1
|
Appetecchia F, Fabbrizi E, Fiorentino F, Consalvi S, Biava M, Poce G, Rotili D. Transmission-Blocking Strategies for Malaria Eradication: Recent Advances in Small-Molecule Drug Development. Pharmaceuticals (Basel) 2024; 17:962. [PMID: 39065810 PMCID: PMC11279868 DOI: 10.3390/ph17070962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Malaria drug research and development efforts have resurged in the last decade following the decelerating rate of mortality and malaria cases in endemic regions. The inefficiency of malaria interventions is largely driven by the spreading resistance of the Plasmodium falciparum parasite to current drug regimens and that of the malaria vector, the Anopheles mosquito, to insecticides. In response to the new eradication agenda, drugs that act by breaking the malaria transmission cycle (transmission-blocking drugs), which has been recognized as an important and additional target for intervention, are being developed. These drugs take advantage of the susceptibility of Plasmodium during population bottlenecks before transmission (gametocytes) and in the mosquito vector (gametes, zygotes, ookinetes, oocysts, sporozoites). To date, compounds targeting stage V gametocytes predominate in the chemical library of transmission-blocking drugs, and some of them have entered clinical trials. The targeting of Plasmodium mosquito stages has recently renewed interest in the development of innovative malaria control tools, which hold promise for the application of compounds effective at these stages. In this review, we highlight the major achievements and provide an update on the research of transmission-blocking drugs, with a particular focus on their chemical scaffolds, antiplasmodial activity, and transmission-blocking potential.
Collapse
Affiliation(s)
| | | | | | | | | | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (E.F.); (F.F.); (S.C.); (M.B.)
| |
Collapse
|
2
|
Legru A, Batista FA, Puszko AK, Bouillon A, Maurel M, Martinez M, Ejjoummany A, Ortega Varga L, Adler P, Méchaly A, Hadjadj M, Sosnowski P, Hopfgartner G, Alzari PM, Blondel A, Haouz A, Barale JC, Hernandez JF. Insights from structure-activity relationships and the binding mode of peptidic α-ketoamide inhibitors of the malaria drug target subtilisin-like SUB1. Eur J Med Chem 2024; 269:116308. [PMID: 38503166 DOI: 10.1016/j.ejmech.2024.116308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/21/2024]
Abstract
Plasmodium multi-resistance, including against artemisinin, seriously threatens malaria treatment and control. Hence, new drugs are urgently needed, ideally targeting different parasitic stages, which are not yet targeted by current drugs. The SUB1 protease is involved in both hepatic and blood stages due to its essential role in the egress of parasites from host cells, and, as potential new target, it would meet the above criteria. We report here the synthesis as well as the biological and structural evaluation of substrate-based α-ketoamide SUB1 pseudopeptidic inhibitors encompassing positions P4-P2'. By individually substituting each position of the reference compound 1 (MAM-117, Ac-Ile-Thr-Ala-AlaCO-Asp-Glu (Oall)-NH2), we better characterized the structural determinants for SUB1 binding. We first identified compound 8 with IC50 values of 50 and 570 nM against Pv- and PfSUB1, respectively (about 3.5-fold higher potency compared to 1). Compound 8 inhibited P. falciparum merozoite egress in culture by 37% at 100 μM. By increasing the overall hydrophobicity of the compounds, we could improve the PfSUB1 inhibition level and antiparasitic activity, as shown with compound 40 (IC50 values of 12 and 10 nM against Pv- and PfSUB1, respectively, IC50 value of 23 μM on P. falciparum merozoite egress). We also found that 8 was highly selective towards SUB1 over three mammalian serine peptidases, supporting the promising value of this compound. Finally, several crystal 3D-structures of SUB1-inhibitor complexes, including with 8, were solved at high resolution to decipher the binding mode of these compounds.
Collapse
Affiliation(s)
- Alice Legru
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Fernando A Batista
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Anna K Puszko
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Anthony Bouillon
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Manon Maurel
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Mariano Martinez
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Abdelaziz Ejjoummany
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Laura Ortega Varga
- Structural Bioinformatic, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Pauline Adler
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Ariel Méchaly
- Cristallography Platform-C2RT, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Margot Hadjadj
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Piotr Sosnowski
- Department of Inorganic and Analytical Chemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Gérard Hopfgartner
- Department of Inorganic and Analytical Chemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Pedro M Alzari
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Arnaud Blondel
- Structural Bioinformatic, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Ahmed Haouz
- Cristallography Platform-C2RT, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Jean-Christophe Barale
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France.
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France.
| |
Collapse
|
3
|
Lisauskaitė M, Nixon GL, Woodley CM, Berry NG, Coninckx A, Qie LC, Leung SC, Taramelli D, Basilico N, Parapini S, Ward SA, Vadas O, Soldati-Favre D, Hong WD, O'Neill PM. Design, synthesis and modelling of photoreactive chemical probes for investigating target engagement of plasmepsin IX and X in Plasmodium falciparum. RSC Chem Biol 2024; 5:19-29. [PMID: 38179191 PMCID: PMC10763550 DOI: 10.1039/d3cb00109a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/27/2023] [Indexed: 01/06/2024] Open
Abstract
The emergence of Plasmodium parasite resistance to current front-line antimalarial treatments poses a serious threat to global malaria control and highlights the necessity for the development of therapeutics with novel targets and mechanisms of action. Plasmepsins IX and X (PMIX/PMX) have been recognised as highly promising targets in Plasmodium due to their contribution to parasite's pathogenicity. Recent research has demonstrated that dual PMIX/PMX inhibition results in the impairment of multiple parasite's life cycle stages, which is an important feature in drug resistance prevention. Herein we report novel hydroxyethylamine photoaffinity labelling (PAL) probes, designed for PMIX/PMX target engagement and proteomics experiments in Plasmodium parasites. The prepared probes have both a photoreactive group (diazirine or benzophenone) for covalent attachment to target proteins, and a terminal alkyne handle allowing their use in bioorthogonal ligation. One of the synthesised benzophenone probes was shown to be highly promising as demonstrated by its outstanding antimalarial potency (IC50 = 15 nM versus D10 P. falciparum) and its inhibitory effect against PfPMX in an enzymatic assay. Molecular docking and molecular dynamics studies show that the inclusion of the benzophenone and alkyne handle does not alter the binding mode compared to the parent compound. The photoaffinity probe can be used in future chemical proteomics studies to allow hydroxyethylamine drug scaffold target identification and validation in Plasmodium. We expect our findings to act as a tool for future investigations on PMIX/PMX inhibition in antimalarial drug discovery.
Collapse
Affiliation(s)
| | - Gemma L Nixon
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | | | - Neil G Berry
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | - Andy Coninckx
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | - L Charlie Qie
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | - Suet C Leung
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFEB), Università degli Studi di Milano 20133 Milano Italy
- Affiliated to Centro Interuniversitario di Ricerche sulla Malaria/Italian Malaria Network (CIRM-IMN), Università degli Studi di Camerino Italy
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano 20133 Milano Italy
- Affiliated to Centro Interuniversitario di Ricerche sulla Malaria/Italian Malaria Network (CIRM-IMN), Università degli Studi di Camerino Italy
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano 20133 Milano Italy
- Affiliated to Centro Interuniversitario di Ricerche sulla Malaria/Italian Malaria Network (CIRM-IMN), Università degli Studi di Camerino Italy
| | - Stephen A Ward
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine Liverpool L3 5QA UK
| | - Oscar Vadas
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, CMU, 1 rue Michel-Servet CH-1211 Genève 4 Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, CMU, 1 rue Michel-Servet CH-1211 Genève 4 Switzerland
| | - W David Hong
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| | - Paul M O'Neill
- Department of Chemistry, University of Liverpool Liverpool L69 7ZD UK
| |
Collapse
|
4
|
Richardson LW, Ashton TD, Dans MG, Nguyen N, Favuzza P, Triglia T, Hodder AN, Ngo A, Jarman KE, Cowman AF, Sleebs BE. Substrate Peptidomimetic Inhibitors of P. falciparum Plasmepsin X with Potent Antimalarial Activity. ChemMedChem 2022; 17:e202200306. [PMID: 35906744 PMCID: PMC9804387 DOI: 10.1002/cmdc.202200306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/29/2022] [Indexed: 01/07/2023]
Abstract
Plasmepsin X (PMX) is an aspartyl protease that processes proteins essential for Plasmodium parasites to invade and egress from host erythrocytes during the symptomatic asexual stage of malaria. PMX substrates possess a conserved cleavage region denoted by the consensus motif, SFhE (h=hydrophobic amino acid). Peptidomimetics reflecting the P3 -P1 positions of the consensus motif were designed and showed potent and selective inhibition of PMX. It was established that PMX prefers Phe in the P1 position, di-substitution at the β-carbon of the P2 moiety and a hydrophobic P3 group which was supported by modelling of the peptidomimetics in complex with PMX. The peptidomimetics were shown to arrest asexual P. falciparum parasites at the schizont stage by impairing PMX substrate processing. Overall, the peptidomimetics described will assist in further understanding PMX substrate specificity and have the potential to act as a template for future antimalarial design.
Collapse
Affiliation(s)
- Lachlan W. Richardson
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Trent D. Ashton
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Madeline G. Dans
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Nghi Nguyen
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Paola Favuzza
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Tony Triglia
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia
| | - Anthony N. Hodder
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Anna Ngo
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia
| | - Kate E. Jarman
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Alan F. Cowman
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| | - Brad E. Sleebs
- Walter and Eliza Hall Institute of Medical ResearchParkville3052VictoriaAustralia,Department of Medical BiologyUniversity of MelbourneParkville3010VictoriaAustralia
| |
Collapse
|
5
|
Kesari P, Deshmukh A, Pahelkar N, Suryawanshi AB, Rathore I, Mishra V, Dupuis JH, Xiao H, Gustchina A, Abendroth J, Labaied M, Yada RY, Wlodawer A, Edwards TE, Lorimer DD, Bhaumik P. Structures of plasmepsin X from Plasmodium falciparum reveal a novel inactivation mechanism of the zymogen and molecular basis for binding of inhibitors in mature enzyme. Protein Sci 2022; 31:882-899. [PMID: 35048450 PMCID: PMC8927862 DOI: 10.1002/pro.4279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 11/06/2022]
Abstract
Plasmodium falciparum plasmepsin X (PfPMX), involved in the invasion and egress of this deadliest malarial parasite, is essential for its survival and hence considered as an important drug target. We report the first crystal structure of PfPMX zymogen containing a novel fold of its prosegment. A unique twisted loop from the prosegment and arginine 244 from the mature enzyme is involved in zymogen inactivation; such mechanism, not previously reported, might be common for apicomplexan proteases similar to PfPMX. The maturation of PfPMX zymogen occurs through cleavage of its prosegment at multiple sites. Our data provide thorough insights into the mode of binding of a substrate and a potent inhibitor 49c to PfPMX. We present molecular details of inactivation, maturation, and inhibition of PfPMX that should aid in the development of potent inhibitors against pepsin-like aspartic proteases from apicomplexan parasites.
Collapse
Affiliation(s)
- Pooja Kesari
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - Anuradha Deshmukh
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - Nikhil Pahelkar
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - Abhishek B. Suryawanshi
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - Ishan Rathore
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - Vandana Mishra
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| | - John H. Dupuis
- Food, Nutrition, and Health Program, Faculty of Land and Food SystemsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Huogen Xiao
- Summerland Research and Development CenterAgriculture and Agri‐Food CanadaSummerlandBritish ColumbiaCanada
| | - Alla Gustchina
- Protein Structure Section, Center for Structural BiologyNational Cancer InstituteFrederickMarylandUSA
| | - Jan Abendroth
- UCB PharmaBainbridge IslandWashingtonUSA
- Seattle Structural Genomics Center for Infectious DiseaseSeattleWashingtonUSA
| | - Mehdi Labaied
- UCB PharmaBainbridge IslandWashingtonUSA
- Seattle Structural Genomics Center for Infectious DiseaseSeattleWashingtonUSA
| | - Rickey Y. Yada
- Food, Nutrition, and Health Program, Faculty of Land and Food SystemsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Alexander Wlodawer
- Protein Structure Section, Center for Structural BiologyNational Cancer InstituteFrederickMarylandUSA
| | - Thomas E. Edwards
- UCB PharmaBainbridge IslandWashingtonUSA
- Seattle Structural Genomics Center for Infectious DiseaseSeattleWashingtonUSA
| | - Donald D. Lorimer
- UCB PharmaBainbridge IslandWashingtonUSA
- Seattle Structural Genomics Center for Infectious DiseaseSeattleWashingtonUSA
| | - Prasenjit Bhaumik
- Department of Biosciences and BioengineeringIndian Institute of Technology BombayMumbaiIndia
| |
Collapse
|
6
|
Bansal M, Upadhyay C, Poonam, Kumar S, Rathi B. Phthalimide analogs for antimalarial drug discovery. RSC Med Chem 2021; 12:1854-1867. [PMID: 34825184 DOI: 10.1039/d1md00244a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/03/2021] [Indexed: 11/21/2022] Open
Abstract
Malaria remains one of the world's most life-threatening diseases and, thus, it is a major public health concern all around the world. The disease can become devastating if not treated with proper medication in a timely manner. Currently, the number of viable treatment therapies is in continuous decline due to compromised effectiveness, probably owing to the complex life cycle of Plasmodium falciparum. The factors responsible for the unclear status of malaria eradication programmes include ever-developing parasite resistance to the most effective treatments used on the frontline (i.e., artemisinin derivatives) and the paucity of new effective therapeutics. Due to these circumstances, the development of novel effective drug candidates with unique modes of action is essential for overcoming the listed obstacles. As such, the discovery of novel chemical compounds based on validated pharmacophores remains an unmet need in the field of medicinal chemistry. In this area, functionalized phthalimide (Pht) analogs have been explored as potential candidates against various diseases, including malaria. Pht presents a promising bioactive scaffold that can be easily functionalized and thus utilized as a starting point for the development of new antimalarial candidates suitable for preclinical and clinical studies. In this short review, we highlight a wide range of Pht analogs that have been investigated for their activity against various strains of Plasmodium falciparum.
Collapse
Affiliation(s)
- Meenakshi Bansal
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi Delhi 110007 India .,Department of Chemistry, Deenbandhu Chhotu Ram University of Science and Technology Murthal Sonepat-131039 Haryana India
| | - Charu Upadhyay
- Department of Chemistry, Miranda House, University of Delhi Delhi 110007 India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi Delhi 110007 India
| | - Sumit Kumar
- Department of Chemistry, Deenbandhu Chhotu Ram University of Science and Technology Murthal Sonepat-131039 Haryana India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi Delhi 110007 India
| |
Collapse
|
7
|
Panda SK, Saxena S, Gupta PSS, Rana MK. Inhibitors of Plasmepsin X Plasmodium falciparum: Structure-based pharmacophore generation and molecular dynamics simulation. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Ding S, Fike KR, Klemba M, Carlier PR. In vitro and in vivo evaluation of the antimalarial MMV665831 and structural analogs. Bioorg Med Chem Lett 2020; 30:127348. [PMID: 32738996 DOI: 10.1016/j.bmcl.2020.127348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
Antimalarial candidates possessing novel mechanisms of action are needed to control drug resistant Plasmodium falciparum. We were drawn to Malaria Box compound 1 (MMV665831) by virtue of its excellent in vitro potency, and twelve analogs were prepared to probe its structure-activity relationship. Modulation of the diethyl amino group was fruitful, producing compound 25, which was twice as potent as 1 against cultured parasites. Efforts were made to modify the phenolic Mannich base functionality of 1, to prevent formation of a reactive quinone methide. Homologated analog 28 had reduced potency relative to 1, but still inhibited growth with EC50 ≤ 200 nM. Thus, the antimalarial activity of 1 does not derive from quinone methide formation. Chemical stability studies on dimethyl analog 2 showed remarkable hydrolytic stability of both the phenolic Mannich base and ethyl ester moieties, and 1 was evaluated for in vivo efficacy in P. berghei-infected mice (40 mg/kg, oral). Unfortunately, no reduction in parasitemia was seen relative to control. These results are discussed in the context of measured plasma and hepatocyte stabilities, with reference to structurally-related, orally-efficacious antimalarials.
Collapse
Affiliation(s)
- Sha Ding
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, United States
| | - Katherine R Fike
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, United States
| | - Michael Klemba
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, United States
| | - Paul R Carlier
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
9
|
Burns AL, Dans MG, Balbin JM, de Koning-Ward TF, Gilson PR, Beeson JG, Boyle MJ, Wilson DW. Targeting malaria parasite invasion of red blood cells as an antimalarial strategy. FEMS Microbiol Rev 2019; 43:223-238. [PMID: 30753425 PMCID: PMC6524681 DOI: 10.1093/femsre/fuz005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Plasmodium spp. parasites that cause malaria disease remain a significant global-health burden. With the spread of parasites resistant to artemisinin combination therapies in Southeast Asia, there is a growing need to develop new antimalarials with novel targets. Invasion of the red blood cell by Plasmodium merozoites is essential for parasite survival and proliferation, thus representing an attractive target for therapeutic development. Red blood cell invasion requires a co-ordinated series of protein/protein interactions, protease cleavage events, intracellular signals, organelle release and engagement of an actin-myosin motor, which provide many potential targets for drug development. As these steps occur in the bloodstream, they are directly susceptible and exposed to drugs. A number of invasion inhibitors against a diverse range of parasite proteins involved in these different processes of invasion have been identified, with several showing potential to be optimised for improved drug-like properties. In this review, we discuss red blood cell invasion as a drug target and highlight a number of approaches for developing antimalarials with invasion inhibitory activity to use in future combination therapies.
Collapse
Affiliation(s)
- Amy L Burns
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005
| | - Madeline G Dans
- Burnet Institute, Melbourne, Victoria, Australia 3004.,Deakin University, School of Medicine, Waurn Ponds, Victoria, Australia 3216
| | - Juan M Balbin
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005
| | | | - Paul R Gilson
- Burnet Institute, Melbourne, Victoria, Australia 3004
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, Australia 3004.,Central Clinical School and Department of Microbiology, Monash University 3004.,Department of Medicine, University of Melbourne, Australia 3052
| | - Michelle J Boyle
- Burnet Institute, Melbourne, Victoria, Australia 3004.,QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia 4006
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia 5005.,Burnet Institute, Melbourne, Victoria, Australia 3004
| |
Collapse
|
10
|
Mishra M, Singh V, Singh S. Structural Insights Into Key Plasmodium Proteases as Therapeutic Drug Targets. Front Microbiol 2019; 10:394. [PMID: 30891019 PMCID: PMC6411711 DOI: 10.3389/fmicb.2019.00394] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
Malaria, caused by protozoan of genus Plasmodium, remains one of the highest mortality infectious diseases. Malaria parasites have a complex life cycle, easily adapt to their host’s immune system and have evolved with an arsenal of unique proteases which play crucial roles in proliferation and survival within the host cells. Owing to the existing knowledge of enzymatic mechanisms, 3D structures and active sites of proteases, they have been proven to be opportune for target based drug development. Here, we discuss in depth the crucial roles of essential proteases in Plasmodium life cycle and particularly focus on highlighting the atypical “structural signatures” of key parasite proteases which have been exploited for drug development. These features, on one hand aid parasites pathogenicity while on the other hand could be effective in designing targeted and very specific inhibitors for counteracting them. We conclude that Plasmodium proteases are suitable as multistage targets for designing novel drugs with new modes of action to combat malaria.
Collapse
Affiliation(s)
- Manasi Mishra
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, India
| | - Vigyasa Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Shailja Singh
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, India.,Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
11
|
Zogota R, Kinena L, Withers-Martinez C, Blackman MJ, Bobrovs R, Pantelejevs T, Kanepe-Lapsa I, Ozola V, Jaudzems K, Suna E, Jirgensons A. Peptidomimetic plasmepsin inhibitors with potent anti-malarial activity and selectivity against cathepsin D. Eur J Med Chem 2018; 163:344-352. [PMID: 30529637 PMCID: PMC6336538 DOI: 10.1016/j.ejmech.2018.11.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 11/18/2022]
Abstract
Following up the open initiative of anti-malarial drug discovery, a GlaxoSmithKline (GSK) phenotypic screening hit was developed to generate hydroxyethylamine based plasmepsin (Plm) inhibitors exhibiting growth inhibition of the malaria parasite Plasmodium falciparum at nanomolar concentrations. Lead optimization studies were performed with the aim of improving Plm inhibition selectivity versus the related human aspartic protease cathepsin D (Cat D). Optimization studies were performed using Plm IV as a readily accessible model protein, the inhibition of which correlates with anti-malarial activity. Guided by sequence alignment of Plms and Cat D, selectivity-inducing structural motifs were modified in the S3 and S4 sub-pocket occupying substituents of the hydroxyethylamine inhibitors. This resulted in potent anti-malarials with an up to 50-fold Plm IV/Cat D selectivity factor. More detailed investigation of the mechanism of action of the selected compounds revealed that they inhibit maturation of the P. falciparum subtilisin-like protease SUB1, and also inhibit parasite egress from erythrocytes. Our results indicate that the anti-malarial activity of the compounds is linked to inhibition of the SUB1 maturase plasmepsin subtype Plm X. Peptidomimimetic plasmepsin inhibitors are developed using Plm IV as a model enzyme. Up to 50-fold selectivity against Cathepsin D is reached. Compounds show growth inhibition of P. falciparum at nanomolar concentrations. Inhibition of SUB1 maturation and parasite egress imply (co)inhibition of Plm X.
Collapse
Affiliation(s)
- Rimants Zogota
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Linda Kinena
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | | | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK; Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Raitis Bobrovs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Vita Ozola
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia
| | - Edgars Suna
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia.
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV, 1006, Latvia.
| |
Collapse
|
12
|
Mathews ES, Odom John AR. Tackling resistance: emerging antimalarials and new parasite targets in the era of elimination. F1000Res 2018; 7. [PMID: 30135714 PMCID: PMC6073090 DOI: 10.12688/f1000research.14874.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2018] [Indexed: 12/27/2022] Open
Abstract
Malaria remains a significant contributor to global human mortality, and roughly half the world’s population is at risk for infection with
Plasmodium spp. parasites. Aggressive control measures have reduced the global prevalence of malaria significantly over the past decade. However, resistance to available antimalarials continues to spread, including resistance to the widely used artemisinin-based combination therapies. Novel antimalarial compounds and therapeutic targets are greatly needed. This review will briefly discuss several promising current antimalarial development projects, including artefenomel, ferroquine, cipargamin, SJ733, KAF156, MMV048, and tafenoquine. In addition, we describe recent large-scale genetic and resistance screens that have been instrumental in target discovery. Finally, we highlight new antimalarial targets, which include essential transporters and proteases. These emerging antimalarial compounds and therapeutic targets have the potential to overcome multi-drug resistance in ongoing efforts toward malaria elimination.
Collapse
Affiliation(s)
- Emily S Mathews
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Audrey R Odom John
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Kinena L, Leitis G, Kanepe-Lapsa I, Bobrovs R, Jaudzems K, Ozola V, Suna E, Jirgensons A. Azole-based non-peptidomimetic plasmepsin inhibitors. Arch Pharm (Weinheim) 2018; 351:e1800151. [DOI: 10.1002/ardp.201800151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/21/2018] [Accepted: 06/29/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Linda Kinena
- Latvian Institute of Organic Synthesis; Riga Latvia
| | | | | | | | | | - Vita Ozola
- Latvian Institute of Organic Synthesis; Riga Latvia
| | - Edgars Suna
- Latvian Institute of Organic Synthesis; Riga Latvia
| | | |
Collapse
|
14
|
Kumar Singh A, Rajendran V, Singh S, Kumar P, Kumar Y, Singh A, Miller W, Potemkin V, Poonam, Grishina M, Gupta N, Kempaiah P, Durvasula R, Singh BK, Dunn BM, Rathi B. Antiplasmodial activity of hydroxyethylamine analogs: Synthesis, biological activity and structure activity relationship of plasmepsin inhibitors. Bioorg Med Chem 2018; 26:3837-3844. [PMID: 29983285 DOI: 10.1016/j.bmc.2018.06.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 01/08/2023]
Abstract
Malaria, particularly in endemic countries remains a threat to the human health and is the leading the cause of mortality in the tropical and sub-tropical areas. Herein, we explored new C2 symmetric hydroxyethylamine analogs as the potential inhibitors of Plasmodium falciparum (P. falciparum; 3D7) in in-vitro cultures. All the listed compounds were also evaluated against crucial drug targets, plasmepsin II (Plm II) and IV (Plm IV), enzymes found in the digestive vacuole of the P. falciparum. Analog 10f showed inhibitory activities against both the enzymes Plm II and Plm IV (Ki, 1.93 ± 0.29 µM for Plm II; Ki, 1.99 ± 0.05 µM for Plm IV). Among all these analogs, compounds 10g selectively inhibited the activity of Plm IV (Ki, 0.84 ± 0.08 µM). In the in vitro screening assay, the growth inhibition of P. falciparum by both the analogs (IC50, 2.27 ± 0.95 µM for 10f; IC50, 3.11 ± 0.65 µM for 10g) displayed marked killing effect. A significant growth inhibition of the P. falciparum was displayed by analog 12c with IC50 value of 1.35 ± 0.85 µM, however, it did not show inhibitory activity against either Plms. The hemolytic assay suggested that the active compounds selectively inhibit the growth of the parasite. Further, potent analogs (10f and 12c) were evaluated for their cytotoxicity towards mammalian HepG2 and vero cells. The selectivity index (SI) values were noticed greater than 10 for both the analogs that suggested their poor toxicity. The present study indicates these analogs as putative lead structures and could serve as crucial for the development of new drug molecules.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India; Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Prashant Kumar
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Yogesh Kumar
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Archana Singh
- Department of Botany, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
| | - Whelton Miller
- Department of Chemistry & Physics, Lincoln University, Lincoln University, PA 19352, USA; Department of Chemical & Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Potemkin
- South Ural State University, Laboratory of Computational Modeling of Drugs, 454080, Russia
| | - Poonam
- South Ural State University, Laboratory of Computational Modeling of Drugs, 454080, Russia; Department of Chemistry, Miranda House University Enclave, University of Delhi, Delhi 110007 India
| | - Maria Grishina
- South Ural State University, Laboratory of Computational Modeling of Drugs, 454080, Russia
| | - Nikesh Gupta
- Special Centre for Nanosciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Prakasha Kempaiah
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Ravi Durvasula
- Department of Medicine, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | | | - Ben M Dunn
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, FL, USA
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India; South Ural State University, Laboratory of Computational Modeling of Drugs, 454080, Russia.
| |
Collapse
|
15
|
Pino P, Caldelari R, Mukherjee B, Vahokoski J, Klages N, Maco B, Collins CR, Blackman MJ, Kursula I, Heussler V, Brochet M, Soldati-Favre D. A multistage antimalarial targets the plasmepsins IX and X essential for invasion and egress. Science 2018; 358:522-528. [PMID: 29074775 DOI: 10.1126/science.aaf8675] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Regulated exocytosis by secretory organelles is important for malaria parasite invasion and egress. Many parasite effector proteins, including perforins, adhesins, and proteases, are extensively proteolytically processed both pre- and postexocytosis. Here we report the multistage antiplasmodial activity of the aspartic protease inhibitor hydroxyl-ethyl-amine-based scaffold compound 49c. This scaffold inhibits the preexocytosis processing of several secreted rhoptry and microneme proteins by targeting the corresponding maturases plasmepsins IX (PMIX) and X (PMX), respectively. Conditional excision of PMIX revealed its crucial role in invasion, and recombinantly active PMIX and PMX cleave egress and invasion factors in a 49c-sensitive manner.
Collapse
Affiliation(s)
- Paco Pino
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Budhaditya Mukherjee
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Natacha Klages
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Christine R Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK.,Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| |
Collapse
|
16
|
Rasina D, Stakanovs G, Borysov OV, Pantelejevs T, Bobrovs R, Kanepe-Lapsa I, Tars K, Jaudzems K, Jirgensons A. 2-Aminoquinazolin-4(3H)-one based plasmepsin inhibitors with improved hydrophilicity and selectivity. Bioorg Med Chem 2018; 26:2488-2500. [PMID: 29636223 DOI: 10.1016/j.bmc.2018.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/29/2018] [Accepted: 04/03/2018] [Indexed: 11/15/2022]
Abstract
2-Aminoquinazolin-4(3H)-ones were previously discovered as perspective leads for antimalarial drug development targeting the plasmepsins. Here we report the lead optimization studies with the aim to reduce inhibitor lipophilicity and increase selectivity versus the human aspartic protease Cathepsin D. Exploiting the solvent exposed area of the enzyme provides an option to install polar groups (R1) the 5-position of 2-aminoquinazolin-4(3H)-one to inhibitors such as carboxylic acid without scarifying enzymatic potency. Moreover, introduction of R1 substituents increased selectivity factors of compounds in this series up to 100-fold for Plm II, IV vs CatD inhibition. The introduction of flap pocket substituent (R2) at 7-postion of 2-aminoquinazolin-4(3H)-one allows to remove Ph group from THF ring without notably impairing Plm inhibitory potency. Based on these findings, inhibitors were developed, which show Plm II and IV inhibitory potency in low nanomolar range and remarkable selectivity against Cathepsin D along with decreased lipophilicity and increased solubility.
Collapse
Affiliation(s)
- Dace Rasina
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Georgijs Stakanovs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Oleksandr V Borysov
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Raitis Bobrovs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Kaspars Tars
- Biomedical Research and Study Centre, Ratsupites 1, Riga LV-1067, Latvia
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia.
| |
Collapse
|
17
|
Mukherjee B, Tessaro F, Vahokoski J, Kursula I, Marq JB, Scapozza L, Soldati-Favre D. Modeling and resistant alleles explain the selectivity of antimalarial compound 49c towards apicomplexan aspartyl proteases. EMBO J 2018. [PMID: 29519896 DOI: 10.15252/embj.201798047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Toxoplasma gondii aspartyl protease 3 (TgASP3) phylogenetically clusters with Plasmodium falciparum Plasmepsins IX and X (PfPMIX, PfPMX). These proteases are essential for parasite survival, acting as key maturases for secreted proteins implicated in invasion and egress. A potent antimalarial peptidomimetic inhibitor (49c) originally developed against Plasmepsin II selectively targets TgASP3, PfPMIX, and PfPMX To unravel the molecular basis for the selectivity of 49c, we constructed homology models of PfPMIX, PfPMX, and TgASP3 that were first validated by identifying the determinants of microneme and rhoptry substrate recognition. The flap and flap-like structures of several reported Plasmepsins are highly flexible and critically modulate the access to the binding cavity. Molecular docking of 49c to TgASP3, PfPMIX, and PfPMX models predicted that the conserved phenylalanine residues in the flap, F344, F291, and F305, respectively, account for the sensitivity toward 49c. Concordantly, phenylalanine mutations in the flap of the three proteases increase twofold to 15-fold the IC50 values of 49c. Compellingly the selection of mutagenized T. gondii resistant strains to 49c reproducibly converted F344 to a cysteine residue.
Collapse
Affiliation(s)
- Budhaditya Mukherjee
- Department of Microbiology and Molecular Medicine, University of Geneva CMU, Geneva 4, Switzerland
| | - Francesca Tessaro
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Lausanne University of Geneva CMU, Geneva, Switzerland
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jean-Baptiste Marq
- Department of Microbiology and Molecular Medicine, University of Geneva CMU, Geneva 4, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Lausanne University of Geneva CMU, Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva CMU, Geneva 4, Switzerland
| |
Collapse
|
18
|
Paul AS, Duraisingh MT. Targeting Plasmodium Proteases to Block Malaria Parasite Escape and Entry. Trends Parasitol 2017; 34:95-97. [PMID: 29269028 DOI: 10.1016/j.pt.2017.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 10/18/2022]
Abstract
Proliferation of malaria parasites in a host requires mechanisms to spread between red blood cells (RBCs). We discuss here the implications for biology and antimalarial drug development of companion studies that establish the requirement of two Plasmodium spp. proteases of the plasmepsin family in parasite egress from, and invasion into, RBCs.
Collapse
Affiliation(s)
- Aditya S Paul
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, FXB, Room 202, Boston, Massachusetts 02115, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 651 Huntington Avenue, FXB, Room 202, Boston, Massachusetts 02115, USA.
| |
Collapse
|
19
|
Dogga SK, Mukherjee B, Jacot D, Kockmann T, Molino L, Hammoudi PM, Hartkoorn RC, Hehl AB, Soldati-Favre D. A druggable secretory protein maturase of Toxoplasma essential for invasion and egress. eLife 2017; 6. [PMID: 28898199 PMCID: PMC5595437 DOI: 10.7554/elife.27480] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/08/2017] [Indexed: 12/19/2022] Open
Abstract
Micronemes and rhoptries are specialized secretory organelles that deploy their contents at the apical tip of apicomplexan parasites in a regulated manner. The secretory proteins participate in motility, invasion, and egress and are subjected to proteolytic maturation prior to organellar storage and discharge. Here we establish that Toxoplasma gondii aspartyl protease 3 (ASP3) resides in the endosomal-like compartment and is crucially associated to rhoptry discharge during invasion and to host cell plasma membrane lysis during egress. A comparison of the N-terminome, by terminal amine isotopic labelling of substrates between wild type and ASP3 depleted parasites identified microneme and rhoptry proteins as repertoire of ASP3 substrates. The role of ASP3 as a maturase for previously described and newly identified secretory proteins is confirmed in vivo and in vitro. An antimalarial compound based on a hydroxyethylamine scaffold interrupts the lytic cycle of T. gondii at submicromolar concentration by targeting ASP3.
Collapse
Affiliation(s)
- Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Budhaditya Mukherjee
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Damien Jacot
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Luca Molino
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Ruben C Hartkoorn
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland.,Chemical Biology of Antibiotics, Center for Infection and Immunity, Inserm U1019, CNRS UMR8204, Institut Pasteur de Lille, Lille, France
| | - Adrian B Hehl
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
20
|
In Silico Mining for Antimalarial Structure-Activity Knowledge and Discovery of Novel Antimalarial Curcuminoids. Molecules 2016; 21:molecules21070853. [PMID: 27367660 PMCID: PMC6273176 DOI: 10.3390/molecules21070853] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/04/2016] [Accepted: 06/10/2016] [Indexed: 11/17/2022] Open
Abstract
Malaria is a parasitic tropical disease that kills around 600,000 patients every year. The emergence of resistant Plasmodium falciparum parasites to artemisinin-based combination therapies (ACTs) represents a significant public health threat, indicating the urgent need for new effective compounds to reverse ACT resistance and cure the disease. For this, extensive curation and homogenization of experimental anti-Plasmodium screening data from both in-house and ChEMBL sources were conducted. As a result, a coherent strategy was established that allowed compiling coherent training sets that associate compound structures to the respective antimalarial activity measurements. Seventeen of these training sets led to the successful generation of classification models discriminating whether a compound has a significant probability to be active under the specific conditions of the antimalarial test associated with each set. These models were used in consensus prediction of the most likely active from a series of curcuminoids available in-house. Positive predictions together with a few predicted as inactive were then submitted to experimental in vitro antimalarial testing. A large majority from predicted compounds showed antimalarial activity, but not those predicted as inactive, thus experimentally validating the in silico screening approach. The herein proposed consensus machine learning approach showed its potential to reduce the cost and duration of antimalarial drug discovery.
Collapse
|
21
|
Rasina D, Otikovs M, Leitans J, Recacha R, Borysov OV, Kanepe-Lapsa I, Domraceva I, Pantelejevs T, Tars K, Blackman MJ, Jaudzems K, Jirgensons A. Fragment-Based Discovery of 2-Aminoquinazolin-4(3H)-ones As Novel Class Nonpeptidomimetic Inhibitors of the Plasmepsins I, II, and IV. J Med Chem 2015; 59:374-87. [PMID: 26670264 DOI: 10.1021/acs.jmedchem.5b01558] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
2-Aminoquinazolin-4(3H)-ones were identified as a novel class of malaria digestive vacuole plasmepsin inhibitors by using NMR-based fragment screening against Plm II. Initial fragment hit optimization led to a submicromolar inhibitor, which was cocrystallized with Plm II to produce an X-ray structure of the complex. The structure showed that 2-aminoquinazolin-4(3H)-ones bind to the open flap conformation of the enzyme and provided clues to target the flap pocket. Further improvement in potency was achieved via introduction of hydrophobic substituents occupying the flap pocket. Most of the 2-aminoquinazolin-4(3H)-one based inhibitors show a similar activity against digestive Plms I, II, and IV and >10-fold selectivity versus CatD, although varying the flap pocket substituent led to one Plm IV selective inhibitor. In cell-based assays, the compounds show growth inhibition of Plasmodium falciparum 3D7 with IC50 ∼ 1 μM. Together, these results suggest 2-aminoquinazolin-4(3H)-ones as perspective leads for future development of an antimalarial agent.
Collapse
Affiliation(s)
- Dace Rasina
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Martins Otikovs
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Janis Leitans
- Biomedical Research and Study Centre , Ratsupites 1, Riga LV-1067, Latvia
| | - Rosario Recacha
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Oleksandr V Borysov
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Ilona Domraceva
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Teodors Pantelejevs
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Kaspars Tars
- Biomedical Research and Study Centre , Ratsupites 1, Riga LV-1067, Latvia
| | - Michael J Blackman
- The Francis Crick Institute, Mill Hill Laboratory , The Ridgeway, Mill Hill, London NW7 1AA, U.K
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis , Aizkraukles 21, Riga LV-1006, Latvia
| |
Collapse
|
22
|
Recacha R, Leitans J, Akopjana I, Aprupe L, Trapencieris P, Jaudzems K, Jirgensons A, Tars K. Structures of plasmepsin II from Plasmodium falciparum in complex with two hydroxyethylamine-based inhibitors. Acta Crystallogr F Struct Biol Commun 2015; 71:1531-9. [PMID: 26625296 PMCID: PMC4666482 DOI: 10.1107/s2053230x15022049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/18/2015] [Indexed: 11/10/2022] Open
Abstract
Plasmepsin II (PMII) is one of the ten plasmepsins (PMs) identified in the genome of Plasmodium falciparum, the causative agent of the most severe and deadliest form of malaria. Owing to the emergence of P. falciparum strains that are resistant to current antimalarial agents such as chloroquine and sulfadoxine/pyrimethamine, there is a constant pressure to find new and lasting chemotherapeutic drug therapies. Previously, the crystal structure of PMII in complex with NU655, a potent antimalarial hydroxyethylamine-based inhibitor, and the design of new compounds based on it have been reported. In the current study, two of these newly designed hydroxyethylamine-based inhibitors, PG418 and PG394, were cocrystallized with PMII and their structures were solved, analyzed and compared with that of the PMII-NU655 complex. Structural analysis of the PMII-PG418 complex revealed that the flap loop can adopt a fully closed conformation, stabilized by interactions with the inhibitor, and a fully open conformation, causing an overall expansion in the active-site cavity, which in turn causes unstable binding of the inhibitor. PG418 also stabilizes the flexible loop Gln275-Met286 of another monomer in the asymmetric unit of PMII, which is disordered in the PMII-NU655 complex structure. The crystal structure of PMII in complex with the inhibitor PG418 demonstrates the conformational flexibility of the active-site cavity of the plasmepsins. The interactions of the different moieties in the P1' position of PG418 and PG394 with Thr217 have to be taken into account in the design of new potent plasmepsin inhibitors.
Collapse
Affiliation(s)
- Rosario Recacha
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia
| | - Janis Leitans
- Biomedical Research and Study Centre, Ratsupites 1, Riga, LV-1067, Latvia
| | - Inara Akopjana
- Biomedical Research and Study Centre, Ratsupites 1, Riga, LV-1067, Latvia
| | - Lilija Aprupe
- Biomedical Research and Study Centre, Ratsupites 1, Riga, LV-1067, Latvia
| | | | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia
| | - Kaspars Tars
- Biomedical Research and Study Centre, Ratsupites 1, Riga, LV-1067, Latvia
| |
Collapse
|
23
|
Singh AK, Rathore S, Tang Y, Goldfarb NE, Dunn BM, Rajendran V, Ghosh PC, Singh N, Latha N, Singh BK, Rawat M, Rathi B. Hydroxyethylamine Based Phthalimides as New Class of Plasmepsin Hits: Design, Synthesis and Antimalarial Evaluation. PLoS One 2015; 10:e0139347. [PMID: 26502278 PMCID: PMC4621027 DOI: 10.1371/journal.pone.0139347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 09/12/2015] [Indexed: 11/18/2022] Open
Abstract
A novel class of phthalimides functionalized with privileged scaffolds was designed, synthesized and evaluated as potential inhibitors of plasmepsin 2 (Ki: 0.99 ± 0.1 μM for 6u) and plasmepsin 4 (Ki: 3.3 ± 0.3 μM for 6t), enzymes found in the digestive vacuole of the plasmodium parasite and considered as crucial drug targets. Three compounds were identified as potential candidates for further development. The listed compounds were also assayed for their antimalarial efficacy against chloroquine (CQ) sensitive strain (3D7) of Plasmodium falciparum. Assay of twenty seven hydroxyethylamine derivatives revealed four (5e, 6j, 6o and 6s) as strongly active, which were further evaluated against CQ resistant strain (7GB) of P. falciparum. Compound 5e possessing the piperidinopiperidine moiety exhibited promising antimalarial activity with an IC50 of 1.16 ± 0.04 μM. Further, compounds 5e, 6j, 6o and 6s exhibited low cytotoxic effect on MCF-7 cell line. Compound 6s possessing C2 symmetry was identified as the least cytotoxic with significant antimalarial activity (IC50: 1.30 ± 0.03 μM). The combined presence of hydroxyethylamine and cyclic amines (piperazines and piperidines) was observed as crucial for the activity. The current studies suggest that hydroxyethylamine based molecules act as potent antimalarial agent and may be helpful in drug development.
Collapse
Affiliation(s)
- Anil K. Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Delhi, Delhi, India
| | - Sumit Rathore
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Yan Tang
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, FL, United States of America
| | - Nathan E. Goldfarb
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, FL, United States of America
| | - Ben M. Dunn
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, FL, United States of America
| | - Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, Dhaula Kuan, New Delhi, India
| | - Prahlad C. Ghosh
- Department of Biochemistry, University of Delhi South Campus, Dhaula Kuan, New Delhi, India
| | - Neelu Singh
- Bioinformatics Center, Sri Venkateswara College, University of Delhi South Campus, Dhaula Kuan, New Delhi, India
| | - N. Latha
- Bioinformatics Center, Sri Venkateswara College, University of Delhi South Campus, Dhaula Kuan, New Delhi, India
| | - Brajendra K. Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Delhi, Delhi, India
| | - Manmeet Rawat
- Department of Internal Medicine, 1 University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Brijesh Rathi
- Bioorganic Research Laboratory, Department of Chemistry, University of Delhi, Delhi, India
- * E-mail:
| |
Collapse
|
24
|
Abstract
Despite substantial scientific progress over the past two decades, malaria remains a worldwide burden that causes hundreds of thousands of deaths every year. New, affordable and safe drugs are required to overcome increasing resistance against artemisinin-based treatments, treat vulnerable populations, interrupt the parasite life cycle by blocking transmission to the vectors, prevent infection and target malaria species that transiently remain dormant in the liver. In this Review, we discuss how the antimalarial drug discovery pipeline has changed over the past 10 years, grouped by the various target compound or product profiles, to assess progress and gaps, and to recommend priorities.
Collapse
|
25
|
Cioc RC, van der Niet DJH, Janssen E, Ruijter E, Orru RVA. One-Pot Synthesis of N-Substituted β-Amino Alcohols from Aldehydes and Isocyanides. Chemistry 2015; 21:7808-13. [PMID: 25868115 DOI: 10.1002/chem.201500210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Indexed: 12/12/2022]
Abstract
A practical two-stage one-pot synthesis of N-substituted β-amino alcohols using aldehydes and isocyanides as starting materials has been developed. This method features mild reaction conditions, broad scope, and general tolerance of functional groups. Based on a less common central carbon-carbon bond disconnection, this protocol complements traditional approaches that involve amines and various carbon electrophiles (epoxides, α-halo ketones, β-halohydrins). Medicinally relevant products can be prepared in a concise and efficient way from simple building blocks, as demonstrated in the synthesis of the antiasthma drug salbutamol. Upgrading the synthesis to an enantioselective variant is also feasible.
Collapse
Affiliation(s)
- Răzvan C Cioc
- Department of Chemistry & Pharmaceutical Sciences and Amsterdam Institute for Molecules Medicines and Systems (AIMMS), VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam (The Netherlands)
| | | | | | | | | |
Collapse
|
26
|
Kumar S, Kumari R, Pandey R. New insight-guided approaches to detect, cure, prevent and eliminate malaria. PROTOPLASMA 2015; 252:717-753. [PMID: 25323622 DOI: 10.1007/s00709-014-0697-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/01/2014] [Indexed: 06/04/2023]
Abstract
New challenges posed by the development of resistance against artemisinin-based combination therapies (ACTs) as well as previous first-line therapies, and the continuing absence of vaccine, have given impetus to research in all areas of malaria control. This review portrays the ongoing progress in several directions of malaria research. The variants of RTS,S and apical membrane antigen 1 (AMA1) are being developed and test adapted as multicomponent and multistage malaria control vaccines, while many other vaccine candidates and methodologies to produce antigens are under experimentation. To track and prevent the spread of artemisinin resistance from Southeast Asia to other parts of the world, rolling circle-enhanced enzyme activity detection (REEAD), a time- and cost-effective malaria diagnosis in field conditions, and a DNA marker associated with artemisinin resistance have become available. Novel mosquito repellents and mosquito trapping and killing techniques much more effective than the prevalent ones are undergoing field testing. Mosquito lines stably infected with their symbiotic wild-type or genetically engineered bacteria that kill sympatric malaria parasites are being constructed and field tested for stopping malaria transmission. A complementary approach being pursued is the addition of ivermectin-like drug molecules to ACTs to cure malaria and kill mosquitoes. Experiments are in progress to eradicate malaria mosquito by making it genetically male sterile. High-throughput screening procedures are being developed and used to discover molecules that possess long in vivo half life and are active against liver and blood stages for the fast cure of malaria symptoms caused by simple or relapsing and drug-sensitive and drug-resistant types of varied malaria parasites, can stop gametocytogenesis and sporogony and could be given in one dose. Target-based antimalarial drug designing has begun. Some of the putative next-generation antimalarials that possess in their scaffold structure several of the desired properties of malaria cure and control are exemplified by OZ439, NITD609, ELQ300 and tafenoquine that are already undergoing clinical trials, and decoquinate, usnic acid, torin-2, ferroquine, WEHI-916, MMV396749 and benzothiophene-type N-myristoyltransferase (NMT) inhibitors, which are candidates for future clinical usage. Among these, NITD609, ELQ300, decoquinate, usnic acid, torin-2 and NMT inhibitors not only cure simple malaria and are prophylactic against simple malaria, but they also cure relapsing malaria.
Collapse
Affiliation(s)
- Sushil Kumar
- SKA Institution for Research, Education and Development (SKAIRED), 4/11 SarvPriya Vihar, New Delhi, 110016, India,
| | | | | |
Collapse
|
27
|
Meyers MJ, Anderson EJ, McNitt SA, Krenning TM, Singh M, Xu J, Zeng W, Qin L, Xu W, Zhao S, Qin L, Eickhoff CS, Oliva J, Campbell MA, Arnett SD, Prinsen MJ, Griggs DW, Ruminski PG, Goldberg DE, Ding K, Liu X, Tu Z, Tortorella MD, Sverdrup FM, Chen X. Evaluation of spiropiperidine hydantoins as a novel class of antimalarial agents. Bioorg Med Chem 2015; 23:5144-50. [PMID: 25797165 DOI: 10.1016/j.bmc.2015.02.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 02/07/2023]
Abstract
Given the rise of parasite resistance to all currently used antimalarial drugs, the identification of novel chemotypes with unique mechanisms of action is of paramount importance. Since Plasmodium expresses a number of aspartic proteases necessary for its survival, we have mined antimalarial datasets for drug-like aspartic protease inhibitors. This effort led to the identification of spiropiperidine hydantoins, bearing similarity to known inhibitors of the human aspartic protease β-secretase (BACE), as new leads for antimalarial drug discovery. Spiropiperidine hydantoins have a dynamic structure-activity relationship profile with positions identified as being tolerant of a variety of substitution patterns as well as a key piperidine N-benzyl phenol pharmacophore. Lead compounds 4e (CWHM-123) and 12k (CWHM-505) are potent antimalarials with IC50 values against Plasmodium falciparum 3D7 of 0.310 μM and 0.099 μM, respectively, and the former features equivalent potency on the chloroquine-resistant Dd2 strain. Remarkably, these compounds do not inhibit human aspartic proteases BACE, cathepsins D and E, or Plasmodium plasmepsins II and IV despite their similarity to known BACE inhibitors. Although the current leads suffer from poor metabolic stability, they do fit into a drug-like chemical property space and provide a new class of potent antimalarial agents for further study.
Collapse
Affiliation(s)
- Marvin J Meyers
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA.
| | - Elizabeth J Anderson
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Sarah A McNitt
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Thomas M Krenning
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Megh Singh
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Jing Xu
- Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wentian Zeng
- Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Limei Qin
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190, Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Wanwan Xu
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190, Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Siting Zhao
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190, Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Li Qin
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190, Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China
| | - Christopher S Eickhoff
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Jonathan Oliva
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Mary A Campbell
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Stacy D Arnett
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Michael J Prinsen
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - David W Griggs
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Peter G Ruminski
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Daniel E Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University in St. Louis, Saint Louis, MO, USA
| | - Ke Ding
- Key Laboratory of Regenerative Biology, Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaorong Liu
- Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhengchao Tu
- Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Micky D Tortorella
- Drug Discovery Pipeline at the Guangzhou Institutes for Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Francis M Sverdrup
- Center for World Health and Medicine, Saint Louis University School of Medicine, 1402 South Grand Blvd, M132 Schwitalla Hall, Saint Louis, MO 63104, USA
| | - Xiaoping Chen
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190, Kaiyuan Avenue, Guangzhou Science Park, Guangzhou 510530, China.
| |
Collapse
|
28
|
Roman G. Mannich bases in medicinal chemistry and drug design. Eur J Med Chem 2015; 89:743-816. [PMID: 25462280 PMCID: PMC7115492 DOI: 10.1016/j.ejmech.2014.10.076] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 01/18/2023]
Abstract
The biological activity of Mannich bases, a structurally heterogeneous class of chemical compounds that are generated from various substrates through the introduction of an aminomethyl function by means of the Mannich reaction, is surveyed, with emphasis on the relationship between structure and biological activity. The review covers extensively the literature reports that have disclosed Mannich bases as anticancer and cytotoxic agents, or compounds with potential antibacterial and antifungal activity in the last decade. The most relevant studies on the activity of Mannich bases as antimycobacterial agents, antimalarials, or antiviral candidates have been included as well. The review contains also a thorough coverage of anticonvulsant, anti-inflammatory, analgesic and antioxidant activities of Mannich bases. In addition, several minor biological activities of Mannich bases, such as their ability to regulate blood pressure or inhibit platelet aggregation, their antiparasitic and anti-ulcer effects, as well as their use as agents for the treatment of mental disorders have been presented. The review gives in the end a brief overview of the potential of Mannich bases as inhibitors of various enzymes or ligands for several receptors.
Collapse
Affiliation(s)
- Gheorghe Roman
- Petru Poni Institute of Macromolecular Chemistry, Department of Inorganic Polymers, 41A Aleea Gr. Ghica Vodă, Iaşi 700487, Romania.
| |
Collapse
|
29
|
Jaudzems K, Tars K, Maurops G, Ivdra N, Otikovs M, Leitans J, Kanepe-Lapsa I, Domraceva I, Mutule I, Trapencieris P, Blackman MJ, Jirgensons A. Plasmepsin inhibitory activity and structure-guided optimization of a potent hydroxyethylamine-based antimalarial hit. ACS Med Chem Lett 2014; 5:373-7. [PMID: 24900843 DOI: 10.1021/ml4004952] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/13/2014] [Indexed: 11/29/2022] Open
Abstract
Antimalarial hit 1 SR (TCMDC-134674) identified in a GlaxoSmithKline cell based screening campaign was evaluated for inhibitory activity against the digestive vacuole plasmepsins (Plm I, II, and IV). It was found to be a potent Plm IV inhibitor with no selectivity over Cathepsin D. A cocrystal structure of 1 SR bound to Plm II was solved, providing structural insight for the design of more potent and selective analogues. Structure-guided optimization led to the identification of structurally simplified analogues 17 and 18 as low nanomolar inhibitors of both, plasmepsin Plm IV activity and P. falciparum growth in erythrocytes.
Collapse
Affiliation(s)
- Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Kaspars Tars
- Biomedical Research and Study Centre, Ratsupites 1, Riga LV-1067, Latvia
| | - Gundars Maurops
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Natalija Ivdra
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Martins Otikovs
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Janis Leitans
- Biomedical Research and Study Centre, Ratsupites 1, Riga LV-1067, Latvia
| | - Iveta Kanepe-Lapsa
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Ilona Domraceva
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Ilze Mutule
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | | | - Michael J. Blackman
- Division
of Parasitology, MRC National Institute for Medical Research, The Ridgeway, Mill, Hill, London NW7 1AA, U.K
| | - Aigars Jirgensons
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| |
Collapse
|
30
|
Meyers MJ, Tortorella MD, Xu J, Qin L, He Z, Lang X, Zeng W, Xu W, Qin L, Prinsen MJ, Sverdrup FM, Eickhoff CS, Griggs DW, Oliva J, Ruminski PG, Jacobsen EJ, Campbell MA, Wood DC, Goldberg DE, Liu X, Lu Y, Lu X, Tu Z, Lu X, Ding K, Chen X. Evaluation of aminohydantoins as a novel class of antimalarial agents. ACS Med Chem Lett 2014; 5:89-93. [PMID: 24900778 DOI: 10.1021/ml400412x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/06/2013] [Indexed: 11/30/2022] Open
Abstract
Given the threat of drug resistance, there is an acute need for new classes of antimalarial agents that act via a unique mechanism of action relative to currently used drugs. We have identified a set of druglike compounds within the Tres Cantos Anti-Malarial Set (TCAMS) which likely act via inhibition of a Plasmodium aspartic protease. Structure-activity relationship analysis and optimization of these aminohydantoins demonstrate that these compounds are potent nanomolar inhibitors of the Plasmodium aspartic proteases PM-II and PM-IV and likely one or more other Plasmodium aspartic proteases. Incorporation of a bulky group, such as a cyclohexyl group, on the aminohydantion N-3 position gives enhanced antimalarial potency while reducing inhibition of human aspartic proteases such as BACE. We have identified compound 8p (CWHM-117) as a promising lead for optimization as an antimalarial drug with a low molecular weight, modest lipophilicity, oral bioavailability, and in vivo antimalarial activity in mice.
Collapse
Affiliation(s)
- Marvin J. Meyers
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Micky D. Tortorella
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Jing Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Limei Qin
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Zhengxiang He
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Xingfen Lang
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Wentian Zeng
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Wanwan Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Li Qin
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Michael J. Prinsen
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Francis M. Sverdrup
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Christopher S. Eickhoff
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - David W. Griggs
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Jonathan Oliva
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Peter G. Ruminski
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - E. Jon Jacobsen
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Mary A. Campbell
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - David C. Wood
- Center
for World Health and Medicine, Saint Louis University, Saint Louis, Missouri 63104, United States
| | - Daniel E. Goldberg
- Howard Hughes Medical Institute, Washington University School of Medicine, Departments of Molecular Microbiology and Medicine,
Saint Louis, Missouri 63110, United States
| | - Xiaorong Liu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Yongzhi Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Xin Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Zhengchao Tu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Xiaoyun Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Ke Ding
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| | - Xiaoping Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese
Academy of Sciences, Guangzhou, China
| |
Collapse
|