1
|
Ferreira LM, García-García P, García PA, Castro MÁ. A review on quinolines: New green synthetic methods and bioactive potential. Eur J Pharm Sci 2025; 209:107097. [PMID: 40221058 DOI: 10.1016/j.ejps.2025.107097] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Quinolines have been an interest of study for a few decades due to the importance of this system in natural and pharmaceutical products. Since their discovery in the nineteenth century, many medicinal properties have been found for quinoline compounds. Firstly, as an anti-parasitic agent against malaria and then against many other diseases, such as, other parasitic infections, HIV, bacterial infections and cancer. Consequently, many synthetic methods have been developed to afford the quinoline ring. In this review we look back at traditional methods and look forward to the most recent and promising "green" methods for the synthesis of quinolines. Also, we review the newest advances in therapeutic compounds based on the quinoline skeleton for the treatment of parasitic and cancer diseases and the most recent applications of quinoline derivatives in drug delivery systems.
Collapse
Affiliation(s)
- Laura M Ferreira
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno Salamanca, 37007, Spain
| | - Pilar García-García
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno Salamanca, 37007, Spain.
| | - Pablo A García
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno Salamanca, 37007, Spain
| | - María Ángeles Castro
- Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, CIETUS/IBSAL, Universidad de Salamanca, Campus Miguel de Unamuno Salamanca, 37007, Spain.
| |
Collapse
|
2
|
Probst AS, Paton DG, Appetecchia F, Bopp S, Adams KL, Rinvee TA, Pou S, Winter R, Du EW, Yahiya S, Vidoudez C, Singh N, Rodrigues J, Castañeda-Casado P, Tammaro C, Chen D, Godinez-Macias KP, Jaramillo JL, Poce G, Rubal MJ, Nilsen A, Winzeler EA, Baum J, Burrows JN, Riscoe MK, Wirth DF, Catteruccia F. In vivo screen of Plasmodium targets for mosquito-based malaria control. Nature 2025:10.1038/s41586-025-09039-2. [PMID: 40399670 DOI: 10.1038/s41586-025-09039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Accepted: 04/17/2025] [Indexed: 05/23/2025]
Abstract
The decline in malaria deaths has recently stalled owing to several factors, including the widespread resistance of Anopheles vectors to the insecticides used in long-lasting insecticide-treated nets (LLINs)1,2. One way to mitigate insecticide resistance is to directly kill parasites during their mosquito-stage of development by incorporating antiparasitic compounds into LLINs. This strategy can prevent onward parasite transmission even when insecticides lose efficacy3,4. Here, we performed an in vivo screen of compounds against the mosquito stages of Plasmodium falciparum development. Of the 81 compounds tested, which spanned 28 distinct modes of action, 22 were active against early parasite stages in the mosquito midgut lumen, which in turn prevented establishment of infection. Medicinal chemistry was then used to improve antiparasitic activity of the top hits from the screen. We generated several endochin-like quinolones (ELQs) that inhibited the P. falciparum cytochrome bc1 complex (CytB). Two lead compounds that targeted separate sites in CytB (Qo and Qi) showed potent, long-lasting and stable activity when incorporated and/or extruded into bed net-like polyethylene films. ELQ activity was fully preserved in insecticide-resistant mosquitoes, and parasites resistant to these compounds had impaired development at the mosquito stage. These data demonstrate the promise of incorporating ELQ compounds into LLINs to counteract insecticide resistance and to reduce malaria transmission.
Collapse
Affiliation(s)
- Alexandra S Probst
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Douglas G Paton
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Infectious Disease, University of Georgia, Athens, GA, USA
| | - Federico Appetecchia
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Kelsey L Adams
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Tasneem A Rinvee
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Esrah W Du
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sabrina Yahiya
- Department of Life Sciences, Imperial College London, London, UK
| | | | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Chiara Tammaro
- Department of Chemistry and Pharmaceutical Technologies, Sapienza University of Rome, Rome, Italy
| | - Daisy Chen
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Giovanna Poce
- Department of Chemistry and Pharmaceutical Technologies, Sapienza University of Rome, Rome, Italy
| | | | - Aaron Nilsen
- VA Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London, UK
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Michael K Riscoe
- VA Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA.
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
3
|
Isern JA, Carlucci R, Labadie GR, Porta EOJ. Progress and Prospects of Triazoles in Advanced Therapies for Parasitic Diseases. Trop Med Infect Dis 2025; 10:142. [PMID: 40423371 DOI: 10.3390/tropicalmed10050142] [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: 03/24/2025] [Revised: 05/14/2025] [Accepted: 05/18/2025] [Indexed: 05/28/2025] Open
Abstract
Parasitic diseases represent a severe global burden, with current treatments often limited by toxicity, drug resistance, and suboptimal efficacy in chronic infections. This review examines the emerging role of triazole-based compounds, originally developed as antifungals, in advanced antiparasitic therapy. Their unique structural properties, particularly those of 1,2,3- and 1,2,4-triazole isomers, facilitate diverse binding interactions and favorable pharmacokinetics. By leveraging innovative synthetic approaches, such as click chemistry (copper-catalyzed azide-alkyne cycloaddition) and structure-based design, researchers have repurposed and optimized triazole scaffolds to target essential parasite pathways, including sterol biosynthesis via CYP51 and other novel enzymatic routes. Preclinical studies in models of Chagas disease, leishmaniasis, malaria, and helminth infections demonstrate that derivatives like posaconazole, ravuconazole, and DSM265 exhibit potent in vitro and in vivo activity, although their primarily static effects have limited their success as monotherapies in chronic cases. Combination strategies and hybrid molecules have demonstrated the potential to enhance efficacy and mitigate drug resistance. Despite challenges in achieving complete parasite clearance and managing potential toxicity, interdisciplinary efforts across medicinal chemistry, parasitology, and clinical research highlight the significant potential of triazoles as components of next-generation, patient-friendly antiparasitic regimens. These findings support the further optimization and clinical evaluation of triazole-based agents to improve treatments for neglected parasitic diseases.
Collapse
Affiliation(s)
- Jaime A Isern
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Renzo Carlucci
- Instituto de Química Rosario, Universidad Nacional de Rosario, CONICET, S2002LRK Rosario, Argentina
| | - Guillermo R Labadie
- Instituto de Química Rosario, Universidad Nacional de Rosario, CONICET, S2002LRK Rosario, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2002LRK Rosario, Argentina
| | - Exequiel O J Porta
- UCL School of Pharmacy, Faculty of Life Sciences, University College London (UCL), 29-39 Brunswick Square, London WC1N 1AX, UK
| |
Collapse
|
4
|
Chen S, Mondile Q, Du X, Wang C, Mukim M, Wrenger C, Dömling ASS, Tastan Bishop Ö, Groves MR. Exploring Aspartate Transcarbamoylase: A Promising Broad-Spectrum Target for Drug Development. Chembiochem 2025; 26:e202401009. [PMID: 39937588 PMCID: PMC12002100 DOI: 10.1002/cbic.202401009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
Pyrimidine nucleotides are essential for a wide variety of cellular processes and are synthesized either via a salvage pathway or through de novo biosynthesis. The latter is particularly important in proliferating cells, such as infectious diseases and cancer cells. Aspartate transcarbamoylase (ATCase) catalyzes the first committed and rate-limiting step in the de novo pyrimidine biosynthesis pathway, making it an attractive therapeutic target for various diseases. This review summarizes the development of a series of allosteric ATCase inhibitors, advancing them as potential candidates for malarial, tuberculosis and cancer therapies. Furthermore, it explores the potential for these compounds to be expanded into drugs targeting neglected tropical diseases, antimicrobial-resistant infections caused by the ESKAPE pathogens, and their possible application as herbicides. We identify the likely equivalent allosteric pocket in these systems and perform a structure and sequence-based analysis of the residues comprising it, providing a rationale for continued exploration of this compound series as both specific and broad-range inhibitors. The review concludes by emphasizing the importance of continued research into ATCase inhibitors, given their potential broad applicability in treating diverse diseases to enhance both human health and agricultural practices.
Collapse
Affiliation(s)
- Siyao Chen
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
| | - Queenie Mondile
- Research Unit in Bioinformatics (RUBi)Department of BiochemistryMicrobiology and BiochemistryRhodes University
| | - XiaoChen Du
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
| | - Chao Wang
- NeurobiologyMRC-Laboratory of Molecular Biology Cambridge Biomedical CampusFrancis Crick Ave, TrumpingtonCambridgeCB2 0QH
| | - Mayur Mukim
- Czech Advanced Technology and Research Institute (CATRIN)and Institute of Molecular and Translational Medicine (IMTMFaculty of Medicine and DentistryPalacky UniversityŠlechtitelů 27779 00OlomoucCzech Republic
| | - Carsten Wrenger
- Unit for Drug DiscoveryDepartment of ParasitologyInstitute of Biomedical SciencesUniversity of São PauloAvenida Professor Lineu Prestes 137405508-000São Paulo-SPBrazil
| | - Alexander S. S. Dömling
- Czech Advanced Technology and Research Institute (CATRIN)and Institute of Molecular and Translational Medicine (IMTMFaculty of Medicine and DentistryPalacky UniversityŠlechtitelů 27779 00OlomoucCzech Republic
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi)Department of BiochemistryMicrobiology and BiochemistryRhodes University
- National Institute for Theoretical and Computational Sciences (NITheCS)South Africa
- Genomics for Health in Africa (GHA)Africa-Europe Cluster of Research Excellence (CoRE)
| | - Matthew R. Groves
- Department of Chemical and Pharmaceutical BiologyUniversity of GroningenAntonius Deusinglaan 19731AVGroningenThe Netherlands
- Genomics for Health in Africa (GHA)Africa-Europe Cluster of Research Excellence (CoRE)
| |
Collapse
|
5
|
Cui L. Predicting Plasmodium drug resistance through in vitro evolution. Trends Parasitol 2025; 41:170-171. [PMID: 39933988 PMCID: PMC11884984 DOI: 10.1016/j.pt.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025]
Abstract
Knowledge about the resistibility and targets of compounds critically informs antimalarial drug development. Luth et al. systematically analyzed the resistome of Plasmodium falciparum through in vitro evolution and whole-genome analysis of 724 clones resistant to 118 compounds, revealing novel drug targets, multidrug resistance loci, and key determinants of resistance.
Collapse
Affiliation(s)
- Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
6
|
Evaristo J, de Laia E, Tavares B, Mendonça E, Grisostenes L, Rodrigues C, do Nascimento W, Garcia C, Guterres S, Nogueira F, Zanchi F, Evaristo G. Identification of Bioactive Metabolites of Capirona macrophylla by Metabolomic Analysis, Molecular Docking, and In Vitro Antiparasitic Assays. Metabolites 2025; 15:157. [PMID: 40137122 PMCID: PMC11943490 DOI: 10.3390/metabo15030157] [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: 01/15/2025] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 03/27/2025] Open
Abstract
Capirona macrophylla is a Rubiaceae known as "mulateiro". Ethnobotanical extracts have been used for skin treatment and in the management of leishmaniasis and malaria. OBJECTIVES The metabolites in aqueous extracts from wood bark, leaves, and stems were identified, and their in silico docking and in vitro cellular efficacy against Leishmania amazonensis and Plasmodium falciparum were evaluated. METHODS The extracts were analyzed by UHPLC/HRMSn using untargeted metabolomics approach with MSDial, MSFinder, and GNPS software for metabolite identification and spectra clustering. The most abundant metabolites underwent molecular docking using AutoDock via PyRx, targeting the dihydroorotate dehydrogenase from Leishmania and P. falciparum, and evaluated through molecular dynamics simulations using Gromacs. In vitro biological assays were conducted on 60 HPLC-fractions against these parasites. RESULTS Metabolomics analysis identified 5100 metabolites in ESI+ and 2839 in ESI- spectra among the "mulateiro" samples. GNPS clustering highlighted large clusters of quercetin and chlorogenic acid groups. The most abundant metabolites were isofraxidin, scopoletin, 5(S)-5-carboxystrictosidine, loliolide, quercetin, quinic acid, caffeoylquinic acid (and isomers), chlorogenic acid, neochlorogenic acid, tryptophan, N-acetyltryptophan, epicatechin, procyanidin, and kaempferol-3-O-robinoside-7-O-rhamnoside. Molecular docking pointed to 3,4-dicaffeoylquinic acid and kaempferol as promising inhibitors. The in vitro assays yielded four active HPLC-fractions against L. amazonensis with IC50 values ranging from 175.2 μg/mL to 194.8 μg/mL, and fraction G29 showed an IC50 of 119.8 μg/mL against P. falciparum. CONCLUSIONS The ethnobotanical use of "mulateiro" wood bark tea as an antimalarial and antileishmanial agent was confirmed through in vitro assays. We speculate that these activities are attributed to linoleic acids and quinic acids.
Collapse
Affiliation(s)
- Joseph Evaristo
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| | - Elise de Laia
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| | - Bruna Tavares
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| | - Esdras Mendonça
- Bioinformatic and Medicinal Chemistry Laboratory, Fiocruz/RO, Porto Velho 76812-245, Rondônia, Brazil; (E.M.); (F.Z.)
- National Institute of Epidemiology in the Western Amazon (INCT-EPIAMO), Porto Velho 76812-245, Rondônia, Brazil
| | - Larissa Grisostenes
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| | - Caroline Rodrigues
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| | - Welington do Nascimento
- Malaria and Leishmaniasis Bioassays Platform Laboratory, Fiocruz/RO, Porto Velho 76812-245, Rondônia, Brazil; (W.d.N.); (C.G.)
| | - Carolina Garcia
- Malaria and Leishmaniasis Bioassays Platform Laboratory, Fiocruz/RO, Porto Velho 76812-245, Rondônia, Brazil; (W.d.N.); (C.G.)
| | - Sheila Guterres
- Chemistry Department, Federal University of Rondônia (UNIR), Rio de Janeiro City 21941-598, Rio de Janeiro State, Brazil;
| | - Fábio Nogueira
- Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro City 21941-598, Rio de Janeiro State, Brazil;
- Laboratory of Protein Chemistry-Proteomic Unit, Center for Research in Precision Medicine (CPMP), Carlos Chagas Filho Biophysics Institute, UFRJ, Rio de Janeiro City 21941-902, Rio de Janeiro State, Brazil
| | - Fernando Zanchi
- Bioinformatic and Medicinal Chemistry Laboratory, Fiocruz/RO, Porto Velho 76812-245, Rondônia, Brazil; (E.M.); (F.Z.)
- National Institute of Epidemiology in the Western Amazon (INCT-EPIAMO), Porto Velho 76812-245, Rondônia, Brazil
| | - Geisa Evaristo
- Center for the Study of Biomolecules Applied to Health (Cebio), Oswaldo Cruz Foundation Rondônia Unity (Fiocruz/RO), Porto Velho 76812-245, Rondônia, Brazil; (J.E.); (E.d.L.); (B.T.); (L.G.); (C.R.)
| |
Collapse
|
7
|
Nie Z, Bonnert R, Tsien J, Deng X, Higgs C, El Mazouni F, Zhang X, Li R, Ho N, Feher V, Paulsen J, Shackleford DM, Katneni K, Chen G, Ng ACF, McInerney M, Wang W, Saunders J, Collins D, Yan D, Li P, Campbell M, Patil R, Ghoshal A, Mondal P, Kundu A, Chittimalla R, Mahadeva M, Kokkonda S, White J, Das R, Mukherjee P, Angulo-Barturen I, Jiménez-Díaz MB, Malmstrom R, Lawrenz M, Rodriguez-Granillo A, Rathod PK, Tomchick DR, Palmer MJ, Laleu B, Qin T, Charman SA, Phillips MA. Structure-Based Discovery and Development of Highly Potent Dihydroorotate Dehydrogenase Inhibitors for Malaria Chemoprevention. J Med Chem 2025; 68:590-637. [PMID: 39710971 PMCID: PMC11726676 DOI: 10.1021/acs.jmedchem.4c02394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024]
Abstract
Malaria remains a serious global health challenge, yet treatment and control programs are threatened by drug resistance. Dihydroorotate dehydrogenase (DHODH) was clinically validated as a target for treatment and prevention of malaria through human studies with DSM265, but currently no drugs against this target are in clinical use. We used structure-based computational tools including free energy perturbation (FEP+) to discover highly ligand efficient, potent, and selective pyrazole-based Plasmodium DHODH inhibitors through a scaffold hop from a pyrrole-based series. Optimized pyrazole-based compounds were identified with low nM-to-pM Plasmodium falciparum cell potency and oral activity in a humanized SCID mouse malaria infection model. The lead compound DSM1465 is more potent and has improved absorption, distribution, metabolism and excretion/pharmacokinetic (ADME/PK) properties compared to DSM265 that support the potential for once-monthly chemoprevention at a low dose. This compound meets the objective of identifying compounds with potential to be used for monthly chemoprevention in Africa to support malaria elimination efforts.
Collapse
Affiliation(s)
- Zhe Nie
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | - Roger Bonnert
- MMV
Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Jet Tsien
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Xiaoyi Deng
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Christopher Higgs
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | - Farah El Mazouni
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Xiaoyu Zhang
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Renzhe Li
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Nhi Ho
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Victoria Feher
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | - Janet Paulsen
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | - David M. Shackleford
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kasiram Katneni
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Gong Chen
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Alice C. F. Ng
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Mitchell McInerney
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Wen Wang
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jessica Saunders
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Daniel Collins
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Dandan Yan
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Peng Li
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michael Campbell
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Rahul Patil
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Atanu Ghoshal
- TCGLS, Block BN, Plot
7, Salt Lake Electronics
Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Pallab Mondal
- TCGLS, Block BN, Plot
7, Salt Lake Electronics
Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Abhijit Kundu
- TCGLS, Block BN, Plot
7, Salt Lake Electronics
Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Rajesh Chittimalla
- Syngene
Scientific Solutions Limited, KSP 9000 Campus, Plot No. 7, Neovantage, Synergy Square 2, Kolthur
Village, Shameerpet Mandal, Medchal Malkajgiri District, Hyderabad 500 078, Telangana, India
| | - Muralikumar Mahadeva
- Syngene
Scientific Solutions Limited, KSP 9000 Campus, Plot No. 7, Neovantage, Synergy Square 2, Kolthur
Village, Shameerpet Mandal, Medchal Malkajgiri District, Hyderabad 500 078, Telangana, India
| | - Sreekanth Kokkonda
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - John White
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rishi Das
- TCGLS, Block BN, Plot
7, Salt Lake Electronics
Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Partha Mukherjee
- TCGLS, Block BN, Plot
7, Salt Lake Electronics
Complex, Sector V, Kolkata 700091, West Bengal, India
| | - Iñigo Angulo-Barturen
- The
Art of Discovery, Biscay Science and Technology
Park, Astondo Bidea,
BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - María Belén Jiménez-Díaz
- The
Art of Discovery, Biscay Science and Technology
Park, Astondo Bidea,
BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - Robert Malmstrom
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | - Morgan Lawrenz
- Schrödinger
Inc., 1540 Broadway, New York, New York 10036, United States
| | | | - Pradipsinh K. Rathod
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Diana R. Tomchick
- Department
of Biophysics, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Michael J. Palmer
- MMV
Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Benoît Laleu
- MMV
Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Tian Qin
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Susan A. Charman
- Centre
for Drug Candidate Optimisation, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Margaret A. Phillips
- Department
of Biochemistry, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
8
|
Brown N, Luniewski A, Yu X, Warthan M, Liu S, Zulawinska J, Ahmad S, Congdon M, Santos W, Xiao F, Guler JL. Replication stress increases de novo CNVs across the malaria parasite genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629492. [PMID: 39803504 PMCID: PMC11722320 DOI: 10.1101/2024.12.19.629492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Changes in the copy number of large genomic regions, termed copy number variations (CNVs), contribute to important phenotypes in many organisms. CNVs are readily identified using conventional approaches when present in a large fraction of the cell population. However, CNVs that are present in only a few genomes across a population are often overlooked but important; if beneficial under specific conditions, a de novo CNV that arises in a single genome can expand during selection to create a larger population of cells with novel characteristics. While the reach of single cell methods to study de novo CNVs is increasing, we continue to lack information about CNV dynamics in rapidly evolving microbial populations. Here, we investigated de novo CNVs in the genome of the Plasmodium parasite that causes human malaria. The highly AT-rich P. falciparum genome readily accumulates CNVs that facilitate rapid adaptation to new drugs and host environments. We employed a low-input genomics approach optimized for this unique genome as well as specialized computational tools to evaluate the de novo CNV rate both before and after the application of stress. We observed a significant increase in genomewide de novo CNVs following treatment with a replication inhibitor. These stress-induced de novo CNVs encompassed genes that contribute to various cellular pathways and tended to be altered in clinical parasite genomes. This snapshot of CNV dynamics emphasizes the connection between replication stress, DNA repair, and CNV generation in this important microbial pathogen.
Collapse
Affiliation(s)
- Noah Brown
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | | | - Xuanxuan Yu
- Unifersity of Florida, Department of Biostatistics, Gainesville, FL, USA
- Unifersity of Florida, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Michelle Warthan
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Shiwei Liu
- University of Virginia, Department of Biology, Charlottesville, VA, USA
- Current affiliation: Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julia Zulawinska
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Syed Ahmad
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Molly Congdon
- Virginia Tech, Department of Chemistry, Blacksburg, VA, USA
| | - Webster Santos
- Virginia Tech, Department of Chemistry, Blacksburg, VA, USA
| | - Feifei Xiao
- Unifersity of Florida, Department of Biostatistics, Gainesville, FL, USA
| | - Jennifer L Guler
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| |
Collapse
|
9
|
Awalt JK, Su W, Nguyen W, Loi K, Jarman KE, Penington JS, Ramesh S, Fairhurst KJ, Yeo T, Park H, Uhlemann AC, Chandra Maity B, De N, Mukherjee P, Chakraborty A, Churchyard A, Famodimu MT, Delves MJ, Baum J, Mittal N, Winzeler EA, Papenfuss AT, Chowdury M, de Koning-Ward TF, Maier AG, van Dooren GG, Baud D, Brand S, Fidock DA, Jackson PF, Cowman AF, Dans MG, Sleebs BE. Exploration and characterization of the antimalarial activity of cyclopropyl carboxamides that target the mitochondrial protein, cytochrome b. Eur J Med Chem 2024; 280:116921. [PMID: 39388903 PMCID: PMC11609934 DOI: 10.1016/j.ejmech.2024.116921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Drug resistance against antimalarials is rendering them increasingly ineffective and so there is a need for the development of new antimalarials. To discover new antimalarial chemotypes a phenotypic screen of the Janssen Jumpstarter library against the P. falciparum asexual stage was undertaken, uncovering the cyclopropyl carboxamide structural hit class. Structure-activity analysis revealed that each structural moiety was largely resistant to change, although small changes led to the frontrunner compound, WJM280, which has potent asexual stage activity (EC50 40 nM) and no human cell cytotoxicity. Forward genetics uncovered that cyclopropyl carboxamide resistant parasites have mutations and an amplification in the cytochrome b gene. Cytochrome b was then verified as the target with profiling against cytochrome b drug-resistant parasites and a mitochondrial oxygen consumption assay. Accordingly, the cyclopropyl carboxamide class was shown to have slow-acting asexual stage activity and activity against male gametes and exoerythrocytic forms. Enhancing metabolic stability to attain efficacy in malaria mouse models remains a challenge in the future development of this antimalarial chemotype.
Collapse
Affiliation(s)
- Jon Kyle Awalt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Wenyin Su
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - William Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Katie Loi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia
| | - Kate E Jarman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Jocelyn S Penington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Saishyam Ramesh
- Research School of Biology, The Australian National University, Canberra, 2600, Australia
| | - Kate J Fairhurst
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Tomas Yeo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Nirupam De
- TCG Lifesciences, Kolkata, West Bengal, 700091, India
| | | | | | - Alisje Churchyard
- Department of Life Sciences, Imperial College London, South Kensington, SW7 2AZ, UK
| | - Mufuliat T Famodimu
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Michael J Delves
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, South Kensington, SW7 2AZ, UK; School of Biomedical Sciences, University of New South Wales, Sydney, 2031, Australia
| | - Nimisha Mittal
- School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Elizabeth A Winzeler
- School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Anthony T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Mrittika Chowdury
- School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia; Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, 3216, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia; Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, 3216, Australia
| | - Alexander G Maier
- Research School of Biology, The Australian National University, Canberra, 2600, Australia
| | - Giel G van Dooren
- Research School of Biology, The Australian National University, Canberra, 2600, Australia
| | - Delphine Baud
- Medicines for Malaria Venture, Geneva, 1215, Switzerland
| | - Stephen Brand
- Medicines for Malaria Venture, Geneva, 1215, Switzerland
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Paul F Jackson
- Global Public Health, Janssen R&D LLC, La Jolla, 92121, USA
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia
| | - Madeline G Dans
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia.
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
10
|
Sharma M, Lolli ML, Vyas VK. A comprehensive review of synthetic strategies and SAR studies for the discovery of PfDHODH inhibitors as antimalarial agents. Part 2: Non-DSM compounds. Bioorg Chem 2024; 153:107754. [PMID: 39241585 DOI: 10.1016/j.bioorg.2024.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Malaria remains a severe global health concern, with 249 million cases reported in 2022, according to the World Health Organization (WHO) [1]. PfDHODH is an essential enzyme in malaria parasites that helps to synthesize certain building blocks for their growth and development. It has been confirmed that targeting Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) enzyme could lead to new and effective antimalarial drugs. Inhibitors of PfDHODH have shown potential for slowing down parasite growth during both the blood and liver stages. Over the last two decades, many species selective PfDHODH inhibitors have been designed, including DSM compounds and other non-DSM compounds. In the first chapter [2] of this review, we have reviewed all synthetic schemes and structure-activity relationship (SAR) studies of DSM compounds. In this second chapter, we have compiled all the other non-DSM PfDHODH inhibitors based on dihydrothiophenones, thiazoles, hydroxyazoles, and N-alkyl-thiophene-2-carboxamides. The review not only offers an insightful overview of the synthetic methods employed but also explores into alternative routes and innovative strategies involving different catalysts and chemical reagents. A critical aspect covered in the review is the SAR studies, which provide a comprehensive understanding of how structural modifications impact the efficacy of PfDHODH inhibitors and challenges related to the discovery of PfDHODH inhibitors. This information is invaluable for scientists engaged in the development of new antimalarial drugs, offering insights into the most promising scaffolds and their synthetic techniques.
Collapse
Affiliation(s)
- Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Marco L Lolli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
11
|
Biswas P, Roy R, Ghosh K, Nath D, Samadder A, Nandi S. To quest new targets of Plasmodium parasite and their potential inhibitors to combat antimalarial drug resistance. J Parasit Dis 2024; 48:671-722. [PMID: 39493470 PMCID: PMC11527868 DOI: 10.1007/s12639-024-01687-x] [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: 11/22/2023] [Accepted: 05/08/2024] [Indexed: 11/05/2024] Open
Abstract
Malaria remains a global health challenge with significant mortality and morbidity annually, with resistant parasite strains complicating treatment efforts. There is an acute need for novel antimalarial drugs that can put a stop to the future public health crisis caused by the multi-drug resistance strains of the Plasmodium parasite. However, the discovery of these new components is very challenging in the context of the generation of multi-drug resistance properties of malaria. The novel drugs also need to have several properties involving enhanced therapeutic prospects, successful treatment capabilities, and novel mechanisms of action that will forestall the resistance. To successfully achieve this aim researchers are trying to focus on exploring promising malaria targets. Various approaches have been made for the development of drugs for malaria including the remodelling of existing drugs and the development of novel inhibitors which acts on new targets. Advancement in the study provides more information on the biology of parasites and the new targets which help in the development of novel drugs. The present review focuses on the study of novel targets of malaria parasites and subsequent inhibitors of those particular targets. Some of these targets include malarial protease, various transporter proteins, enzymes involved in the synthesis of DNA, and nucleic acids like dihydroorotate dehydrogenase, dihydrofolate reductase, apicoplast and dihydropteroate synthase. Other potential targets are also included in this review such as isoprenoid biosynthesis, farnesyl transferase of parasite, P. falciparum translational elongation factor 2, and phosphatidyl inositol 4 kinase. These promising targets have also been summed up along with their corresponding inhibitors for combating multi-drug resistance malaria.
Collapse
Affiliation(s)
- Pratyusa Biswas
- Department of Zoology, University of Kalyani, Kalyani, Nadia 741235 India
| | - Rini Roy
- Department of Zoology, University of Kalyani, Kalyani, Nadia 741235 India
| | - Kuldip Ghosh
- Department of Zoology, University of Kalyani, Kalyani, Nadia 741235 India
| | - Debjani Nath
- Department of Zoology, University of Kalyani, Kalyani, Nadia 741235 India
| | - Asmita Samadder
- Department of Zoology, University of Kalyani, Kalyani, Nadia 741235 India
| | - Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research (Affiliated to Veer Madho Singh Bhandari Uttarakhand Technical University), Kashipur, 244713 India
| |
Collapse
|
12
|
Nguyen GB, Cooper CA, McWhorter O, Sharma R, Elliot A, Ruberto A, Freitas R, Pathak AK, Kyle DE, Maher SP. Screening the Global Health Priority Box against Plasmodium berghei liver stage parasites using an inexpensive luciferase detection protocol. Malar J 2024; 23:357. [PMID: 39580415 PMCID: PMC11585928 DOI: 10.1186/s12936-024-05155-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/24/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Malaria, a disease caused by parasites of the genus Plasmodium, continues to impact many regions globally. The rise in resistance to artemisinin-based anti-malarial drugs highlights the need for new treatments. Ideally, new anti-malarials will kill the asymptomatic liver stages as well as the symptomatic blood stages. While blood stage screening assays are routine and efficient, liver stage screening assays are more complex and costly. To decrease the cost of liver stage screening, a previously reported luciferase detection protocol requiring only common laboratory reagents was adapted for testing against luciferase-expressing Plasmodium berghei liver stage parasites. METHODS After optimizing cell lysis conditions, the concentration of reagents, and the density of host hepatocytes (HepG2), the protocol was validated with 28 legacy anti-malarials to show this simple protocol produces a stable signal useful for obtaining quality small molecule potency data similar to that obtained from a high content imaging endpoint. The protocol was then used to screen the Global Health Priority Box (GHPB) and confirm the potency of hits in dose-response assays. Selectivity was determined using a galactose-based, 72 h HepG2 assay to avoid missing mitochondrial-toxic compounds due to the Crabtree effect. Receiver-operator characteristic plots were used to retroactively characterize the screens' predictive value. RESULTS Optimal luciferase signal was achieved using a lower HepG2 seed density (5 × 103 cells/well of a 384-well microtitre plate) compared to many previously reported luciferase-based screens. While producing lower signal compared to a commercial alternative, this luciferase detection method was found much more stable, with a > 3 h half-life, and robust enough for producing dose-response plots with as few as 500 sporozoites/well. A screen of the GHPB resulted in 9 hits with selective activity against P. berghei liver schizonts, including MMV674132 which exhibited 30.2 nM potency. Retrospective analyses show excellent predictive value for both anti-malarial activity and cytotoxicity. CONCLUSIONS This method is suitable for high-throughput screening at a cost nearly 20-fold less than using commercial luciferase detection kits, thereby enabling larger liver stage anti-malarial screens and hit optimization make-test cycles. Further optimization of the hits detected using this protocol is ongoing.
Collapse
Affiliation(s)
- Gia-Bao Nguyen
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Caitlin A Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Olivia McWhorter
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Ritu Sharma
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Anne Elliot
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Anthony Ruberto
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Rafael Freitas
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Ashutosh K Pathak
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr, Athens, GA, 30602, USA.
| |
Collapse
|
13
|
Sparkes PC, Famodimu MT, Alves E, Springer E, Przyborski J, Delves MJ. Mitochondrial ATP synthesis is essential for efficient gametogenesis in Plasmodium falciparum. Commun Biol 2024; 7:1525. [PMID: 39550509 PMCID: PMC11569237 DOI: 10.1038/s42003-024-07240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Plasmodium male and female gametocytes are the gatekeepers of human-to-mosquito transmission, therefore essential for propagation of malaria within a population. Whilst dormant in humans, their divergent roles during transmission become apparent soon after mosquito feeding with a rapid transformation into gametes - males forming eight motile sperm-like cells aiming to fertilise a single female gamete. Little is known about how the parasite fuels this abrupt change, and the potential role played by their large and elaborate cristate mitochondrion. Using a sex-specific antibody and functional mitochondrial labelling, we show that the male gametocyte mitochondrion is less active than that of female gametocytes and more sensitive to antimalarials targeting mitochondrial energy metabolism. Rather than a vestigial organelle discarded during male gametogenesis, we demonstrate that mitochondrial ATP synthesis is essential for its completion. Additionally, using a genetically encoded ratiometric ATP sensor, we show that gametocytes can maintain cytoplasmic ATP homeostasis in the absence of mitochondrial respiration, indicating the essentiality of the gametocyte mitochondrion for transmission alone. Together, this reveals how gametocytes responsively balance the conflicting demands of a dormant and active lifestyle, highlighting the mitochondria as a rich source of transmission-blocking targets for future drug development.
Collapse
Affiliation(s)
- Penny C Sparkes
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Eduardo Alves
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Eric Springer
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Jude Przyborski
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Michael J Delves
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
14
|
Naude M, van Heerden A, Reader J, van der Watt M, Niemand J, Joubert D, Siciliano G, Alano P, Njoroge M, Chibale K, Herreros E, Leroy D, Birkholtz LM. Eliminating malaria transmission requires targeting immature and mature gametocytes through lipoidal uptake of antimalarials. Nat Commun 2024; 15:9896. [PMID: 39548094 PMCID: PMC11568134 DOI: 10.1038/s41467-024-54144-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Novel antimalarial compounds targeting both the pathogenic and transmissible stages of the human malaria parasite, Plasmodium falciparum, would greatly benefit malaria elimination strategies. However, most compounds affecting asexual blood stage parasites show severely reduced activity against gametocytes. The impact of this activity loss on a compound's transmission-blocking activity is unclear. Here, we report the systematic evaluation of the activity loss against gametocytes and investigate the confounding factors contributing to this. A threshold for acceptable activity loss between asexual blood stage parasites and gametocytes was defined, with near-equipotent compounds required to prevent continued gametocyte maturation and onward transmission. Target abundance is not predictive of gametocytocidal activity, but instead, lipoidal uptake is the main barrier of dual activity and is influenced by distinct physicochemical properties. This study provides guidelines for the required profiles of potential dual-active antimalarial agents and facilitates the development of effective transmission-blocking compounds.
Collapse
Affiliation(s)
- Mariska Naude
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Ashleigh van Heerden
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Mariëtte van der Watt
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Jandeli Niemand
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Dorè Joubert
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Giulia Siciliano
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, viale Regina Elena 299, 00161, Rome, Italy
| | - Mathew Njoroge
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town, 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, Cape Town, 7701, 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, Cape Town, 7701, South Africa
| | | | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Hatfield, Pretoria, 0028, South Africa.
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria, 0028, South Africa.
- Department of Biochemistry, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa.
| |
Collapse
|
15
|
Calit J, Prajapati SK, Benavente ED, Araújo JE, Deng B, Miura K, Annunciato Y, Moura IMR, Usui M, Medeiros JF, Andrade CH, Silva-Mendonça S, Simeonov A, Eastman RT, Long CA, da Silva Araujo M, Williamson KC, Aguiar ACC, Bargieri DY. Pyrimidine Azepine Targets the Plasmodium bc 1 Complex and Displays Multistage Antimalarial Activity. JACS AU 2024; 4:3942-3952. [PMID: 39483245 PMCID: PMC11522906 DOI: 10.1021/jacsau.4c00674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024]
Abstract
Malaria control and elimination efforts would benefit from the identification and validation of new malaria chemotherapeutics. Recently, a transgenic Plasmodium berghei line was used to perform a series of high-throughput in vitro screens for new antimalarials acting against the parasite sexual stages. The screens identified pyrimidine azepine chemotypes with potent activity. Here, we validate the activity of PyAz90, the most potent pyrimidine azepine chemotype identified, against P. falciparum and P. vivax in the asexual and sexual stages. PyAz90 blocked parasite transmission to the mosquito vector at nanomolar concentrations and inhibited in vitro asexual parasite multiplication with a fast-action profile. Through the generation of P. falciparum PyAz90-resistant parasites and in vitro assays of mitochondrial activity, we identified cytochrome b as a molecular target of PyAz90. This work characterizes a promising chemotype that can be explored for the future development of new antimalarials targeting the Plasmodium cytochrome bc 1 complex.
Collapse
Affiliation(s)
- Juliana Calit
- Department
of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Surendra K. Prajapati
- Department
of Microbiology and Immunology, Uniformed
Services University of the Health Sciences, Bethesda, Maryland 20814-4712, United
States
| | - Ernest D. Benavente
- Laboratory
of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CS, The Netherlands
| | - Jessica E. Araújo
- Plataforma
de Produção e Infecção de Vetores da Malária−PIVEM, Laboratório de Entomologia, Fundação
Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, RO 76812-245, Brazil
- Programa
de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia/Fiocruz Rondônia, Porto Velho, RO 76812-245, Brazil
| | - Bingbing Deng
- Laboratory
of Malaria and Vector Research, National
Institute of Allergy and Infectious Diseases, National Institutes
of Health, Rockville, Maryland 20852, United States
| | - Kazutoyo Miura
- Laboratory
of Malaria and Vector Research, National
Institute of Allergy and Infectious Diseases, National Institutes
of Health, Rockville, Maryland 20852, United States
| | - Yasmin Annunciato
- Department
of Bioscience, Federal University of São
Paulo, São Paulo, SP 04021-001, Brazil
| | - Igor M. R. Moura
- Institute
of Physics of São Carlos, University
of São Paulo, São
Carlos, SP 13566-590, Brazil
| | - Miho Usui
- Department
of Microbiology and Immunology, Uniformed
Services University of the Health Sciences, Bethesda, Maryland 20814-4712, United
States
| | - Jansen F. Medeiros
- Plataforma
de Produção e Infecção de Vetores da Malária−PIVEM, Laboratório de Entomologia, Fundação
Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, RO 76812-245, Brazil
- Programa
de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia/Fiocruz Rondônia, Porto Velho, RO 76812-245, Brazil
| | - Carolina H. Andrade
- LabMol−Laboratory
for Molecular Modeling and Drug Design−Faculty of Pharmacy, Federal University of Goias, Goiania, GO 74605-220, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Institute of Informatics, Universidade Federal de Goiás, Goiania, GO 74605-170, Brazil
| | - Sabrina Silva-Mendonça
- LabMol−Laboratory
for Molecular Modeling and Drug Design−Faculty of Pharmacy, Federal University of Goias, Goiania, GO 74605-220, Brazil
- Center
for Excellence in Artificial Intelligence (CEIA), Institute of Informatics, Universidade Federal de Goiás, Goiania, GO 74605-170, Brazil
| | - Anton Simeonov
- Division
of Preclinical Innovation, National Center
for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Richard T. Eastman
- Division
of Preclinical Innovation, National Center
for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Carole A. Long
- Laboratory
of Malaria and Vector Research, National
Institute of Allergy and Infectious Diseases, National Institutes
of Health, Rockville, Maryland 20852, United States
| | - Maisa da Silva Araujo
- Plataforma
de Produção e Infecção de Vetores da Malária−PIVEM, Laboratório de Entomologia, Fundação
Oswaldo Cruz-Fiocruz Rondônia, Porto Velho, RO 76812-245, Brazil
| | - Kim C. Williamson
- Department
of Microbiology and Immunology, Uniformed
Services University of the Health Sciences, Bethesda, Maryland 20814-4712, United
States
| | - Anna Caroline C. Aguiar
- Department
of Bioscience, Federal University of São
Paulo, São Paulo, SP 04021-001, Brazil
- Department
of Microbiology, Immunology, and Parasitology.
Federal University of São Paulo, São Paulo, SP 13563-120, Brazil
| | - Daniel Y. Bargieri
- Department
of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
16
|
Li J, Docile HJ, Fisher D, Pronyuk K, Zhao L. Current Status of Malaria Control and Elimination in Africa: Epidemiology, Diagnosis, Treatment, Progress and Challenges. J Epidemiol Glob Health 2024; 14:561-579. [PMID: 38656731 PMCID: PMC11442732 DOI: 10.1007/s44197-024-00228-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
The African continent carries the greatest malaria burden in the world. Falciparum malaria especially has long been the leading cause of death in Africa. Climate, economic factors, geographical location, human intervention and unstable security are factors influencing malaria transmission. Due to repeated infections and early interventions, the proportion of clinically atypical malaria or asymptomatic plasmodium carriers has increased significantly, which easily lead to misdiagnosis and missed diagnosis. African countries have made certain progress in malaria control and elimination, including rapid diagnosis of malaria, promotion of mosquito nets and insecticides, intermittent prophylactic treatment in high-risk groups, artemisinin based combination therapies, and the development of vaccines. Between 2000 and 2022, there has been a 40% decrease in malaria incidence and a 60% reduction in mortality rate in the WHO African Region. However, many challenges are emerging in the fight against malaria in Africa, such as climate change, poverty, substandard health services and coverage, increased outdoor transmission and the emergence of new vectors, and the growing threat of resistance to antimalarial drugs and insecticides. Joint prevention and treatment, identifying molecular determinants of resistance, new drug development, expanding seasonal malaria chemo-prevention intervention population, and promoting the vaccination of RTS, S/AS01 and R21/Matrix-M may help to solve the dilemma. China's experience in eliminating malaria is conducive to Africa's malaria prevention and control, and China-Africa cooperation needs to be constantly deepened and advanced. Our review aims to help the global public develop a comprehensive understanding of malaria in Africa, thereby contributing to malaria control and elimination.
Collapse
Affiliation(s)
- Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Haragakiza Jean Docile
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | - Khrystyna Pronyuk
- Department of Infectious Diseases, O. Bogomolets National Medical University, Kyiv, Ukraine
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
17
|
Ashton T, Calic PPS, Dans MG, Ooi ZK, Zhou Q, Palandri J, Loi K, Jarman KE, Qiu D, Lehane AM, Maity BC, De N, Giannangelo C, MacRaild CA, Creek DJ, Mao EY, Gancheva MR, Wilson DW, Chowdury M, de Koning-Ward TF, Famodimu MT, Delves MJ, Pollard H, Sutherland CJ, Baud D, Brand S, Jackson PF, Cowman AF, Sleebs BE. Property and Activity Refinement of Dihydroquinazolinone-3-carboxamides as Orally Efficacious Antimalarials that Target PfATP4. J Med Chem 2024; 67:14493-14523. [PMID: 39134060 PMCID: PMC11345840 DOI: 10.1021/acs.jmedchem.4c01241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
To contribute to the global effort to develop new antimalarial therapies, we previously disclosed initial findings on the optimization of the dihydroquinazolinone-3-carboxamide class that targets PfATP4. Here we report on refining the aqueous solubility and metabolic stability to improve the pharmacokinetic profile and consequently in vivo efficacy. We show that the incorporation of heterocycle systems in the 8-position of the scaffold was found to provide the greatest attainable balance between parasite activity, aqueous solubility, and metabolic stability. Optimized analogs, including the frontrunner compound S-WJM992, were shown to inhibit PfATP4-associated Na+-ATPase activity, gave rise to a metabolic signature consistent with PfATP4 inhibition, and displayed altered activities against parasites with mutations in PfATP4. Finally, S-WJM992 showed appreciable efficacy in a malaria mouse model and blocked gamete development preventing transmission to mosquitoes. Importantly, further optimization of the dihydroquinazolinone class is required to deliver a candidate with improved pharmacokinetic and risk of resistance profiles.
Collapse
Affiliation(s)
- Trent
D. Ashton
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Petar P. S. Calic
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Madeline G. Dans
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Zi Kang Ooi
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Qingmiao Zhou
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Josephine Palandri
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Katie Loi
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Kate E. Jarman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Deyun Qiu
- Research
School of Biology, Australian National University, Canberra 2601, Australia
| | - Adele M. Lehane
- Research
School of Biology, Australian National University, Canberra 2601, Australia
| | | | - Nirupam De
- TCG
Lifesciences, Kolkata, West Bengal 700091, India
| | - Carlo Giannangelo
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal
Parade, Parkville, Victoria 3052, Australia
| | - Christopher A. MacRaild
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal
Parade, Parkville, Victoria 3052, Australia
| | - Darren J. Creek
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal
Parade, Parkville, Victoria 3052, Australia
| | - Emma Y. Mao
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Maria R. Gancheva
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Danny W. Wilson
- Research
Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Mrittika Chowdury
- School
of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
- Institute
for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria 3216, Australia
| | - Tania F. de Koning-Ward
- School
of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
- Institute
for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria 3216, Australia
| | - Mufuliat T. Famodimu
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, London WC1E 7HT, U.K.
| | - Michael J. Delves
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, London WC1E 7HT, U.K.
| | - Harry Pollard
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, London WC1E 7HT, U.K.
| | - Colin J. Sutherland
- Department
of Infection Biology, London School of Hygiene
and Tropical Medicine, London WC1E 7HT, U.K.
| | - Delphine Baud
- MMV Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, Geneva 1215, Switzerland
| | - Stephen Brand
- MMV Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, Geneva 1215, Switzerland
| | - Paul F. Jackson
- Emerging Science & Innovation, Discovery
Sciences, Janssen R&D LLC, La Jolla, California 92121, United States
| | - Alan F. Cowman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Brad E. Sleebs
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
18
|
Lawong A, Gahalawat S, Ray S, Ho N, Han Y, Ward KE, Deng X, Chen Z, Kumar A, Xing C, Hosangadi V, Fairhurst KJ, Tashiro K, Liszczak G, Shackleford DM, Katneni K, Chen G, Saunders J, Crighton E, Casas A, Robinson JJ, Imlay LS, Zhang X, Lemoff A, Zhao Z, Angulo-Barturen I, Jiménez-Díaz MB, Wittlin S, Campbell SF, Fidock DA, Laleu B, Charman SA, Ready JM, Phillips MA. Identification of potent and reversible piperidine carboxamides that are species-selective orally active proteasome inhibitors to treat malaria. Cell Chem Biol 2024; 31:1503-1517.e19. [PMID: 39084225 PMCID: PMC11531662 DOI: 10.1016/j.chembiol.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
Malaria remains a global health concern as drug resistance threatens treatment programs. We identified a piperidine carboxamide (SW042) with anti-malarial activity by phenotypic screening. Selection of SW042-resistant Plasmodium falciparum (Pf) parasites revealed point mutations in the Pf_proteasome β5 active-site (Pfβ5). A potent analog (SW584) showed efficacy in a mouse model of human malaria after oral dosing. SW584 had a low propensity to generate resistance (minimum inoculum for resistance [MIR] >109) and was synergistic with dihydroartemisinin. Pf_proteasome purification was facilitated by His8-tag introduction onto β7. Inhibition of Pfβ5 correlated with parasite killing, without inhibiting human proteasome isoforms or showing cytotoxicity. The Pf_proteasome_SW584 cryoelectron microscopy (cryo-EM) structure showed that SW584 bound non-covalently distal from the catalytic threonine, in an unexplored pocket at the β5/β6/β3 subunit interface that has species differences between Pf and human proteasomes. Identification of a reversible, species selective, orally active series with low resistance propensity provides a path for drugging this essential target.
Collapse
Affiliation(s)
- Aloysus Lawong
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Sneha Ray
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Nhi Ho
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yan Han
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Kurt E Ward
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaoyi Deng
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhe Chen
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Ashwani Kumar
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Chao Xing
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Varun Hosangadi
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kate J Fairhurst
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kyuto Tashiro
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Glen Liszczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Arturo Casas
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Joshua J Robinson
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Leah S Imlay
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Xiaoyu Zhang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Iñigo Angulo-Barturen
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - María Belén Jiménez-Díaz
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; University of Basel Kreuzstrasse 2, 4123 Allschwil, Switzerland
| | | | - David A Fidock
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benoît Laleu
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| | - Margaret A Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Dziwornu G, Seanego D, Fienberg S, Clements M, Ferreira J, Sypu VS, Samanta S, Bhana AD, Korkor CM, Garnie LF, Teixeira N, Wicht KJ, Taylor D, Olckers R, Njoroge M, Gibhard L, Salomane N, Wittlin S, Mahato R, Chakraborty A, Sevilleno N, Coyle R, Lee MCS, Godoy LC, Pasaje CF, Niles JC, Reader J, van der Watt M, Birkholtz LM, Bolscher JM, de Bruijni MHC, Coulson LB, Basarab GS, Ghorpade SR, Chibale K. 2,8-Disubstituted-1,5-naphthyridines as Dual Inhibitors of Plasmodium falciparum Phosphatidylinositol-4-kinase and Hemozoin Formation with In Vivo Efficacy. J Med Chem 2024; 67:11401-11420. [PMID: 38918002 PMCID: PMC11247499 DOI: 10.1021/acs.jmedchem.4c01154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Structure-activity relationship studies of 2,8-disubstituted-1,5-naphthyridines, previously reported as potent inhibitors of Plasmodium falciparum (Pf) phosphatidylinositol-4-kinase β (PI4K), identified 1,5-naphthyridines with basic groups at 8-position, which retained Plasmodium PI4K inhibitory activity but switched primary mode of action to the host hemoglobin degradation pathway through inhibition of hemozoin formation. These compounds showed minimal off-target inhibitory activity against the human phosphoinositide kinases and MINK1 and MAP4K kinases, which were associated with the teratogenicity and testicular toxicity observed in rats for the PfPI4K inhibitor clinical candidate MMV390048. A representative compound from the series retained activity against field isolates and lab-raised drug-resistant strains of Pf. It was efficacious in the humanized NSG mouse malaria infection model at a single oral dose of 32 mg/kg. This compound was nonteratogenic in the zebrafish embryo model of teratogenicity and has a low predicted human dose, indicating that this series has the potential to deliver a preclinical candidate for malaria.
Collapse
Affiliation(s)
- Godwin
Akpeko Dziwornu
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Donald Seanego
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Stephen Fienberg
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Monica Clements
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Jasmin Ferreira
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Venkata S. Sypu
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Sauvik Samanta
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Ashlyn D. Bhana
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Constance M. Korkor
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Larnelle F. Garnie
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Nicole Teixeira
- Department
of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kathryn J. Wicht
- Drug
Discovery and Development Centre (H3D), Department of Chemistry and
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Dale Taylor
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Ronald Olckers
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Mathew Njoroge
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Liezl Gibhard
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Nicolaas Salomane
- Drug
Discovery and Development Centre (H3D), Institute of Infectious Disease
and Molecular Medicine, University of Cape
Town, Observatory, Cape Town 7925, South Africa
| | - Sergio Wittlin
- Swiss Tropical
and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, 4001 Basel, Switzerland
| | | | | | - Nicole Sevilleno
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Rachael Coyle
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Marcus C. S. Lee
- Wellcome
Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, U.K.
| | - Luiz C. Godoy
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Charisse Flerida Pasaje
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jacquin C. Niles
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Janette Reader
- Department
of Biochemistry, Genetics and Microbiology, Institute
for Sustainable Malaria Control, University
of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Mariette van der Watt
- Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department
of Biochemistry, Genetics and Microbiology, Institute
for Sustainable Malaria Control, University
of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Judith M. Bolscher
- TropIQ Health Sciences, Transistorweg 5, 6534 AT Nijmegen, The Netherlands
| | | | - Lauren B. Coulson
- Drug
Discovery and Development Centre (H3D), Institute of Infectious Disease
and Molecular Medicine, University of Cape
Town, Observatory, Cape Town 7925, South Africa
| | - Gregory S. Basarab
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Drug
Discovery and Development Centre (H3D), Division of Clinical Pharmacology,
Department of Medicine, University of Cape
Town, Observatory 7925, South Africa
| | - Sandeep R. Ghorpade
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug
Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, 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 7701, South Africa
| |
Collapse
|
20
|
Edgar RCS, Malcolm TR, Siddiqui G, Giannangelo C, Counihan NA, Challis M, Duffy S, Chowdhury M, Marfurt J, Dans M, Wirjanata G, Noviyanti R, Daware K, Suraweera CD, Price RN, Wittlin S, Avery VM, Drinkwater N, Charman SA, Creek DJ, de Koning-Ward TF, Scammells PJ, McGowan S. On-target, dual aminopeptidase inhibition provides cross-species antimalarial activity. mBio 2024; 15:e0096624. [PMID: 38717141 PMCID: PMC11237774 DOI: 10.1128/mbio.00966-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 06/13/2024] Open
Abstract
To combat the global burden of malaria, development of new drugs to replace or complement current therapies is urgently required. Here, we show that the compound MMV1557817 is a selective, nanomolar inhibitor of both Plasmodium falciparum and Plasmodium vivax aminopeptidases M1 and M17, leading to inhibition of end-stage hemoglobin digestion in asexual parasites. MMV1557817 can kill sexual-stage P. falciparum, is active against murine malaria, and does not show any shift in activity against a panel of parasites resistant to other antimalarials. MMV1557817-resistant P. falciparum exhibited a slow growth rate that was quickly outcompeted by wild-type parasites and were sensitized to the current clinical drug, artemisinin. Overall, these results confirm MMV1557817 as a lead compound for further drug development and highlights the potential of dual inhibition of M1 and M17 as an effective multi-species drug-targeting strategy.IMPORTANCEEach year, malaria infects approximately 240 million people and causes over 600,000 deaths, mostly in children under 5 years of age. For the past decade, artemisinin-based combination therapies have been recommended by the World Health Organization as the standard malaria treatment worldwide. Their widespread use has led to the development of artemisinin resistance in the form of delayed parasite clearance, alongside the rise of partner drug resistance. There is an urgent need to develop and deploy new antimalarial agents with novel targets and mechanisms of action. Here, we report a new and potent antimalarial compound, known as MMV1557817, and show that it targets multiple stages of the malaria parasite lifecycle, is active in a preliminary mouse malaria model, and has a novel mechanism of action. Excitingly, resistance to MMV15578117 appears to be self-limiting, suggesting that development of the compound may provide a new class of antimalarial.
Collapse
Affiliation(s)
- Rebecca C S Edgar
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Tess R Malcolm
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Natalie A Counihan
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Matthew Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, Griffith University, Nathan, Queensland, Australia
| | - Mrittika Chowdhury
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Jutta Marfurt
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Madeline Dans
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Grennady Wirjanata
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | | | - Kajal Daware
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Chathura D Suraweera
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Ric N Price
- Global Health and Tropical Medicine Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Vicky M Avery
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, Queensland, Australia
| | - Nyssa Drinkwater
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Geelong, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Sheena McGowan
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
21
|
Nandal R, Kumar D, Aggarwal N, Kumar V, Narasimhan B, Marwaha RK, Sharma PC, Kumar S, Bansal N, Chopra H, Deep A. Recent advances, challenges and updates on the development of therapeutics for malaria. EXCLI JOURNAL 2024; 23:672-713. [PMID: 38887396 PMCID: PMC11180964 DOI: 10.17179/excli2023-6856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/03/2024] [Indexed: 06/20/2024]
Abstract
Malaria has developed as a serious worldwide health issue as a result of the introduction of resistant Plasmodium species strains. Because of the common chemo resistance to most of the existing drugs on the market, it poses a severe health problem and significant obstacles in drug research. Malaria treatment has evolved during the last two decades in response to Plasmodium falciparum drug sensitivity and a return of the disease in tropical areas. Plasmodium falciparum is now highly resistant to the majority of antimalarial drugs. The parasite resistance drew focus to developing novel antimalarials to combat parasite resistance. The requirement for many novel antimalarial drugs in the future year necessitates adopting various drug development methodologies. Different innovative strategies for discovering antimalarial drugs are now being examined here. This review is primarily concerned with the description of newly synthesized antimalarial compounds, i.e. Tafenoquine, Cipargamin, Ferroquine, Artefenomel, DSM265, MMV390048 designed to improve the activity of pure antimalarial enantiomers. In this review, we selected the representative malarial drugs in clinical trials, classified them with detailed targets according to their action, discussed the relationship within the human trials, and generated a summative discussion with prospective expectations.
Collapse
Affiliation(s)
- Rimmy Nandal
- Shri Baba MastNath Institute of Pharmaceutical Sciences and Research, Baba Mast Nath University, Asthal Bohar, Rohtak-124001, Haryana, India
| | - Davinder Kumar
- College of Pharmacy, PGIMS University of Health Sciences, Rohtak-124001, Haryana, India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Virender Kumar
- College of Pharmacy, PGIMS University of Health Sciences, Rohtak-124001, Haryana, India
| | | | - Rakesh Kumar Marwaha
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak 124001 Haryana, India
| | - Prabodh Chander Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Surender Kumar
- Department of Chemistry, Chaudhary Bansi Lal University, Bhiwani-127021, India
| | - Nitin Bansal
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai - 602105, Tamil Nadu, India
| | - Aakash Deep
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| |
Collapse
|
22
|
Sharma M, Pandey V, Poli G, Tuccinardi T, Lolli ML, Vyas VK. A comprehensive review of synthetic strategies and SAR studies for the discovery of PfDHODH inhibitors as antimalarial agents. Part 1: triazolopyrimidine, isoxazolopyrimidine and pyrrole-based (DSM) compounds. Bioorg Chem 2024; 146:107249. [PMID: 38493638 DOI: 10.1016/j.bioorg.2024.107249] [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: 12/29/2023] [Revised: 02/10/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
One of the deadliest infectious diseases, malaria, still has a significant impact on global morbidity and mortality. Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) catalyzes the fourth step in de novo pyrimidine nucleotide biosynthesis and has been clinically validated as an innovative and promising target for the development of novel targeted antimalarial drugs. PfDHODH inhibitors have the potential to significantly slow down parasite growth at the blood and liver stages. Several PfDHODH inhibitors based on various scaffolds have been explored over the past two decades. Among them, triazolopyrimidines, isoxazolopyrimidines, and pyrrole-based derivatives known as DSM compounds showed tremendous potential as novel antimalarial agents, and one of the triazolopyrimidine-based compounds (DSM265) was able to reach phase IIa clinical trials. DSM compounds were synthesized as PfDHODH inhibitors with various substitutions based on structure-guided medicinal chemistry approaches and further optimised as well. For the first time, this review provides an overview of all the synthetic approaches used for the synthesis, alternative synthetic routes, and novel strategies involving various catalysts and chemical reagents that have been used to synthesize DSM compounds. We have also summarized SAR study of all these PfDHODH inhibitors. In an attempt to assist readers, scientists, and researchers involved in the development of new PfDHODH inhibitors as antimalarials, this review provides accessibility of all synthetic techniques and SAR studies of the most promising triazolopyrimidines, isoxazolopyrimidines, and pyrrole-based PfDHODH inhibitors.
Collapse
Affiliation(s)
- Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Vinita Pandey
- MIT College of Pharmacy, Ramganga Vihar, Phase-II, Moradabad, UP-244001, India
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco L Lolli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 - Turin, Italy
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
23
|
Pires CV, Cassandra D, Xu S, Laleu B, Burrows JN, Adams JH. Oxidative stress changes the effectiveness of artemisinin in Plasmodium falciparum. mBio 2024; 15:e0316923. [PMID: 38323831 PMCID: PMC10936410 DOI: 10.1128/mbio.03169-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Malaria parasites have adaptive mechanisms to modulate their intracellular redox status to tolerate the enhanced oxidizing effects created by malaria fever, hemoglobinopathies and other stress conditions, including antimalaria drugs. Emerging artemisinin (ART) resistance in Plasmodium falciparum is a complex phenotype linked to the parasite's tolerance of the activated drug's oxidative damage along with changes in vesicular transport, lipid metabolism, DNA repair, and exported proteins. In an earlier study, we discovered that many of these metabolic processes are induced in P. falciparum to respond to the oxidative damage caused by artemisinin, which exhibited a highly significant overlap with the parasite's adaptive response mechanisms to survive febrile temperatures. In addition, there was a significant overlap with the parasite's survival responses to oxidative stress. In this study, we investigated these relationships further using an in vitro model to evaluate if oxidative stress and heat-shock conditions could alter the parasite's response to artemisinin. The results revealed that compared to ideal culture conditions, the antimalarial efficacy of artemisinin was significantly reduced in parasites growing in intraerythrocytic oxidative stress but not in heat-shock condition. In contrast, heat shock significantly reduced the efficacy of lumefantrine that is an important ART combination therapy partner drug. We propose that prolonged exposure to intraerythrocytic microenvironmental oxidative stress, as would occur in endemic regions with high prevalence for sickle trait and other hemoglobinopathies, can predispose malaria parasites to develop tolerance to the oxidative damage caused by antimalarial drugs like artemisinin. IMPORTANCE Emerging resistance to the frontline antimalarial drug artemisinin represents a significant threat to worldwide malaria control and elimination. The patterns of parasite changes associated with emerging resistance represent a complex array of metabolic processes evident in various genetic mutations and altered transcription profiles. Genetic factors identified in regulating P. falciparum sensitivity to artemisinin overlap with the parasite's responses to malarial fever, sickle trait, and other types of oxidative stresses, suggesting conserved inducible survival responses. In this study we show that intraerythrocytic stress conditions, oxidative stress and heat shock, can significantly decrease the sensitivity of the parasite to artemisinin and lumefantrine, respectively. These results indicate that an intraerythrocytic oxidative stress microenvironment and heat-shock condition can alter antimalarial drug efficacy. Evaluating efficacy of antimalarial drugs under ideal in vitro culture conditions may not accurately predict drug efficacy in all malaria patients.
Collapse
Affiliation(s)
- Camilla Valente Pires
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Debora Cassandra
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Shulin Xu
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Benoit Laleu
- Medicines for Malaria Venture, ICC, Geneva, Switzerland
| | | | - John H. Adams
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
24
|
Feitosa LM, Franca RRF, Ferreira MDLG, Aguiar ACC, de Souza GE, Maluf SEC, de Souza JO, Zapata L, Duarte D, Morais I, Nogueira F, Nonato MC, Pinheiro LCS, Guido RVC, Boechat N. Discovery of new piperaquine hybrid analogs linked by triazolopyrimidine and pyrazolopyrimidine scaffolds with antiplasmodial and transmission blocking activities. Eur J Med Chem 2024; 267:116163. [PMID: 38290351 DOI: 10.1016/j.ejmech.2024.116163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
The World Health Organization (WHO) estimated that there were 247 million malaria cases in 2021 worldwide, representing an increase in 2 million cases compared to 2020. The urgent need for the development of new antimalarials is underscored by specific criteria, including the requirement of new modes of action that avoid cross-drug resistance, the ability to provide single-dose cures, and efficacy against both assexual and sexual blood stages. Motivated by the promising results obtained from our research group with [1,2,4]triazolo[1,5-a]pyrimidine and pyrazolo[1,5-a]pyrimidine derivatives, we selected these molecular scaffolds as the foundation for designing two new series of piperaquine analogs as potential antimalarial candidates. The initial series of hybrids was designed by substituting one quinolinic ring of piperaquine with the 1,2,4-triazolo[1,5-a]pyrimidine or pyrazolo[1,5-a]pyrimidine nucleus. To connect the heterocyclic systems, spacers with 3, 4, or 7 methylene carbons were introduced at the 4 position of the quinoline. In the second series, we used piperazine as a spacer to link the 1,2,4-triazolo[1,5-a]pyrimidine or pyrazolo[1,5-a]pyrimidine group to the quinoline core, effectively merging both pharmacophoric groups via a rigid spacer. Our research efforts yielded promising compounds characterized by low cytotoxicity and selectivity indices exceeding 1570. These compounds displayed potent in vitro inhibitory activity in the low nanomolar range against the erythrocytic form of the parasite, encompassing both susceptible and resistant strains. Notably, these compounds did not show cross-resistance with either chloroquine or established P. falciparum inhibitors. Even though they share a pyrazolo- or triazolo-pyrimidine core, enzymatic inhibition assays revealed that these compounds had minimal inhibitory effects on PfDHODH, indicating a distinct mode of action unrelated to targeting this enzyme. We further assessed the compounds' potential to interfere with gametocyte and ookinete infectivity using mature P. falciparum gametocytes cultured in vitro. Four compounds demonstrated significant gametocyte inhibition ranging from 58 % to 86 %, suggesting potential transmission blocking activity. Finally, we evaluated the druggability of these new compounds using in silico methods, and the results indicated that these analogs had favorable physicochemical and ADME (absorption, distribution, metabolism, and excretion) properties. In summary, our research has successfully identified and characterized new piperaquine analogs based on [1,2,4]triazolo[1,5-a]pyrimidine and pyrazolo[1,5-a]pyrimidine scaffolds and has demonstrated their potential as promising candidates for the development of antimalarial drugs with distinct mechanisms of action, considerable selectivity, and P. falciparum transmission blocking activity.
Collapse
Affiliation(s)
- Livia M Feitosa
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - FIOCRUZ, Laboratorio de Sintese de Farmacos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil; Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pós Graduação em Farmacologia e Química Medicinal, Rio de Janeiro, RJ, Brazil
| | - Rodolfo Rodrigo F Franca
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - FIOCRUZ, Laboratorio de Sintese de Farmacos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| | - Maria de Lourdes G Ferreira
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - FIOCRUZ, Laboratorio de Sintese de Farmacos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| | - Anna C C Aguiar
- Universidade de São Paulo, Instituto de Física de São Carlos, Av. João Dagnone, 1.100, Jd. Santa Angelina, São Carlos, SP, Brazil; Universidade Federal de São Paulo, Departamento de Microbiologia, Imunologia e Parasitologia. Rua Botucatu 862, Vila Clementino, 04023-062, São Paulo, SP, Brazil
| | - Guilherme E de Souza
- Universidade de São Paulo, Instituto de Física de São Carlos, Av. João Dagnone, 1.100, Jd. Santa Angelina, São Carlos, SP, Brazil
| | - Sarah El Chamy Maluf
- Universidade de São Paulo, Instituto de Física de São Carlos, Av. João Dagnone, 1.100, Jd. Santa Angelina, São Carlos, SP, Brazil
| | - Juliana O de Souza
- Universidade de São Paulo, Instituto de Física de São Carlos, Av. João Dagnone, 1.100, Jd. Santa Angelina, São Carlos, SP, Brazil
| | - Luana Zapata
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Ciências BioMoleculares, Laboratório de Cristalografia de Proteínas, Avenida do Café s/n Monte Alegre, 14040-903 Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Center for the Research and Advancement of Fragments and Molecular Targets (CRAFT), Avenida do Café s/n Monte Alegre, 14040-903 Ribeirão Preto, SP, Brazil
| | - Denise Duarte
- Universidade NOVA de Lisboa, UNL, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Ines Morais
- Universidade NOVA de Lisboa, UNL, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Fatima Nogueira
- Universidade NOVA de Lisboa, UNL, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Rua da Junqueira 100, 1349-008 Lisboa, Portugal.
| | - M Cristina Nonato
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Ciências BioMoleculares, Laboratório de Cristalografia de Proteínas, Avenida do Café s/n Monte Alegre, 14040-903 Ribeirão Preto, SP, Brazil; Universidade de São Paulo, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Center for the Research and Advancement of Fragments and Molecular Targets (CRAFT), Avenida do Café s/n Monte Alegre, 14040-903 Ribeirão Preto, SP, Brazil.
| | - Luiz C S Pinheiro
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - FIOCRUZ, Laboratorio de Sintese de Farmacos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil; Universidade do Estado do Rio de Janeiro, UERJ, Faculdade de Formação de Professores, Departamento de Ciências, Rua Dr. Francisco Portela, 1470, Patronato, 24435-005, São Gonçalo, RJ, Brazil.
| | - Rafael V C Guido
- Universidade de São Paulo, Instituto de Física de São Carlos, Av. João Dagnone, 1.100, Jd. Santa Angelina, São Carlos, SP, Brazil.
| | - Nubia Boechat
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - FIOCRUZ, Laboratorio de Sintese de Farmacos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil; Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Programa de Pós Graduação em Farmacologia e Química Medicinal, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
25
|
Chery-Karschney L, Patrapuvich R, Mudeppa DG, Kokkonda S, Chakrabarti R, Sriwichai P, O'Connor RM, Rathod PK, White J. Tartrolon E, a secondary metabolite of a marine symbiotic bacterium, is a potent inhibitor of asexual and sexual Plasmodium falciparum. Antimicrob Agents Chemother 2024; 68:e0068423. [PMID: 38193705 PMCID: PMC10848769 DOI: 10.1128/aac.00684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/10/2023] [Indexed: 01/10/2024] Open
Abstract
Due to the spread of resistance to front-line artemisinin derivatives worldwide, there is a need for new antimalarials. Tartrolon E (TrtE), a secondary metabolite of a symbiotic bacterium of marine bivalve mollusks, is a promising antimalarial because it inhibits the growth of sexual and asexual blood stages of Plasmodium falciparum at sub-nanomolar levels. The potency of TrtE warrants further investigation into its mechanism of action, cytotoxicity, and ease with which parasites may evolve resistance to it.
Collapse
Affiliation(s)
| | - Rapatbhorn Patrapuvich
- Drug Research Unit for Malaria, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Sreekanth Kokkonda
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Rimi Chakrabarti
- Department of Chemistry, University of Washington, Seattle, Washington, USA
- Department of Medicine, Goa Medical College and Hospital, Bambolim, Goa, India
| | - Patchara Sriwichai
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Roberta M. O'Connor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - John White
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| |
Collapse
|
26
|
Xie SC, Wang Y, Morton CJ, Metcalfe RD, Dogovski C, Pasaje CFA, Dunn E, Luth MR, Kumpornsin K, Istvan ES, Park JS, Fairhurst KJ, Ketprasit N, Yeo T, Yildirim O, Bhebhe MN, Klug DM, Rutledge PJ, Godoy LC, Dey S, De Souza ML, Siqueira-Neto JL, Du Y, Puhalovich T, Amini M, Shami G, Loesbanluechai D, Nie S, Williamson N, Jana GP, Maity BC, Thomson P, Foley T, Tan DS, Niles JC, Han BW, Goldberg DE, Burrows J, Fidock DA, Lee MCS, Winzeler EA, Griffin MDW, Todd MH, Tilley L. Reaction hijacking inhibition of Plasmodium falciparum asparagine tRNA synthetase. Nat Commun 2024; 15:937. [PMID: 38297033 PMCID: PMC10831071 DOI: 10.1038/s41467-024-45224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024] Open
Abstract
Malaria poses an enormous threat to human health. With ever increasing resistance to currently deployed drugs, breakthrough compounds with novel mechanisms of action are urgently needed. Here, we explore pyrimidine-based sulfonamides as a new low molecular weight inhibitor class with drug-like physical parameters and a synthetically accessible scaffold. We show that the exemplar, OSM-S-106, has potent activity against parasite cultures, low mammalian cell toxicity and low propensity for resistance development. In vitro evolution of resistance using a slow ramp-up approach pointed to the Plasmodium falciparum cytoplasmic asparaginyl-tRNA synthetase (PfAsnRS) as the target, consistent with our finding that OSM-S-106 inhibits protein translation and activates the amino acid starvation response. Targeted mass spectrometry confirms that OSM-S-106 is a pro-inhibitor and that inhibition of PfAsnRS occurs via enzyme-mediated production of an Asn-OSM-S-106 adduct. Human AsnRS is much less susceptible to this reaction hijacking mechanism. X-ray crystallographic studies of human AsnRS in complex with inhibitor adducts and docking of pro-inhibitors into a model of Asn-tRNA-bound PfAsnRS provide insights into the structure-activity relationship and the selectivity mechanism.
Collapse
Affiliation(s)
- Stanley C Xie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Yinuo Wang
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Craig J Morton
- Biomedical Manufacturing Program, CSIRO, Clayton South, VIC, Australia
| | - Riley D Metcalfe
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Con Dogovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Charisse Flerida A Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Elyse Dunn
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Madeline R Luth
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Krittikorn Kumpornsin
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Calibr, Division of the Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Eva S Istvan
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Joon Sung Park
- Research Institute of Pharmaceutical Sciences and Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kate J Fairhurst
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Nutpakal Ketprasit
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Tomas Yeo
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Okan Yildirim
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Dana M Klug
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Peter J Rutledge
- School of Chemistry, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Luiz C Godoy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sumanta Dey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mariana Laureano De Souza
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jair L Siqueira-Neto
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yawei Du
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Tanya Puhalovich
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Mona Amini
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gerry Shami
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | | | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gouranga P Jana
- TCG Lifesciences Private Limited, Salt-Lake Electronics Complex, Kolkata, India
| | - Bikash C Maity
- TCG Lifesciences Private Limited, Salt-Lake Electronics Complex, Kolkata, India
| | - Patrick Thomson
- School of Chemistry, The University of Edinburgh, Edinburgh, EH9 3JJ, UK
| | - Thomas Foley
- School of Chemistry, The University of Edinburgh, Edinburgh, EH9 3JJ, UK
| | - Derek S Tan
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences and Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeremy Burrows
- Medicines for Malaria Venture, 20, Route de Pré-Bois, 1215, Geneva 15, Switzerland
| | - David A Fidock
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Marcus C S Lee
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Wellcome Centre for Anti-Infectives Research, Biological Chemistry and Drug Discovery, University of Dundee, Dundee, DD1 4HN, UK
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Matthew H Todd
- School of Pharmacy, University College London, London, WC1N 1AX, UK.
- Structural Genomics Consortium, University College London, London, WC1N 1AX, UK.
| | - Leann Tilley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
27
|
Cheuka PM, Njaria P, Mayoka G, Funjika E. Emerging Drug Targets for Antimalarial Drug Discovery: Validation and Insights into Molecular Mechanisms of Function. J Med Chem 2024; 67:838-863. [PMID: 38198596 DOI: 10.1021/acs.jmedchem.3c01828] [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: 01/12/2024]
Abstract
Approximately 619,000 malaria deaths were reported in 2021, and resistance to recommended drugs, including artemisinin-combination therapies (ACTs), threatens malaria control. Treatment failure with ACTs has been found to be as high as 93% in northeastern Thailand, and parasite mutations responsible for artemisinin resistance have already been reported in some African countries. Therefore, there is an urgent need to identify alternative treatments with novel targets. In this Perspective, we discuss some promising antimalarial drug targets, including enzymes involved in proteolysis, DNA and RNA metabolism, protein synthesis, and isoprenoid metabolism. Other targets discussed are transporters, Plasmodium falciparum acetyl-coenzyme A synthetase, N-myristoyltransferase, and the cyclic guanosine monophosphate-dependent protein kinase G. We have outlined mechanistic details, where these are understood, underpinning the biological roles and hence druggability of such targets. We believe that having a clear understanding of the underlying chemical interactions is valuable to medicinal chemists in their quest to design appropriate inhibitors.
Collapse
Affiliation(s)
- Peter Mubanga Cheuka
- Department of Chemistry, School of Natural Sciences, University of Zambia, P.O. Box 32379, Lusaka 10101, Zambia
| | - Paul Njaria
- Department of Pharmacognosy and Pharmaceutical Chemistry, Kenyatta University, P.O. Box 14548-00400, Nairobi 00100, Kenya
| | - Godfrey Mayoka
- Department of Pharmacology and Pharmacognosy, School of Pharmacy, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000-00200, Nairobi 00100, Kenya
| | - Evelyn Funjika
- Department of Chemistry, School of Natural Sciences, University of Zambia, P.O. Box 32379, Lusaka 10101, Zambia
| |
Collapse
|
28
|
de Sousa NF, de Araújo IMA, Rodrigues TCML, da Silva PR, de Moura JP, Scotti MT, Scotti L. Proposition of In silico Pharmacophore Models for Malaria: A Review. Comb Chem High Throughput Screen 2024; 27:2525-2543. [PMID: 37815185 DOI: 10.2174/0113862073247691230925062440] [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: 03/05/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 10/11/2023]
Abstract
In the field of medicinal chemistry, the concept of pharmacophore refers to the specific region of a molecule that possesses essential structural and chemical characteristics for binding to a receptor and eliciting biological activity. Understanding the pharmacophore is crucial for drug research and development, as it allows the design of new drugs. Malaria, a widespread disease, is commonly treated with chloroquine and artemisinin, but the emergence of parasite resistance limits their effectiveness. This study aims to explore computer simulations to discover a specific pharmacophore for Malaria, providing new alternatives for its treatment. A literature review was conducted, encompassing articles proposing a pharmacophore for Malaria, gathered from the "Web of Science" database, with a focus on recent publications to ensure up-to-date analysis. The selected articles employed diverse methods, including ligand-based and structurebased approaches, integrating molecular structure and biological activity data to yield comprehensive analyses. Affinity evaluation between the proposed pharmacophore and the target receptor involved calculating free energy to quantify their interaction. Multiple linear regression was commonly utilized, though it is sensitive to multicollinearity issues. Another recurrent methodology was the use of the Schrödinger package, employing tools such as the Phase module and the OPLS force field for interaction analysis. Pharmacophore model proposition allows threedimensional representations guiding the synthesis and design of new biologically active compounds, offering a promising avenue for discovering therapeutic agents to combat Malaria.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Igor Mikael Alves de Araújo
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | | | - Pablo Rayff da Silva
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Jéssica Paiva de Moura
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| | - Luciana Scotti
- Postgraduate Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa-PB, Brazil
| |
Collapse
|
29
|
Abdelkhalek AS, Attia MS, Kamal MA. Triazolopyrimidine Derivatives: An Updated Review on Recent Advances in Synthesis, Biological Activities and Drug Delivery Aspects. Curr Med Chem 2024; 31:1896-1919. [PMID: 36852819 DOI: 10.2174/0929867330666230228120416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 03/01/2023]
Abstract
Molecules containing triazolopyrimidine core showed diverse biological activities, including anti-Alzheimer's, anti-diabetes, anti-cancer, anti-microbial, anti-tuberculosis, anti-viral, anti-malarial, anti-inflammatory, anti-parkinsonism, and anti-glaucoma activities. Triazolopyrimidines have 8 isomeric structures, including the most stable 1,2,4-triazolo[1,5- a] pyrimidine ones. Triazolopyrimidines were obtained by using various chemical reactions, including a) 1,2,4-triazole nucleus annulation to pyrimidine, b) pyrimidines annulation to 1,2,4-triazole structure, c) 1,2,4-triazolo[l,5-a] pyrimidines rearrangement, and d) pyrimidotetrazine rearrangement. This review discusses synthetic methods, recent pharmacological actions and drug delivery perspectives of triazolopyrimidines.
Collapse
Affiliation(s)
- Ahmed S Abdelkhalek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed S Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mohammad A Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia, Bangladesh
- Novel Global Community Educational Foundation, Enzymoics, 7 Peterlee Place, Hebersham, NSW, 2770, Australia
| |
Collapse
|
30
|
McLellan JL, Sausman W, Reers AB, Bunnik EM, Hanson KK. Single-cell quantitative bioimaging of P. berghei liver stage translation. mSphere 2023; 8:e0054423. [PMID: 37909773 PMCID: PMC10732057 DOI: 10.1128/msphere.00544-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
IMPORTANCE Plasmodium parasites cause malaria in humans. New multistage active antimalarial drugs are needed, and a promising class of drugs targets the core cellular process of translation, which has many potential molecular targets. During the obligate liver stage, Plasmodium parasites grow in metabolically active hepatocytes, making it challenging to study core cellular processes common to both host cells and parasites, as the signal from the host typically overwhelms that of the parasite. Here, we present and validate a flexible assay to quantify Plasmodium liver stage translation using a technique to fluorescently label the newly synthesized proteins of both host and parasite followed by computational separation of their respective nascent proteomes in confocal image sets. We use the assay to determine whether a test set of known compounds are direct or indirect liver stage translation inhibitors and show that the assay can also predict the mode of action for novel antimalarial compounds.
Collapse
Affiliation(s)
- James L. McLellan
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - William Sausman
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Ashley B. Reers
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Evelien M. Bunnik
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Kirsten K. Hanson
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
31
|
Luo AP, Giannangelo C, Siddiqui G, Creek DJ. Promising antimalarial hits from phenotypic screens: a review of recently-described multi-stage actives and their modes of action. Front Cell Infect Microbiol 2023; 13:1308193. [PMID: 38162576 PMCID: PMC10757594 DOI: 10.3389/fcimb.2023.1308193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Over the last two decades, global malaria cases caused by Plasmodium falciparum have declined due to the implementation of effective treatments and the use of insecticides. However, the COVID-19 pandemic caused major disruption in the timely delivery of medical goods and diverted public health resources, impairing malaria control. The emergence of resistance to all existing frontline antimalarials underpins an urgent need for new antimalarials with novel mechanisms of action. Furthermore, the need to reduce malaria transmission and/or prevent malaria infection has shifted the focus of antimalarial research towards the discovery of compounds that act beyond the symptomatic blood stage and also impact other parasite life cycle stages. Phenotypic screening has been responsible for the majority of new antimalarial lead compounds discovered over the past 10 years. This review describes recently reported novel antimalarial hits that target multiple parasite stages and were discovered by phenotypic screening during the COVID-19 pandemic. Their modes of action and targets in blood stage parasites are also discussed.
Collapse
Affiliation(s)
| | | | - Ghizal Siddiqui
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Darren J. Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
32
|
Lima Costa AH, Bezerra KS, de Lima Neto JX, Oliveira JIN, Galvão DS, Fulco UL. Deciphering Interactions between Potential Inhibitors and the Plasmodium falciparum DHODH Enzyme: A Computational Perspective. J Phys Chem B 2023; 127:9461-9475. [PMID: 37897437 DOI: 10.1021/acs.jpcb.3c05738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
Abstract
Malaria is a parasitic disease that, in its most severe form, can even lead to death. Insect-resistant vectors, insufficiently effective vaccines, and drugs that cannot stop parasitic infestations are making the fight against the disease increasingly difficult. It is known that the enzyme dihydroorotate dehydrogenase (DHODH) is of paramount importance for the synthesis of pyrimidine from the Plasmodium precursor, that is, for its growth and reproduction. Therefore, its blockade can lead to disruption of the parasite's life cycle in the vertebrate host. In this scenario, PfDHODH inhibitors have been considered candidates for a new therapy to stop the parasitic energy source. Given what is known, in this work, we applied molecular fractionation with conjugated caps (MFCC) in the framework of the quantum formalism of density functional theory (DFT) to evaluate the energies of the interactions between the enzyme and the different triazolopyrimidines (DSM483, DMS557, and DSM1), including a complex carrying the mutation C276F. From these results, it was possible to identify the main features of each system, focusing on the wild-type and mutant PfDHODH and examining the major amino acid residues that are part of the four complexes. Our analysis provides new information that can be used to develop new drugs that could prove to be more effective alternatives to present antimalarial drugs.
Collapse
Affiliation(s)
- Aranthya Hevelly Lima Costa
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970 Natal-RN, Brazil
| | - Katyanna Sales Bezerra
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970 Natal-RN, Brazil
- Applied Physics Department, University of Campinas, 130838-59 Campinas, São Paulo, Brazil
| | - José Xavier de Lima Neto
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970 Natal-RN, Brazil
| | - Jonas Ivan Nobre Oliveira
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970 Natal-RN, Brazil
| | - Douglas Soares Galvão
- Applied Physics Department, University of Campinas, 130838-59 Campinas, São Paulo, Brazil
| | - Umberto Laino Fulco
- Departamento de Biofísica e Farmacologia, Universidade Federal do Rio Grande do Norte, 59072-970 Natal-RN, Brazil
| |
Collapse
|
33
|
Gudla CS, Selvam V, Selvaraj SS, Tripathi R, Joshi P, Shaham SH, Singh M, Shandil RK, Habib S, Narayanan S. Novel Baicalein-Derived Inhibitors of Plasmodium falciparum. Pathogens 2023; 12:1242. [PMID: 37887758 PMCID: PMC10610289 DOI: 10.3390/pathogens12101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Malaria, a life-threatening mosquito-borne disease caused by Plasmodium parasites, continues to pose a significant global health burden. Despite notable progress in combating the disease in recent years, malaria remains prevalent in many regions, particularly in Southeast Asia and most of sub-Saharan Africa, where it claims hundreds of thousands of lives annually. Flavonoids, such as the baicalein class of compounds, are known to have antimalarial properties. In this study, we rationally designed and synthesized a series of baicalein derivatives and identified a lead compound, FNDR-10132, that displayed potent in vitro antimalarial activity against Plasmodium falciparum (P. falciparum), both chloroquine-sensitive (60 nM) and chloroquine-resistant (177 nM) parasites. FNDR-10132 was evaluated for its antimalarial activity in vivo against the chloroquine-resistant strain Plasmodium yoelii N67 in Swiss mice. The oral administration of 100 mg/kg of FNDR-10132 showed 44% parasite suppression on day 4, with a mean survival time of 13.5 ± 2.3 days vs. 8.4 ± 2.3 days of control. Also, FNDR-10132 displayed equivalent activity against the resistant strains of P. falciparum in the 200-300 nM range. This study offers a novel series of antimalarial compounds that could be developed into potent drugs against chloroquine-resistant malarial parasites through further chemistry and DMPK optimization.
Collapse
Affiliation(s)
| | - Vignesh Selvam
- Foundation for Neglected Disease Research, Bangalore 561203, Karnataka, India
| | | | - Renu Tripathi
- Molecular Microbiology and Immunology, CSIR—Central Drug Research Institute, Lucknow 226301, Uttar Pradesh, India
| | - Prince Joshi
- Molecular Microbiology and Immunology, CSIR—Central Drug Research Institute, Lucknow 226301, Uttar Pradesh, India
| | - Salique Hassan Shaham
- Molecular Microbiology and Immunology, CSIR—Central Drug Research Institute, Lucknow 226301, Uttar Pradesh, India
| | - Mayas Singh
- Foundation for Neglected Disease Research, Bangalore 561203, Karnataka, India
| | | | - Saman Habib
- Biochemistry and Structural Biology, CSIR—Central Drug Research Institute, Lucknow 226301, Uttar Pradesh, India
| | - Shridhar Narayanan
- Foundation for Neglected Disease Research, Bangalore 561203, Karnataka, India
| |
Collapse
|
34
|
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
|
35
|
Mandt REK, Luth MR, Tye MA, Mazitschek R, Ottilie S, Winzeler EA, Lafuente-Monasterio MJ, Gamo FJ, Wirth DF, Lukens AK. Diverse evolutionary pathways challenge the use of collateral sensitivity as a strategy to suppress resistance. eLife 2023; 12:e85023. [PMID: 37737220 PMCID: PMC10695565 DOI: 10.7554/elife.85023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
Drug resistance remains a major obstacle to malaria control and eradication efforts, necessitating the development of novel therapeutic strategies to treat this disease. Drug combinations based on collateral sensitivity, wherein resistance to one drug causes increased sensitivity to the partner drug, have been proposed as an evolutionary strategy to suppress the emergence of resistance in pathogen populations. In this study, we explore collateral sensitivity between compounds targeting the Plasmodium dihydroorotate dehydrogenase (DHODH). We profiled the cross-resistance and collateral sensitivity phenotypes of several DHODH mutant lines to a diverse panel of DHODH inhibitors. We focus on one compound, TCMDC-125334, which was active against all mutant lines tested, including the DHODH C276Y line, which arose in selections with the clinical candidate DSM265. In six selections with TCMDC-125334, the most common mechanism of resistance to this compound was copy number variation of the dhodh locus, although we did identify one mutation, DHODH I263S, which conferred resistance to TCMDC-125334 but not DSM265. We found that selection of the DHODH C276Y mutant with TCMDC-125334 yielded additional genetic changes in the dhodh locus. These double mutant parasites exhibited decreased sensitivity to TCMDC-125334 and were highly resistant to DSM265. Finally, we tested whether collateral sensitivity could be exploited to suppress the emergence of resistance in the context of combination treatment by exposing wildtype parasites to both DSM265 and TCMDC-125334 simultaneously. This selected for parasites with a DHODH V532A mutation which were cross-resistant to both compounds and were as fit as the wildtype parent in vitro. The emergence of these cross-resistant, evolutionarily fit parasites highlights the mutational flexibility of the DHODH enzyme.
Collapse
Affiliation(s)
- Rebecca EK Mandt
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Madeline R Luth
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
| | - Mark A Tye
- Center for Systems Biology, Massachusetts General HospitalBostonUnited States
- Harvard Graduate School of Arts and SciencesCambridgeUnited States
| | - Ralph Mazitschek
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Center for Systems Biology, Massachusetts General HospitalBostonUnited States
| | - Sabine Ottilie
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
| | - Elizabeth A Winzeler
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
- Skaggs School of Pharmaceutical Sciences, University of California, San DiegoLa JollaUnited States
| | | | - Francisco Javier Gamo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKlineMadridSpain
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Infectious Disease and Microbiome Program, The Broad InstituteCambridgeUnited States
| | - Amanda K Lukens
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Infectious Disease and Microbiome Program, The Broad InstituteCambridgeUnited States
| |
Collapse
|
36
|
Abstract
Apicomplexan parasites constitute more than 6,000 species infecting a wide range of hosts. These include important pathogens such as those causing malaria and toxoplasmosis. Their evolutionary emergence coincided with the dawn of animals. Mitochondrial genomes of apicomplexan parasites have undergone dramatic reduction in their coding capacity, with genes for only three proteins and ribosomal RNA genes present in scrambled fragments originating from both strands. Different branches of the apicomplexans have undergone rearrangements of these genes, with Toxoplasma having massive variations in gene arrangements spread over multiple copies. The vast evolutionary distance between the parasite and the host mitochondria has been exploited for the development of antiparasitic drugs, especially those used to treat malaria, wherein inhibition of the parasite mitochondrial respiratory chain is selectively targeted with little toxicity to the host mitochondria. We describe additional unique characteristics of the parasite mitochondria that are being investigated and provide greater insights into these deep-branching eukaryotic pathogens.
Collapse
Affiliation(s)
- Ian M Lamb
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA;
| | - Ijeoma C Okoye
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA;
| | - Michael W Mather
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA;
| | - Akhil B Vaidya
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
37
|
Stadler E, Maiga M, Friedrich L, Thathy V, Demarta-Gatsi C, Dara A, Sogore F, Striepen J, Oeuvray C, Djimdé AA, Lee MCS, Dembélé L, Fidock DA, Khoury DS, Spangenberg T. Propensity of selecting mutant parasites for the antimalarial drug cabamiquine. Nat Commun 2023; 14:5205. [PMID: 37626093 PMCID: PMC10457284 DOI: 10.1038/s41467-023-40974-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
We report an analysis of the propensity of the antimalarial agent cabamiquine, a Plasmodium-specific eukaryotic elongation factor 2 inhibitor, to select for resistant Plasmodium falciparum parasites. Through in vitro studies of laboratory strains and clinical isolates, a humanized mouse model, and volunteer infection studies, we identified resistance-associated mutations at 11 amino acid positions. Of these, six (55%) were present in more than one infection model, indicating translatability across models. Mathematical modelling suggested that resistant mutants were likely pre-existent at the time of drug exposure across studies. Here, we estimated a wide range of frequencies of resistant mutants across the different infection models, much of which can be attributed to stochastic differences resulting from experimental design choices. Structural modelling implicates binding of cabamiquine to a shallow mRNA binding site adjacent to two of the most frequently identified resistance mutations.
Collapse
Affiliation(s)
- Eva Stadler
- The Kirby Institute, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Mohamed Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Faculté de Pharmacie, Malaria Research and Training Center (MRTC), Point G, PB1805, Bamako, Mali
| | - Lukas Friedrich
- Medicinal Chemistry & Drug Design Global Research & Development, Discovery Technologies, Merck Healthcare, 64293, Darmstadt, Germany
| | - Vandana Thathy
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Claudia Demarta-Gatsi
- Global Health Institute of Merck, Ares Trading S.A., (an affiliate of Merck KGaA, Darmstadt, Germany), 1262, Eysins, Switzerland
| | - Antoine Dara
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Faculté de Pharmacie, Malaria Research and Training Center (MRTC), Point G, PB1805, Bamako, Mali
| | - Fanta Sogore
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Faculté de Pharmacie, Malaria Research and Training Center (MRTC), Point G, PB1805, Bamako, Mali
| | - Josefine Striepen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Weill Cornell Medical College, New York, NY, 10021, USA
| | - Claude Oeuvray
- Global Health Institute of Merck, Ares Trading S.A., (an affiliate of Merck KGaA, Darmstadt, Germany), 1262, Eysins, Switzerland
| | - Abdoulaye A Djimdé
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Faculté de Pharmacie, Malaria Research and Training Center (MRTC), Point G, PB1805, Bamako, Mali
| | - Marcus C S Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, CB10 1SA, Hinxton, UK
- Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, DD1 4HN, Scotland, UK
| | - Laurent Dembélé
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Faculté de Pharmacie, Malaria Research and Training Center (MRTC), Point G, PB1805, Bamako, Mali.
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - David S Khoury
- The Kirby Institute, UNSW Sydney, Kensington, NSW, 2052, Australia.
| | - Thomas Spangenberg
- Global Health Institute of Merck, Ares Trading S.A., (an affiliate of Merck KGaA, Darmstadt, Germany), 1262, Eysins, Switzerland.
| |
Collapse
|
38
|
Pinder C, Lebedinec R, Levine TP, Birch M, Oliver JD. Characterisation of putative class 1A DHODH-like proteins from Mucorales and dematiaceous mould species. PLoS One 2023; 18:e0289441. [PMID: 37531380 PMCID: PMC10395836 DOI: 10.1371/journal.pone.0289441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Olorofim is a new antifungal in clinical development which has a novel mechanism of action against dihydroorotate dehydrogenase (DHODH). DHODH form a ubiquitous family of enzymes in the de novo pyrimidine biosynthetic pathway and are split into class 1A, class 1B and class 2. Olorofim specifically targets the fungal class 2 DHODH present in a range of pathogenic moulds. The nature and number of DHODH present in many fungal species have not been addressed for large clades of this kingdom. Mucorales species do not respond to olorofim; previous work suggests they have only class 1A DHODH and so lack the class 2 target that olorofim inhibits. The dematiaceous moulds have mixed susceptibility to olorofim, yet previous analyses imply that they have class 2 DHODH. As this is at odds with their intermediate susceptibility to olorofim, we hypothesised that these pathogens may maintain a second class of DHODH, facilitating pyrimidine biosynthesis in the presence of olorofim. The aim of this study was to investigate the DHODH repertoire of clinically relevant species of Mucorales and dematiaceous moulds to further characterise these pathogens and understand variations in olorofim susceptibility. Using bioinformatic analysis, S. cerevisiae complementation and biochemical assays of recombinant protein, we provide the first evidence that two representative members of the Mucorales have only class 1A DHODH, substantiating a lack of olorofim susceptibility. In contrast, bioinformatic analyses initially suggested that seven dematiaceous species appeared to harbour both class 1A-like and class 2-like DHODH genes. However, further experimental investigation of the putative class 1A-like genes through yeast complementation and biochemical assays characterised them as dihydrouracil oxidases rather than DHODHs. These data demonstrate variation in dematiaceous mould olorofim susceptibility is not due to a secondary DHODH and builds on the growing picture of fungal dihydrouracil oxidases as an example of horizontal gene transfer.
Collapse
Affiliation(s)
| | | | - Tim P Levine
- UCL Institute of Ophthalmology, London, United Kingdom
| | | | | |
Collapse
|
39
|
Xie SC, Wang Y, Morton CJ, Metcalfe RD, Dogovski C, Pasaje CFA, Dunn E, Luth MR, Kumpornsin K, Istvan ES, Park JS, Fairhurst KJ, Ketprasit N, Yeo T, Yildirim O, Bhebhe MN, Klug DM, Rutledge PJ, Godoy LC, Dey S, De Souza ML, Siqueira-Neto JL, Du Y, Puhalovich T, Amini M, Shami G, Loesbanluechai D, Nie S, Williamson N, Jana GP, Maity BC, Thomson P, Foley T, Tan DS, Niles JC, Han BW, Goldberg DE, Burrows J, Fidock DA, Lee MC, Winzeler EA, Griffin MDW, Todd MH, Tilley L. Reaction hijacking inhibition of Plasmodium falciparum asparagine tRNA synthetase. RESEARCH SQUARE 2023:rs.3.rs-3198291. [PMID: 37546892 PMCID: PMC10402266 DOI: 10.21203/rs.3.rs-3198291/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Malaria poses an enormous threat to human health. With ever increasing resistance to currently deployed drugs, breakthrough compounds with novel mechanisms of action are urgently needed. Here, we explore pyrimidine-based sulfonamides as a new low molecular weight inhibitor class with drug-like physical parameters and a synthetically accessible scaffold. We show that the exemplar, OSM-S-106, has potent activity against parasite cultures, low mammalian cell toxicity and low propensity for resistance development. In vitro evolution of resistance using a slow ramp-up approach pointed to the Plasmodium falciparum cytoplasmic asparaginyl tRNA synthetase (PfAsnRS) as the target, consistent with our finding that OSM-S-106 inhibits protein translation and activates the amino acid starvation response. Targeted mass spectrometry confirms that OSM-S-106 is a pro-inhibitor and that inhibition of PfAsnRS occurs via enzyme-mediated production of an Asn-OSM-S-106 adduct. Human AsnRS is much less susceptible to this reaction hijacking mechanism. X-ray crystallographic studies of human AsnRS in complex with inhibitor adducts and docking of pro-inhibitors into a model of Asn-tRNA-bound PfAsnRS provide insights into the structure activity relationship and the selectivity mechanism.
Collapse
Affiliation(s)
- Stanley C. Xie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yinuo Wang
- School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Craig J. Morton
- Biomedical Manufacturing Program, CSIRO, Clayton South, Australia
| | - Riley D. Metcalfe
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Con Dogovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Elyse Dunn
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Madeline R Luth
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Krittikorn Kumpornsin
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, CB10 1SA, United Kingdom
- Calibr, Division of the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eva S Istvan
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis, USA
| | - Joon Sung Park
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Kate J. Fairhurst
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Nutpakal Ketprasit
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Tomas Yeo
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Okan Yildirim
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Dana M. Klug
- School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Peter J. Rutledge
- School of Chemistry, University of Sydney, Camperdown, NSW 2006, Australia
| | - Luiz C. Godoy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sumanta Dey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mariana Laureano De Souza
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Jair L. Siqueira-Neto
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Yawei Du
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Tanya Puhalovich
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mona Amini
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gerry Shami
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gouranga P. Jana
- TCG Lifesciences Private Limited, Salt-lake Electronics Complex, Kolkata, India
| | - Bikash C. Maity
- TCG Lifesciences Private Limited, Salt-lake Electronics Complex, Kolkata, India
| | - Patrick Thomson
- School of Chemistry, The University of Edinburgh, Edinburgh EH9 3JJ, United Kingdom
| | - Thomas Foley
- School of Chemistry, The University of Edinburgh, Edinburgh EH9 3JJ, United Kingdom
| | - Derek S. Tan
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine, Washington University in St. Louis, USA
| | - Jeremy Burrows
- Medicines for Malaria Venture, 20, Route de Pré-Bois 1215, Geneva 15, Switzerland
| | - David A. Fidock
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Marcus C.S. Lee
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, CB10 1SA, United Kingdom
- Wellcome Centre for Anti-Infectives Research, Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 4HN, United Kingdom
| | - Elizabeth A. Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Michael D. W. Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Matthew H. Todd
- School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
- Structural Genomics Consortium, University College London, London WC1N 1AX, United Kingdom
| | - Leann Tilley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
40
|
McLellan JL, Sausman W, Reers AB, Bunnik EM, Hanson KK. Single-cell quantitative bioimaging of P. berghei liver stage translation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547872. [PMID: 37461595 PMCID: PMC10350035 DOI: 10.1101/2023.07.05.547872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Plasmodium parasite resistance to existing antimalarial drugs poses a devastating threat to the lives of many who depend on their efficacy. New antimalarial drugs and novel drug targets are in critical need, along with novel assays to accelerate their identification. Given the essentiality of protein synthesis throughout the complex parasite lifecycle, translation inhibitors are a promising drug class, capable of targeting the disease-causing blood stage of infection, as well as the asymptomatic liver stage, a crucial target for prophylaxis. To identify compounds capable of inhibiting liver stage parasite translation, we developed an assay to visualize and quantify translation in the P. berghei-HepG2 infection model. After labeling infected monolayers with o-propargyl puromycin (OPP), a functionalized analog of puromycin permitting subsequent bioorthogonal addition of a fluorophore to each OPP-terminated nascent polypetide, we use automated confocal feedback microscopy followed by batch image segmentation and feature extraction to visualize and quantify the nascent proteome in individual P. berghei liver stage parasites and host cells simultaneously. After validation, we demonstrate specific, concentration-dependent liver stage translation inhibition by both parasite-selective and pan-eukaryotic active compounds, and further show that acute pre-treatment and competition modes of the OPP assay can distinguish between direct and indirect translation inhibitors. We identify a Malaria Box compound, MMV019266, as a direct translation inhibitor in P. berghei liver stages and confirm this potential mode of action in P. falciparum asexual blood stages.
Collapse
Affiliation(s)
- James L McLellan
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - William Sausman
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ashley B Reers
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Evelien M Bunnik
- Department of Microbiology, Immunology, and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Kirsten K Hanson
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
41
|
Vyas VK, Shukla T, Sharma M. Medicinal chemistry approaches for the discovery of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. Future Med Chem 2023; 15:1295-1321. [PMID: 37551689 DOI: 10.4155/fmc-2023-0113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Malaria is a severe human disease and a global health problem because of drug-resistant strains. Drugs reported to prevent the growth of Plasmodium parasites target various phases of the parasites' life cycle. Antimalarial drugs can inhibit key enzymes that are responsible for the cellular growth and development of parasites. Plasmodium falciparum dihydroorotate dehydrogenase is one such enzyme that is necessary for de novo pyrimidine biosynthesis. This review focuses on various medicinal chemistry approaches used for the discovery and identification of selective P. falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. This comprehensive review discusses recent advances in the selective therapeutic activity of distinct chemical classes of compounds as P. falciparum dihydroorotate dehydrogenase inhibitors and antimalarial drugs.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Tanvi Shukla
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| |
Collapse
|
42
|
Gehlot P, Vyas VK. Recent advances on patents of Plasmodium falciparum dihydroorotate dehydrogenase ( PfDHODH) inhibitors as antimalarial agents. Expert Opin Ther Pat 2023; 33:579-596. [PMID: 37942637 DOI: 10.1080/13543776.2023.2280596] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Pyrimidine nucleotides are essential for the parasite's growth and replication. Parasites have only a de novo pathway for the biosynthesis of pyrimidine nucleotides. Dihydroorotate dehydrogenase (DHODH) enzyme is involved in the rate-limiting step of the pyrimidine biosynthesis pathway. DHODH is a biochemical target for the discovery of new antimalarial agents. AREA COVERED This review discussed the development of patented PfDHODH inhibitors published between 2007 and 2023 along with their chemical structures and activities. EXPERT OPINION PfDHODH enzyme is involved in the rate-limiting fourth step of the pyrimidine biosynthesis pathway. Thus, inhibition of PfDHODH using species-selective inhibitors has drawn much attention for treating malaria because they inhibit parasite growth without affecting normal human functions. Looking at the current scenario of antimalarial drug resistance with most of the available antimalarial drugs, there is a huge need for targeted newer agents. Newer agents with unique mechanisms of action may be devoid of drug toxicity, adverse effects, and the ability of parasites to quickly gain resistance, and PfDHODH inhibitors can be those newer agents. Many PfDHODH inhibitors were patented in the past, and the dependency of Plasmodium on de novo pyrimidine provided a new approach for the development of novel antimalarial agents.
Collapse
Affiliation(s)
- Pinky Gehlot
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
43
|
Elamin EM, Eshage SE, Mohmmode SM, Mukhtar RM, Mahjoub M, Sadelin E, Shoaib TH, Edris A, Elshamly EM, Makki AA, Ashour A, Sherif AE, Osman W, Ibrahim SRM, Mohamed GA, Alzain AA. Discovery of dual-target natural antimalarial agents against DHODH and PMT of Plasmodium falciparum: pharmacophore modelling, molecular docking, quantum mechanics, and molecular dynamics simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:709-728. [PMID: 37665563 DOI: 10.1080/1062936x.2023.2251876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Malaria is a lethal disease that claims thousands of lives worldwide annually. The objective of this study was to identify new natural compounds that can target two P. falciparum enzymes; P. falciparum Dihydroorotate dehydrogenase (PfDHODH) and P. falciparum phosphoethanolamine methyltransferase (PfPMT). To accomplish this, e-pharmacophore modelling and molecular docking were employed against PfDHODH. Following this, 1201 natural compounds with docking scores of ≤ -7 kcal/mol were docked into the active site of the second enzyme PMT. The top nine compounds were subjected to further investigation using MM-GBSA free binding energy calculations and ADME analysis. The results revealed favourable free binding energy values better than the references, as well as acceptable pharmacokinetic properties. Compounds ZINC000013377887, ZINC000015113777, and ZINC000085595753 were scrutinized to assess their interaction stability with the PfDHODH enzyme, and chemical stability reactivity using molecular dynamics (MD) simulation and density functional theory (DFT) calculations. These findings indicate that the three natural compounds are potential candidates for dual PfDHODH and PfPMT inhibitors for malaria treatment.
Collapse
Affiliation(s)
- E M Elamin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S E Eshage
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - S M Mohmmode
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - R M Mukhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - M Mahjoub
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - E Sadelin
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - T H Shoaib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - A Edris
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - E M Elshamly
- Department of Molecular Biotechnology, Hochschule Anhalt, Köthen, Germany
| | - A A Makki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| | - A Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Al Mansurah, Egypt
| | - A E Sherif
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Al Mansurah, Egypt
| | - W Osman
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - S R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - G A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - A A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Gezira, Sudan
| |
Collapse
|
44
|
Kreutzfeld O, Tumwebaze PK, Okitwi M, Orena S, Byaruhanga O, Katairo T, Conrad MD, Rasmussen SA, Legac J, Aydemir O, Giesbrecht D, Forte B, Campbell P, Smith A, Kano H, Nsobya SL, Blasco B, Duffey M, Bailey JA, Cooper RA, Rosenthal PJ. Susceptibility of Ugandan Plasmodium falciparum Isolates to the Antimalarial Drug Pipeline. Microbiol Spectr 2023; 11:e0523622. [PMID: 37158739 PMCID: PMC10269555 DOI: 10.1128/spectrum.05236-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Malaria, especially Plasmodium falciparum infection, remains an enormous problem, and its treatment and control are seriously challenged by drug resistance. New antimalarial drugs are needed. To characterize the Medicines for Malaria Venture pipeline of antimalarials under development, we assessed the ex vivo drug susceptibilities to 19 compounds targeting or potentially impacted by mutations in P. falciparum ABC transporter I family member 1, acetyl-CoA synthetase, cytochrome b, dihydroorotate dehydrogenase, elongation factor 2, lysyl-tRNA synthetase, phenylalanyl-tRNA synthetase, plasmepsin X, prodrug activation and resistance esterase, and V-type H+ ATPase of 998 fresh P. falciparum clinical isolates collected in eastern Uganda from 2015 to 2022. Drug susceptibilities were assessed by 72-h growth inhibition (half-maximum inhibitory concentration [IC50]) assays using SYBR green. Field isolates were highly susceptible to lead antimalarials, with low- to midnanomolar median IC50s, near values previously reported for laboratory strains, for all tested compounds. However, outliers with decreased susceptibilities were identified. Positive correlations between IC50 results were seen for compounds with shared targets. We sequenced genes encoding presumed targets to characterize sequence diversity, search for polymorphisms previously selected with in vitro drug pressure, and determine genotype-phenotype associations. We identified many polymorphisms in target genes, generally in <10% of isolates, but none were those previously selected in vitro with drug pressure, and none were associated with significantly decreased ex vivo drug susceptibility. Overall, Ugandan P. falciparum isolates were highly susceptible to 19 compounds under development as next-generation antimalarials, consistent with a lack of preexisting or novel resistance-conferring mutations in circulating Ugandan parasites. IMPORTANCE Drug resistance necessitates the development of new antimalarial drugs. It is important to assess the activities of compounds under development against parasites now causing disease in Africa, where most malaria cases occur, and to determine if mutations in these parasites may limit the efficacies of new agents. We found that African isolates were generally highly susceptible to the 19 studied lead antimalarials. Sequencing of the presumed drug targets identified multiple mutations in these genes, but these mutations were generally not associated with decreased antimalarial activity. These results offer confidence that the activities of the tested antimalarial compounds now under development will not be limited by preexisting resistance-mediating mutations in African malaria parasites.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- University of California, San Francisco, San Francisco, California, USA
| | | | - Martin Okitwi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Stephen Orena
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Melissa D. Conrad
- University of California, San Francisco, San Francisco, California, USA
| | | | - Jennifer Legac
- University of California, San Francisco, San Francisco, California, USA
| | - Ozkan Aydemir
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Barbara Forte
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Peter Campbell
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Alasdair Smith
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, United Kingdom
| | - Hiroki Kano
- Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
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
|
46
|
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: 9] [Impact Index Per Article: 4.5] [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
|
47
|
Espino-Sanchez T, Wienkers H, Marvin R, Nalder SA, García-Guerrero A, VanNatta P, Jami-Alahmadi Y, Mixon Blackwell A, Whitby F, Wohlschlegel J, Kieber-Emmons M, Hill C, A. Sigala P. Direct tests of cytochrome c and c1 functions in the electron transport chain of malaria parasites. Proc Natl Acad Sci U S A 2023; 120:e2301047120. [PMID: 37126705 PMCID: PMC10175771 DOI: 10.1073/pnas.2301047120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/30/2023] [Indexed: 05/03/2023] Open
Abstract
The mitochondrial electron transport chain (ETC) of Plasmodium malaria parasites is a major antimalarial drug target, but critical cytochrome (cyt) functions remain unstudied and enigmatic. Parasites express two distinct cyt c homologs (c and c-2) with unusually sparse sequence identity and uncertain fitness contributions. P. falciparum cyt c-2 is the most divergent eukaryotic cyt c homolog currently known and has sequence features predicted to be incompatible with canonical ETC function. We tagged both cyt c homologs and the related cyt c1 for inducible knockdown. Translational repression of cyt c and cyt c1 was lethal to parasites, which died from ETC dysfunction and impaired ubiquinone recycling. In contrast, cyt c-2 knockdown or knockout had little impact on blood-stage growth, indicating that parasites rely fully on the more conserved cyt c for ETC function. Biochemical and structural studies revealed that both cyt c and c-2 are hemylated by holocytochrome c synthase, but UV-vis absorbance and EPR spectra strongly suggest that cyt c-2 has an unusually open active site in which heme is stably coordinated by only a single axial amino acid ligand and can bind exogenous small molecules. These studies provide a direct dissection of cytochrome functions in the ETC of malaria parasites and identify a highly divergent Plasmodium cytochrome c with molecular adaptations that defy a conserved role in eukaryotic evolution.
Collapse
Affiliation(s)
| | - Henry Wienkers
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Rebecca G. Marvin
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Shai-anne Nalder
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | | | - Peter E. VanNatta
- Department of Chemistry, University of Utah, Salt Lake City, UT84112
| | | | - Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Frank G. Whitby
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | | | | | - Christopher P. Hill
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| |
Collapse
|
48
|
Brown AC, Warthan MD, Aryal A, Liu S, Guler JL. Nutrient Limitation Mimics Artemisinin Tolerance in Malaria. mBio 2023:e0070523. [PMID: 37097173 DOI: 10.1128/mbio.00705-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Mounting evidence demonstrates that nutritional environment can alter pathogen drug sensitivity. While the rich media used for in vitro culture contains supraphysiological nutrient concentrations, pathogens encounter a relatively restrictive environment in vivo. We assessed the effect of nutrient limitation on the protozoan parasite that causes malaria and demonstrated that short-term growth under physiologically relevant mild nutrient stress (or "metabolic priming") triggers increased tolerance of a potent antimalarial drug. We observed beneficial effects using both short-term survival assays and longer-term proliferation studies, where metabolic priming increases parasite survival to a level previously defined as resistant (>1% survival). We performed these assessments by either decreasing single nutrients that have distinct roles in metabolism or using a media formulation that simulates the human plasma environment. We determined that priming-induced tolerance was restricted to parasites that had newly invaded the host red blood cell, but the effect was not dependent on genetic background. The molecular mechanisms of this intrinsic effect mimic aspects of genetic tolerance, including translational repression and protein export. This finding suggests that regardless of the impact on survival rates, environmental stress could stimulate changes that ultimately directly contribute to drug tolerance. Because metabolic stress is likely to occur more frequently in vivo compared to the stable in vitro environment, priming-induced drug tolerance has ramifications for how in vitro results translate to in vivo studies. Improving our understanding of how pathogens adjust their metabolism to impact survival of current and future drugs is an important avenue of research to slow the evolution of resistance. IMPORTANCE There is a dire need for effective treatments against microbial pathogens. Yet, the continuing emergence of drug resistance necessitates a deeper knowledge of how pathogens respond to treatments. We have long appreciated the contribution of genetic evolution to drug resistance, but transient metabolic changes that arise in response to environmental factors are less recognized. Here, we demonstrate that short-term growth of malaria parasites in a nutrient-limiting environment triggers cellular changes that lead to better survival of drug treatment. We found that these strategies are similar to those employed by drug-tolerant parasites, which suggests that starvation "primes" parasites to survive and potentially evolve resistance. Since the environment of the human host is relatively nutrient restrictive compared to growth conditions in standard laboratory culture, this discovery highlights the important connections among nutrient levels, protective cellular pathways, and resistance evolution.
Collapse
Affiliation(s)
- Audrey C Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Michelle D Warthan
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Anush Aryal
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Shiwei Liu
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
49
|
Imlay LS, Lawong AK, Gahalawat S, Kumar A, Xing C, Mittal N, Wittlin S, Churchyard A, Niederstrasser H, Crespo-Fernandez B, Posner BA, Gamo FJ, Baum J, Winzeler EA, LALEU B, Ready JM, Phillips MA. Fast-Killing Tyrosine Amide (( S)-SW228703) with Blood- and Liver-Stage Antimalarial Activity Associated with the Cyclic Amine Resistance Locus ( PfCARL). ACS Infect Dis 2023; 9:527-539. [PMID: 36763526 PMCID: PMC10053980 DOI: 10.1021/acsinfecdis.2c00527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Current malaria treatments are threatened by drug resistance, and new drugs are urgently needed. In a phenotypic screen for new antimalarials, we identified (S)-SW228703 ((S)-SW703), a tyrosine amide with asexual blood and liver stage activity and a fast-killing profile. Resistance to (S)-SW703 is associated with mutations in the Plasmodium falciparum cyclic amine resistance locus (PfCARL) and P. falciparum acetyl CoA transporter (PfACT), similarly to several other compounds that share features such as fast activity and liver-stage activity. Compounds with these resistance mechanisms are thought to act in the ER, though their targets are unknown. The tyramine of (S)-SW703 is shared with some reported PfCARL-associated compounds; however, we observed that strict S-stereochemistry was required for the activity of (S)-SW703, suggesting differences in the mechanism of action or binding mode. (S)-SW703 provides a new chemical series with broad activity for multiple life-cycle stages and a fast-killing mechanism of action, available for lead optimization to generate new treatments for malaria.
Collapse
Affiliation(s)
- Leah S. Imlay
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Aloysus K. Lawong
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nimisha Mittal
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland
- University of Basel, 4002, Basel, Switzerland
| | - Alisje Churchyard
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Hanspeter Niederstrasser
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | | | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | | | - Jake Baum
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- School of Biomedical Sciences, University of New South Wales, Kensington, NSW, Australia
| | - Elizabeth A. Winzeler
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Benoît LALEU
- Medicines for Malaria Venture, 1215 Geneva 15, Switzerland
| | - Joseph M. Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Margaret A. Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
50
|
Nguyen W, Dans MG, Currie I, Awalt JK, Bailey BL, Lumb C, Ngo A, Favuzza P, Palandri J, Ramesh S, Penington J, Jarman KE, Mukherjee P, Chakraborty A, Maier AG, van Dooren GG, Papenfuss T, Wittlin S, Churchyard A, Baum J, Winzeler EA, Baud D, Brand S, Jackson PF, Cowman AF, Sleebs BE. 7- N-Substituted-3-oxadiazole Quinolones with Potent Antimalarial Activity Target the Cytochrome bc1 Complex. ACS Infect Dis 2023; 9:668-691. [PMID: 36853190 PMCID: PMC10012268 DOI: 10.1021/acsinfecdis.2c00607] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Indexed: 03/01/2023]
Abstract
The development of new antimalarials is required because of the threat of resistance to current antimalarial therapies. To discover new antimalarial chemotypes, we screened the Janssen Jumpstarter library against the P. falciparum asexual parasite and identified the 7-N-substituted-3-oxadiazole quinolone hit class. We established the structure-activity relationship and optimized the antimalarial potency. The optimized analog WJM228 (17) showed robust metabolic stability in vitro, although the aqueous solubility was limited. Forward genetic resistance studies uncovered that WJM228 targets the Qo site of cytochrome b (cyt b), an important component of the mitochondrial electron transport chain (ETC) that is essential for pyrimidine biosynthesis and an established antimalarial target. Profiling against drug-resistant parasites confirmed that WJM228 confers resistance to the Qo site but not Qi site mutations, and in a biosensor assay, it was shown to impact the ETC via inhibition of cyt b. Consistent with other cyt b targeted antimalarials, WJM228 prevented pre-erythrocytic parasite and male gamete development and reduced asexual parasitemia in a P. berghei mouse model of malaria. Correcting the limited aqueous solubility and the high susceptibility to cyt b Qo site resistant parasites found in the clinic will be major obstacles in the future development of the 3-oxadiazole quinolone antimalarial class.
Collapse
Affiliation(s)
- William Nguyen
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Madeline G. Dans
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Iain Currie
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Jon Kyle Awalt
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Brodie L. Bailey
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Chris Lumb
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Anna Ngo
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Paola Favuzza
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Josephine Palandri
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Saishyam Ramesh
- Research
School of Biology, The Australian National
University, Canberra 2600, Australia
| | - Jocelyn Penington
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Kate E. Jarman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | | | | | - Alexander G. Maier
- Research
School of Biology, The Australian National
University, Canberra 2600, Australia
| | - Giel G. van Dooren
- Research
School of Biology, The Australian National
University, Canberra 2600, Australia
| | - Tony Papenfuss
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Sergio Wittlin
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, 4003 Basel, Switzerland
| | - Alisje Churchyard
- Department
of Life Sciences, Imperial College London, South Kensington, SW7
2AZ U.K.
| | - Jake Baum
- Department
of Life Sciences, Imperial College London, South Kensington, SW7
2AZ U.K.
- School
of Biomedical Sciences, University of New
South Wales, Sydney 2031, Australia
| | - Elizabeth A. Winzeler
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive
0760, La Jolla, California 92093, United States
| | - Delphine Baud
- Medicines
for Malaria Venture, Geneva 1215, Switzerland
| | - Stephen Brand
- Medicines
for Malaria Venture, Geneva 1215, Switzerland
| | - Paul F. Jackson
- Global
Public Health, Janssen R&D LLC, La Jolla, California 92121, United States
| | - Alan F. Cowman
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Brad E. Sleebs
- The
Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department
of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|