries have led to a deeper understanding of how antimalarial drugs act, the identification of a new generation of drug targets, and multiple structure-based chemistry initiatives. The limited pool of funding means it is vital to prioritize new drug candidates. They should exhibit high potency, a low propensity for resistance, a pharmacokinetic profile that favours infrequent dosing, low cost, preclinical results that demonstrate safety and tolerability in women and infants, and preferably the ability to block Plasmodium transmission to Anopheles mosquito vectors. In this Review, we describe the approaches that have been successful, progress in preclinical and clinical development, and existing challenges. We illustrate how antimalarial drug discovery can serve as a model for drug discovery in diseases of poverty.
Collapse
Affiliation(s)
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | | | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
9
|
Hoarau M, Sermmai P, Varatthan T, Thiabma R, Jantra T, Rattanajak R, Vitsupakorn D, Vanichtanankul J, Saepua S, Yuthavong Y, Thongpanchang C, Kamchonwongpaisan S. Discovery of rigid biphenyl Plasmodium falciparum DHFR inhibitors using a fragment linking strategy. RSC Med Chem 2023; 14:1755-1766. [PMID: 37731689 PMCID: PMC10507804 DOI: 10.1039/d3md00242j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/24/2023] [Indexed: 09/22/2023] Open
Abstract
Plasmodium falciparum dihydrofolate reductase (PfDHFR), a historical target for antimalarials, has been considered compromised due to resistance inducing mutations caused by pyrimethamine (PYR) overexposure. The clinical candidate P218 has demonstrated that inhibitors could efficiently target both PYR-sensitive and PYR-resistant parasites through careful drug design. Yet, P218 clinical development has been limited by its pharmacokinetic profile, incompatible with single dose regimen. Herein, we report the design of new PfDHFR inhibitors using fragment-based design, aiming at improved lipophilicity and overall drug-like properties. Fragment-based screening identified hits binding in the pABA site of the enzyme. Using structure-guided design, hits were elaborated into leads by fragment linking with 2,4-diaminopyrimidine. Resulting compounds display nM range inhibition of both drug-sensitive and resistant PfDHFR, high selectivity against the human isoform, drug-like lipophilicity and metabolic stability. Compound 4 and its ester derivative 3 kill blood stage TM4/8.2 parasite at nM concentrations while showing no toxicity against Vero cells.
Collapse
Affiliation(s)
- Marie Hoarau
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Patpanat Sermmai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Thaveechai Varatthan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Ratthiya Thiabma
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Tararat Jantra
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Roonglawan Rattanajak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Danoo Vitsupakorn
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Jarunee Vanichtanankul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Siriporn Saepua
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Chawanee Thongpanchang
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency Pathum Thani 12120 Thailand
| |
Collapse
|
10
|
Nyandele JP, Kibondo UA, Issa F, Van Geertruyden JP, Warimwe G, Jongo S, Abdulla S, Olotu A. Pre-vaccination monocyte-to-lymphocyte ratio as a biomarker for the efficacy of malaria candidate vaccines: A subgroup analysis of pooled clinical trial data. PLoS One 2023; 18:e0291244. [PMID: 37708143 PMCID: PMC10501550 DOI: 10.1371/journal.pone.0291244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/24/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Pre-vaccination monocyte-to-lymphocyte ratio was previously suggested as a marker for malaria vaccine effectiveness. We investigated the potential of this cell ratio as a marker for malaria vaccine efficacy and effectiveness. Effectiveness was investigated by using clinical malaria endpoint, and efficacy was investigated by using surrogate endpoints of Plasmodium falciparum prepatent period, parasite density, and multiplication rates in a controlled human malaria infection trial (CHMI). METHODS We evaluated the correlation between monocyte-to-lymphocyte ratio and RTS,S vaccine effectiveness using Cox regression modeling with clinical malaria as the primary endpoint. Of the 1704 participants in the RTS,S field trial, data on monocyte-to-lymphocyte ratio was available for 842 participants, of whom our analyses were restricted. We further used Spearman Correlations and Cox regression modeling to evaluate the correlation between monocyte-to-lymphocyte ratio and Whole Sporozoite malaria vaccine efficacy using the surrogate endpoints. Of the 97 participants in the controlled human malaria infection vaccine trials, hematology and parasitology information were available for 82 participants, of whom our analyses were restricted. RESULTS The unadjusted efficacy of RTS,S malaria vaccine was 54% (95% CI: 37%-66%, p <0.001). No correlation was observed between monocyte-to-lymphocyte ratio and RTS,S vaccine efficacy (Hazard Rate (HR):0.90, 95%CI:0.45-1.80; p = 0.77). The unadjusted efficacy of Whole Sporozoite malaria vaccine in the appended dataset was 17.6% (95%CI:10%-28.5%, p<0.001). No association between monocyte-to-lymphocyte ratio and the Whole Sporozoite malaria vaccine was found against either the prepatent period (HR = 1.16; 95%CI:0.51-2.62, p = 0.72), parasite density (rho = 0.004, p = 0.97) or multiplication rates (rho = 0.031, p = 0.80). CONCLUSION Monocyte-to-lymphocyte ratio alone may not be an adequate marker for malaria vaccine efficacy. Further investigations on immune correlates and underlying mechanisms of immune protection against malaria could provide a clearer explanation of the differences between those protected in comparison with those not protected against malaria by vaccination.
Collapse
Affiliation(s)
- Jane Paula Nyandele
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ummi Abdul Kibondo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Fatuma Issa
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | | | | | - Said Jongo
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Ally Olotu
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| |
Collapse
|
11
|
Alaithan H, Kumar N, Islam MZ, Liappis AP, Nava VE. Novel Therapeutics for Malaria. Pharmaceutics 2023; 15:1800. [PMID: 37513987 PMCID: PMC10383744 DOI: 10.3390/pharmaceutics15071800] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Malaria is a potentially fatal disease caused by protozoan parasites of the genus Plasmodium. It is responsible for significant morbidity and mortality in endemic countries of the tropical and subtropical world, particularly in Africa, Southeast Asia, and South America. It is estimated that 247 million malaria cases and 619,000 deaths occurred in 2021 alone. The World Health Organization's (WHO) global initiative aims to reduce the burden of disease but has been massively challenged by the emergence of parasitic strains resistant to traditional and emerging antimalarial therapy. Therefore, development of new antimalarial drugs with novel mechanisms of action that overcome resistance in a safe and efficacious manner is urgently needed. Based on the evolving understanding of the physiology of Plasmodium, identification of potential targets for drug intervention has been made in recent years, resulting in more than 10 unique potential anti-malaria drugs added to the pipeline for clinical development. This review article will focus on current therapies as well as novel targets and therapeutics against malaria.
Collapse
Affiliation(s)
- Haitham Alaithan
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Medicine, George Washington University, Washington, DC 20037, USA
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute of Public Health, George Washington University, Washington, DC 20037, USA
| | - Mohammad Z Islam
- Department of Pathology and Translational Pathology, Louisiana State University Health Science Center, Shreveport, LA 71103, USA
| | - Angelike P Liappis
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Medicine, George Washington University, Washington, DC 20037, USA
| | - Victor E Nava
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Pathology, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
12
|
Miglianico M, Bolscher JM, Vos MW, Koolen KJM, de Bruijni M, Rajagopal DS, Chen E, Kiczun M, Gray D, Campo B, Sauerwein RW, Dechering KJ. Assessment of the drugability of initial malaria infection through miniaturized sporozoite assays and high-throughput screening. Commun Biol 2023; 6:216. [PMID: 36823266 PMCID: PMC9950425 DOI: 10.1038/s42003-023-04599-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
The sporozoite stages of malaria parasites are the primary cause of infection of the vertebrate host and are targeted by (experimental) vaccines. Yet, little is known about their susceptibility to chemical intervention. Phenotypic high-throughput screens have not been feasible due to a lack of in vitro systems. Here we tested 78 marketed and experimental antimalarial compounds in miniaturized assays addressing sporozoite viability, gliding motility, hepatocyte traversal, and intrahepatocytic schizogony. None potently interfered with sporozoite viability or motility but ten compounds acted at the level of schizogony with IC50s < 100 nM. To identify compounds directly targeting sporozoites, we screened 81,000 compounds from the Global Health Diversity and reFRAME libraries in a sporozoite viability assay using a parasite expressing a luciferase reporter driven by the circumsporozoite promoter. The ionophore gramicidin emerged as the single hit from this screening campaign. Its effect on sporozoite viability translated into reduced gliding motility and an inability of sporozoites to invade human primary hepatocytes and develop into hepatic schizonts. While providing proof of concept for a small molecule sporontocidal mode of action, our combined data indicate that liver schizogony is more accessible to chemical intervention by (candidate) antimalarials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emily Chen
- Calibr, a division of The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael Kiczun
- Drug Discovery Unit, University of Dundee, Dundee, United Kingdom
| | - David Gray
- Drug Discovery Unit, University of Dundee, Dundee, United Kingdom
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | | | | |
Collapse
|
13
|
Ibraheem W, Makki AA, Alzain AA. Phthalide derivatives as dihydrofolate reductase inhibitors for malaria: molecular docking and molecular dynamics studies. J Biomol Struct Dyn 2022:1-11. [DOI: 10.1080/07391102.2022.2080114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Walaa Ibraheem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Medani, Gezira, Sudan
| | - Alaa A. Makki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Medani, Gezira, Sudan
| | - Abdulrahim Altoam Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Medani, Gezira, Sudan
| |
Collapse
|
14
|
New insights into antimalarial chemopreventive activity of antifolates. Antimicrob Agents Chemother 2021; 66:e0153821. [PMID: 34930029 DOI: 10.1128/aac.01538-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antifolates targeting dihydrofolate reductase (DHFR) are antimalarial compounds that have long been used for malaria treatment and chemoprevention (inhibition of infection from mosquitoes to humans). Despite their extensive applications, the thorough understanding of antifolate activity against hepatic malaria parasites, especially resistant parasites, have yet to be achieved. Using a transgenic P. berghei harboring quadruple mutant dhfr from P. falciparum (Pb::Pfdhfr-4M), we demonstrate that quadruple mutations on Pfdhfr confer complete chemoprevention resistance to pyrimethamine, the previous generation of antifolate, but not to a new class of antifolate designed to overcome the resistance such as P218. Detailed investigation to pin-point stage-specific chemoprevention further demonstrated that it is unnecessary for the drug to be present throughout hepatic development. The drug is most potent against the developmental stages from early hepatic trophozoite to late hepatic trophozoite, but is not effective at inhibiting sporozoite and early hepatic stage development from sporozoite to early trophozoite. Our data shows that P218 also inhibited the late hepatic stage development, from trophozoite to mature schizonts to a lesser extent. With a single dose of 15 mg/kg, P218 prevented infection from up to 25,000 pyrimethamine-resistant sporozoites, a number equal to thousands of infectious mosquito bites. Additionally, the hepatic stage of malaria parasite is much more susceptible to antifolates than the asexual blood stage. This study provides important insights into the activity of antifolates, as a chemopreventive therapeutic which could lead to a more efficient and cost effective treatment regime.
Collapse
|
15
|
Review of the Current Landscape of the Potential of Nanotechnology for Future Malaria Diagnosis, Treatment, and Vaccination Strategies. Pharmaceutics 2021; 13:pharmaceutics13122189. [PMID: 34959470 PMCID: PMC8706932 DOI: 10.3390/pharmaceutics13122189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Malaria eradication has for decades been on the global health agenda, but the causative agents of the disease, several species of the protist parasite Plasmodium, have evolved mechanisms to evade vaccine-induced immunity and to rapidly acquire resistance against all drugs entering clinical use. Because classical antimalarial approaches have consistently failed, new strategies must be explored. One of these is nanomedicine, the application of manipulation and fabrication technology in the range of molecular dimensions between 1 and 100 nm, to the development of new medical solutions. Here we review the current state of the art in malaria diagnosis, prevention, and therapy and how nanotechnology is already having an incipient impact in improving them. In the second half of this review, the next generation of antimalarial drugs currently in the clinical pipeline is presented, with a definition of these drugs' target product profiles and an assessment of the potential role of nanotechnology in their development. Opinions extracted from interviews with experts in the fields of nanomedicine, clinical malaria, and the economic landscape of the disease are included to offer a wider scope of the current requirements to win the fight against malaria and of how nanoscience can contribute to achieve them.
Collapse
|
16
|
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: 11] [Impact Index Per Article: 2.8] [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
|
17
|
Efforts Made to Eliminate Drug-Resistant Malaria and Its Challenges. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5539544. [PMID: 34497848 PMCID: PMC8421183 DOI: 10.1155/2021/5539544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023]
Abstract
Since 2000, a good deal of progress has been made in malaria control. However, there is still an unacceptably high burden of the disease and numerous challenges limiting advancement towards its elimination and ultimate eradication. Among the challenges is the antimalarial drug resistance, which has been documented for almost all antimalarial drugs in current use. As a result, the malaria research community is working on the modification of existing treatments as well as the discovery and development of new drugs to counter the resistance challenges. To this effect, many products are in the pipeline and expected to be marketed soon. In addition to drug and vaccine development, mass drug administration (MDA) is under scientific scrutiny as an important strategy for effective utilization of the developed products. This review discusses the challenges related to malaria elimination, ongoing approaches to tackle the impact of drug-resistant malaria, and upcoming antimalarial drugs.
Collapse
|
18
|
Kreutzfeld O, Tumwebaze PK, Byaruhanga O, Katairo T, Okitwi M, Orena S, Rasmussen SA, Legac J, Conrad MD, Nsobya SL, Aydemir O, Bailey JA, Duffey M, Cooper RA, Rosenthal PJ. Decreased Susceptibility to Dihydrofolate Reductase Inhibitors Associated With Genetic Polymorphisms in Ugandan Plasmodium falciparum Isolates. J Infect Dis 2021; 225:696-704. [PMID: 34460932 PMCID: PMC8844592 DOI: 10.1093/infdis/jiab435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/27/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The Plasmodium falciparum dihydrofolate reductase (PfDHFR) inhibitors pyrimethamine and cycloguanil (the active metabolite of proguanil) have important roles in malaria chemoprevention, but drug resistance challenges their efficacies. A new compound, P218, was designed to overcome resistance, but drug-susceptibility data for P falciparum field isolates are limited. METHODS We studied ex vivo PfDHFR inhibitor susceptibilities of 559 isolates from Tororo and Busia districts, Uganda, from 2016 to 2020, sequenced 383 isolates, and assessed associations between genotypes and drug-susceptibility phenotypes. RESULTS Median half-maximal inhibitory concentrations (IC50s) were 42 100 nM for pyrimethamine, 1200 nM for cycloguanil, 13000 nM for proguanil, and 0.6 nM for P218. Among sequenced isolates, 3 PfDHFR mutations, 51I (100%), 59R (93.7%), and 108N (100%), were very common, as previously seen in Uganda, and another mutation, 164L (12.8%), had moderate prevalence. Increasing numbers of mutations were associated with decreasing susceptibility to pyrimethamine, cycloguanil, and P218, but not proguanil, which does not act directly against PfDHFR. Differences in P218 susceptibilities were modest, with median IC50s of 1.4 nM for parasites with mixed genotype at position 164 and 5.7 nM for pure quadruple mutant (51I/59R/108N/164L) parasites. CONCLUSIONS Resistance-mediating PfDHFR mutations were common in Ugandan isolates, but P218 retained excellent activity against mutant parasites.
Collapse
Affiliation(s)
| | | | | | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Martin Okitwi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Stephen Orena
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Jennifer Legac
- University of California, San Francisco, California, USA
| | | | - Sam L Nsobya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | | | - Roland A Cooper
- Dominican University of California, San Rafael, California, USA
| | - Philip J Rosenthal
- Correspondence: Philip J. Rosenthal, MD, Department of Medicine, University of California, Box 0811, San Francisco, CA 94143 USA ()
| |
Collapse
|
19
|
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
|