1
|
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
|
2
|
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
|
3
|
Stopper D, de Carvalho LP, de Souza ML, Kponomaizoun CE, Winzeler EA, Held J, Hansen FK. Development of peptoid-based heteroaryl-decorated histone deacetylase (HDAC) inhibitors with dual-stage antiplasmodial activity. Eur J Med Chem 2024; 277:116782. [PMID: 39208744 DOI: 10.1016/j.ejmech.2024.116782] [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: 04/10/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Dynamics of epigenetic modifications such as acetylation and deacetylation of histone proteins have been shown to be crucial for the life cycle development and survival of Plasmodium falciparum, the deadliest malaria parasite. In this study, we present a novel series of peptoid-based histone deacetylase (HDAC) inhibitors incorporating nitrogen-containing bicyclic heteroaryl residues as a new generation of antiplasmodial peptoid-based HDAC inhibitors. We synthesized the HDAC inhibitors by an efficient multicomponent protocol based on the Ugi four-component reaction. The subsequent screening of 16 compounds from our mini-library identified 6i as the most promising candidate, demonstrating potent activity against asexual blood-stage parasites (IC50Pf3D7 = 30 nM; IC50PfDd2 = 98 nM), low submicromolar activity against liver-stage parasites (IC50PbEEF = 0.25 μM), excellent microsomal stability (t1/2 > 60 min), and low cytotoxicity to HEK293 cells (IC50 = 136 μM).
Collapse
Affiliation(s)
- Daniel Stopper
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | | | - Mariana Laureano de Souza
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Cindy-Esther Kponomaizoun
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, 72074, Tübingen, Germany; German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Finn K Hansen
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany.
| |
Collapse
|
4
|
Ferreira LT, Cassiano GC, Alvarez LCS, Okombo J, Calit J, Fontinha D, Gil-Iturbe E, Coyle R, Andrade CH, Sunnerhagen P, Bargieri DY, Prudêncio M, Quick M, Cravo PV, Lee MCS, Fidock DA, Costa FTM. A novel 4-aminoquinoline chemotype with multistage antimalarial activity and lack of cross-resistance with PfCRT and PfMDR1 mutants. PLoS Pathog 2024; 20:e1012627. [PMID: 39471233 PMCID: PMC11521309 DOI: 10.1371/journal.ppat.1012627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 11/01/2024] Open
Abstract
Artemisinin-based combination therapy (ACT) is the mainstay of effective treatment of Plasmodium falciparum malaria. However, the long-term utility of ACTs is imperiled by widespread partial artemisinin resistance in Southeast Asia and its recent emergence in parts of East Africa. This underscores the need to identify chemotypes with new modes of action (MoAs) to circumvent resistance to ACTs. In this study, we characterized the asexual blood stage antiplasmodial activity and resistance mechanisms of LDT-623, a 4-aminoquinoline (4-AQ). We also detected LDT-623 activity against multiple stages (liver schizonts, stage IV-V gametocytes, and ookinetes) of Plasmodium's life cycle, a feature unlike other 4-AQs such as chloroquine (CQ) and piperaquine (PPQ). Using heme fractionation profiling and drug uptake studies in PfCRT-containing proteoliposomes, we observed inhibition of hemozoin formation and PfCRT-mediated transport, which constitute characteristic features of 4-AQs' MoA. We also found minimal cross-resistance to LDT-623 in a panel of mutant pfcrt or pfmdr1 lines, but not the PfCRT F145I mutant that is highly resistant to PPQ resistance yet is very unfit. No P. falciparum parasites were recovered in an in vitro resistance selection study, suggesting a high barrier for resistance to emerge. Finally, a competitive growth assay comprising >50 barcoded parasite lines with mutated resistance mediators or major drug targets found no evidence of cross-resistance. Our findings support further exploration of this promising 4-AQ.
Collapse
Affiliation(s)
- Letícia Tiburcio Ferreira
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Gustavo Capatti Cassiano
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Luis Carlos Salazar Alvarez
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Juliana Calit
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | - Carolina Horta Andrade
- Laboratory of Molecular Modeling and Drug Design, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
- Center for the Research and Advancement in Fragments and molecular Targets, School of Pharmaceutical Sciences at Ribeirao Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Center for Excellence in Artificial Intelligence, Institute of Informatics, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Youssef Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Physiology & Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States of America
- New York State Psychiatric Institute, Area Neuroscience – Molecular Therapeutics, New York, New York, United States of America
| | - Pedro V. Cravo
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Marcus C. S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, United States of America
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
5
|
Mogire RM, Miruka SA, Juma DW, McNamara CW, Andagalu B, Burrows JN, Chenu E, Duffy J, Ogutu BR, Akala HM. Protein target similarity is positive predictor of in vitro antipathogenic activity: a drug repurposing strategy for Plasmodium falciparum. J Cheminform 2024; 16:63. [PMID: 38831351 PMCID: PMC11145868 DOI: 10.1186/s13321-024-00856-7] [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: 11/26/2023] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Drug discovery is an intricate and costly process. Repurposing existing drugs and active compounds offers a viable pathway to develop new therapies for various diseases. By leveraging publicly available biomedical information, it is possible to predict compounds' activity and identify their potential targets across diverse organisms. In this study, we aimed to assess the antiplasmodial activity of compounds from the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library using in vitro and bioinformatics approaches. We assessed the in vitro antiplasmodial activity of the compounds using blood-stage and liver-stage drug susceptibility assays. We used protein sequences of known targets of the ReFRAME compounds with high antiplasmodial activity (EC50 < 10 uM) to conduct a protein-pairwise search to identify similar Plasmodium falciparum 3D7 proteins (from PlasmoDB) using NCBI protein BLAST. We further assessed the association between the compounds' in vitro antiplasmodial activity and level of similarity between their known and predicted P. falciparum target proteins using simple linear regression analyses. BLAST analyses revealed 735 P. falciparum proteins that were similar to the 226 known protein targets associated with the ReFRAME compounds. Antiplasmodial activity of the compounds was positively associated with the degree of similarity between the compounds' known targets and predicted P. falciparum protein targets (percentage identity, E value, and bit score), the number of the predicted P. falciparum targets, and their respective mutagenesis index and fitness scores (R2 between 0.066 and 0.92, P < 0.05). Compounds predicted to target essential P. falciparum proteins or those with a druggability index of 1 showed the highest antiplasmodial activity.
Collapse
Affiliation(s)
- Reagan M Mogire
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya.
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya.
| | - Silviane A Miruka
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya
| | - Dennis W Juma
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Department of Emerging Infections Diseases (DEID), Walter Reed Army Institute of Research - Africa, Kisumu, Kenya
| | - Case W McNamara
- Calibr-Skaggs Institute for Innovative Medicine, a division of The Scripps Research Institute, La Jolla, CA, USA
| | - Ben Andagalu
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
| | | | - Elodie Chenu
- Medicines for Malaria Venture, Geneva, Switzerland
| | - James Duffy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Bernhards R Ogutu
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya
| | - Hoseah M Akala
- Center for Clinical Research, Kenya Medical Research Institute (KEMRI), P. O. Box 54, Kisumu, 40100, Kenya.
- Center for Research in Therapeutic Sciences, Strathmore University, P.O. Box 59857-00200, Nairobi, Kenya.
- Department of Emerging Infections Diseases (DEID), Walter Reed Army Institute of Research - Africa, Kisumu, Kenya.
| |
Collapse
|
6
|
Maher SP, Bakowski MA, Vantaux A, Flannery EL, Andolina C, Gupta M, Antonova-Koch Y, Argomaniz M, Cabrera-Mora M, Campo B, Chao AT, Chatterjee AK, Cheng WT, Chuenchob E, Cooper CA, Cottier K, Galinski MR, Harupa-Chung A, Ji H, Joseph SB, Lenz T, Lonardi S, Matheson J, Mikolajczak SA, Moeller T, Orban A, Padín-Irizarry V, Pan K, Péneau J, Prudhomme J, Roesch C, Ruberto AA, Sabnis SS, Saney CL, Sattabongkot J, Sereshki S, Suriyakan S, Ubalee R, Wang Y, Wasisakun P, Yin J, Popovici J, McNamara CW, Joyner CJ, Nosten F, Witkowski B, Le Roch KG, Kyle DE. A Drug Repurposing Approach Reveals Targetable Epigenetic Pathways in Plasmodium vivax Hypnozoites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.31.526483. [PMID: 36778461 PMCID: PMC9915689 DOI: 10.1101/2023.01.31.526483] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Radical cure of Plasmodium vivax malaria must include elimination of quiescent 'hypnozoite' forms in the liver; however, the only FDA-approved treatments are contraindicated in many vulnerable populations. To identify new drugs and drug targets for hypnozoites, we screened the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library and a collection of epigenetic inhibitors against P. vivax liver stages. From both libraries, we identified inhibitors targeting epigenetics pathways as selectively active against P. vivax and P. cynomolgi hypnozoites. These include DNA methyltransferase (DNMT) inhibitors as well as several inhibitors targeting histone post-translational modifications. Immunofluorescence staining of Plasmodium liver forms showed strong nuclear 5-methylcystosine signal, indicating liver stage parasite DNA is methylated. Using bisulfite sequencing, we mapped genomic DNA methylation in sporozoites, revealing DNA methylation signals in most coding genes. We also demonstrated that methylation level in proximal promoter regions as well as in the first exon of the genes may affect, at least partially, gene expression in P. vivax. The importance of selective inhibitors targeting epigenetic features on hypnozoites was validated using MMV019721, an acetyl-CoA synthetase inhibitor that affects histone acetylation and was previously reported as active against P. falciparum blood stages. In summary, our data indicate that several epigenetic mechanisms are likely modulating hypnozoite formation or persistence and provide an avenue for the discovery and development of improved radical cure antimalarials.
Collapse
Affiliation(s)
- S. P. Maher
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - M. A. Bakowski
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - A. Vantaux
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - E. L. Flannery
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. Andolina
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - M. Gupta
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - Y. Antonova-Koch
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - M. Argomaniz
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - M. Cabrera-Mora
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - B. Campo
- Medicines for Malaria Venture (MMV); Geneva, 1215, Switzerland
| | - A. T. Chao
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - A. K. Chatterjee
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - W. T. Cheng
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - E. Chuenchob
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. A. Cooper
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | | | - M. R. Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
- Division of Infectious Diseases, Department of Medicine, Emory University; Atlanta, GA, 30329, USA
| | - A. Harupa-Chung
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - H. Ji
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - S. B. Joseph
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - T. Lenz
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - S. Lonardi
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - J. Matheson
- Department of Microbiology and Immunology, University of Otago; Dunedin, 9016, New Zealand
| | - S. A. Mikolajczak
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | | | - A. Orban
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - V. Padín-Irizarry
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
- School of Sciences, Clayton State University; Morrow, GA, 30260, USA
| | - K. Pan
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - J. Péneau
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - J. Prudhomme
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - C. Roesch
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - A. A. Ruberto
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - S. S. Sabnis
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - C. L. Saney
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - J. Sattabongkot
- Mahidol Vivax Research Unit, Mahidol University; Bangkok, 10400, Thailand
| | - S. Sereshki
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - S. Suriyakan
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - R. Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Sciences (AFRIMS); Bangkok, 10400, Thailand
| | - Y. Wang
- Department of Chemistry, University of California; Riverside, CA, 92521
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - P. Wasisakun
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - J. Yin
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - J. Popovici
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - C. W. McNamara
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - C. J. Joyner
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - F. Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford; Oxford, OX3 7LG, UK
| | - B. Witkowski
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - K. G. Le Roch
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - D. E. Kyle
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| |
Collapse
|
7
|
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
|
8
|
Voorberg-van der Wel A, Zeeman AM, Kocken CHM. Transfection Models to Investigate Plasmodium vivax-Type Dormant Liver Stage Parasites. Pathogens 2023; 12:1070. [PMID: 37764878 PMCID: PMC10534883 DOI: 10.3390/pathogens12091070] [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] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Plasmodium vivax causes the second highest number of malaria morbidity and mortality cases in humans. Several biological traits of this parasite species, including the formation of dormant stages (hypnozoites) that persist inside the liver for prolonged periods of time, present an obstacle for intervention measures and create a barrier for the elimination of malaria. Research into the biology of hypnozoites requires efficient systems for parasite transmission, liver stage cultivation and genetic modification. However, P. vivax research is hampered by the lack of an in vitro blood stage culture system, rendering it reliant on in vivo-derived, mainly patient, material for transmission and liver stage culture. This has also resulted in limited capability for genetic modification, creating a bottleneck in investigations into the mechanisms underlying the persistence of the parasite inside the liver. This bottleneck can be overcome through optimal use of the closely related and experimentally more amenable nonhuman primate (NHP) parasite, Plasmodium cynomolgi, as a model system. In this review, we discuss the genetic modification tools and liver stage cultivation platforms available for studying P. vivax persistent stages and highlight how their combined use may advance our understanding of hypnozoite biology.
Collapse
Affiliation(s)
- Annemarie Voorberg-van der Wel
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands; (A.-M.Z.); (C.H.M.K.)
| | | | | |
Collapse
|
9
|
Baykova SO, Geyl KK, Baykov SV, Boyarskiy VP. Synthesis of 3-(Pyridin-2-yl)quinazolin-2,4(1 H,3 H)-diones via Annulation of Anthranilic Esters with N-pyridyl Ureas. Int J Mol Sci 2023; 24:ijms24087633. [PMID: 37108796 PMCID: PMC10142796 DOI: 10.3390/ijms24087633] [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: 03/29/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
A new route for the synthesis of quinazolin-2,4(1H,3H)-diones and thieno [2,3-d]pyrimidine-2,4(1H,3H)-diones substituted by pyridyl/quinolinyl moiety in position 3 has been developed. The proposed method concluded in an annulation of substituted anthranilic esters or 2-aminothiophene-3-carboxylates with 1,1-dimethyl-3-(pyridin-2-yl) ureas. The process consists of the formation of N-aryl-N'-pyridyl ureas followed by their cyclocondensation into the corresponding fused heterocycles. The reaction does not require the use of metal catalysts and proceeds with moderate to good yields (up to 89%). The scope of the method is more than 30 examples, including compounds with both electron-withdrawing and electron-donating groups, as well as diverse functionalities. At the same time, strong electron-acceptor substituents in the pyridine ring of the starting ureas reduce the product yield or even prevent the cyclocondensation step. The reaction can be easily scaled to gram quantities.
Collapse
Affiliation(s)
- Svetlana O Baykova
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., Saint Petersburg 199034, Russia
| | - Kirill K Geyl
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., Saint Petersburg 199034, Russia
| | - Sergey V Baykov
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., Saint Petersburg 199034, Russia
| | - Vadim P Boyarskiy
- Institute of Chemistry, Saint Petersburg State University, 7/9 Universitetskaya Nab., Saint Petersburg 199034, Russia
| |
Collapse
|
10
|
Almaliti J, Fajtová P, Calla J, LaMonte GM, Feng M, Rocamora F, Ottilie S, Glukhov E, Boura E, Suhandynata RT, Momper JD, Gilson MK, Winzeler EA, Gerwick WH, O'Donoghue AJ. Development of Potent and Highly Selective Epoxyketone-Based Plasmodium Proteasome Inhibitors. Chemistry 2023; 29:e202203958. [PMID: 36617500 PMCID: PMC10894724 DOI: 10.1002/chem.202203958] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023]
Abstract
Here, we present remarkable epoxyketone-based proteasome inhibitors with low nanomolar in vitro potency for blood-stage Plasmodium falciparum and low cytotoxicity for human cells. Our best compound has more than 2,000-fold greater selectivity for erythrocytic-stage P. falciparum over HepG2 and H460 cells, which is largely driven by the accommodation of the parasite proteasome for a D-amino acid in the P3 position and the preference for a difluorobenzyl group in the P1 position. We isolated the proteasome from P. falciparum cell extracts and determined that the best compound is 171-fold more potent at inhibiting the β5 subunit of P. falciparum proteasome when compared to the same subunit of the human constitutive proteasome. These compounds also significantly reduce parasitemia in a P. berghei mouse infection model and prolong survival of animals by an average of 6 days. The current epoxyketone inhibitors are ideal starting compounds for orally bioavailable anti-malarial drugs.
Collapse
Affiliation(s)
- Jehad Almaliti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Department Pharmaceutical Sciences, College of Pharmacy, University of Jordan, Amman, 11942, Jordan
| | - Pavla Fajtová
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Jaeson Calla
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Gregory M LaMonte
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Mudong Feng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Calibr, a division of The Scripps Research Institute, 11119 N Torrey Pines Rd, La Jolla, California, 92093, USA
| | - Evgenia Glukhov
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Raymond T Suhandynata
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - William H Gerwick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| |
Collapse
|
11
|
Nourani L, Mehrizi AA, Pirahmadi S, Pourhashem Z, Asadollahi E, Jahangiri B. CRISPR/Cas advancements for genome editing, diagnosis, therapeutics, and vaccine development for Plasmodium parasites, and genetic engineering of Anopheles mosquito vector. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 109:105419. [PMID: 36842543 DOI: 10.1016/j.meegid.2023.105419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Malaria as vector-borne disease remains important health concern with over 200 million cases globally. Novel antimalarial medicines and more effective vaccines must be developed to eliminate and eradicate malaria. Appraisal of preceding genome editing approaches confirmed the CRISPR/Cas nuclease system as a novel proficient genome editing system and a tool for species-specific diagnosis, and drug resistance researches for Plasmodium species, and gene drive to control Anopheles population. CRISPR/Cas technology, as a handy tool for genome editing can be justified for the production of transgenic malaria parasites like Plasmodium transgenic lines expressing Cas9, chimeric Plasmodium transgenic lines, knockdown and knockout transgenic parasites, and transgenic parasites expressing alternative alleles, and also mutant strains of Anopheles such as only male mosquito populations, generation of wingless mosquitoes, and creation of knock-out/ knock-in mutants. Though, the incorporation of traditional methods and novel molecular techniques could noticeably enhance the quality of results. The striking development of a CRISPR/Cas-based diagnostic kit that can specifically diagnose the Plasmodium species or drug resistance markers is highly required in malaria settings with affordable cost and high-speed detection. Furthermore, the advancement of genome modifications by CRISPR/Cas technologies resolves contemporary restrictions to culturing, maintaining, and analyzing these parasites, and the aptitude to investigate parasite genome functions opens up new vistas in the better understanding of pathogenesis.
Collapse
Affiliation(s)
- Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Elahe Asadollahi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Babak Jahangiri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Istvan ES, Guerra F, Abraham M, Huang KS, Rocamora F, Zhao H, Xu L, Pasaje C, Kumpornsin K, Luth MR, Cui H, Yang T, Diaz SP, Gomez-Lorenzo MG, Qahash T, Mittal N, Ottilie S, Niles J, Lee MCS, Llinas M, Kato N, Okombo J, Fidock DA, Schimmel P, Gamo FJ, Goldberg DE, Winzeler EA. Cytoplasmic isoleucyl tRNA synthetase as an attractive multistage antimalarial drug target. Sci Transl Med 2023; 15:eadc9249. [PMID: 36888694 PMCID: PMC10286833 DOI: 10.1126/scitranslmed.adc9249] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
Development of antimalarial compounds into clinical candidates remains costly and arduous without detailed knowledge of the target. As resistance increases and treatment options at various stages of disease are limited, it is critical to identify multistage drug targets that are readily interrogated in biochemical assays. Whole-genome sequencing of 18 parasite clones evolved using thienopyrimidine compounds with submicromolar, rapid-killing, pan-life cycle antiparasitic activity showed that all had acquired mutations in the P. falciparum cytoplasmic isoleucyl tRNA synthetase (cIRS). Engineering two of the mutations into drug-naïve parasites recapitulated the resistance phenotype, and parasites with conditional knockdowns of cIRS became hypersensitive to two thienopyrimidines. Purified recombinant P. vivax cIRS inhibition, cross-resistance, and biochemical assays indicated a noncompetitive, allosteric binding site that is distinct from that of known cIRS inhibitors mupirocin and reveromycin A. Our data show that Plasmodium cIRS is an important chemically and genetically validated target for next-generation medicines for malaria.
Collapse
Affiliation(s)
- Eva S. Istvan
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Francisco Guerra
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Matthew Abraham
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | - Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | - Lan Xu
- The Global Health Drug Discovery Institute, Tsinghua University 30 Shuangqing Rd, Haidian District, Beijing, China
| | - Charisse Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Madeline R. Luth
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Haissi Cui
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tuo Yang
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Sara Palomo Diaz
- Global Health Medicines, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Spain
| | | | - Tarrick Qahash
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Huck Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA
| | - Nimisha Mittal
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Jacquin Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Marcus C. S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Manuel Llinas
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Huck Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Nobutaka Kato
- The Global Health Drug Discovery Institute, Tsinghua University 30 Shuangqing Rd, Haidian District, Beijing, China
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Paul Schimmel
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Elizabeth A. Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
15
|
Schnyder JL, de Jong HK, Bache EB, van Hest RM, Schlagenhauf P, Borrmann S, Hanscheid T, Grobusch MP. On the potential for discontinuing atovaquone-proguanil (AP) ad-hoc post-exposure and other abbreviated AP-regimens: Pharmacology, pharmacokinetics and perspectives. Travel Med Infect Dis 2022; 52:102520. [PMID: 36526126 DOI: 10.1016/j.tmaid.2022.102520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
According to current guidelines, atovaquone-proguanil (AP) malaria chemoprophylaxis should be taken once daily starting one day before travel and continued for seven days post-exposure. However, drug-sparing regimens, including discontinuing AP after leaving malaria-endemic areas are cost-saving and probably more attractive to travelers, and may thus enhance adherence. AP has causal prophylactic effects, killing malaria parasites during the hepatic stage. If early hepatic stages were already targeted by AP, AP could possibly be discontinued upon return. Pharmacokinetic data and studies on drug-sparing AP regimens suggest this to be the case. Nevertheless, the evidence is weak and considered insufficient to modify current recommendations. Field trials require large numbers of travelers and inherently suffer from the lack of a control group. Safely-designed controlled human malaria infection trials could significantly reduce study participant numbers and safely establish an effective AP abbreviated regimen which we propose as the optimal trial design to test this concept.
Collapse
Affiliation(s)
- Jenny L Schnyder
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Hanna K de Jong
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Emmanuel B Bache
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Reinier M van Hest
- Department of Hospital Pharmacy & Clinical Pharmacology, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands
| | - Patricia Schlagenhauf
- University of Zurich Centre for Travel Medicine, WHO Collaborating Centre for Travelers' Health, Department of Public and Global Health, Military Medicine Biology Competence Centre, Institute for Epidemiology, Biostatistics and Prevention, Zurich, Switzerland
| | - Steffen Borrmann
- Institute of Tropical Medicine, German Centre for Infection Research (DZIF), University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales en Lambaréné (CERMEL), Lambaréné, Gabon
| | - Thomas Hanscheid
- Instituto de Microbiologia, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Martin P Grobusch
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Location University of Amsterdam, Amsterdam, Netherlands; Institute of Tropical Medicine, German Centre for Infection Research (DZIF), University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales en Lambaréné (CERMEL), Lambaréné, Gabon; Masanga Medical Research Unit (MMRU), Masanga, Sierra Leone; Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
16
|
Challis MP, Devine SM, Creek DJ. Current and emerging target identification methods for novel antimalarials. Int J Parasitol Drugs Drug Resist 2022; 20:135-144. [PMID: 36410177 PMCID: PMC9771836 DOI: 10.1016/j.ijpddr.2022.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
New antimalarial compounds with novel mechanisms of action are urgently needed to combat the recent rise in antimalarial drug resistance. Phenotypic high-throughput screens have proven to be a successful method for identifying new compounds, however, do not provide mechanistic information about the molecular target(s) responsible for antimalarial action. Current and emerging target identification methods such as in vitro resistance generation, metabolomics screening, chemoproteomic approaches and biophysical assays measuring protein stability across the whole proteome have successfully identified novel drug targets. This review provides an overview of these techniques, comparing their strengths and weaknesses and how they can be utilised for antimalarial target identification.
Collapse
Affiliation(s)
- Matthew P. Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Shane M. Devine
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia,Corresponding author. Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
17
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
18
|
Mughal H, Bell EC, Mughal K, Derbyshire ER, Freundlich JS. Random Forest Model Predictions Afford Dual-Stage Antimalarial Agents. ACS Infect Dis 2022; 8:1553-1562. [PMID: 35894649 PMCID: PMC9987178 DOI: 10.1021/acsinfecdis.2c00189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The need for novel antimalarials is apparent given the continuing disease burden worldwide, despite significant drug discovery advances from the bench to the bedside. In particular, small-molecule agents with potent efficacy against both the liver and blood stages of Plasmodium parasite infection are critical for clinical settings as they would simultaneously prevent and treat malaria with a reduced selection pressure for resistance. While experimental screens for such dual-stage inhibitors have been conducted, the time and cost of these efforts limit their scope. Here, we have focused on leveraging machine learning approaches to discover novel antimalarials with such properties. A random forest modeling approach was taken to predict small molecules with in vitro efficacy versus liver-stage Plasmodium berghei parasites and a lack of human liver cell cytotoxicity. Empirical validation of the model was achieved with the realization of hits with liver-stage efficacy after prospective scoring of a commercial diversity library and consideration of structural diversity. A subset of these hits also demonstrated promising blood-stage Plasmodium falciparum efficacy. These 18 validated dual-stage antimalarials represent novel starting points for drug discovery and mechanism of action studies with significant potential for seeding a new generation of therapies.
Collapse
Affiliation(s)
- Haseeb Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
| | - Elise C. Bell
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Khadija Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
| | - Emily R. Derbyshire
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Joel S. Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
- Department of Medicine, Center for Emerging and Re-emerging Pathogens, Rutgers University – New Jersey Medical School, Newark, NJ, 07103
| |
Collapse
|
19
|
Ottilie S, Luth MR, Hellemann E, Goldgof GM, Vigil E, Kumar P, Cheung AL, Song M, Godinez-Macias KP, Carolino K, Yang J, Lopez G, Abraham M, Tarsio M, LeBlanc E, Whitesell L, Schenken J, Gunawan F, Patel R, Smith J, Love MS, Williams RM, McNamara CW, Gerwick WH, Ideker T, Suzuki Y, Wirth DF, Lukens AK, Kane PM, Cowen LE, Durrant JD, Winzeler EA. Adaptive laboratory evolution in S. cerevisiae highlights role of transcription factors in fungal xenobiotic resistance. Commun Biol 2022; 5:128. [PMID: 35149760 PMCID: PMC8837787 DOI: 10.1038/s42003-022-03076-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022] Open
Abstract
In vitro evolution and whole genome analysis were used to comprehensively identify the genetic determinants of chemical resistance in Saccharomyces cerevisiae. Sequence analysis identified many genes contributing to the resistance phenotype as well as numerous amino acids in potential targets that may play a role in compound binding. Our work shows that compound-target pairs can be conserved across multiple species. The set of 25 most frequently mutated genes was enriched for transcription factors, and for almost 25 percent of the compounds, resistance was mediated by one of 100 independently derived, gain-of-function SNVs found in a 170 amino acid domain in the two Zn2C6 transcription factors YRR1 and YRM1 (p < 1 × 10−100). This remarkable enrichment for transcription factors as drug resistance genes highlights their important role in the evolution of antifungal xenobiotic resistance and underscores the challenge to develop antifungal treatments that maintain potency. Ottilie et al. employ an experimental evolution approach to investigate the role of transcription factors in yeast chemical resistance. Most emergent mutations in resistant strains were enriched in transcription factor coding genes, highlighting their importance in drug resistance.
Collapse
Affiliation(s)
- Sabine Ottilie
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Madeline R Luth
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Erich Hellemann
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Gregory M Goldgof
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Eddy Vigil
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Prianka Kumar
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Andrea L Cheung
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Miranda Song
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Karla P Godinez-Macias
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Krypton Carolino
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Jennifer Yang
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Gisel Lopez
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Matthew Abraham
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Maureen Tarsio
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, NY, 13210, USA
| | - Emmanuelle LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Jake Schenken
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Felicia Gunawan
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Reysha Patel
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Joshua Smith
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA
| | - Melissa S Love
- Calibr, a division of The Scripps Research Institutes, La Jolla, CA, 92037, USA
| | - Roy M Williams
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA.,Aspen Neuroscience, San Diego, CA, 92121, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institutes, La Jolla, CA, 92037, USA
| | - William H Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, La Jolla, CA, 92037, USA
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yo Suzuki
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, La Jolla, CA, 92037, 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, Broad Institute, Cambridge, MA, 02142, USA
| | - Amanda K Lukens
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, NY, 13210, USA
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, University of California, San Diego, Gilman Dr, La Jolla, CA, 92093, USA.
| |
Collapse
|
20
|
New insights into antimalarial chemopreventive activity of antifolates. Antimicrob Agents Chemother 2021; 66:e0153821. [PMID: 34930029 DOI: 10.1128/aac.01538-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antifolates targeting dihydrofolate reductase (DHFR) are antimalarial compounds that have long been used for malaria treatment and chemoprevention (inhibition of infection from mosquitoes to humans). Despite their extensive applications, the thorough understanding of antifolate activity against hepatic malaria parasites, especially resistant parasites, have yet to be achieved. Using a transgenic P. berghei harboring quadruple mutant dhfr from P. falciparum (Pb::Pfdhfr-4M), we demonstrate that quadruple mutations on Pfdhfr confer complete chemoprevention resistance to pyrimethamine, the previous generation of antifolate, but not to a new class of antifolate designed to overcome the resistance such as P218. Detailed investigation to pin-point stage-specific chemoprevention further demonstrated that it is unnecessary for the drug to be present throughout hepatic development. The drug is most potent against the developmental stages from early hepatic trophozoite to late hepatic trophozoite, but is not effective at inhibiting sporozoite and early hepatic stage development from sporozoite to early trophozoite. Our data shows that P218 also inhibited the late hepatic stage development, from trophozoite to mature schizonts to a lesser extent. With a single dose of 15 mg/kg, P218 prevented infection from up to 25,000 pyrimethamine-resistant sporozoites, a number equal to thousands of infectious mosquito bites. Additionally, the hepatic stage of malaria parasite is much more susceptible to antifolates than the asexual blood stage. This study provides important insights into the activity of antifolates, as a chemopreventive therapeutic which could lead to a more efficient and cost effective treatment regime.
Collapse
|
21
|
Mourot L, Schmitt M, Mouray E, Spichty M, Florent I, Albrecht S. Structure-activity relationship and molecular modelling studies of quinazolinedione derivatives MMV665916 as potential antimalarial agent. Bioorg Med Chem 2021; 51:116513. [PMID: 34798379 DOI: 10.1016/j.bmc.2021.116513] [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: 06/23/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
A series of new quinazolinedione derivatives have been readily synthesized and evaluated for their in vitro antiplasmodial growth inhibition activity. Most of the compounds inhibited P. falciparum FcB1 strain in the low to medium micromolar concentration. The 2-ethoxy 8ag', 2-trifluoromethoxy 8ai' and 4-fluoro-2-methoxy 8ak' showed the best inhibitory activity with EC50 values around 5 µM and were non-toxic to the primary human fibroblast cell line AB943. However, these compounds were less potent than the original hit MMV665916, which showed remarkable growth inhibition with EC50 value of 0.4 µM and presented the highest selectivity index (SI > 250). In addition, a novel approach for determining the docking poses of these quinazolinedione derivatives with their potential protein target, the P. falciparum farnesyltransferase PfFT, was investigated.
Collapse
Affiliation(s)
- Laura Mourot
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Marjorie Schmitt
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Elisabeth Mouray
- Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, Muséum National d'Histoire Naturelle, CNRS, Sorbonne Universités, Paris, France
| | - Martin Spichty
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France
| | - Isabelle Florent
- Unité Molécules de Communication et Adaptation des Micro-organismes, UMR7245, Muséum National d'Histoire Naturelle, CNRS, Sorbonne Universités, Paris, France
| | - Sébastien Albrecht
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, F-68000 Mulhouse, France.
| |
Collapse
|
22
|
The Novel bis-1,2,4-Triazine MIPS-0004373 Demonstrates Rapid and Potent Activity against All Blood Stages of the Malaria Parasite. Antimicrob Agents Chemother 2021; 65:e0031121. [PMID: 34460304 DOI: 10.1128/aac.00311-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Novel bis-1,2,4-triazine compounds with potent in vitro activity against Plasmodium falciparum parasites were recently identified. The bis-1,2,4-triazines represent a unique antimalarial pharmacophore and are proposed to act by a novel but as-yet-unknown mechanism of action. This study investigated the activity of the bis-1,2,4-triazine MIPS-0004373 across the mammalian life cycle stages of the parasite and profiled the kinetics of activity against blood and transmission stage parasites in vitro and in vivo. MIPS-0004373 demonstrated rapid and potent activity against P. falciparum, with excellent in vitro activity against all asexual blood stages. Prolonged in vitro drug exposure failed to generate stable resistance de novo, suggesting a low propensity for the emergence of resistance. Excellent activity was observed against sexually committed ring stage parasites, but activity against mature gametocytes was limited to inhibiting male gametogenesis. Assessment of liver stage activity demonstrated good activity in an in vitro P. berghei model but no activity against Plasmodium cynomolgi hypnozoites or liver schizonts. The bis-1,2,4-triazine MIPS-0004373 efficiently cleared an established P. berghei infection in vivo, with efficacy similar to that of artesunate and chloroquine and a recrudescence profile comparable to that of chloroquine. This study demonstrates the suitability of bis-1,2,4-triazines for further development toward a novel treatment for acute malaria.
Collapse
|
23
|
Design of proteasome inhibitors with oral efficacy in vivo against Plasmodium falciparum and selectivity over the human proteasome. Proc Natl Acad Sci U S A 2021; 118:2107213118. [PMID: 34548400 PMCID: PMC8488693 DOI: 10.1073/pnas.2107213118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
Here, we describe inhibitors of the Plasmodium proteasome, an enzymatic complex that malaria parasites rely on to degrade proteins. Starting from inhibitors developed to treat cancer, derivatives were designed and synthesized with the aim of increasing potency against the Plasmodium proteasome and decreasing activity against the human enzyme. Biochemical and cellular assays identified compounds that exhibit selectivity and potency, both in vitro and in vivo, at different stages of the parasite’s lifecycle. Cryo-electron microscopy revealed that the inhibitors bind in a hydrophobic pocket that is structurally different in the human proteasome—underpinning their selectivity. The work will help develop antimalarial therapeutics, which are desperately needed to treat a disease that kills nearly half a million people annually. The Plasmodium falciparum proteasome is a potential antimalarial drug target. We have identified a series of amino-amide boronates that are potent and specific inhibitors of the P. falciparum 20S proteasome (Pf20S) β5 active site and that exhibit fast-acting antimalarial activity. They selectively inhibit the growth of P. falciparum compared with a human cell line and exhibit high potency against field isolates of P. falciparum and Plasmodium vivax. They have a low propensity for development of resistance and possess liver stage and transmission-blocking activity. Exemplar compounds, MPI-5 and MPI-13, show potent activity against P. falciparum infections in a SCID mouse model with an oral dosing regimen that is well tolerated. We show that MPI-5 binds more strongly to Pf20S than to human constitutive 20S (Hs20Sc). Comparison of the cryo-electron microscopy (EM) structures of Pf20S and Hs20Sc in complex with MPI-5 and Pf20S in complex with the clinically used anti-cancer agent, bortezomib, reveal differences in binding modes that help to explain the selectivity. Together, this work provides insights into the 20S proteasome in P. falciparum, underpinning the design of potent and selective antimalarial proteasome inhibitors.
Collapse
|
24
|
Moraes Barros RR, Thawnashom K, Gibson TJ, Armistead JS, Caleon RL, Kaneko M, Kite WA, Mershon JP, Brockhurst JK, Engels T, Lambert L, Orr-Gonzalez S, Adams JH, Sá JM, Kaneko O, Wellems TE. Activity of Plasmodium vivax promoter elements in Plasmodium knowlesi, and a centromere-containing plasmid that expresses NanoLuc throughout the parasite life cycle. Malar J 2021; 20:247. [PMID: 34090438 PMCID: PMC8180018 DOI: 10.1186/s12936-021-03773-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/16/2021] [Indexed: 12/23/2022] Open
Abstract
Background Plasmodium knowlesi is now the major cause of human malaria in Malaysia, complicating malaria control efforts that must attend to the elimination of multiple Plasmodium species. Recent advances in the cultivation of P. knowlesi erythrocytic-stage parasites in vitro, transformation with exogenous DNA, and infection of mosquitoes with gametocytes from culture have opened up studies of this pathogen without the need for resource-intensive and costly non-human primate (NHP) models. For further understanding and development of methods for parasite transformation in malaria research, this study examined the activity of various trans-species transcriptional control sequences and the influence of Plasmodium vivax centromeric (pvcen) repeats in plasmid-transfected P. knowlesi parasites. Methods In vitro cultivated P. knowlesi parasites were transfected with plasmid constructs that incorporated Plasmodium vivax or Plasmodium falciparum 5′ UTRs driving the expression of bioluminescence markers (firefly luciferase or Nanoluc). Promoter activities were assessed by bioluminescence, and parasites transformed with human resistant allele dihydrofolate reductase-expressing plasmids were selected using antifolates. The stability of transformants carrying pvcen-stabilized episomes was assessed by bioluminescence over a complete parasite life cycle through a rhesus macaque monkey, mosquitoes, and a second rhesus monkey. Results Luciferase expression assessments show that certain P. vivax promoter regions, not functional in the more evolutionarily-distant P. falciparum, can drive transgene expression in P. knowlesi. Further, pvcen repeats may improve the stability of episomal plasmids in P. knowlesi and support detection of NanoLuc-expressing elements over the full parasite life cycle from rhesus macaque monkeys to Anopheles dirus mosquitoes and back again to monkeys. In assays of drug responses to chloroquine, G418 and WR9910, anti-malarial half-inhibitory concentration (IC50) values of blood stages measured by NanoLuc activity proved comparable to IC50 values measured by the standard SYBR Green method. Conclusion All three P. vivax promoters tested in this study functioned in P. knowlesi, whereas two of the three were inactive in P. falciparum. NanoLuc-expressing, centromere-stabilized plasmids may support high-throughput screenings of P. knowlesi for new anti-malarial agents, including compounds that can block the development of mosquito- and/or liver-stage parasites. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03773-4.
Collapse
Affiliation(s)
- Roberto R Moraes Barros
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. .,Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Kittisak Thawnashom
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Tyler J Gibson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer S Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Ramoncito L Caleon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Miho Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Whitney A Kite
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J Patrick Mershon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline K Brockhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theresa Engels
- Division of Veterinary Research, National Institutes of Health, Bethesda, MD, USA
| | - Lynn Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Hermann T, Hochegger P, Dolensky J, Seebacher W, Saf R, Kaiser M, Mäser P, Weis R. New Acyl Derivatives of 3-Aminofurazanes and Their Antiplasmodial Activities. Pharmaceuticals (Basel) 2021; 14:ph14050412. [PMID: 33925485 PMCID: PMC8145535 DOI: 10.3390/ph14050412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 11/16/2022] Open
Abstract
An N-acylated furazan-3-amine of a Medicines for Malaria Venture (MMV) project has shown activity against different strains of Plasmodium falciparum. Seventeen new derivatives were prepared and tested in vitro for their activities against blood stages of two strains of Plasmodium falciparum. Several structure-activity relationships were revealed. The activity strongly depended on the nature of the acyl moiety. Only benzamides showed promising activity. The substitution pattern of their phenyl ring affected the activity and the cytotoxicity of compounds. In addition, physicochemical parameters were calculated (log P, log D, ligand efficiency) or determined experimentally (permeability) via a PAMPA. The N-(4-(3,4-diethoxyphenyl)-1,2,5-oxadiazol-3-yl)-3-(trifluoromethyl)benzamide possessed good physicochemical properties and showed high antiplasmodial activity against a chloroquine-sensitive strain (IC50(NF54) = 0.019 µM) and even higher antiplasmodial activity against a multiresistant strain (IC50(K1) = 0.007 µM). Compared to the MMV compound, the permeability and the activity against the multiresistant strain were improved.
Collapse
Affiliation(s)
- Theresa Hermann
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Schubertstraße 1, A-8010 Graz, Austria; (T.H.); (J.D.); (W.S.); (R.W.)
| | - Patrick Hochegger
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Schubertstraße 1, A-8010 Graz, Austria; (T.H.); (J.D.); (W.S.); (R.W.)
- Correspondence: ; Tel.: +43-316-380-5379; Fax: +43-316-380-9846
| | - Johanna Dolensky
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Schubertstraße 1, A-8010 Graz, Austria; (T.H.); (J.D.); (W.S.); (R.W.)
| | - Werner Seebacher
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Schubertstraße 1, A-8010 Graz, Austria; (T.H.); (J.D.); (W.S.); (R.W.)
| | - Robert Saf
- Institute for Chemistry and Technology of Materials (ICTM), Graz University of Technology, Stremayrgasse 9, A-8010 Graz, Austria;
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Socinstraße 57, CH-4002 Basel, Switzerland; (M.K.); (P.M.)
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Socinstraße 57, CH-4002 Basel, Switzerland; (M.K.); (P.M.)
| | - Robert Weis
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Schubertstraße 1, A-8010 Graz, Austria; (T.H.); (J.D.); (W.S.); (R.W.)
| |
Collapse
|
26
|
Le Manach C, Dam J, Woodland JG, Kaur G, Khonde LP, Brunschwig C, Njoroge M, Wicht KJ, Horatscheck A, Paquet T, Boyle GA, Gibhard L, Taylor D, Lawrence N, Yeo T, Mok S, Eastman RT, Dorjsuren D, Talley DC, Guo H, Simeonov A, Reader J, van der Watt M, Erlank E, Venter N, Zawada JW, Aswat A, Nardini L, Coetzer TL, Lauterbach SB, Bezuidenhout BC, Theron A, Mancama D, Koekemoer LL, Birkholtz LM, Wittlin S, Delves M, Ottilie S, Winzeler EA, von Geldern TW, Smith D, Fidock DA, Street LJ, Basarab GS, Duffy J, Chibale K. Identification and Profiling of a Novel Diazaspiro[3.4]octane Chemical Series Active against Multiple Stages of the Human Malaria Parasite Plasmodium falciparum and Optimization Efforts. J Med Chem 2021; 64:2291-2309. [PMID: 33573376 DOI: 10.1021/acs.jmedchem.1c00034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (<50 nM) together with strong gametocyte sterilizing properties that translated to transmission-blocking activity in the standard membrane feeding assay. Mechanistic studies through resistance selection with one of the analogues followed by whole-genome sequencing implicated the P. falciparum cyclic amine resistance locus in the mode of resistance.
Collapse
Affiliation(s)
- Claire Le Manach
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Jean Dam
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - John G Woodland
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Gurminder Kaur
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Lutete P Khonde
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kathryn J Wicht
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - André Horatscheck
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Tanya Paquet
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Grant A Boyle
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Dale Taylor
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Nina Lawrence
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Richard T Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dorjbal Dorjsuren
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Daniel C Talley
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hui Guo
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Erica Erlank
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Nelius Venter
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Jacek W Zawada
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Ayesha Aswat
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Luisa Nardini
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Theresa L Coetzer
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Sonja B Lauterbach
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Belinda C Bezuidenhout
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Anjo Theron
- Biosciences, Council for Scientific and Industrial Research, P.O. Box 395, Pretoria 0001, South Africa
| | - Dalu Mancama
- Biosciences, Council for Scientific and Industrial Research, P.O. Box 395, Pretoria 0001, South Africa
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland.,University of Basel, 4003 Basel, Switzerland
| | - Michael Delves
- Imperial College London, South Kensington, London SW7 2AZ, U.K.,London School of Hygiene and Tropical Medicine, London WC1E 7HT, U.K
| | - Sabine Ottilie
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093-076, United States
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093-076, United States
| | | | | | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States.,Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Leslie J Street
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Gregory S Basarab
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - James Duffy
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, P.O. Box 1826, 1215 Geneva, Switzerland
| | - Kelly Chibale
- Drug Discovery and Development Center (H3D), 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
|
27
|
Chemoprotective antimalarials identified through quantitative high-throughput screening of Plasmodium blood and liver stage parasites. Sci Rep 2021; 11:2121. [PMID: 33483532 PMCID: PMC7822874 DOI: 10.1038/s41598-021-81486-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration–response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.
Collapse
|
28
|
Multistage and transmission-blocking targeted antimalarials discovered from the open-source MMV Pandemic Response Box. Nat Commun 2021; 12:269. [PMID: 33431834 PMCID: PMC7801607 DOI: 10.1038/s41467-020-20629-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022] Open
Abstract
Chemical matter is needed to target the divergent biology associated with the different life cycle stages of Plasmodium. Here, we report the parallel de novo screening of the Medicines for Malaria Venture (MMV) Pandemic Response Box against Plasmodium asexual and liver stage parasites, stage IV/V gametocytes, gametes, oocysts and as endectocides. Unique chemotypes were identified with both multistage activity or stage-specific activity, including structurally diverse gametocyte-targeted compounds with potent transmission-blocking activity, such as the JmjC inhibitor ML324 and the antitubercular clinical candidate SQ109. Mechanistic investigations prove that ML324 prevents histone demethylation, resulting in aberrant gene expression and death in gametocytes. Moreover, the selection of parasites resistant to SQ109 implicates the druggable V-type H+-ATPase for the reduced sensitivity. Our data therefore provides an expansive dataset of compounds that could be redirected for antimalarial development and also point towards proteins that can be targeted in multiple parasite life cycle stages. Here, Reader et al. screen the Medicines for Malaria Venture Pandemic Response Box in parallel against Plasmodiumasexual and liver stage parasites, stage IV/V gametocytes, gametes, oocysts and as endectocides. They identify two potent transmission-blocking drugs: a histone demethylase inhibitor ML324 and the antitubercular SQ109.
Collapse
|
29
|
Mackwitz MKW, Hesping E, Eribez K, Schöler A, Antonova-Koch Y, Held J, Winzeler EA, Andrews KT, Hansen FK. Investigation of the in vitro and in vivo efficacy of peptoid-based HDAC inhibitors with dual-stage antiplasmodial activity. Eur J Med Chem 2020; 211:113065. [PMID: 33360801 DOI: 10.1016/j.ejmech.2020.113065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 12/20/2022]
Abstract
Histone deacetylases (HDACs) have been identified as emerging antiplasmodial drug targets. In this work, we report on the synthesis, structure-activity relationships, metabolic stability and in vivo efficacy of new peptoid-based HDAC inhibitors with dual-stage antiplasmodial activity. A mini library of HDAC inhibitors was synthesized using a one-pot, multi-component protocol or submonomer pathways. The screening of the target compounds for their activity against asexual blood stage parasites, human cell cytotoxicity, liver stage parasites, and selected human HDAC isoforms provided important structure-activity relationship data. The most promising HDAC inhibitor from this series, compound 3n, demonstrated potent activity against drug-sensitive and drug-resistant asexual stage P. falciparum parasites and was selective for the parasite versus human cells (Pf3D7 IC50 0.016 μM; SIHepG2/Pf3D7 573; PfDd2 IC50 0.002 μM; SIHepG2/PfDd2 4580) combined with activity against P. berghei exoerythrocytic liver stages (PbEEF IC50 0.48 μM). While compound 3n displayed high stability in human (Clint 5 μL/min/mg) and mouse (Clint 6 μL/min/mg) liver microsomes, only modest oral in vivo efficacy was observed in P. berghei infected mice. Together these data provide a foundation for future work to improve the properties of these dual-stage inhibitors as drug leads for malaria.
Collapse
Affiliation(s)
- Marcel K W Mackwitz
- Institute for Drug Discovery, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| | - Eva Hesping
- Griffith Institute for Drug Discovery, 46 Don Young Road, Nathan Campus, Griffith University, QLD, 4111, Australia
| | - Korina Eribez
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, United States
| | - Andrea Schöler
- Institute for Drug Discovery, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| | - Yevgeniya Antonova-Koch
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, United States
| | - Jana Held
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, United States
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, 46 Don Young Road, Nathan Campus, Griffith University, QLD, 4111, Australia.
| | - Finn K Hansen
- Institute for Drug Discovery, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany; Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
30
|
Eagon S, Hammill JT, Sigal M, Ahn KJ, Tryhorn JE, Koch G, Belanger B, Chaplan CA, Loop L, Kashtanova AS, Yniguez K, Lazaro H, Wilkinson SP, Rice AL, Falade MO, Takahashi R, Kim K, Cheung A, DiBernardo C, Kimball JJ, Winzeler EA, Eribez K, Mittal N, Gamo FJ, Crespo B, Churchyard A, García-Barbazán I, Baum J, Anderson MO, Laleu B, Guy RK. Synthesis and Structure-Activity Relationship of Dual-Stage Antimalarial Pyrazolo[3,4- b]pyridines. J Med Chem 2020; 63:11902-11919. [PMID: 32945666 DOI: 10.1021/acs.jmedchem.0c01152] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Malaria remains one of the most deadly infectious diseases, causing hundreds of thousands of deaths each year, primarily in young children and pregnant mothers. Here, we report the discovery and derivatization of a series of pyrazolo[3,4-b]pyridines targeting Plasmodium falciparum, the deadliest species of the malaria parasite. Hit compounds in this series display sub-micromolar in vitro activity against the intraerythrocytic stage of the parasite as well as little to no toxicity against the human fibroblast BJ and liver HepG2 cell lines. In addition, our hit compounds show good activity against the liver stage of the parasite but little activity against the gametocyte stage. Parasitological profiles, including rate of killing, docking, and molecular dynamics studies, suggest that our compounds may target the Qo binding site of cytochrome bc1.
Collapse
Affiliation(s)
- Scott Eagon
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Jared T Hammill
- Department of Pharmaceutical Sciences, University of Kentucky , Lexington, Kentucky 40508, United States
| | - Martina Sigal
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Kevin J Ahn
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Julia E Tryhorn
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Grant Koch
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California 95064, United States
| | - Briana Belanger
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Cory A Chaplan
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Lauren Loop
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Anna S Kashtanova
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Kenya Yniguez
- SRI International, 333 Ravenswood Avenue, Menlo Park, California 94025, United States
| | - Horacio Lazaro
- Promega Biosciences, 277 Granada Drive, San Luis Obispo, California 93401, United States
| | - Steven P Wilkinson
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Amy L Rice
- Department of Pharmaceutical Sciences, University of Kentucky , Lexington, Kentucky 40508, United States
| | - Mofolusho O Falade
- Department of Pharmaceutical Sciences, University of Kentucky , Lexington, Kentucky 40508, United States
| | - Rei Takahashi
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Katie Kim
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Ashley Cheung
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Celine DiBernardo
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California 93407, United States
| | - Joshua J Kimball
- Promega Biosciences, 277 Granada Drive, San Luis Obispo, California 93401, United States
| | - Elizabeth A Winzeler
- School of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Korina Eribez
- School of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | - Nimisha Mittal
- School of Medicine, University of California San Diego, La Jolla, California 92093, United States
| | | | - Benigno Crespo
- GlaxoSmithKline, Global Health, DDW, Tres Cantos, Madrid 28760, Spain
| | - Alisje Churchyard
- Department of Life Sciences, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Irene García-Barbazán
- Department of Life Sciences, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Jake Baum
- Department of Life Sciences, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Marc O Anderson
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California 94132, United States
| | - Benoît Laleu
- Medicines for Malaria Venture (MMV), P.O. Box 1826, 20, Route de Pré-Bois, Geneva 1215, Switzerland
| | - R Kiplin Guy
- Department of Pharmaceutical Sciences, University of Kentucky , Lexington, Kentucky 40508, United States
| |
Collapse
|
31
|
Coley CW, Eyke NS, Jensen KF. Autonomous Discovery in the Chemical Sciences Part I: Progress. Angew Chem Int Ed Engl 2020; 59:22858-22893. [DOI: 10.1002/anie.201909987] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Connor W. Coley
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Natalie S. Eyke
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Klavs F. Jensen
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| |
Collapse
|
32
|
Coley CW, Eyke NS, Jensen KF. Autonome Entdeckung in den chemischen Wissenschaften, Teil I: Fortschritt. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201909987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Connor W. Coley
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Natalie S. Eyke
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| | - Klavs F. Jensen
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge MA 02139 USA
| |
Collapse
|
33
|
Malebo HM, D'Alessandro S, Ebstie YA, Sorè H, Tenoh Guedoung AR, Katani SJ, Parapini S, Taramelli D, Habluetzel A. In vitro Multistage Malaria Transmission Blocking Activity of Selected Malaria Box Compounds. Drug Des Devel Ther 2020; 14:1593-1607. [PMID: 32425505 PMCID: PMC7196193 DOI: 10.2147/dddt.s242883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/20/2020] [Indexed: 12/04/2022] Open
Abstract
Purpose Continuous efforts into the discovery and development of new antimalarials are required to face the emerging resistance of the parasite to available treatments. Thus, new effective drugs, ideally able to inhibit the Plasmodium life-cycle stages that cause the disease as well as those responsible for its transmission, are needed. Eight compounds from the Medicines for Malaria Venture (MMV) Malaria Box, potentially interfering with the parasite polyamine biosynthesis were selected and assessed in vitro for activity against malaria transmissible stages, namely mature gametocytes and early sporogonic stages. Methods Compound activity against asexual blood stages of chloroquine-sensitive 3D7 and chloroquine-resistant W2 strains of Plasmodium falciparum was tested measuring the parasite lactate dehydrogenase activity. The gametocytocidal effect was determined against the P. falciparum 3D7elo1-pfs16-CBG99 strain with a luminescent method. The murine P. berghei CTRP.GFP strain was employed to assess compounds activities against early sporogonic stage development in an in vitro assay simulating mosquito midgut conditions. Results Among the eight tested molecules, MMV000642, MMV000662 and MMV006429, containing a 1,2,3,4-tetrahydroisoquinoline-4-carboxamide chemical skeleton substituted at N-2, C-3 and C-4, displayed multi-stage activity. Activity against asexual blood stages of both strains was confirmed with values of IC50 (50% inhibitory concentration) in the range of 0.07–0.13 µM. They were also active against mature stage V gametocytes with IC50 values below 5 µM (range: 3.43–4.42 µM). These molecules exhibited moderate effects on early sporogonic stage development, displaying IC50 values between 20 and 40 µM. Conclusion Given the multi-stage, transmission-blocking profiles of MMV000642, MMV000662, MMV006429, and their chemical characteristics, these compounds can be considered worthy for further optimisation toward a TCP5 or TCP6 target product profile proposed by MMV for transmission-blocking antimalarials.
Collapse
Affiliation(s)
- Hamisi M Malebo
- Department of Traditional Medicine Research, National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Sarah D'Alessandro
- Dipartimento di Scienze Biomediche per la Salute , University of Milan, Milan, Italy.,Centro Interuniversitario di Ricerca Sulla Malaria/Italian Malaria Network, University of Milan, Milan, Italy
| | | | - Harouna Sorè
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou, Burkina Faso
| | | | - Shaaban J Katani
- Department of Traditional Medicine Research, National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute , University of Milan, Milan, Italy.,Centro Interuniversitario di Ricerca Sulla Malaria/Italian Malaria Network, University of Milan, Milan, Italy
| | - Donatella Taramelli
- Centro Interuniversitario di Ricerca Sulla Malaria/Italian Malaria Network, University of Milan, Milan, Italy.,Dipartimento di Scienze Farmacologiche e Biomolecolari, University of Milan, Milan, Italy
| | - Annette Habluetzel
- Centro Interuniversitario di Ricerca Sulla Malaria/Italian Malaria Network, University of Milan, Milan, Italy.,School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
34
|
Abstract
As the world gets closer to eliminating malaria, the scientific community worldwide has begun to realize the importance of malaria transmission-blocking interventions. The onus of breaking the life cycle of the human malaria parasite Plasmodium falciparum predominantly rests upon transmission-blocking drugs because of emerging resistance to commonly used schizonticides and insecticides. This third part of our review series on malaria transmission-blocking entails transmission-blocking potential of preclinical transmission-blocking antimalarials and other non-malaria drugs/experimental compounds that are not in clinical or preclinical development for malaria but possess transmission-blocking potential. Collective analysis of the structure and the activity of these experimental compounds might pave the way toward generation of novel prototypes of next-generation transmission-blocking drugs.
Collapse
|
35
|
Voorberg-van der Wel A, Zeeman AM, Nieuwenhuis IG, van der Werff NM, Klooster EJ, Klop O, Vermaat LC, Kocken CHM. Dual-Luciferase-Based Fast and Sensitive Detection of Malaria Hypnozoites for the Discovery of Antirelapse Compounds. Anal Chem 2020; 92:6667-6675. [PMID: 32267675 PMCID: PMC7203758 DOI: 10.1021/acs.analchem.0c00547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/08/2020] [Indexed: 01/24/2023]
Abstract
Efforts to eradicate Plasmodium vivax malaria are hampered by the presence of hypnozoites, persisting stages in the liver that can reactivate after prolonged periods of time enabling further transmission and causing renewed disease. Large-scale drug screening is needed to identify compounds with antihypnozoite activity, but current platforms rely on time-consuming high-content fluorescence imaging as read-out, limiting assay throughput. We here report an ultrafast and sensitive dual-luciferase-based method to differentiate hypnozoites from liver stage schizonts using a transgenic P. cynomolgi parasite line that contains Nanoluc driven by the constitutive hsp70 promoter, as well as firefly luciferase driven by the schizont-specific lisp2 promoter. The transgenic parasite line showed similar fitness and drug sensitivity profiles of selected compounds to wild type. We demonstrate robust bioluminescence-based detection of hypnozoites in 96-well and 384-well plate formats, setting the stage for implementation in large scale drug screens.
Collapse
Affiliation(s)
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Ivonne G. Nieuwenhuis
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Nicole M. van der Werff
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Els J. Klooster
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Onny Klop
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Lars C. Vermaat
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Clemens H. M. Kocken
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
36
|
Abraham M, Gagaring K, Martino ML, Vanaerschot M, Plouffe DM, Calla J, Godinez-Macias KP, Du AY, Wree M, Antonova-Koch Y, Eribez K, Luth MR, Ottilie S, Fidock DA, McNamara CW, Winzeler EA. Probing the Open Global Health Chemical Diversity Library for Multistage-Active Starting Points for Next-Generation Antimalarials. ACS Infect Dis 2020; 6:613-628. [PMID: 32078764 PMCID: PMC7155171 DOI: 10.1021/acsinfecdis.9b00482] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Indexed: 12/13/2022]
Abstract
Most phenotypic screens aiming to discover new antimalarial chemotypes begin with low cost, high-throughput tests against the asexual blood stage (ABS) of the malaria parasite life cycle. Compounds active against the ABS are then sequentially tested in more difficult assays that predict whether a compound has other beneficial attributes. Although applying this strategy to new chemical libraries may yield new leads, repeated iterations may lead to diminishing returns and the rediscovery of chemotypes hitting well-known targets. Here, we adopted a different strategy to find starting points, testing ∼70,000 open source small molecules from the Global Health Chemical Diversity Library for activity against the liver stage, mature sexual stage, and asexual blood stage malaria parasites in parallel. In addition, instead of using an asexual assay that measures accumulated parasite DNA in the presence of compound (SYBR green), a real time luciferase-dependent parasite viability assay was used that distinguishes slow-acting (delayed death) from fast-acting compounds. Among 382 scaffolds with the activity confirmed by dose response (<10 μM), we discovered 68 novel delayed-death, 84 liver stage, and 68 stage V gametocyte inhibitors as well. Although 89% of the evaluated compounds had activity in only a single life cycle stage, we discovered six potent (half-maximal inhibitory concentration of <1 μM) multistage scaffolds, including a novel cytochrome bc1 chemotype. Our data further show the luciferase-based assays have higher sensitivity. Chemoinformatic analysis of positive and negative compounds identified scaffold families with a strong enrichment for activity against specific or multiple stages.
Collapse
Affiliation(s)
- Matthew Abraham
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Kerstin Gagaring
- Calibr, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Marisa L. Martino
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Manu Vanaerschot
- Department
of Microbiology and Immunology, Columbia
University Irving Medical Center, New York, New York 10032, United States
| | - David M. Plouffe
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Jaeson Calla
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Karla P. Godinez-Macias
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Alan Y. Du
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Melanie Wree
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yevgeniya Antonova-Koch
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Korina Eribez
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Madeline R. Luth
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Sabine Ottilie
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - David A. Fidock
- Department
of Microbiology and Immunology, Columbia
University Irving Medical Center, New York, New York 10032, United States
- Division
of Infectious Disease, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Case W. McNamara
- Calibr, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Elizabeth A. Winzeler
- School
of Medicine, University of California San
Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
37
|
Álvarez-Bardón M, Pérez-Pertejo Y, Ordóñez C, Sepúlveda-Crespo D, Carballeira NM, Tekwani BL, Murugesan S, Martinez-Valladares M, García-Estrada C, Reguera RM, Balaña-Fouce R. Screening Marine Natural Products for New Drug Leads against Trypanosomatids and Malaria. Mar Drugs 2020; 18:E187. [PMID: 32244488 PMCID: PMC7230869 DOI: 10.3390/md18040187] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Neglected Tropical Diseases (NTD) represent a serious threat to humans, especially for those living in poor or developing countries. Almost one-sixth of the world population is at risk of suffering from these diseases and many thousands die because of NTDs, to which we should add the sanitary, labor and social issues that hinder the economic development of these countries. Protozoan-borne diseases are responsible for more than one million deaths every year. Visceral leishmaniasis, Chagas disease or sleeping sickness are among the most lethal NTDs. Despite not being considered an NTD by the World Health Organization (WHO), malaria must be added to this sinister group. Malaria, caused by the apicomplexan parasite Plasmodium falciparum, is responsible for thousands of deaths each year. The treatment of this disease has been losing effectiveness year after year. Many of the medicines currently in use are obsolete due to their gradual loss of efficacy, their intrinsic toxicity and the emergence of drug resistance or a lack of adherence to treatment. Therefore, there is an urgent and global need for new drugs. Despite this, the scant interest shown by most of the stakeholders involved in the pharmaceutical industry makes our present therapeutic arsenal scarce, and until recently, the search for new drugs has not been seriously addressed. The sources of new drugs for these and other pathologies include natural products, synthetic molecules or repurposing drugs. The most frequent sources of natural products are microorganisms, e.g., bacteria, fungi, yeasts, algae and plants, which are able to synthesize many drugs that are currently in use (e.g. antimicrobials, antitumor, immunosuppressants, etc.). The marine environment is another well-established source of bioactive natural products, with recent applications against parasites, bacteria and other pathogens which affect humans and animals. Drug discovery techniques have rapidly advanced since the beginning of the millennium. The combination of novel techniques that include the genetic modification of pathogens, bioimaging and robotics has given rise to the standardization of High-Performance Screening platforms in the discovery of drugs. These advancements have accelerated the discovery of new chemical entities with antiparasitic effects. This review presents critical updates regarding the use of High-Throughput Screening (HTS) in the discovery of drugs for NTDs transmitted by protozoa, including malaria, and its application in the discovery of new drugs of marine origin.
Collapse
Affiliation(s)
- María Álvarez-Bardón
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - César Ordóñez
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Daniel Sepúlveda-Crespo
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Nestor M. Carballeira
- Department of Chemistry, University of Puerto Rico, Río Piedras 00925-2537, San Juan, Puerto Rico;
| | - Babu L. Tekwani
- Department of Infectious Diseases, Division of Drug Discovery, Southern Research, Birmingham, AL 35205, USA;
| | - Sankaranarayanan Murugesan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani 333031, India;
| | - Maria Martinez-Valladares
- Department of Animal Health, Instituto de Ganadería de Montaña (CSIC-Universidad de León), Grulleros, 24346 León, Spain;
| | - Carlos García-Estrada
- INBIOTEC (Instituto de Biotecnología de León), Avda. Real 1-Parque Científico de León, 24006 León, Spain;
| | - Rosa M. Reguera
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| | - Rafael Balaña-Fouce
- Department of Biomedical Sciences; University of León, 24071 León, Spain; (M.Á.-B.); (Y.P.-P.); (C.O.); (D.S.-C.); (R.M.R.)
| |
Collapse
|
38
|
Keller L, Siqueira-Neto JL, Souza JM, Eribez K, LaMonte GM, Smith JE, Gerwick WH. Palstimolide A: A Complex Polyhydroxy Macrolide with Antiparasitic Activity. Molecules 2020; 25:molecules25071604. [PMID: 32244512 PMCID: PMC7180531 DOI: 10.3390/molecules25071604] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022] Open
Abstract
Marine Cyanobacteria (blue-green algae) have been shown to possess an enormous potential to produce structurally diverse natural products that exhibit a broad spectrum of potent biological activities, including cytotoxic, antifungal, antiparasitic, antiviral, and antibacterial activities. Here, we report the isolation and structure determination of palstimolide A, a complex polyhydroxy macrolide with a 40-membered ring that was isolated from a tropical marine cyanobacterium collected at Palmyra Atoll. NMR-guided fractionation in combination with MS2-based molecular networking and isolation via HPLC yielded 0.7 mg of the pure compound. The small quantity isolated along with the presence of significant signal degeneracy in both the 1H and 13C-NMR spectra complicated the structure elucidation of palstimolide A. Various NMR experiments and solvent systems were employed, including the LR-HSQMBC experiment that allows the detection of long-range 1H–13C correlation data across 4-, 5-, and even 6-bonds. This expanded NMR data set enabled the elucidation of the palstimolide’s planar structure, which is characterized by several 1,5-disposed hydroxy groups as well as a tert-butyl group. The compound showed potent antimalarial activity with an IC50 of 223 nM as well as interesting anti-leishmanial activity with an IC50 of 4.67 µM.
Collapse
Affiliation(s)
- Lena Keller
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92037, USA;
| | - Jair L. Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (J.L.S.-N.); (J.M.S.)
| | - Julia M. Souza
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (J.L.S.-N.); (J.M.S.)
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP 14404-600, Brazil;
| | - Korina Eribez
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA;
| | - Gregory M. LaMonte
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP 14404-600, Brazil;
| | - Jennifer E. Smith
- Marine Biology Division, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92037, USA;
| | - William H. Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92037, USA;
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (J.L.S.-N.); (J.M.S.)
- Correspondence: ; Tel.: +1-858-534-0576
| |
Collapse
|
39
|
Sweeney-Jones AM, Gagaring K, Antonova-Koch J, Zhou H, Mojib N, Soapi K, Skolnick J, McNamara CW, Kubanek J. Antimalarial Peptide and Polyketide Natural Products from the Fijian Marine Cyanobacterium Moorea producens. Mar Drugs 2020; 18:E167. [PMID: 32197482 PMCID: PMC7142784 DOI: 10.3390/md18030167] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
A new cyclic peptide, kakeromamide B (1), and previously described cytotoxic cyanobacterial natural products ulongamide A (2), lyngbyabellin A (3), 18E-lyngbyaloside C (4), and lyngbyaloside (5) were identified from an antimalarial extract of the Fijian marine cyanobacterium Moorea producens. Compounds 1 and 1 exhibited moderate activity against Plasmodium falciparum blood-stages with EC50 values of 0.89 and 0.99 µM, respectively, whereas 3 was more potent with an EC50 value of 0.15 nM, respectively. Compounds 1, 4, and 5 displayed moderate liver-stage antimalarial activity against P. berghei liver schizonts with EC50 values of 1.1, 0.71, and 0.45 µM, respectively. The threading-based computational method FINDSITEcomb2.0 predicted the binding of 1 and 2 to potentially druggable proteins of Plasmodiumfalciparum, prompting formulation of hypotheses about possible mechanisms of action. Kakeromamide B (1) was predicted to bind to several Plasmodium actin-like proteins and a sortilin protein suggesting possible interference with parasite invasion of host cells. When 1 was tested in a mammalian actin polymerization assay, it stimulated actin polymerization in a dose-dependent manner, suggesting that 1 does, in fact, interact with actin.
Collapse
Affiliation(s)
| | - Kerstin Gagaring
- Calibr, a division of The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jenya Antonova-Koch
- Calibr, a division of The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hongyi Zhou
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Nazia Mojib
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Katy Soapi
- Institute of Applied Sciences, University of the South Pacific, Suva, Fiji
| | - Jeffrey Skolnick
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Case W. McNamara
- Calibr, a division of The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Julia Kubanek
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
40
|
Wong HN, Padín-Irizarry V, van der Watt ME, Reader J, Liebenberg W, Wiesner L, Smith P, Eribez K, Winzeler EA, Kyle DE, Birkholtz LM, Coertzen D, Haynes RK. Optimal 10-Aminoartemisinins With Potent Transmission-Blocking Capabilities for New Artemisinin Combination Therapies-Activities Against Blood Stage P. falciparum Including PfKI3 C580Y Mutants and Liver Stage P. berghei Parasites. Front Chem 2020; 7:901. [PMID: 31998692 PMCID: PMC6967409 DOI: 10.3389/fchem.2019.00901] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022] Open
Abstract
We have demonstrated previously that amino-artemisinins including artemiside and artemisone in which an amino group replaces the oxygen-bearing substituents attached to C-10 of the current clinical artemisinin derivatives dihydroartemisinin (DHA), artemether and artesunate, display potent activities in vitro against the asexual blood stages of Plasmodium falciparum (Pf). In particular, the compounds are active against late blood stage Pf gametocytes, and are strongly synergistic in combination with the redox active drug methylene blue. In order to fortify the eventual selection of optimum amino-artemisinins for development into new triple combination therapies also active against artemisinin-resistant Pf mutants, we have prepared new amino-artemisinins based on the easily accessible and inexpensive DHA-piperazine. The latter was converted into alkyl- and aryl sulfonamides, ureas and amides. These derivatives were screened together with the comparator drugs DHA and the hitherto most active amino-artemisinins artemiside and artemisone against asexual and sexual blood stages of Pf and liver stage P. berghei (Pb) sporozoites. Several of the new amino-artemisinins bearing aryl-urea and -amide groups are potently active against both asexual, and late blood stage gametocytes (IC50 0.4-1.0 nM). Although the activities are superior to those of artemiside (IC50 1.5 nM) and artemisone (IC50 42.4 nM), the latter are more active against the liver stage Pb sporozoites (IC50 artemisone 28 nM). In addition, early results indicate these compounds tend not to display reduced susceptibility against parasites bearing the Pf Kelch 13 propeller domain C580Y mutation characteristic of artemisinin-resistant Pf. Thus, the advent of the amino-artemisinins including artemiside and artemisone will enable the development of new combination therapies that by virtue of the amino-artemisinin component itself will possess intrinsic transmission-blocking capabilities and may be effective against artemisinin resistant falciparum malaria.
Collapse
Affiliation(s)
- Ho Ning Wong
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Vivian Padín-Irizarry
- Center for Tropical & Emerging Global Diseases, Coverdell Center, University of Georgia, Athens, GA, United States
| | - Mariëtte E van der Watt
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Janette Reader
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Wilna Liebenberg
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Peter Smith
- Division of Clinical Pharmacology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Korina Eribez
- School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elizabeth A Winzeler
- School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Dennis E Kyle
- Center for Tropical & Emerging Global Diseases, Coverdell Center, University of Georgia, Athens, GA, United States
| | - Lyn-Marie Birkholtz
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Dina Coertzen
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Richard K Haynes
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
41
|
Alam MM, Sanchez-Azqueta A, Janha O, Flannery EL, Mahindra A, Mapesa K, Char AB, Sriranganadane D, Brancucci NMB, Antonova-Koch Y, Crouch K, Simwela NV, Millar SB, Akinwale J, Mitcheson D, Solyakov L, Dudek K, Jones C, Zapatero C, Doerig C, Nwakanma DC, Vázquez MJ, Colmenarejo G, Lafuente-Monasterio MJ, Leon ML, Godoi PHC, Elkins JM, Waters AP, Jamieson AG, Álvaro EF, Ranford-Cartwright LC, Marti M, Winzeler EA, Gamo FJ, Tobin AB. Validation of the protein kinase PfCLK3 as a multistage cross-species malarial drug target. Science 2019; 365:365/6456/eaau1682. [PMID: 31467193 DOI: 10.1126/science.aau1682] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 03/15/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022]
Abstract
The requirement for next-generation antimalarials to be both curative and transmission-blocking necessitates the identification of previously undiscovered druggable molecular pathways. We identified a selective inhibitor of the Plasmodium falciparum protein kinase PfCLK3, which we used in combination with chemogenetics to validate PfCLK3 as a drug target acting at multiple parasite life stages. Consistent with a role for PfCLK3 in RNA splicing, inhibition resulted in the down-regulation of more than 400 essential parasite genes. Inhibition of PfCLK3 mediated rapid killing of asexual liver- and blood-stage P. falciparum and blockade of gametocyte development, thereby preventing transmission, and also showed parasiticidal activity against P. berghei and P. knowlesi Hence, our data establish PfCLK3 as a target for drugs, with the potential to offer a cure-to be prophylactic and transmission blocking in malaria.
Collapse
Affiliation(s)
- Mahmood M Alam
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ana Sanchez-Azqueta
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Omar Janha
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Erika L Flannery
- Novartis Institute for Biomedical Research, Emeryville, CA 94608, USA
| | - Amit Mahindra
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, UK
| | - Kopano Mapesa
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, UK
| | - Aditya B Char
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow G12 8QQ, UK
| | - Dev Sriranganadane
- Structural Genomics Consortium, Universidade Estadual de Campinas, Campinas, São Paulo 13083-886, Brazil
| | - Nicolas M B Brancucci
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
| | - Yevgeniya Antonova-Koch
- Skaggs School of Pharmaceutical Sciences, UC Health Sciences Center for Immunology, Infection and Inflammation, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Kathryn Crouch
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Nelson Victor Simwela
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Scott B Millar
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jude Akinwale
- Medical Research Council Toxicology Unit, University of Leicester, Leicester LE1 9HN, UK
| | - Deborah Mitcheson
- Department of Molecular Cell Biology, University of Leicester, Leicester LE1 9HN, UK
| | - Lev Solyakov
- Medical Research Council Toxicology Unit, University of Leicester, Leicester LE1 9HN, UK
| | - Kate Dudek
- Medical Research Council Toxicology Unit, University of Leicester, Leicester LE1 9HN, UK
| | - Carolyn Jones
- Medical Research Council Toxicology Unit, University of Leicester, Leicester LE1 9HN, UK
| | - Cleofé Zapatero
- Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
| | - Christian Doerig
- Biomedical Science Cluster, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology, Melbourne, VIC 3000, Australia
| | | | - Maria Jesús Vázquez
- Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
| | - Gonzalo Colmenarejo
- Biostatistics and Bioinformatics Unit, IMDEA Food Institute, 28049 Madrid, Spain
| | | | - Maria Luisa Leon
- Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain
| | - Paulo H C Godoi
- Structural Genomics Consortium, Universidade Estadual de Campinas, Campinas, São Paulo 13083-886, Brazil
| | - Jon M Elkins
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Andrew P Waters
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | | | | | - Lisa C Ranford-Cartwright
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow G12 8QQ, UK
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow G12 8QQ, UK
| | - Elizabeth A Winzeler
- Skaggs School of Pharmaceutical Sciences, UC Health Sciences Center for Immunology, Infection and Inflammation, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | - Andrew B Tobin
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
42
|
Chua ACY, Ong JJY, Malleret B, Suwanarusk R, Kosaisavee V, Zeeman AM, Cooper CA, Tan KSW, Zhang R, Tan BH, Abas SN, Yip A, Elliot A, Joyner CJ, Cho JS, Breyer K, Baran S, Lange A, Maher SP, Nosten F, Bodenreider C, Yeung BKS, Mazier D, Galinski MR, Dereuddre-Bosquet N, Le Grand R, Kocken CHM, Rénia L, Kyle DE, Diagana TT, Snounou G, Russell B, Bifani P. Robust continuous in vitro culture of the Plasmodium cynomolgi erythrocytic stages. Nat Commun 2019; 10:3635. [PMID: 31406175 PMCID: PMC6690977 DOI: 10.1038/s41467-019-11332-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 06/28/2019] [Indexed: 01/19/2023] Open
Abstract
The ability to culture pathogenic organisms substantially enhances the quest for fundamental knowledge and the development of vaccines and drugs. Thus, the elaboration of a protocol for the in vitro cultivation of the erythrocytic stages of Plasmodium falciparum revolutionized research on this important parasite. However, for P. vivax, the most widely distributed and difficult to treat malaria parasite, a strict preference for reticulocytes thwarts efforts to maintain it in vitro. Cultivation of P. cynomolgi, a macaque-infecting species phylogenetically close to P. vivax, was briefly reported in the early 1980s, but not pursued further. Here, we define the conditions under which P. cynomolgi can be adapted to long term in vitro culture to yield parasites that share many of the morphological and phenotypic features of P. vivax. We further validate the potential of this culture system for high-throughput screening to prime and accelerate anti-P. vivax drug discovery efforts. Present understanding of Plasmodium vivax biology is hampered by its inability to grow in vitro. Here, the authors developed an in vitro culture of its simian counterpart, P. cynomolgi, which shares morphological and phenotypic similarities with P. vivax, initiating a new phase in vivax research.
Collapse
Affiliation(s)
- Adeline C Y Chua
- Singapore Immunology Network, A*STAR, Singapore, 138648, Singapore.,Department of Microbiology and Immunology, University of Otago, Dunedin, 9054, New Zealand.,Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | - Jessica Jie Ying Ong
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9054, New Zealand.,Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | - Benoit Malleret
- Singapore Immunology Network, A*STAR, Singapore, 138648, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Rossarin Suwanarusk
- Singapore Immunology Network, A*STAR, Singapore, 138648, Singapore.,Department of Microbiology and Immunology, University of Otago, Dunedin, 9054, New Zealand
| | - Varakorn Kosaisavee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore.,Department of Parasitology and Entomology, Faculty of Public Health, Mahidol University, Bangkok, 10400, Thailand
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288, The Netherlands
| | - Caitlin A Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602, USA
| | - Kevin S W Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Rou Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Bee Huat Tan
- Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | | | - Andy Yip
- Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | - Anne Elliot
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602, USA
| | - Chester J Joyner
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, 30322, USA.,Emory Vaccine Center, Emory University, Atlanta, 30317, USA
| | - Jee Sun Cho
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Kate Breyer
- Laboratory Animal Services, Scientific Operations, Novartis Institutes for Biomedical Research, East Hanover, 07936-1080, USA
| | - Szczepan Baran
- Laboratory Animal Services, Scientific Operations, Novartis Institutes for Biomedical Research, East Hanover, 07936-1080, USA
| | - Amber Lange
- Laboratory Animal Services, Scientific Operations, Novartis Institutes for Biomedical Research, East Hanover, 07936-1080, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602, USA
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, 63110, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford Old Road Campus, Oxford, OX3 7FZ, UK
| | | | - Bryan K S Yeung
- Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | - Dominique Mazier
- Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, F-75013, France.,CIMI-Paris, INSERM, U1135, CNRS, Paris, F-75013, France
| | - Mary R Galinski
- Emory Vaccine Center, Emory University, Atlanta, 30317, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, 30322, USA
| | - Nathalie Dereuddre-Bosquet
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBJF, DRF, Fontenay-aux-Roses, 92265, France
| | - Roger Le Grand
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBJF, DRF, Fontenay-aux-Roses, 92265, France
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288, The Netherlands
| | - Laurent Rénia
- Singapore Immunology Network, A*STAR, Singapore, 138648, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, 30602, USA
| | - Thierry T Diagana
- Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore
| | - Georges Snounou
- Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, F-75013, France.,CIMI-Paris, INSERM, U1135, CNRS, Paris, F-75013, France.,CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, IBJF, DRF, Fontenay-aux-Roses, 92265, France
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9054, New Zealand.
| | - Pablo Bifani
- Singapore Immunology Network, A*STAR, Singapore, 138648, Singapore. .,Novartis Institute for Tropical Diseases, Singapore, 138670, Singapore. .,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore. .,Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
43
|
Ashton TD, Devine SM, Möhrle JJ, Laleu B, Burrows JN, Charman SA, Creek DJ, Sleebs BE. The Development Process for Discovery and Clinical Advancement of Modern Antimalarials. J Med Chem 2019; 62:10526-10562. [DOI: 10.1021/acs.jmedchem.9b00761] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Trent D. Ashton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Shane M. Devine
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jörg J. Möhrle
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Benoît Laleu
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Jeremy N. Burrows
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Susan A. Charman
- 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
| | - Brad E. Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
44
|
Norcross NR, Wilson C, Baragaña B, Hallyburton I, Osuna‐Cabello M, Norval S, Riley J, Fletcher D, Sinden R, Delves M, Ruecker A, Duffy S, Meister S, Antonova‐Koch Y, Crespo B, de Cózar C, Sanz LM, Gamo FJ, Avery VM, Frearson JA, Gray DW, Fairlamb AH, Winzeler EA, Waterson D, Campbell SF, Willis PA, Read KD, Gilbert IH. Substituted Aminoacetamides as Novel Leads for Malaria Treatment. ChemMedChem 2019; 14:1329-1335. [PMID: 31188540 PMCID: PMC6899483 DOI: 10.1002/cmdc.201900329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Indexed: 01/29/2023]
Abstract
Herein we describe the optimization of a phenotypic hit against Plasmodium falciparum based on an aminoacetamide scaffold. This led to N-(3-chloro-4-fluorophenyl)-2-methyl-2-{[4-methyl-3-(morpholinosulfonyl)phenyl]amino}propanamide (compound 28) with low-nanomolar activity against the intraerythrocytic stages of the malaria parasite, and which was found to be inactive in a mammalian cell counter-screen up to 25 μm. Inhibition of gametes in the dual gamete activation assay suggests that this family of compounds may also have transmission blocking capabilities. Whilst we were unable to optimize the aqueous solubility and microsomal stability to a point at which the aminoacetamides would be suitable for in vivo pharmacokinetic and efficacy studies, compound 28 displayed excellent antimalarial potency and selectivity; it could therefore serve as a suitable chemical tool for drug target identification.
Collapse
Affiliation(s)
- Neil R. Norcross
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Caroline Wilson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Beatriz Baragaña
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Irene Hallyburton
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Maria Osuna‐Cabello
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Suzanne Norval
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Jennifer Riley
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Daniel Fletcher
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | | | | | | | - Sandra Duffy
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Stephan Meister
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Yevgeniya Antonova‐Koch
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Benigno Crespo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Cristina de Cózar
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Laura M. Sanz
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Francisco Javier Gamo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Vicky M. Avery
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Julie A. Frearson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - David W. Gray
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Alan H. Fairlamb
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Elizabeth A. Winzeler
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - David Waterson
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Simon F. Campbell
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Paul A. Willis
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Kevin D. Read
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Ian H. Gilbert
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| |
Collapse
|
45
|
Khatri Chhetri B, Lavoie S, Sweeney-Jones AM, Mojib N, Raghavan V, Gagaring K, Dale B, McNamara CW, Soapi K, Quave CL, Polavarapu PL, Kubanek J. Peyssonnosides A-B, Unusual Diterpene Glycosides with a Sterically Encumbered Cyclopropane Motif: Structure Elucidation Using an Integrated Spectroscopic and Computational Workflow. J Org Chem 2019; 84:8531-8541. [PMID: 31244158 PMCID: PMC6614789 DOI: 10.1021/acs.joc.9b00884] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Indexed: 11/30/2022]
Abstract
Two sulfated diterpene glycosides featuring a highly substituted and sterically encumbered cyclopropane ring have been isolated from the marine red alga Peyssonnelia sp. Combination of a wide array of 2D NMR spectroscopic experiments, in a systematic structure elucidation workflow, revealed that peyssonnosides A-B (1-2) represent a new class of diterpene glycosides with a tetracyclo [7.5.0.01,10.05,9] tetradecane architecture. A salient feature of this workflow is the unique application of quantitative interproton distances obtained from the rotating frame Overhauser effect spectroscopy (ROESY) NMR experiment, wherein the β-d-glucose moiety of 1 was used as an internal probe to unequivocally determine the absolute configuration, which was also supported by optical rotatory dispersion (ORD). Peyssonnoside A (1) exhibited promising activity against liver stage Plasmodium berghei and moderate antimethicillin-resistant Staphylococcus aureus (MRSA) activity, with no cytotoxicity against human keratinocytes. Additionally, 1 showed strong growth inhibition of the marine fungus Dendryphiella salina indicating an antifungal ecological role in its natural environment. The high natural abundance and novel carbon skeleton of 1 suggests a rare terpene cyclase machinery, exemplifying the chemical diversity in this phylogenetically distinct marine red alga.
Collapse
Affiliation(s)
- Bhuwan Khatri Chhetri
- School
of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, and School of Biological
Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Serge Lavoie
- School
of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, and School of Biological
Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Institut
des Sciences de la Forêt Tempérée, Université du Québec en Outaouais, 58, rue Principale, Ripon, Québec J0V 1V0, Canada
| | - Anne Marie Sweeney-Jones
- School
of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, and School of Biological
Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Nazia Mojib
- School
of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, and School of Biological
Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Vijay Raghavan
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Kerstin Gagaring
- Calibr
at
The Scripps Research Institute, La Jolla, California 92037, United States
| | - Brandon Dale
- Department
of Dermatology, Center for the Study of Human Health, and Antibiotic
Resistance Center, Emory University, Atlanta, Georgia 30322, United States
| | - Case W. McNamara
- Calibr
at
The Scripps Research Institute, La Jolla, California 92037, United States
| | - Katy Soapi
- Institute
of Applied Sciences, University of South
Pacific, Suva, Fiji
| | - Cassandra L. Quave
- Department
of Dermatology, Center for the Study of Human Health, and Antibiotic
Resistance Center, Emory University, Atlanta, Georgia 30322, United States
| | - Prasad L. Polavarapu
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Julia Kubanek
- School
of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, and School of Biological
Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
46
|
Mackwitz MKW, Hesping E, Antonova-Koch Y, Diedrich D, Woldearegai TG, Skinner-Adams T, Clarke M, Schöler A, Limbach L, Kurz T, Winzeler EA, Held J, Andrews KT, Hansen FK. Structure-Activity and Structure-Toxicity Relationships of Peptoid-Based Histone Deacetylase Inhibitors with Dual-Stage Antiplasmodial Activity. ChemMedChem 2019; 14:912-926. [PMID: 30664827 PMCID: PMC6502651 DOI: 10.1002/cmdc.201800808] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Indexed: 12/17/2022]
Abstract
Novel malaria intervention strategies are of great importance, given the development of drug resistance in malaria-endemic countries. In this regard, histone deacetylases (HDACs) have emerged as new and promising malaria drug targets. In this work, we present the design, synthesis, and biological evaluation of 20 novel HDAC inhibitors with antiplasmodial activity. Based on a previously discovered peptoid-based hit compound, we modified all regions of the peptoid scaffold by using a one-pot multicomponent pathway and submonomer routes to gain a deeper understanding of the structure-activity and structure-toxicity relationships. Most compounds displayed potent activity against asexual blood-stage P. falciparum parasites, with IC50 values in the range of 0.0052-0.25 μm and promising selectivity over mammalian cells (SIPf3D7/HepG2 : 170-1483). In addition, several compounds showed encouraging sub-micromolar activity against P. berghei exo-erythrocytic forms (PbEEF). Our study led to the discovery of the hit compound N-(2-(benzylamino)-2-oxoethyl)-N-(4-(hydroxycarbamoyl)benzyl)-4-isopropylbenzamide (2 h) as a potent and parasite-specific dual-stage antiplasmodial HDAC inhibitor (IC50 Pf3D7=0.0052 μm, IC50 PbEEF=0.016 μm).
Collapse
Affiliation(s)
- Marcel K W Mackwitz
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| | - Eva Hesping
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
| | - Yevgeniya Antonova-Koch
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, USA
| | - Daniela Diedrich
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Tamirat Gebru Woldearegai
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Tina Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
| | - Mary Clarke
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
| | - Andrea Schöler
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| | - Laura Limbach
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA, 92093, USA
| | - Jana Held
- Institut für Tropenmedizin, Eberhard Karls Universität Tübingen, Wilhelmstraße 27, 72074, Tübingen, Germany
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD, 4111, Australia
| | - Finn K Hansen
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Medical Faculty, Leipzig University, Brüderstraße 34, 04103, Leipzig, Germany
| |
Collapse
|
47
|
Marin-Mogollon C, Salman AM, Koolen KMJ, Bolscher JM, van Pul FJA, Miyazaki S, Imai T, Othman AS, Ramesar J, van Gemert GJ, Kroeze H, Chevalley-Maurel S, Franke-Fayard B, Sauerwein RW, Hill AVS, Dechering KJ, Janse CJ, Khan SM. A P. falciparum NF54 Reporter Line Expressing mCherry-Luciferase in Gametocytes, Sporozoites, and Liver-Stages. Front Cell Infect Microbiol 2019; 9:96. [PMID: 31058097 PMCID: PMC6477837 DOI: 10.3389/fcimb.2019.00096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/22/2019] [Indexed: 12/16/2022] Open
Abstract
Transgenic malaria parasites expressing fluorescent and bioluminescent proteins are valuable tools to interrogate malaria-parasite biology and to evaluate drugs and vaccines. Using CRISPR/Cas9 methodology a transgenic Plasmodium falciparum (Pf) NF54 line was generated that expresses a fusion of mCherry and luciferase genes under the control of the Pf etramp10.3 gene promoter (line mCherry-luc@etramp10.3). Pf etramp10.3 is related to rodent Plasmodium uis4 and the uis4 promoter has been used to drive high transgene expression in rodent parasite sporozoites and liver-stages. We examined transgene expression throughout the complete life cycle and compared this expression to transgenic lines expressing mCherry-luciferase and GFP-luciferase under control of the constitutive gapdh and eef1a promoters. The mCherry-luc@etramp10.3 parasites express mCherry in gametocytes, sporozoites, and liver-stages. While no mCherry signal was detected in asexual blood-stage parasites above background levels, luciferase expression was detected in asexual blood-stages, as well as in gametocytes, sporozoites and liver-stages, with the highest levels of reporter expression detected in stage III-V gametocytes and in sporozoites. The expression of mCherry and luciferase in gametocytes and sporozoites makes this transgenic parasite line suitable to use in in vitro assays that examine the effect of transmission blocking inhibitors and to analyse gametocyte and sporozoite biology.
Collapse
Affiliation(s)
| | - Ahmed M Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Fiona J A van Pul
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ahmad Syibli Othman
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Robert W Sauerwein
- TropIQ Health Sciences, Nijmegen, Netherlands.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
48
|
Yahiya S, Rueda-Zubiaurre A, Delves MJ, Fuchter MJ, Baum J. The antimalarial screening landscape-looking beyond the asexual blood stage. Curr Opin Chem Biol 2019; 50:1-9. [PMID: 30875617 PMCID: PMC6591700 DOI: 10.1016/j.cbpa.2019.01.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022]
Abstract
In recent years, the research agenda to tackle global morbidity and mortality from malaria disease has shifted towards innovation, in the hope that efforts at the frontiers of scientific research may re-invigorate gains made towards eradication. Discovery of new antimalarial drugs with novel chemotypes or modes of action lie at the heart of these efforts. There is a particular interest in drug candidates that target stages of the malaria parasite lifecycle beyond the symptomatic asexual blood stages. This is especially important given the spectre of emerging drug resistance to all current frontline antimalarials. One approach gaining increased interest is the potential of designing novel drugs that target parasite passage from infected individual to feeding mosquito and back again. Action of such therapeutics is geared much more at the population level rather than just concerned with the infected individual. The search for novel drugs active against these stages has been helped by improvements to in vitro culture of transmission and pre-erythrocytic parasite lifecycle stages, robotic automation and high content imaging, methodologies that permit the high-throughput screening (HTS) of compound libraries for drug discovery. Here, we review recent advances in the antimalarial screening landscape, focussed on transmission blocking as a key aim for drug-treatment campaigns of the future.
Collapse
Affiliation(s)
- Sabrina Yahiya
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Ainoa Rueda-Zubiaurre
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Michael J Delves
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Matthew J Fuchter
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 OBZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK.
| |
Collapse
|
49
|
Antonova-Koch Y, Meister S, Abraham M, Luth MR, Ottilie S, Lukens AK, Sakata-Kato T, Vanaerschot M, Owen E, Jado JC, Maher SP, Calla J, Plouffe D, Zhong Y, Chen K, Chaumeau V, Conway AJ, McNamara CW, Ibanez M, Gagaring K, Serrano FN, Eribez K, Taggard CM, Cheung AL, Lincoln C, Ambachew B, Rouillier M, Siegel D, Nosten F, Kyle DE, Gamo FJ, Zhou Y, Llinás M, Fidock DA, Wirth DF, Burrows J, Campo B, Winzeler EA. Open-source discovery of chemical leads for next-generation chemoprotective antimalarials. Science 2019; 362:362/6419/eaat9446. [PMID: 30523084 PMCID: PMC6516198 DOI: 10.1126/science.aat9446] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/18/2018] [Indexed: 11/19/2022]
Abstract
To discover leads for next-generation chemoprotective antimalarial drugs,we tested more than 500,000 compounds for their ability to inhibit liver-stage development of luciferase-expressing Plasmodium spp. parasites (681 compounds showed a half-maximal inhibitory concentration of less than 1micromolar).Cluster analysis identified potent and previously unreported scaffold families as well as other series previously associated with chemoprophylaxis. Further testing through multiple phenotypic assays that predict stage-specific and multispecies antimalarial activity distinguished compound classes that are likely to provide symptomatic relief by reducing asexual blood-stage parasitemia from those which are likely to only prevent malaria. Target identification by using functional assays, in vitro evolution, or metabolic profiling revealed 58 mitochondrial inhibitors but also many chemotypes possibly with previously unidentified mechanisms of action.
Collapse
Affiliation(s)
- Yevgeniya Antonova-Koch
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Stephan Meister
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Matthew Abraham
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Madeline R Luth
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Sabine Ottilie
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Amanda K Lukens
- Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA.,The Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | | | - Manu Vanaerschot
- Division of Infectious Diseases, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Edward Owen
- Department of Biochemistry and Molecular Biology and Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA
| | - Juan Carlos Jado
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D. W. Brooks Drive, Athens, GA 30602, USA.,Department of Global Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Jaeson Calla
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - David Plouffe
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Yang Zhong
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Kaisheng Chen
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Victor Chaumeau
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Amy J Conway
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D. W. Brooks Drive, Athens, GA 30602, USA.,Department of Global Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Case W McNamara
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Maureen Ibanez
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Kerstin Gagaring
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Fernando Neria Serrano
- Tres Cantos Medicines Development Campus, Malaria DPU, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Korina Eribez
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Cullin McLean Taggard
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Andrea L Cheung
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Christie Lincoln
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Biniam Ambachew
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA
| | - Melanie Rouillier
- Medicines for Malaria Venture, Post Office Box 1826, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA 92093, USA
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D. W. Brooks Drive, Athens, GA 30602, USA.,Department of Global Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL 33612, USA
| | - Francisco-Javier Gamo
- Medicines for Malaria Venture, Post Office Box 1826, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Yingyao Zhou
- The Genomics Institute of the Novartis Research Foundation, 10675 John J Hopkins Drive, San Diego, CA 92121, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology and Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA.,Department of Chemistry and Center for Infectious Diseases Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - David A Fidock
- Division of Infectious Diseases, Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Dyann F Wirth
- Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA.,The Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Jeremy Burrows
- Medicines for Malaria Venture, Post Office Box 1826, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Brice Campo
- Medicines for Malaria Venture, Post Office Box 1826, 20 Route de Pre-Bois, 1215 Geneva 15, Switzerland
| | - Elizabeth A Winzeler
- School of Medicine, University of California, San Diego, 9500 Gilman Drive 0760, La Jolla, CA 92093, USA. .,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive 0741, La Jolla, CA 92093, USA
| |
Collapse
|
50
|
Phenotypic Screening of Small Molecules with Antimalarial Activity for Three Different Parasitic Life Stages. Methods Mol Biol 2019. [PMID: 29736708 DOI: 10.1007/978-1-4939-7847-2_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Malaria remains one of the deadliest infectious diseases globally. Available therapeutic agents are already limited in their efficacy, and drug resistance threatens to diminish further our ability to prevent and treat the disease. Despite a renewed effort to identify compounds with antimalarial activity, the drug discovery and development pipeline lacks target diversity and availability of compounds that target liver- and gametocyte-stage parasites. Phenotypic screens are a powerful and valuable tool for identifying new chemical compounds with antimalarial activity. This chapter highlights recent phenotypic screening methodologies for all three parasitic life stages.
Collapse
|