1
|
Liu X, Thistlethwaite S, Kholiya R, Pierscianowski J, Saliba KJ, Auclair K. Chemical synthesis and enzymatic late-stage diversification of novel pantothenate analogues with antiplasmodial activity. Eur J Med Chem 2024; 280:116902. [PMID: 39423490 DOI: 10.1016/j.ejmech.2024.116902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 10/21/2024]
Abstract
The emergence of resistance to nearly every therapeutic agent directed against malaria-causing Plasmodium parasites emphasises the dire need for new antimalarials. Despite their high potency and low cytotoxicity in vitro, the clinical use of pantothenamides is hindered by pantetheinase-mediated hydrolysis in human serum. We herein report the chemical synthesis and biological activity of a new series of pantothenamide analogues in which the labile amide group is replaced with an isoxazole ring. In addition, we utilised, for the first time, enzymatic late-stage diversification to generate additional isoxazole-containing pantothenamide-mimics. Thirteen novel isoxazole-containing pantothenamide-mimics were generated, several of which display nanomolar antiplasmodial activity against Plasmodium falciparum and are non-toxic to human cells in vitro. Although the derivatives generated via late-stage diversification are less potent than the parent compounds, the most potent still exerted its activity via a mechanism that interferes with the pantothenate-utilising process and appears to be nontoxic to human cells. This increases the appeal of using late-stage diversification to modify pantothenamide-mimics, potentially leading to compounds with improved antiplasmodial and/or pharmacological properties.
Collapse
Affiliation(s)
- Xiangning Liu
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Sian Thistlethwaite
- Department of Chemistry, McGill University, Montreal, Quebec, Canada, H3A 0B8
| | - Rohit Kholiya
- Department of Chemistry, McGill University, Montreal, Quebec, Canada, H3A 0B8
| | | | - Kevin J Saliba
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | - Karine Auclair
- Department of Chemistry, McGill University, Montreal, Quebec, Canada, H3A 0B8.
| |
Collapse
|
2
|
Redway A, Spry C, Brown A, Wiedemann U, Fathoni I, Garnie LF, Qiu D, Egan TJ, Lehane AM, Jackson Y, Saliba KJ, Downer-Riley N. Discovery of antiplasmodial pyridine carboxamides and thiocarboxamides. Int J Parasitol Drugs Drug Resist 2024; 25:100536. [PMID: 38663046 PMCID: PMC11068522 DOI: 10.1016/j.ijpddr.2024.100536] [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: 06/01/2023] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024]
Abstract
Malaria continues to be a significant burden, particularly in Africa, which accounts for 95% of malaria deaths worldwide. Despite advances in malaria treatments, malaria eradication is hampered by insecticide and antimalarial drug resistance. Consequently, the need to discover new antimalarial lead compounds remains urgent. To help address this need, we evaluated the antiplasmodial activity of twenty-two amides and thioamides with pyridine cores and their non-pyridine analogues. Twelve of these compounds showed in vitro anti-proliferative activity against the intraerythrocytic stage of Plasmodium falciparum, the most virulent species of Plasmodium infecting humans. Thiopicolinamide 13i was found to possess submicromolar activity (IC50 = 142 nM) and was >88-fold less active against a human cell line. The compound was equally effective against chloroquine-sensitive and -resistant parasites and did not inhibit β-hematin formation, pH regulation or PfATP4. Compound 13i may therefore possess a novel mechanism of action.
Collapse
Affiliation(s)
- Alexa Redway
- Department of Chemistry, The University of the West Indies, Mona, Kingston 7, Jamaica; Chemistry Divison, University of Technology, 237 Old Hope Road, Kingston 6, Jamaica
| | - Christina Spry
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Ainka Brown
- Department of Chemistry, The University of the West Indies, Mona, Kingston 7, Jamaica
| | - Ursula Wiedemann
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Imam Fathoni
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Larnelle F Garnie
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Deyun Qiu
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, South Africa
| | - Adele M Lehane
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Yvette Jackson
- Department of Chemistry, The University of the West Indies, Mona, Kingston 7, Jamaica
| | - Kevin J Saliba
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nadale Downer-Riley
- Department of Chemistry, The University of the West Indies, Mona, Kingston 7, Jamaica.
| |
Collapse
|
3
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
4
|
Khairnar P, Aleshire SL, Kumar Ongolu R, Jin L, Laidlaw MG, Donsbach KO, Gupton BF, Nelson RC, Shanahan CS. Highly Regioselective Protecting-Group-Free Synthesis of the Antimalarial Drug MMV693183. Org Process Res Dev 2024; 28:273-280. [PMID: 38268773 PMCID: PMC10804412 DOI: 10.1021/acs.oprd.3c00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024]
Abstract
MMV693183 is a promising antimalarial drug candidate that works for uncomplicated malaria treatment and resistance management. Herein, we report an efficient and highly regioselective synthesis of MMV693183. This novel synthetic method highlights a three-step route with an overall yield of 46% from readily available starting materials. The key to the success lies in (1) utilizing the subtle difference of the two amino groups in the starting material (S)-propane-1,2-diamine dihydrochloride without amino protection and (2) identifying the L-(+)-tartaric acid as the counter acid for the organic salt formation, yielding the desired regioisomer up to 100:0. The efficient and scalable three-step protocol operates under mild conditions with a high chemo/regioselectivity, providing effective access to MMV693183.
Collapse
Affiliation(s)
- Pankaj
V. Khairnar
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Sarah L. Aleshire
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Ravi Kumar Ongolu
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Limei Jin
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Michael G. Laidlaw
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Kai O. Donsbach
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - B. Frank Gupton
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Ryan C. Nelson
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| | - Charles S. Shanahan
- Medicines for All Institute, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| |
Collapse
|
5
|
Riske BF, Luckhart S, Riehle MA. Starving the Beast: Limiting Coenzyme A Biosynthesis to Prevent Disease and Transmission in Malaria. Int J Mol Sci 2023; 24:13915. [PMID: 37762222 PMCID: PMC10530615 DOI: 10.3390/ijms241813915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Malaria parasites must acquire all necessary nutrients from the vertebrate and mosquito hosts to successfully complete their life cycle. Failure to acquire these nutrients can limit or even block parasite development and presents a novel target for malaria control. One such essential nutrient is pantothenate, also known as vitamin B5, which the parasite cannot synthesize de novo and is required for the synthesis of coenzyme A (CoA) in the parasite. This review examines pantothenate and the CoA biosynthesis pathway in the human-mosquito-malaria parasite triad and explores possible approaches to leverage the CoA biosynthesis pathway to limit malaria parasite development in both human and mosquito hosts. This includes a discussion of sources for pantothenate for the mosquito, human, and parasite, examining the diverse strategies used by the parasite to acquire substrates for CoA synthesis across life stages and host resource pools and a discussion of drugs and alternative approaches being studied to disrupt CoA biosynthesis in the parasite. The latter includes antimalarial pantothenate analogs, known as pantothenamides, that have been developed to target this pathway during the human erythrocytic stages. In addition to these parasite-targeted drugs, we review studies of mosquito-targeted allosteric enzymatic regulators known as pantazines as an approach to limit pantothenate availability in the mosquito and subsequently deprive the parasite of this essential nutrient.
Collapse
Affiliation(s)
- Brendan F. Riske
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA;
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID 83843, USA;
- Department of Biological Sciences, University of Idaho, Moscow, ID 83843, USA
| | - Michael A. Riehle
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA;
| |
Collapse
|
6
|
Brigg SE, Koekemoer L, Brand LA, Strauss E. Multifaceted Target Specificity Analysis as a Tool in Antimicrobial Drug Development: Type III Pantothenate Kinases as a Case Study. ChemMedChem 2023; 18:e202200630. [PMID: 36749500 DOI: 10.1002/cmdc.202200630] [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: 11/20/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/08/2023]
Abstract
The research and development of a new antimicrobial drug using a target-based approach raises the question of whether any resulting hits will also show activity against the homologous target in other closely related organisms. While an assessment of the similarities of the predicted interactions between the identified inhibitor and the various targets is an obvious first step in answering this question, no clear and consistent framework has been proposed for how this should be done. Here we developed Multifaceted Target Specificity Analysis (MTSA) and applied it to type III pantothenate kinase (PanKIII ) - an essential enzyme required for coenzyme A biosynthesis in a wide range of pathogenic bacteria - as a case study to establish if targeting a specific organism's PanKIII would lead to a narrow- or broad-spectrum agent. We propose that MTSA is a useful tool and aid for directing new target-based antimicrobial drug development initiatives.
Collapse
Affiliation(s)
- Siobhan Ernan Brigg
- Department of Biochemistry, Stellenbosch University, Matieland, 7602, South Africa
| | - Lizbé Koekemoer
- Department of Biochemistry, Stellenbosch University, Matieland, 7602, South Africa
| | - Leisl A Brand
- Department of Biochemistry, Stellenbosch University, Matieland, 7602, South Africa
| | - Erick Strauss
- Department of Biochemistry, Stellenbosch University, Matieland, 7602, South Africa
| |
Collapse
|
7
|
Roseoflavin, a Natural Riboflavin Analogue, Possesses In Vitro and In Vivo Antiplasmodial Activity. Antimicrob Agents Chemother 2022; 66:e0054022. [PMID: 36094195 PMCID: PMC9578400 DOI: 10.1128/aac.00540-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability of the human malaria parasite Plasmodium falciparum to access and utilize vital nutrients is critical to its growth and proliferation. Molecules that interfere with these processes could potentially serve as antimalarials. We found that two riboflavin analogues, roseoflavin and 8-aminoriboflavin, inhibit malaria parasite proliferation by targeting riboflavin metabolism and/or the utilization of the riboflavin metabolites flavin mononucleotide and flavin adenine dinucleotide. An additional eight riboflavin analogues were evaluated, but none were found to be more potent than roseoflavin, nor was their activity on target. Focusing on roseoflavin, we tested its antimalarial activity in vivo against Plasmodium vinckei vinckei in mice. We found that roseoflavin decreased the parasitemia by 46-fold following a 4 day suppression test and, on average, increased the survival of mice by 4 to 5 days. Our data are consistent with riboflavin metabolism and/or the utilization of riboflavin-derived cofactors being viable drug targets for the development of new antimalarials and that roseoflavin could serve as a potential starting point.
Collapse
|
8
|
Olivier T, Loots L, Kok M, de Villiers M, Reader J, Birkholtz LM, Arnott GE, de Villiers KA. Adsorption to the Surface of Hemozoin Crystals: Structure-Based Design and Synthesis of Amino-Phenoxazine β-Hematin Inhibitors. ChemMedChem 2022; 17:e202200139. [PMID: 35385211 PMCID: PMC9119941 DOI: 10.1002/cmdc.202200139] [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: 03/16/2022] [Revised: 04/05/2022] [Indexed: 11/07/2022]
Abstract
In silico adsorption of eight antimalarials that inhibit β-hematin (synthetic hemozoin) formation identified a primary binding site on the (001) face, which accommodates inhibitors via formation of predominantly π-π interactions. A good correlation (r2 =0.64, P=0.017) between adsorption energies and the logarithm of β-hematin inhibitory activity was found for this face. Of 53 monocyclic, bicyclic and tricyclic scaffolds, the latter yielded the most favorable adsorption energies. Five new amino-phenoxazine compounds were pursued as β-hematin inhibitors based on adsorption behaviour. The 2-substituted phenoxazines show good to moderate β-hematin inhibitory activity (<100 μM) and Plasmodium falciparum blood stage activity against the 3D7 strain. N1 ,N1 -diethyl-N4 -(10H-phenoxazin-2-yl)pentane-1,4-diamine (P2a) is the most promising hit with IC50 values of 4.7±0.6 and 0.64±0.05 μM, respectively. Adsorption energies are predictive of β-hematin inhibitory activity, and thus the in silico approach is a beneficial tool for structure-based development of new non-quinoline inhibitors.
Collapse
Affiliation(s)
- Tania Olivier
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Leigh Loots
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Michélle Kok
- Department of Biochemistry, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Marianne de Villiers
- Department of Biochemistry, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, 0028, South Africa
| | - Gareth E Arnott
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Katherine A de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| |
Collapse
|
9
|
de Vries LE, Jansen PAM, Barcelo C, Munro J, Verhoef JMJ, Pasaje CFA, Rubiano K, Striepen J, Abla N, Berning L, Bolscher JM, Demarta-Gatsi C, Henderson RWM, Huijs T, Koolen KMJ, Tumwebaze PK, Yeo T, Aguiar ACC, Angulo-Barturen I, Churchyard A, Baum J, Fernández BC, Fuchs A, Gamo FJ, Guido RVC, Jiménez-Diaz MB, Pereira DB, Rochford R, Roesch C, Sanz LM, Trevitt G, Witkowski B, Wittlin S, Cooper RA, Rosenthal PJ, Sauerwein RW, Schalkwijk J, Hermkens PHH, Bonnert RV, Campo B, Fidock DA, Llinás M, Niles JC, Kooij TWA, Dechering KJ. Preclinical characterization and target validation of the antimalarial pantothenamide MMV693183. Nat Commun 2022; 13:2158. [PMID: 35444200 PMCID: PMC9021288 DOI: 10.1038/s41467-022-29688-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
Drug resistance and a dire lack of transmission-blocking antimalarials hamper malaria elimination. Here, we present the pantothenamide MMV693183 as a first-in-class acetyl-CoA synthetase (AcAS) inhibitor to enter preclinical development. Our studies demonstrate attractive drug-like properties and in vivo efficacy in a humanized mouse model of Plasmodium falciparum infection. The compound shows single digit nanomolar in vitro activity against P. falciparum and P. vivax clinical isolates, and potently blocks P. falciparum transmission to Anopheles mosquitoes. Genetic and biochemical studies identify AcAS as the target of the MMV693183-derived antimetabolite, CoA-MMV693183. Pharmacokinetic-pharmacodynamic modelling predict that a single 30 mg oral dose is sufficient to cure a malaria infection in humans. Toxicology studies in rats indicate a > 30-fold safety margin in relation to the predicted human efficacious exposure. In conclusion, MMV693183 represents a promising candidate for further (pre)clinical development with a novel mode of action for treatment of malaria and blocking transmission. Here, de Vries et al. perform a pre-clinical characterization of the antimalarial compound MMV693183: the compound targets acetyl-CoA synthetase, has efficacy in humanized mice against Plasmodium falciparum infection, blocks transmission to mosquito vectors, is safe in rats, and pharmacokinetic-pharmacodynamic modeling informs about a potential oral human dosing regimen.
Collapse
Affiliation(s)
- Laura E de Vries
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Patrick A M Jansen
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Justin Munro
- Department of Chemistry and Huck Center for Malaria Research, The Pennsylvania State University, University Park, PA, USA
| | - Julie M J Verhoef
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Kelly Rubiano
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Josefine Striepen
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nada Abla
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Luuk Berning
- TropIQ Health Sciences, Nijmegen, The Netherlands
| | | | | | | | - Tonnie Huijs
- TropIQ Health Sciences, Nijmegen, The Netherlands
| | | | | | - Tomas Yeo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna C C Aguiar
- Sao Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, Brazil, São Carlos, SP, Brazil
| | | | - Alisje Churchyard
- Department of Life Sciences, Imperial College London, South Kensington, London, United Kingdom
| | - Jake Baum
- Department of Life Sciences, Imperial College London, South Kensington, London, United Kingdom
| | | | - Aline Fuchs
- Medicines for Malaria Venture, Geneva, Switzerland
| | | | - Rafael V C Guido
- Sao Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, Brazil, São Carlos, SP, Brazil
| | | | - Dhelio B Pereira
- Research Center for Tropical Medicine of Rondonia, Porto Velho, Brazil
| | - Rosemary Rochford
- Department of Immunology and Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Camille Roesch
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.,Malaria Translational Research Unit, Institut Pasteur, Paris & Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Laura M Sanz
- Global Health, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | | | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.,Malaria Translational Research Unit, Institut Pasteur, Paris & Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Roland A Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,TropIQ Health Sciences, Nijmegen, The Netherlands
| | - Joost Schalkwijk
- Department of Dermatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY, USA.,Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Manuel Llinás
- Department of Chemistry and Huck Center for Malaria Research, The Pennsylvania State University, University Park, PA, USA.,Department of Biochemistry & Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taco W A Kooij
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | |
Collapse
|
10
|
Duncan D, Auclair K. Itaconate: an antimicrobial metabolite of macrophages. CAN J CHEM 2022. [DOI: 10.1139/cjc-2021-0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Itaconate is a conjugated 1,4-dicarboxylate produced by macrophages. This small molecule has recently received increasing attention due to its role in modulating the immune response of macrophages upon exposure to pathogens. Itaconate has also been proposed to play an antimicrobial function; however, this has not been explored as intensively. Consistent with the latter, itaconate is known to show antibacterial activity in vitro and was reported to inhibit isocitrate lyase, an enzyme required for survival of bacterial pathogens in mammalian systems. Recent studies have revealed bacterial growth inhibition under biologically relevant conditions. In addition, an antimicrobial role for itaconate is substantiated by the high concentration of itaconate found in bacteria-containing vacuoles, and by the production of itaconate-degrading enzymes in pathogens such as Salmonella enterica ser. Typhimurium, Pseudomonas aeruginosa, and Yersinia pestis. This review describes the current state of literature in understanding the role of itaconate as an antimicrobial agent in host–pathogen interactions.
Collapse
Affiliation(s)
- Dustin Duncan
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| | - Karine Auclair
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
11
|
de Vries LE, Lunghi M, Krishnan A, Kooij TWA, Soldati-Favre D. Pantothenate and CoA biosynthesis in Apicomplexa and their promise as antiparasitic drug targets. PLoS Pathog 2021; 17:e1010124. [PMID: 34969059 PMCID: PMC8717973 DOI: 10.1371/journal.ppat.1010124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Apicomplexa phylum comprises thousands of distinct intracellular parasite species, including coccidians, haemosporidians, piroplasms, and cryptosporidia. These parasites are characterized by complex and divergent life cycles occupying a variety of host niches. Consequently, they exhibit distinct adaptations to the differences in nutritional availabilities, either relying on biosynthetic pathways or by salvaging metabolites from their host. Pantothenate (Pan, vitamin B5) is the precursor for the synthesis of an essential cofactor, coenzyme A (CoA), but among the apicomplexans, only the coccidian subgroup has the ability to synthesize Pan. While the pathway to synthesize CoA from Pan is largely conserved across all branches of life, there are differences in the redundancy of enzymes and possible alternative pathways to generate CoA from Pan. Impeding the scavenge of Pan and synthesis of Pan and CoA have been long recognized as potential targets for antimicrobial drug development, but in order to fully exploit these critical pathways, it is important to understand such differences. Recently, a potent class of pantothenamides (PanAms), Pan analogs, which target CoA-utilizing enzymes, has entered antimalarial preclinical development. The potential of PanAms to target multiple downstream pathways make them a promising compound class as broad antiparasitic drugs against other apicomplexans. In this review, we summarize the recent advances in understanding the Pan and CoA biosynthesis pathways, and the suitability of these pathways as drug targets in Apicomplexa, with a particular focus on the cyst-forming coccidian, Toxoplasma gondii, and the haemosporidian, Plasmodium falciparum.
Collapse
Affiliation(s)
- Laura E. de Vries
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Matteo Lunghi
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aarti Krishnan
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Taco W. A. Kooij
- Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dominique Soldati-Favre
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Tjhin ET, Howieson VM, Spry C, van Dooren GG, Saliba KJ. A novel heteromeric pantothenate kinase complex in apicomplexan parasites. PLoS Pathog 2021; 17:e1009797. [PMID: 34324601 PMCID: PMC8366970 DOI: 10.1371/journal.ppat.1009797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 08/16/2021] [Accepted: 07/13/2021] [Indexed: 11/19/2022] Open
Abstract
Coenzyme A is synthesised from pantothenate via five enzyme-mediated steps. The first step is catalysed by pantothenate kinase (PanK). All PanKs characterised to date form homodimers. Many organisms express multiple PanKs. In some cases, these PanKs are not functionally redundant, and some appear to be non-functional. Here, we investigate the PanKs in two pathogenic apicomplexan parasites, Plasmodium falciparum and Toxoplasma gondii. Each of these organisms express two PanK homologues (PanK1 and PanK2). We demonstrate that PfPanK1 and PfPanK2 associate, forming a single, functional PanK complex that includes the multi-functional protein, Pf14-3-3I. Similarly, we demonstrate that TgPanK1 and TgPanK2 form a single complex that possesses PanK activity. Both TgPanK1 and TgPanK2 are essential for T. gondii proliferation, specifically due to their PanK activity. Our study constitutes the first examples of heteromeric PanK complexes in nature and provides an explanation for the presence of multiple PanKs within certain organisms.
Collapse
Affiliation(s)
- Erick T. Tjhin
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Vanessa M. Howieson
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Christina Spry
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Giel G. van Dooren
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Kevin J. Saliba
- Research School of Biology, The Australian National University, Canberra, Australia
- Medical School, The Australian National University, Canberra, Australia
- * E-mail:
| |
Collapse
|
13
|
Guan J, Spry C, Tjhin ET, Yang P, Kittikool T, Howieson VM, Ling H, Starrs L, Duncan D, Burgio G, Saliba KJ, Auclair K. Exploring Heteroaromatic Rings as a Replacement for the Labile Amide of Antiplasmodial Pantothenamides. J Med Chem 2021; 64:4478-4497. [PMID: 33792339 DOI: 10.1021/acs.jmedchem.0c01755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Malaria-causing Plasmodium parasites are developing resistance to antimalarial drugs, providing the impetus for new antiplasmodials. Although pantothenamides show potent antiplasmodial activity, hydrolysis by pantetheinases/vanins present in blood rapidly inactivates them. We herein report the facile synthesis and biological activity of a small library of pantothenamide analogues in which the labile amide group is replaced with a heteroaromatic ring. Several of these analogues display nanomolar antiplasmodial activity against Plasmodium falciparum and/or Plasmodium knowlesi, and are stable in the presence of pantetheinase. Both a known triazole and a novel isoxazole derivative were further characterized and found to possess high selectivity indices, medium or high Caco-2 permeability, and medium or low microsomal clearance in vitro. Although they fail to suppress Plasmodium berghei proliferation in vivo, the pharmacokinetic and contact time data presented provide a benchmark for the compound profile likely required to achieve antiplasmodial activity in mice and should facilitate lead optimization.
Collapse
Affiliation(s)
- Jinming Guan
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| | - Christina Spry
- Research School of Biology, The Australian National University, Acton, ACT 2601, Australia
| | - Erick T Tjhin
- Research School of Biology, The Australian National University, Acton, ACT 2601, Australia
| | - Penghui Yang
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada.,College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an 710065, China
| | - Tanakorn Kittikool
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| | - Vanessa M Howieson
- Research School of Biology, The Australian National University, Acton, ACT 2601, Australia
| | - Harriet Ling
- Research School of Biology, The Australian National University, Acton, ACT 2601, Australia
| | - Lora Starrs
- John Curtin School of Medical Research, The Australian National University, Acton, ACT 2601, Australia
| | - Dustin Duncan
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| | - Gaetan Burgio
- John Curtin School of Medical Research, The Australian National University, Acton, ACT 2601, Australia
| | - Kevin J Saliba
- Research School of Biology, The Australian National University, Acton, ACT 2601, Australia.,Medical School, The Australian National University, Acton, ACT 2601, Australia
| | - Karine Auclair
- Department of Chemistry, McGill University, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
14
|
Cools F, Delputte P, Cos P. The search for novel treatment strategies for Streptococcus pneumoniae infections. FEMS Microbiol Rev 2021; 45:6064299. [PMID: 33399826 PMCID: PMC8371276 DOI: 10.1093/femsre/fuaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/01/2021] [Indexed: 12/13/2022] Open
Abstract
This review provides an overview of the most important novel treatment strategies against Streptococcus pneumoniae infections published over the past 10 years. The pneumococcus causes the majority of community-acquired bacterial pneumonia cases, and it is one of the prime pathogens in bacterial meningitis. Over the last 10 years, extensive research has been conducted to prevent severe pneumococcal infections, with a major focus on (i) boosting the host immune system and (ii) discovering novel antibacterials. Boosting the immune system can be done in two ways, either by actively modulating host immunity, mostly through administration of selective antibodies, or by interfering with pneumococcal virulence factors, thereby supporting the host immune system to effectively overcome an infection. While several of such experimental therapies are promising, few have evolved to clinical trials. The discovery of novel antibacterials is hampered by the high research and development costs versus the relatively low revenues for the pharmaceutical industry. Nevertheless, novel enzymatic assays and target-based drug design, allow the identification of targets and the development of novel molecules to effectively treat this life-threatening pathogen.
Collapse
Affiliation(s)
- F Cools
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|