1
|
Ouji M, Reyser T, Yamaryo-Botté Y, Nguyen M, Rengel D, Dutreuil A, Marcellin M, Burlet-Schiltz O, Augereau JM, Riscoe MK, Paloque L, Botté C, Benoit-Vical F. In artemisinin-resistant falciparum malaria parasites, mitochondrial metabolic pathways are essential for survival but not those of apicoplast. Int J Parasitol Drugs Drug Resist 2024; 26:100565. [PMID: 39332236 PMCID: PMC11466614 DOI: 10.1016/j.ijpddr.2024.100565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Emergence and spread of parasite resistance to artemisinins, the first-line antimalarial therapy, threaten the malaria eradication policy. To identify therapeutic targets to eliminate artemisinin-resistant parasites, the functioning of the apicoplast and the mitochondrion was studied, focusing on the fatty acid synthesis type II (FASII) pathway in the apicoplast and the electron transfer chain in the mitochondrion. A significant enrichment of the FASII pathway among the up-regulated genes in artemisinin-resistant parasites under dihydroartemisinin treatment was found, in agreement with published transcriptomic data. However, using GC-MS analyzes of fatty acids, we demonstrated for the first time that the FASII pathway is non-functional, ruling out the use of FASII inhibitors to target artemisinin-resistant parasites. Conversely, by assessing the modulation of the oxygen consumption rate, we evidenced that mitochondrial respiration remains functional and flexible in artemisinin-resistant parasites and even at the quiescent stage. Two novel compounds targeting electron transport chain (ELQ300, ELQ400) efficiently killed quiescent artemisinin-resistant parasites. Therefore, mitochondrial respiration represents a key target for the elimination of artemisinin-resistant persistent Plasmodium falciparum parasites.
Collapse
Affiliation(s)
- Manel Ouji
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Thibaud Reyser
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Yoshiki Yamaryo-Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Michel Nguyen
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - David Rengel
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Axelle Dutreuil
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Marlène Marcellin
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France; Infrastructure nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France; Infrastructure nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Jean-Michel Augereau
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Michael K Riscoe
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Lucie Paloque
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Cyrille Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Françoise Benoit-Vical
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
2
|
El Gaaloul M, Tchouatieu AM, Kayentao K, Campo B, Buffet B, Ramachandruni H, Ndiaye JL, Wells TNC, Audibert C, Achan J, Donini C, Barsosio HC, Tinto H. Chemoprevention of malaria with long-acting oral and injectable drugs: an updated target product profile. Malar J 2024; 23:315. [PMID: 39425110 PMCID: PMC11490162 DOI: 10.1186/s12936-024-05128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Malaria is preventable, but the burden of disease remains high with over 249 million cases and 608,000 deaths reported in 2022. Historically, the most important protective interventions have been vector control and chemopreventive medicines with over 50 million children receiving seasonal malaria chemoprevention in the year 2023. Two vaccines are approved and starting to be deployed, bringing additional protection for children up to 36 months. However, the impact of these currently available tools is somewhat limited on various fronts. Vaccines exhibit partial efficacy, are relatively costly, and not accessible in all settings. The challenges encountered with chemoprevention are barriers to acceptability and feasibility, including frequency of dosing, and the lack of options in the first trimester of pregnancy and for women living with HIV. Also, the emergence of resistance against chemopreventive medicines is concerning. To address these limitations, a target product profile (TPP) is proposed as a road map to guide innovation and to boost the quest for novel chemopreventive alternatives. This TPP describes the ideal product attributes, while acknowledging potential trade-offs that may be needed. Critically, it considers the target populations most at risk; primarily infants, children, and pregnant women. Malaria control and elimination requires appropriate chemoprevention, not only in areas of high endemicity and transmission, but also in lower transmission areas where immunity is declining, as well as for travellers from areas where malaria has been eliminated. New medicines should show acceptable safety and tolerability, with high and long protective efficacy. Formulations and costs need to support operational adherence, access, and effectiveness. Next generation long-acting oral and injectable drugs are likely to constitute the backbone of malaria prevention. Therefore, the perspectives of front-line experts in malaria prevention, researchers, and those involved in drug development are captured in the TPP. This inclusive approach aims at concentrating efforts and aligning responses across the community to develop new and transformative medicines.
Collapse
Affiliation(s)
- Myriam El Gaaloul
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland.
| | - Andre Marie Tchouatieu
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland.
| | - Kassoum Kayentao
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Brice Campo
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Benedicte Buffet
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Hanu Ramachandruni
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | | | - Timothy N C Wells
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Celine Audibert
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | | | - Cristina Donini
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Hellen C Barsosio
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| |
Collapse
|
3
|
Pou S, Winter RW, Dodean RA, Liebman K, Li Y, Mather MW, Nepal B, Nilsen A, Handford MJ, Riscoe TM, Laxson S, Kirtley PJ, Aleshnick M, Zakharov LN, Kelly JX, Smilkstein MJ, Wilder BK, Kortagere S, Vaidya AB, Alday PH, Doggett JS, Riscoe MK. 3-Position Biaryl Endochin-like Quinolones with Enhanced Antimalarial Performance. ACS Infect Dis 2024; 10:2419-2442. [PMID: 38862127 PMCID: PMC11245370 DOI: 10.1021/acsinfecdis.4c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 06/13/2024]
Abstract
ELQ-300 is a potent antimalarial drug with activity against blood, liver, and vector stages of the disease. A prodrug, ELQ-331, exhibits reduced crystallinity and improved in vivo efficacy in preclinical testing, and currently, it is in the developmental pipeline for once-a-week dosing for oral prophylaxis against malaria. Because of the high cost of developing a new drug for human use and the high risk of drug failure, it is prudent to have a back-up plan in place. Here we describe ELQ-596, a member of a new subseries of 3-biaryl-ELQs, with enhanced potency in vitro against multidrug-resistant Plasmodium falciparum parasites. ELQ-598, a prodrug of ELQ-596 with diminished crystallinity, is more effective vs murine malaria than its progenitor ELQ-331 by 4- to 10-fold, suggesting that correspondingly lower doses could be used to protect and cure humans of malaria. With a longer bloodstream half-life in mice compared to its progenitor, ELQ-596 highlights a novel series of next-generation ELQs with the potential for once-monthly dosing for protection against malaria infection. Advances in the preparation of 3-biaryl-ELQs are presented along with preliminary results from experiments to explore key structure-activity relationships for drug potency, selectivity, pharmacokinetics, and safety.
Collapse
Affiliation(s)
- Sovitj Pou
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rolf W. Winter
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rozalia A. Dodean
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Katherine Liebman
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Yuexin Li
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Michael W. Mather
- Department
of Microbiology and Immunology, Drexel University
College of Medicine, 2900 Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Binod Nepal
- Department
of Microbiology and Immunology, Drexel University
College of Medicine, 2900 Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Aaron Nilsen
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- Department
of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Mason J. Handford
- Department
of Chemical Physiology and Biochemistry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Teresa M. Riscoe
- Department
of Microbiology and Molecular Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Sydney Laxson
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Payton J. Kirtley
- Vaccine
& Gene Therapy Institute (VGTI), Oregon
Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, Oregon 97006, United States
| | - Maya Aleshnick
- Vaccine
& Gene Therapy Institute (VGTI), Oregon
Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, Oregon 97006, United States
| | - Lev N. Zakharov
- Center
for Advanced Materials Characterization in Oregon (CAMCOR), 1443 E. 13th Avenue, Eugene, Oregon 97403, United States
| | - Jane X. Kelly
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Martin J. Smilkstein
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Brandon K. Wilder
- Vaccine
& Gene Therapy Institute (VGTI), Oregon
Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, Oregon 97006, United States
| | - Sandhya Kortagere
- Department
of Microbiology and Immunology, Drexel University
College of Medicine, 2900 Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Akhil B. Vaidya
- Department
of Microbiology and Immunology, Drexel University
College of Medicine, 2900 Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - P. Holland Alday
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- School of
Medicine Division of Infectious Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - J. Stone Doggett
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- School of
Medicine Division of Infectious Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Michael K. Riscoe
- VA
Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- Department
of Microbiology and Molecular Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| |
Collapse
|
4
|
Hu Y, Yuan M, Julian A, Tuz K, Juárez O. Identification of complex III, NQR, and SDH as primary bioenergetic enzymes during the stationary phase of Pseudomonas aeruginosa cultured in urine-like conditions. Front Microbiol 2024; 15:1347466. [PMID: 38468849 PMCID: PMC10926992 DOI: 10.3389/fmicb.2024.1347466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/13/2024] Open
Abstract
Pseudomonas aeruginosa is a common cause of urinary tract infections by strains that are often multidrug resistant, representing a major challenge to the world's health care system. This microorganism has a highly adaptable metabolism that allows it to colonize many environments, including the urinary tract. In this work, we have characterized the metabolic strategies used by stationary phase P. aeruginosa cells cultivated in urine-like media to understand the adaptations used by this microorganism to survive and produce disease. Our proteomics results show that cells rely on the Entner-Duodoroff pathway, pentose phosphate pathway, the Krebs cycle/ glyoxylate shunt and the aerobic oxidative phosphorylation to survive in urine-like media and other conditions. A deep characterization of the oxidative phosphorylation showed that the respiratory rate of stationary phase cells is increased 3-4 times compared to cells in the logarithmic phase of growth, indicating that the aerobic metabolism plays critical roles in the stationary phase of cells grown in urine like media. Moreover, the data show that respiratory complex III, succinate dehydrogenase and the NADH dehydrogenase NQR have important functions and could be used as targets to develop new antibiotics against this bacterium.
Collapse
Affiliation(s)
| | | | | | | | - Oscar Juárez
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, United States
| |
Collapse
|
5
|
Ranjith P, Ignatious A, Panicker CY, Sureshkumar B, Armakovic S, Armakovic SJ, Van Alsenoy C, Anto P. Spectroscopic investigations, DFT calculations, molecular docking and MD simulations of 3-[(4-Carboxyphenyl) carbamoyl]-4-hydroxy-2-oxo-1, 2-dihydroxy quinoline-6-carboxylic acid. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
6
|
Alday PH, Nilsen A, Doggett JS. Structure-activity relationships of Toxoplasma gondii cytochrome bc1 inhibitors. Expert Opin Drug Discov 2022; 17:997-1011. [PMID: 35772172 PMCID: PMC9561756 DOI: 10.1080/17460441.2022.2096588] [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: 04/18/2022] [Accepted: 06/28/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Toxoplasma gondii is a prolific apicomplexan parasite that infects human and nonhuman animals worldwide and can cause severe brain and eye disease. Safer, more effective therapies for toxoplasmosis are needed. Cytochrome bc1 inhibitors are remarkably effective against toxoplasmosis and other apicomplexan-caused diseases. AREAS COVERED This work reviews T. gondii cytochrome bc1 inhibitors. Emphasis is placed on the structure-activity relationships of these inhibitors with regard to efficacy, pharmacokinetics, selectivity of T. gondii cytochrome bc1 over host, safety, and potential therapeutic strategies. EXPERT OPINION Cytochrome bc1 inhibitors are highly promising compounds for toxoplasmosis that have been effective in clinical and preclinical studies. Clinical experience with atovaquone previously validated cytochrome bc1 as a tractable drug target and, over the past decade, optimization of cytochrome bc1 inhibitors has resulted in improved bioavailability, metabolic stability, potency, blood-brain barrier penetration, and selectivity for the T. gondii cytochrome bc1 over the mammalian bc1. Recent studies have demonstrated preclinical safety, identified novel therapeutic strategies for toxoplasmosis using synergistic combinations or long-acting administration and provided insight into their role in chronic infection. This research has identified drug candidates that are more effective than clinically used drugs in preclinical measures of efficacy.
Collapse
Affiliation(s)
- Phil Holland Alday
- Portland VA Medical Center, Portland, Oregon, USA
- Oregon Health & Science University, Portland, Oregon, USA
| | - Aaron Nilsen
- Portland VA Medical Center, Portland, Oregon, USA
- Oregon Health & Science University, Portland, Oregon, USA
| | | |
Collapse
|
7
|
Amporndanai K, Pinthong N, O’Neill PM, Hong WD, Amewu RK, Pidathala C, Berry NG, Leung SC, Ward SA, Biagini GA, Hasnain SS, Antonyuk SV. Targeting the Ubiquinol-Reduction (Q i) Site of the Mitochondrial Cytochrome bc1 Complex for the Development of Next Generation Quinolone Antimalarials. BIOLOGY 2022; 11:biology11081109. [PMID: 35892964 PMCID: PMC9330653 DOI: 10.3390/biology11081109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022]
Abstract
Antimalarials targeting the ubiquinol-oxidation (Qo) site of the Plasmodium falciparum bc1 complex, such as atovaquone, have become less effective due to the rapid emergence of resistance linked to point mutations in the Qo site. Recent findings showed a series of 2-aryl quinolones mediate inhibitions of this complex by binding to the ubiquinone-reduction (Qi) site, which offers a potential advantage in circumventing drug resistance. Since it is essential to understand how 2-aryl quinolone lead compounds bind within the Qi site, here we describe the co-crystallization and structure elucidation of the bovine cytochrome bc1 complex with three different antimalarial 4(1H)-quinolone sub-types, including two 2-aryl quinolone derivatives and a 3-aryl quinolone analogue for comparison. Currently, no structural information is available for Plasmodial cytochrome bc1. Our crystallographic studies have enabled comparison of an in-silico homology docking model of P. falciparum with the mammalian's equivalent, enabling an examination of how binding compares for the 2- versus 3-aryl analogues. Based on crystallographic and computational modeling, key differences in human and P. falciparum Qi sites have been mapped that provide new insights that can be exploited for the development of next-generation antimalarials with greater selective inhibitory activity against the parasite bc1 with improved antimalarial properties.
Collapse
Affiliation(s)
- Kangsa Amporndanai
- Molecular Biophysics Group, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK; (K.A.); (N.P.); (S.S.H.)
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | - Nattapon Pinthong
- Molecular Biophysics Group, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK; (K.A.); (N.P.); (S.S.H.)
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
- Correspondence: (P.M.O.); (S.V.A.); Tel.: +44-(0)-1517955145 (S.V.A.); +44-(0)-1517943552 (P.M.O.)
| | - W. David Hong
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
| | - Richard K. Amewu
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
- Department of Chemistry, School of Physical and Mathematical Sciences, University of Ghana, Accra P.O. Box LG 586, Ghana
| | - Chandrakala Pidathala
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
- Composite Interceptive Med-Science Laboratories Pvt. Ltd., Bengaluru 60099, Karnataka, India
| | - Neil G. Berry
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
| | - Suet C. Leung
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK; (W.D.H.); (R.K.A.); (C.P.); (N.G.B.); (S.C.L.)
| | - Stephen A. Ward
- Centre for Drugs and Diagnostics, Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (S.A.W.); (G.A.B.)
| | - Giancarlo A. Biagini
- Centre for Drugs and Diagnostics, Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK; (S.A.W.); (G.A.B.)
| | - S. Samar Hasnain
- Molecular Biophysics Group, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK; (K.A.); (N.P.); (S.S.H.)
| | - Svetlana V. Antonyuk
- Molecular Biophysics Group, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK; (K.A.); (N.P.); (S.S.H.)
- Correspondence: (P.M.O.); (S.V.A.); Tel.: +44-(0)-1517955145 (S.V.A.); +44-(0)-1517943552 (P.M.O.)
| |
Collapse
|
8
|
van der Watt ME, Reader J, Birkholtz LM. Adapt or Die: Targeting Unique Transmission-Stage Biology for Malaria Elimination. Front Cell Infect Microbiol 2022; 12:901971. [PMID: 35755845 PMCID: PMC9218253 DOI: 10.3389/fcimb.2022.901971] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Plasmodium parasites have a complex life cycle that includes development in the human host as well as the Anopheles vector. Successful transmission of the parasite between its host and vector therefore requires the parasite to balance its investments in asexual replication and sexual reproduction, varying the frequency of sexual commitment to persist within the human host and generate future opportunities for transmission. The transmission window is extended further by the ability of stage V gametocytes to circulate in peripheral blood for weeks, whereas immature stage I to IV gametocytes sequester in the bone marrow and spleen until final maturation. Due to the low gametocyte numbers in blood circulation and with the ease of targeting such life cycle bottlenecks, transmission represents an efficient target for therapeutic intervention. The biological process of Plasmodium transmission is a multistage, multifaceted process and the past decade has seen a much deeper understanding of the molecular mechanisms and regulators involved. Clearly, specific and divergent processes are used during transmission compared to asexual proliferation, which both poses challenges but also opportunities for discovery of transmission-blocking antimalarials. This review therefore presents an update of our molecular understanding of gametocyte and gamete biology as well as the status of transmission-blocking activities of current antimalarials and lead development compounds. By defining the biological components associated with transmission, considerations for the development of new transmission-blocking drugs to target such untapped but unique biology is suggested as an important, main driver for transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Mariëtte E van der Watt
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Janette Reader
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
9
|
Renard I, Ben Mamoun C. Treatment of Human Babesiosis: Then and Now. Pathogens 2021; 10:pathogens10091120. [PMID: 34578153 PMCID: PMC8469882 DOI: 10.3390/pathogens10091120] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Babesiosis is an emerging tick-borne disease caused by apicomplexan parasites of the genus Babesia. With its increasing incidence worldwide and the risk of human-to-human transmission through blood transfusion, babesiosis is becoming a rising public health concern. The current arsenal for the treatment of human babesiosis is limited and consists of combinations of atovaquone and azithromycin or clindamycin and quinine. These combination therapies were not designed based on biological criteria unique to Babesia parasites, but were rather repurposed based on their well-established efficacy against other apicomplexan parasites. However, these compounds are associated with mild or severe adverse events and a rapid emergence of drug resistance, thus highlighting the need for new therapeutic strategies that are specifically tailored to Babesia parasites. Herein, we review ongoing babesiosis therapeutic and management strategies and their limitations, and further review current efforts to develop new, effective, and safer therapies for the treatment of this disease.
Collapse
|
10
|
Anghel N, Imhof D, Winzer P, Balmer V, Ramseier J, Haenggeli K, Choi R, Hulverson MA, Whitman GR, Arnold SL, Ojo KK, Van Voorhis WC, Doggett JS, Ortega-Mora LM, Hemphill A. Endochin-like quinolones (ELQs) and bumped kinase inhibitors (BKIs): Synergistic and additive effects of combined treatments against Neospora caninum infection in vitro and in vivo. Int J Parasitol Drugs Drug Resist 2021; 17:92-106. [PMID: 34482255 PMCID: PMC8416643 DOI: 10.1016/j.ijpddr.2021.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 12/30/2022]
Abstract
The apicomplexan parasite Neospora caninum is an important causative agent of congenital neosporosis, resulting in abortion, birth of weak offspring and neuromuscular disorders in cattle, sheep, and many other species. Among several compound classes that are currently being developed, two have been reported to limit the effects of congenital neosporosis: (i) bumped kinase inhibitors (BKIs) target calcium dependent protein kinase 1 (CDPK1), an enzyme that is encoded by an apicoplast-derived gene and found only in apicomplexans and plants. CDPK1 is essential for host cell invasion and egress; (ii) endochin-like quinolones (ELQs) are inhibitors of the cytochrome bc1 complex of the mitochondrial electron transport chain and thus inhibit oxidative phosphorylation. We here report on the in vitro and in vivo activities of BKI-1748, and of ELQ-316 and its respective prodrugs ELQ-334 and ELQ-422, applied either as single-compounds or ELQ-BKI-combinations. In vitro, BKI-1748 and ELQ-316, as well as BKI-1748 and ELQ-334, acted synergistically, while this was not observed for the BKI-1748/ELQ-422 combination treatment. In a N. caninum-infected pregnant BALB/c mouse model, the synergistic effects observed in vitro were not entirely reproduced, but 100% postnatal survival and 100% inhibition of vertical transmission was noted in the group treated with the BKI-1748/ELQ-334 combination. In addition, the combined drug applications resulted in lower neonatal mortality compared to treatments with single drugs.
Collapse
Affiliation(s)
- Nicoleta Anghel
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland,Corresponding author. Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland.
| | - Dennis Imhof
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Pablo Winzer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Vreni Balmer
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Jessica Ramseier
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Kai Haenggeli
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Ryan Choi
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew A. Hulverson
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Grant R. Whitman
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Samuel L.M. Arnold
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA,Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Kayode K. Ojo
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA,Departments of Global Health and Microbiology, University of Washington, Seattle, WA, USA
| | - J. Stone Doggett
- VA Portland Health Care System, Research and Development Service, Portland, OR, USA
| | - Luis M. Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, Madrid, Spain
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Switzerland,Corresponding author.
| |
Collapse
|
11
|
Pou S, Dodean RA, Frueh L, Liebman KM, Gallagher RT, Jin H, Jacobs RT, Nilsen A, Stuart DR, Doggett JS, Riscoe MK, Winter RW. A New Scalable Synthesis of ELQ-300, ELQ-316, and other Antiparasitic Quinolones. Org Process Res Dev 2021; 25:1841-1852. [PMID: 35110959 DOI: 10.1021/acs.oprd.1c00099] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Endochin-Like Quinolone (ELQ) compound class may yield effective, safe treatments for a range of important human and animal afflictions. However, to access the public health potential of this compound series, a synthetic route needed to be devised that lowers costs and is amenable to large scale production. In the new synthetic route described here, a substituted β-keto ester, formed by an Ullmann reaction and subsequent acylation, is reacted with an aniline via a Conrad-Limpach reaction to produce 3-substituted 4(1H)-quinolones such as ELQ-300 and ELQ-316. This synthetic route, the first described to be truly amenable to industrial scale production, is relatively short (5 reaction steps), does not require palladium, chromatographic separation or protecting group chemistry, and may be performed without high vacuum distillation.
Collapse
Affiliation(s)
- Sovitj Pou
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rozalia A Dodean
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Lisa Frueh
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Katherine M Liebman
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rory T Gallagher
- Department of Chemistry, Portland State University, 1719 SW 10 Avenue, Portland, Oregon 97201, United States
| | - Haihong Jin
- Medicinal Chemistry Core, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Robert T Jacobs
- Medicines for Malaria Venture, ICC, route de Pré-Bois 20, P.O. Box 1826, 1215 Geneva 15, Switzerland
| | - Aaron Nilsen
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,Medicinal Chemistry Core, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - David R Stuart
- Department of Chemistry, Portland State University, 1719 SW 10 Avenue, Portland, Oregon 97201, United States
| | - J Stone Doggett
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,School of Medicine Division of Infectious Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Michael K Riscoe
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States.,Department of Microbiology and Molecular Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Rolf W Winter
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| |
Collapse
|
12
|
Effective Therapy Targeting Cytochrome bc1 Prevents Babesia Erythrocytic Development and Protects from Lethal Infection. Antimicrob Agents Chemother 2021; 65:e0066221. [PMID: 34152821 PMCID: PMC8370247 DOI: 10.1128/aac.00662-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
An effective strategy to control blood-borne diseases and prevent outbreak recrudescence involves targeting conserved metabolic processes that are essential for pathogen viability. One such target for Plasmodium and Babesia, the infectious agents of malaria and babesiosis, respectively, is the mitochondrial cytochrome bc1 protein complex, which can be inhibited by endochin-like quinolones (ELQ) and atovaquone. We used the tick-transmitted and culturable blood-borne pathogen Babesia duncani to evaluate the structure-activity relationship, safety, efficacy, and mode of action of ELQs. We identified a potent and highly selective ELQ prodrug (ELQ-502), which, alone or in combination with atovaquone, eliminates B. microti and B. duncani infections in vitro and in mouse models of parasitemia and lethal infection. The strong efficacy at low dose, excellent safety, bioavailability, and long half-life of this experimental therapy make it an ideal clinical candidate for the treatment of human infections caused by Babesia and its closely related apicomplexan parasites.
Collapse
|
13
|
Rocamora F, Gupta P, Istvan ES, Luth MR, Carpenter EF, Kümpornsin K, Sasaki E, Calla J, Mittal N, Carolino K, Owen E, Llinás M, Ottilie S, Goldberg DE, Lee MCS, Winzeler EA. PfMFR3: A Multidrug-Resistant Modulator in Plasmodium falciparum. ACS Infect Dis 2021; 7:811-825. [PMID: 33715347 PMCID: PMC8042660 DOI: 10.1021/acsinfecdis.0c00676] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
In
malaria, chemical genetics is a powerful method for assigning
function to uncharacterized genes. MMV085203 and GNF-Pf-3600 are two
structurally related napthoquinone phenotypic screening hits that
kill both blood- and sexual-stage P. falciparum parasites in the low nanomolar to low micromolar range. In order
to understand their mechanism of action, parasites from two different
genetic backgrounds were exposed to sublethal concentrations of MMV085203
and GNF-Pf-3600 until resistance emerged. Whole genome sequencing
revealed all 17 resistant clones acquired nonsynonymous mutations
in the gene encoding the orphan apicomplexan transporter PF3D7_0312500
(pfmfr3) predicted to encode a member of the major
facilitator superfamily (MFS). Disruption of pfmfr3 and testing against a panel of antimalarial compounds showed decreased
sensitivity to MMV085203 and GNF-Pf-3600 as well as other compounds
that have a mitochondrial mechanism of action. In contrast, mutations
in pfmfr3 provided no protection against compounds
that act in the food vacuole or the cytosol. A dihydroorotate dehydrogenase
rescue assay using transgenic parasite lines, however, indicated a
different mechanism of action for both MMV085203 and GNF-Pf-3600 than
the direct inhibition of cytochrome bc1. Green fluorescent protein
(GFP) tagging of PfMFR3 revealed that it localizes to the parasite
mitochondrion. Our data are consistent with PfMFR3 playing roles in
mitochondrial transport as well as drug resistance for clinically
relevant antimalarials that target the mitochondria. Furthermore,
given that pfmfr3 is naturally polymorphic, naturally
occurring mutations may lead to differential sensitivity to clinically
relevant compounds such as atovaquone.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Purva Gupta
- VA San Diego Healthcare System, Medical and Research Sections, La Jolla, California 92161, United States
- Department of Medicine, Division of Pulmonary and Critical Care, University of California, San Diego, La Jolla, California 92037, United States
| | - Eva S. Istvan
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Madeline R. Luth
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | | | | | - Erika Sasaki
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Jaeson Calla
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Nimisha Mittal
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Krypton Carolino
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Edward Owen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | | | - Elizabeth A. Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
14
|
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
|
15
|
Rawat R, Verma SM. High-throughput virtual screening approach involving pharmacophore mapping, ADME filtering, molecular docking and MM-GBSA to identify new dual target inhibitors of PfDHODH and PfCytbc1 complex to combat drug resistant malaria. J Biomol Struct Dyn 2020; 39:5148-5159. [PMID: 32579074 DOI: 10.1080/07391102.2020.1784288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Emerging cases of drug resistance against Artemisinin combination therapies which are the current and the last line of defense against malaria makes the situation very alarming. Due to the liability of single-target drugs to be more prone to drug resistance, the trend of development of dual or multi-target inhibitors is emerging. Recently, a malaria box molecule, MMV007571 which is a well known new permeability pathways inhibitor was investigated to be also multi-targeting Plasmodium falciparum dihydroorotate dehydrogenase and cytochrome bc1 complex. The aspiration behind this study was to use the information of its pharmacophoric features essential for binding as two of its new targets. In this regard, high throughput virtual screening involving pharmacophore mapping, ADME filtering, molecular docking, and MM-GBSA calculations were carried out. This approach has lead to the identification of two new hits namely DT00V1902 and DT00V1922 which binds with -37.85 and -24.65 kcal/mol of more stable ΔG Bind energy at two targets than the lead molecule, MMV007571. The screened compounds are indicated to be carry improvement in binding potential and pharmacokinetic characters as per in silico studies. The authors propose that DT00V1902 and DT00V1922 can be forwarded for experimental validation and clinical studies for antimalarial chemotherapy. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ravi Rawat
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Saurabh M Verma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| |
Collapse
|
16
|
Basto AP, Anghel N, Rubbiani R, Müller J, Stibal D, Giannini F, Süss-Fink G, Balmer V, Gasser G, Furrer J, Hemphill A. Targeting of the mitochondrion by dinuclear thiolato-bridged arene ruthenium complexes in cancer cells and in the apicomplexan parasite Neospora caninum. Metallomics 2020; 11:462-474. [PMID: 30620038 DOI: 10.1039/c8mt00307f] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A library of 18 dinuclear-thiolato bridged arene ruthenium complexes, some of which with demonstrated activity against cancer cells, was screened for activity against a transgenic Neospora caninum strain that constitutively expresses beta-galactosidase. Initial assessments were done at concentrations of 2500, 250, 25 and 2.5 nM, and 5 compounds were further evaluated with regard to their half maximal proliferation-inhibiting concentration (IC50). Among those, [(η6-p-MeC6H4Pri)2Ru2(μ2-SC6H4-p-CH3)3]Cl (1), [(η6-p-MeC6H4Pri)2Ru2(μ2-SC6H4-p-But)3]Cl (2) and [(η6-p-MeC6H4Pri)2Ru2(μ2-SCH2C6H4-p-But)2(μ2-SC6H4-p-OH)]BF4 (9) inhibited N. caninum proliferation with low C50 values of 15, 5 and 1 nM, respectively, while [(η6-p-MeC6H4Pri)2Ru2(μ2-SC6H4-p-OH)3]Cl (3) and [(η6-p-MeC6H4Pri)2Ru2(μ2-SC6H4-p-mco)3]Cl (5, mco = 4-methylcoumarinyl) were less active (IC50 = 280 and 108 nM, respectively). These compounds did not affect human foreskin fibroblast (HFF) host cells at dosages of 5 μM and above, but impaired proliferation of the human ovarian carcinoma cell line A2780 (IC50 values of 130 nM (1), 30 nM (2), 530 nM (3), 7730 nM (5), 130 nM (9)). A2780 cancer cells were treated with complexes 1, 2, and 5, and biodistribution analysis using inductively coupled plasma mass spectrometry (ICP-MS) showed that most of the drugs accumulated in the mitochondrial fractions. Transmission electron microscopy showed that the parasite mitochondrion is the primary target also in N. caninum tachyzoites, but these compounds, when applied at 200 nM for 15 days in vitro, did not act parasiticidal. Complexes 1, 2 and 9 applied orally at 2 and 10 mg kg-1 day-1 during 5 days in a neosporosis mouse model did not reduce parasite load and did not limit parasite dissemination to the central nervous system. In accordance with these results, ICP-MS carried out on different organs of mice orally administrated with complexes 1 and 9, demonstrated that the drugs were readily absorbed, and after 3 and 48 h, were mainly detected in liver and kidney, but were largely absent from the brain. Thus, dinuclear thiolato-bridged arene ruthenium complexes exhibit interesting activities against N. caninum in vitro, but further modifications of these promising molecules are required to improve their bioavailability and pharmacokinetic properties in order to exert a pronounced and selective effect against N. caninum in vivo.
Collapse
Affiliation(s)
- Afonso P Basto
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, CH-3012 Berne, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Novel Endochin-Like Quinolones Exhibit Potent In Vitro Activity against Plasmodium knowlesi but Do Not Synergize with Proguanil. Antimicrob Agents Chemother 2020; 64:AAC.02549-19. [PMID: 32094134 DOI: 10.1128/aac.02549-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
Quinolones, such as the antimalarial atovaquone, are inhibitors of the malarial mitochondrial cytochrome bc 1 complex, a target critical to the survival of both liver- and blood-stage parasites, making these drugs useful as both prophylaxis and treatment. Recently, several derivatives of endochin have been optimized to produce novel quinolones that are active in vitro and in animal models. While these quinolones exhibit potent ex vivo activity against Plasmodium falciparum and Plasmodium vivax, their activity against the zoonotic agent Plasmodium knowlesi is unknown. We screened several of these novel endochin-like quinolones (ELQs) for their activity against P. knowlesi in vitro and compared this with their activity against P. falciparum tested under identical conditions. We demonstrated that ELQs are potent against P. knowlesi (50% effective concentration, <117 nM) and equally effective against P. falciparum We then screened selected quinolones and partner drugs using a longer exposure (2.5 life cycles) and found that proguanil is 10-fold less potent against P. knowlesi than P. falciparum, while the quinolones demonstrate similar potency. Finally, we used isobologram analysis to compare combinations of the ELQs with either proguanil or atovaquone. We show that all quinolone combinations with proguanil are synergistic against P. falciparum However, against P. knowlesi, no evidence of synergy between proguanil and the quinolones was found. Importantly, the combination of the novel quinolone ELQ-300 with atovaquone was synergistic against both species. Our data identify potentially important species differences in proguanil susceptibility and in the interaction of proguanil with quinolones and support the ongoing development of novel quinolones as potent antimalarials that target multiple species.
Collapse
|
18
|
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
|
19
|
Secrieru A, Costa ICC, O’Neill PM, Cristiano MLS. Antimalarial Agents as Therapeutic Tools Against Toxoplasmosis-A Short Bridge between Two Distant Illnesses. Molecules 2020; 25:E1574. [PMID: 32235463 PMCID: PMC7181032 DOI: 10.3390/molecules25071574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 11/16/2022] Open
Abstract
Toxoplasmosis is an infectious disease with paramount impact worldwide, affecting many vulnerable populations and representing a significant matter of concern. Current therapies used against toxoplasmosis are based essentially on old chemotypes, which fail in providing a definitive cure for the disease, placing the most sensitive populations at risk for irreversible damage in vital organs, culminating in death in the most serious cases. Antimalarial drugs have been shown to possess key features for drug repurposing, finding application in the treatment of other parasite-borne illnesses, including toxoplasmosis. Antimalarials provide the most effective therapeutic solutions against toxoplasmosis and make up for the majority of currently available antitoxoplasmic drugs. Additionally, other antiplasmodial drugs have been scrutinized and many promising candidates have emanated in recent developments. Available data demonstrate that it is worthwhile to explore the activity of classical and most recent antimalarial chemotypes, such as quinolines, endoperoxides, pyrazolo[1,5-a]pyrimidines, and nature-derived peptide-based parasiticidal agents, in the context of toxoplasmosis chemotherapy, in the quest for encountering more effective and safer tools for toxoplasmosis control or eradication.
Collapse
Affiliation(s)
- Alina Secrieru
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK;
| | - Inês C. C. Costa
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
| | - Paul M. O’Neill
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK;
| | - Maria L. S. Cristiano
- Center of Marine Sciences, CCMAR, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal; (A.S.); (I.C.C.C.)
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, FCT, Gambelas Campus, University of Algarve, UAlg, 8005-139 Faro, Portugal
| |
Collapse
|
20
|
Dodean RA, Kancharla P, Li Y, Melendez V, Read L, Bane CE, Vesely B, Kreishman-Deitrick M, Black C, Li Q, Sciotti RJ, Olmeda R, Luong TL, Gaona H, Potter B, Sousa J, Marcsisin S, Caridha D, Xie L, Vuong C, Zeng Q, Zhang J, Zhang P, Lin H, Butler K, Roncal N, Gaynor-Ohnstad L, Leed SE, Nolan C, Huezo SJ, Rasmussen SA, Stephens MT, Tan JC, Cooper RA, Smilkstein MJ, Pou S, Winter RW, Riscoe MK, Kelly JX. Discovery and Structural Optimization of Acridones as Broad-Spectrum Antimalarials. J Med Chem 2019; 62:3475-3502. [PMID: 30852885 DOI: 10.1021/acs.jmedchem.8b01961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Malaria remains one of the deadliest diseases in the world today. Novel chemoprophylactic and chemotherapeutic antimalarials are needed to support the renewed eradication agenda. We have discovered a novel antimalarial acridone chemotype with dual-stage activity against both liver-stage and blood-stage malaria. Several lead compounds generated from structural optimization of a large library of novel acridones exhibit efficacy in the following systems: (1) picomolar inhibition of in vitro Plasmodium falciparum blood-stage growth against multidrug-resistant parasites; (2) curative efficacy after oral administration in an erythrocytic Plasmodium yoelii murine malaria model; (3) prevention of in vitro Plasmodium berghei sporozoite-induced development in human hepatocytes; and (4) protection of in vivo P. berghei sporozoite-induced infection in mice. This study offers the first account of liver-stage antimalarial activity in an acridone chemotype. Details of the design, chemistry, structure-activity relationships, safety, metabolic/pharmacokinetic studies, and mechanistic investigation are presented herein.
Collapse
Affiliation(s)
- Rozalia A Dodean
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Papireddy Kancharla
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States
| | - Yuexin Li
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Victor Melendez
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lisa Read
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Charles E Bane
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Brian Vesely
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Mara Kreishman-Deitrick
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Chad Black
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Qigui Li
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Richard J Sciotti
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Raul Olmeda
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Thu-Lan Luong
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Heather Gaona
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Brittney Potter
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Jason Sousa
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Sean Marcsisin
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Diana Caridha
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lisa Xie
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Chau Vuong
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Qiang Zeng
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Jing Zhang
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Ping Zhang
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Hsiuling Lin
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Kirk Butler
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Norma Roncal
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Lacy Gaynor-Ohnstad
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Susan E Leed
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Christina Nolan
- Division of Experimental Therapeutics , Walter Reed Army Institute of Research , Silver Spring , Maryland 20910 , United States
| | - Stephanie J Huezo
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | - Stephanie A Rasmussen
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | | | | | - Roland A Cooper
- Department of Natural Sciences and Mathematics , Dominican University of California , San Rafael , California 94901 , United States
| | - Martin J Smilkstein
- Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Sovitj Pou
- Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Rolf W Winter
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Michael K Riscoe
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| | - Jane X Kelly
- Department of Chemistry , Portland State University , Portland , Oregon 97201 , United States.,Department of Veterans Affairs Medical Center , Portland , Oregon 97239 , United States
| |
Collapse
|
21
|
Singh S, Rajendran V, He J, Singh AK, Achieng AO, Vandana, Pant A, Nasamu AS, Pandit M, Singh J, Quadiri A, Gupta N, Poonam, Ghosh PC, Singh BK, Narayanan L, Kempaiah P, Chandra R, Dunn BM, Pandey KC, Goldberg DE, Singh AP, Rathi B. Fast-Acting Small Molecules Targeting Malarial Aspartyl Proteases, Plasmepsins, Inhibit Malaria Infection at Multiple Life Stages. ACS Infect Dis 2019; 5:184-198. [PMID: 30554511 DOI: 10.1021/acsinfecdis.8b00197] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The eradication of malaria remains challenging due to the complex life cycle of Plasmodium and the rapid emergence of drug-resistant forms of Plasmodium falciparum and Plasmodium vivax. New, effective, and inexpensive antimalarials against multiple life stages of the parasite are urgently needed to combat the spread of malaria. Here, we synthesized a set of novel hydroxyethylamines and investigated their activities in vitro and in vivo. All of the compounds tested had an inhibitory effect on the blood stage of P. falciparum at submicromolar concentrations, with the best showing 50% inhibitory concentrations (IC50) of around 500 nM against drug-resistant P. falciparum parasites. These compounds showed inhibitory actions against plasmepsins, a family of malarial aspartyl proteases, and exhibited a marked killing effect on blood stage Plasmodium. In chloroquine-resistant Plasmodium berghei and P. berghei ANKA infected mouse models, treating mice with both compounds led to a significant decrease in blood parasite load. Importantly, two of the compounds displayed an inhibitory effect on the gametocyte stages (III-V) of P. falciparum in culture and the liver-stage infection of P. berghei both in in vitro and in vivo. Altogether, our findings suggest that fast-acting hydroxyethylamine-phthalimide analogs targeting multiple life stages of the parasite could be a valuable chemical lead for the development of novel antimalarial drugs.
Collapse
Affiliation(s)
- Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Vinoth Rajendran
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | - Jiang He
- Institute for Medical Engineering and Science, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Amit K. Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Angela O. Achieng
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
| | - Vandana
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Akansha Pant
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
| | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Mansi Pandit
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Jyoti Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Afshana Quadiri
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Nikesh Gupta
- Special Centre for Nanosciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi North Campus, Delhi 110007, India
| | - Prahlad C. Ghosh
- Department of Biochemistry, University of Delhi South Campus, New Delhi 110021, India
| | | | - Latha Narayanan
- Bioinformatics Infrastructure Facility, Sri Venkateswara College, University of Delhi South Campus, New Delhi 110021, India
| | - Prakasha Kempaiah
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
- Department of Medicine, Loyola University Stritch School of Medicine, 2160 South First Avenue, Chicago, Illinois 60153, United States
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Ben M. Dunn
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, P.O. Box 100245, Gainesville, Florida 32610, United States
| | - Kailash C. Pandey
- Host−Parasite Interaction Biology Group, National Institute of Malaria Research, Lab. No. 219, Sector-8 Dwarka, New Delhi 110077, India
- Department of Biochemistry, National Institute for Research in Environmental Health, ICMR, Bhopal 462001, India
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Agam P. Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi 110007, India
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
22
|
McConnell EV, Bruzual I, Pou S, Winter R, Dodean RA, Smilkstein MJ, Krollenbrock A, Nilsen A, Zakharov LN, Riscoe MK, Doggett JS. Targeted Structure-Activity Analysis of Endochin-like Quinolones Reveals Potent Qi and Qo Site Inhibitors of Toxoplasma gondii and Plasmodium falciparum Cytochrome bc 1 and Identifies ELQ-400 as a Remarkably Effective Compound against Acute Experimental Toxoplasmosis. ACS Infect Dis 2018; 4:1574-1584. [PMID: 30117728 DOI: 10.1021/acsinfecdis.8b00133] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cytochrome bc1 inhibitors have been broadly studied as human and veterinary medicines and agricultural fungicides. For the most part, cytochrome bc1 inhibitors compete with ubiquinol at the ubiquinol oxidation (Qo) site or with ubiquinone at the quinone reduction (Qi) site. 4(1 H)-Quinolones with 3-position substituents may inhibit either site based on quinolone ring substituents. 4(1 H)-Quinolones that inhibit the Qi site are highly effective against toxoplasmosis, malaria, and babesiosis and do not inhibit human cytochrome bc1. We tested a series of 4(1 H)-Quinolones against wild-type and drug resistant strains of Toxoplasma gondii and Plasmodium falciparum. These experiments identified very potent compounds that inhibit T. gondii proliferation at picomolar concentrations. The most potent compounds target the Qo site, and for these compounds, an alkyl side chain confers potency against T. gondii greater than that of bulkier side chains. Our experiments also show that substituents on the quinolone ring influenced selectivity between T. gondii and P. falciparum and between Qo and Qi site-mediated activity. Comparison of the parasite cytochrome b sequences identified amino acids that are associated with drug resistance in P. falciparum that exist naturally in wild-type T. gondii. These underlying differences may influence drug susceptibility. Finally, a Qo site active 4(1 H)-quinolone-3-diarylether tested in a murine model of toxoplasmosis was superior to atovaquone, resulting in survival from Type I strain T. gondii infection. These experiments identify highly effective compounds for toxoplasmosis and provide valuable insight into the structure-activity relationship of cytochrome bc1 inhibitors.
Collapse
Affiliation(s)
- Erin V. McConnell
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Igor Bruzual
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Sovitj Pou
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rolf Winter
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rozalia A. Dodean
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Martin J. Smilkstein
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Alina Krollenbrock
- Oregon Health & Science University Department of Physiology and Pharmacology, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Aaron Nilsen
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Lev N. Zakharov
- Department of Chemistry, University of Oregon, 1585 E 13th Avenue, Eugene, Oregon 97403, United States
| | - Michael K. Riscoe
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - J. Stone Doggett
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- Division of Infectious Diseases, Oregon Health & Science University Department of Medicine 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| |
Collapse
|
23
|
Poonam, Gupta Y, Gupta N, Singh S, Wu L, Chhikara BS, Rawat M, Rathi B. Multistage inhibitors of the malaria parasite: Emerging hope for chemoprotection and malaria eradication. Med Res Rev 2018; 38:1511-1535. [PMID: 29372568 DOI: 10.1002/med.21486] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/09/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
Over time, several exciting advances have been made in the treatment and prevention of malaria; however, this devastating disease continues to be a major global health problem and affects millions of people every year. Notably, the paucity of new efficient drug molecules and the inevitable drug resistance of the malaria parasite, Plasmodium falciparum, against frontline therapeutics are the foremost struggles facing malaria eradication initiatives. According to the malaria eradication agenda, the discovery of new chemical entities that can destroy the parasite at the liver stage, the asexual blood stage, the gametocyte stage, and the insect ookinete stage of the parasite life cycle (i.e., compounds exhibiting multistage activity) are in high demand, preferably with novel and multiple modes of action. Phenotypic screening of chemical libraries against the malaria parasite is certainly a crucial step toward overcoming these crises. In the last few years, various research groups, including industrial research laboratories, have performed large-scale phenotypic screenings that have identified a wealth of chemical entities active against multiple life stages of the malaria parasite. Vital scientific and technological developments have led to the discovery of multistage inhibitors of the malaria parasite; these compounds, considered highly valuable starting points for subsequent drug discovery and eradication of malaria, are reviewed.
Collapse
Affiliation(s)
- Poonam
- Department of Chemistry, Miranda House, University of Delhi, India
| | - Yash Gupta
- National Institute of Malaria Research (ICMR), New Delhi, India
| | - Nikesh Gupta
- Special Centre for Nanosciences, Jawaharlal Nehru University, New Delhi, India
| | - Snigdha Singh
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, India
| | - Lidong Wu
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Key Laboratory of Control of Quality and Safety for Aquatic Products, Ministry of Agriculture, Chinese Academy of Fishery Sciences, Beijing, China
| | | | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, India
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
24
|
Macedo TS, Villarreal W, Couto CC, Moreira DRM, Navarro M, Machado M, Prudêncio M, Batista AA, Soares MBP. Platinum(ii)-chloroquine complexes are antimalarial agents against blood and liver stages by impairing mitochondrial function. Metallomics 2018; 9:1548-1561. [PMID: 28960224 DOI: 10.1039/c7mt00196g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chloroquine is an antimalarial agent with strong activity against the blood stage of Plasmodium infection, but with low activity against the parasite's liver stage. In addition, the resistance to chloroquine limits its clinical use. The discovery of new molecules possessing multistage activity and overcoming drug resistance is needed. One possible strategy to achieve this lies in combining antimalarial quinolones with the pharmacological effects of transition metals. We investigated the antimalarial activity of four platinum(ii) complexes composed of chloroquine and phosphine ligands, denoted as WV-90, WV-92, WV-93 and WV-94. In comparison with chloroquine, the complexes were less potent against the chloroquine-sensitive 3D7 strain but they were as active as chloroquine in inhibiting the chloroquine-resistant W2 strain of P. falciparum. Regarding selectivity, the complexes WV-90 and WV-93 displayed higher indexes. Unlike chloroquine, the complexes act as irreversible parasiticidal agents against trophozoites and the WV-93 complex displayed activity against the hepatic stage of P. berghei. The in vivo suppression activity against P. berghei in the Peters 4 day test displayed by the complexes was similar to that of chloroquine. However, the efficacy in an established P. berghei infection in the Thompson test was superior for the WV-93 complex compared to chloroquine. The complexes' antimalarial mechanism of action is initiated by inhibiting the hemozoin formation. While chloroquine efficiently inhibits hemozoin, parasites treated with the platinum complexes display residual hemozoin crystals. This is explained since the interaction of the platinum complexes with ferriprotoporphyrin is weaker than that of chloroquine. However, the complexes caused a loss of mitochondrial integrity and subsequent reduction in mitochondrial activity, and their effects on mitochondria were more pronounced than those in the chloroquine-treated parasites. The dual effect of the platinum complexes may explain their activity against the hemozoin-lacking parasites (hepatic stage), where chloroquine has no activity. Our findings indicate that the platinum(ii)-chloroquine complexes are multifunctional antimalarial compounds and reinforce the importance of metal complexes in antimalarial drug discovery.
Collapse
Affiliation(s)
- Taís S Macedo
- FIOCRUZ, Instituto Gonçalo Moniz, CEP 40296-710, Salvador, BA, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Aguiar ACC, Panciera M, Simão dos Santos EF, Singh MK, Garcia ML, de Souza GE, Nakabashi M, Costa JL, Garcia CRS, Oliva G, Correia CRD, Guido RVC. Discovery of Marinoquinolines as Potent and Fast-Acting Plasmodium falciparum Inhibitors with in Vivo Activity. J Med Chem 2018; 61:5547-5568. [DOI: 10.1021/acs.jmedchem.8b00143] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Anna Caroline Campos Aguiar
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Michele Panciera
- Institute of Chemistry, State University of Campinas, Josue de Castro St., Campinas, SP 13083-970, Brazil
| | | | - Maneesh Kumar Singh
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - Mariana Lopes Garcia
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Guilherme Eduardo de Souza
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Myna Nakabashi
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - José Luiz Costa
- Faculty of Pharmaceutical Sciences, State University of Campinas, Rua Oswaldo Cruz, 2° Andar, Bloco F3, Cidade Universitária, Campinas, SP 13083-859, Brazil
| | - Célia R. S. Garcia
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
| | - Glaucius Oliva
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | | | - Rafael Victorio Carvalho Guido
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| |
Collapse
|
26
|
Selection of Plasmodium falciparum cytochrome B mutants by putative PfNDH2 inhibitors. Proc Natl Acad Sci U S A 2018; 115:6285-6290. [PMID: 29844160 DOI: 10.1073/pnas.1804492115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malaria control is threatened by a limited pipeline of effective pharmaceuticals against drug-resistant strains of Plasmodium falciparum Components of the mitochondrial electron transport chain (ETC) are attractive targets for drug development, owing to exploitable differences between the parasite and human ETC. Disruption of ETC function interferes with metabolic processes including de novo pyrimidine synthesis, essential for nucleic acid replication. We investigated the effects of ETC inhibitor selection on two distinct P. falciparum clones, Dd2 and 106/1. Compounds CK-2-68 and RYL-552, substituted quinolones reported to block P. falciparum NADH dehydrogenase 2 (PfNDH2; a type II NADH:quinone oxidoreductase), unexpectedly selected mutations at the quinol oxidation (Qo) pocket of P. falciparum cytochrome B (PfCytB). Selection experiments with atovaquone (ATQ) on 106/1 parasites yielded highly resistant PfCytB Y268S mutants seen in clinical infections that fail ATQ-proguanil treatment. In contrast, ATQ pressure on Dd2 yielded moderately resistant parasites carrying a PfCytB M133I or K272R mutation. Strikingly, all ATQ-selected mutants demonstrated little change or slight increase of sensitivity to CK-2-68 or RYL-552. Molecular docking studies demonstrated binding of all three ETC inhibitors to the Qo pocket of PfCytB, where Y268 forms strong van der Waals interactions with the hydroxynaphthoquinone ring of ATQ but not the quinolone ring of CK-2-68 or RYL-552. Our results suggest that combinations of suitable ETC inhibitors may be able to subvert or delay the development of P. falciparum drug resistance.
Collapse
|
27
|
Song Z, Iorga BI, Mounkoro P, Fisher N, Meunier B. The antimalarial compound
ELQ
‐400 is an unusual inhibitor of the
bc
1
complex, targeting both
Q
o
and
Q
i
sites. FEBS Lett 2018; 592:1346-1356. [DOI: 10.1002/1873-3468.13035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Zehua Song
- Translational Research Institute Henan Provincial People's Hospital School of Medicine Henan University Zhengzhou China
- Institute for Integrative Biology of the Cell (I2BC) CEA CNRS Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Bogdan I. Iorga
- Institut de Chimie des Substances Naturelles CNRS UPR 2301 Labex LERMIT Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Pierre Mounkoro
- Institute for Integrative Biology of the Cell (I2BC) CEA CNRS Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
| | - Nicholas Fisher
- MSU‐DOE Plant Research Laboratory Michigan State University East Lansing MI USA
| | - Brigitte Meunier
- Institute for Integrative Biology of the Cell (I2BC) CEA CNRS Université Paris‐Sud Université Paris‐Saclay Gif‐sur‐Yvette France
| |
Collapse
|
28
|
Frueh L, Li Y, Mather MW, Li Q, Pou S, Nilsen A, Winter RW, Forquer IP, Pershing AM, Xie LH, Smilkstein MJ, Caridha D, Koop DR, Campbell RF, Sciotti RJ, Kreishman-Deitrick M, Kelly JX, Vesely B, Vaidya AB, Riscoe MK. Alkoxycarbonate Ester Prodrugs of Preclinical Drug Candidate ELQ-300 for Prophylaxis and Treatment of Malaria. ACS Infect Dis 2017; 3:728-735. [PMID: 28927276 DOI: 10.1021/acsinfecdis.7b00062] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
ELQ-300 is a preclinical antimalarial drug candidate that is active against liver, blood, and transmission stages of Plasmodium falciparum. While ELQ-300 is highly effective when administered in a low multidose regimen, poor aqueous solubility and high crystallinity have hindered its clinical development. To overcome its challenging physiochemical properties, a number of bioreversible alkoxycarbonate ester prodrugs of ELQ-300 were synthesized. These bioreversible prodrugs are converted to ELQ-300 by host and parasite esterase action in the liver and bloodstream of the host. One such alkoxycarbonate prodrug, ELQ-331, is curative against Plasmodium yoelii with a single low dose of 3 mg/kg in a murine model of patent malaria infection. ELQ-331 is at least as fully protective as ELQ-300 in a murine malaria prophylaxis model when delivered 24 h before sporozoite inoculation at an oral dose of 1 mg/kg. Here, we show that ELQ-331 is a promising prodrug of ELQ-300 with improved physiochemical and metabolic properties and excellent potential for clinical formulation.
Collapse
Affiliation(s)
- Lisa Frueh
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
- Department of Molecular Microbiology and
Immunology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Yuexin Li
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Michael W. Mather
- Department
of Microbiology and Immunology, Drexel University, 2900 W. Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Qigui Li
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Sovitj Pou
- Department of Molecular Microbiology and
Immunology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Aaron Nilsen
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Rolf W. Winter
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Isaac P. Forquer
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - April M. Pershing
- Department
of Microbiology and Immunology, Drexel University, 2900 W. Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Lisa H. Xie
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Martin J. Smilkstein
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Diana Caridha
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Dennis R. Koop
- Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Robert F. Campbell
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Richard J. Sciotti
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Mara Kreishman-Deitrick
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Jane X. Kelly
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| | - Brian Vesely
- Experimental
Therapeutics Branch, Military Malaria Research Program (MMRP), Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Akhil B. Vaidya
- Department
of Microbiology and Immunology, Drexel University, 2900 W. Queen Lane, Philadelphia, Pennsylvania 19129, United States
| | - Michael K. Riscoe
- Experimental Chemotherapy Laboratory, VA Medical Center (Mail code RD-33), 3710 SW US Veterans Hospital Road, Portland, Oregon 97239, United States
| |
Collapse
|
29
|
Müller J, Aguado A, Laleu B, Balmer V, Ritler D, Hemphill A. In vitro screening of the open source Pathogen Box identifies novel compounds with profound activities against Neospora caninum. Int J Parasitol 2017; 47:801-809. [PMID: 28751177 DOI: 10.1016/j.ijpara.2017.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/01/2017] [Accepted: 06/03/2017] [Indexed: 11/25/2022]
Abstract
Neospora caninum is a major cause of abortion in cattle and represents an important veterinary health problem of great economic significance. The Medicines for Malaria Venture (MMV) Pathogen Box, an open-source collection of 400 compounds with proven anti-infective properties against a wide range of pathogens, was screened against a N. caninum beta-galactosidase reporter strain grown in human foreskin fibroblasts. A primary screening carried out at 1µM yielded 40 compounds that were effective against N. caninum tachyzoites. However, 30 of these compounds also affected the viability of the host cells. The 10 remaining compounds exhibited IC50 values between 4 and 43nM. Three compounds with IC50 values below 10nM, namely MMV676602, MMV688762 and MMV671636, were further characterized in vitro in more detail with respect to inhibition of invasion versus intracellular proliferation, and only MMV671636 had an impact on intracellular proliferation of tachyzoites. This was confirmed by transmission electron microscopy, showing that the primary target of MMV671636 was the mitochondrion. MMV671636 treatment of experimentally infected mice significantly reduced the number of animals with lung and brain infection, and these mice also exhibited a significantly reduced titer of antibodies directed against N. caninum antigens. Thus, MMV671636 is a promising starting point for the development of a future neosporosis therapy.
Collapse
Affiliation(s)
- Joachim Müller
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Adriana Aguado
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Benoît Laleu
- Medicines for Malaria Venture (MMV), PO Box 1826, 20, Route de Pré-Bois, 1215 Geneva 15, Switzerland
| | - Vreni Balmer
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Dominic Ritler
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Andrew Hemphill
- Institute for Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
30
|
Is the Mitochondrion a Good Malaria Drug Target? Trends Parasitol 2017; 33:185-193. [DOI: 10.1016/j.pt.2016.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/25/2016] [Accepted: 10/06/2016] [Indexed: 01/21/2023]
|
31
|
Targeting the Cytochrome bc1 Complex of Leishmania Parasites for Discovery of Novel Drugs. Antimicrob Agents Chemother 2016; 60:4972-82. [PMID: 27297476 DOI: 10.1128/aac.00850-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/02/2016] [Indexed: 11/20/2022] Open
Abstract
Endochin-like quinolones (ELQs) are potent and specific inhibitors of cytochrome bc1 from Plasmodium falciparum and Toxoplasma gondii and show promise for novel antiparasitic drug development. To determine whether the mitochondrial electron transport chain of Leishmania parasites could be targeted similarly for drug development, we investigated the activity of 134 structurally diverse ELQs. A cohort of ELQs was selectively toxic to amastigotes of Leishmania mexicana and L. donovani, with 50% inhibitory concentrations (IC50s) in the low micromolar range, but the structurally similar hydroxynaphthoquinone buparvaquone was by far the most potent inhibitor of electron transport, ATP production, and intracellular amastigote growth. Cytochrome bc1 is thus a promising target for novel antileishmanial drugs, and further improvements on the buparvaquone scaffold are warranted for development of enhanced therapeutics.
Collapse
|
32
|
Atovaquone and ELQ-300 Combination Therapy as a Novel Dual-Site Cytochrome bc1 Inhibition Strategy for Malaria. Antimicrob Agents Chemother 2016; 60:4853-9. [PMID: 27270285 DOI: 10.1128/aac.00791-16] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/24/2016] [Indexed: 12/31/2022] Open
Abstract
Antimalarial combination therapies play a crucial role in preventing the emergence of drug-resistant Plasmodium parasites. Although artemisinin-based combination therapies (ACTs) comprise the majority of these formulations, inhibitors of the mitochondrial cytochrome bc1 complex (cyt bc1) are among the few compounds that are effective for both acute antimalarial treatment and prophylaxis. There are two known sites for inhibition within cyt bc1: atovaquone (ATV) blocks the quinol oxidase (Qo) site of cyt bc1, while some members of the endochin-like quinolone (ELQ) family, including preclinical candidate ELQ-300, inhibit the quinone reductase (Qi) site and retain full potency against ATV-resistant Plasmodium falciparum strains with Qo site mutations. Here, we provide the first in vivo comparison of ATV, ELQ-300, and combination therapy consisting of ATV plus ELQ-300 (ATV:ELQ-300), using P. yoelii murine models of malaria. In our monotherapy assessments, we found that ATV functioned as a single-dose curative compound in suppressive tests whereas ELQ-300 demonstrated a unique cumulative dosing effect that successfully blocked recrudescence even in a high-parasitemia acute infection model. ATV:ELQ-300 therapy was highly synergistic, and the combination was curative with a single combined dose of 1 mg/kg of body weight. Compared to the ATV:proguanil (Malarone) formulation, ATV:ELQ-300 was more efficacious in multiday, acute infection models and was equally effective at blocking the emergence of ATV-resistant parasites. Ultimately, our data suggest that dual-site inhibition of cyt bc1 is a valuable strategy for antimalarial combination therapy and that Qi site inhibitors such as ELQ-300 represent valuable partner drugs for the clinically successful Qo site inhibitor ATV.
Collapse
|
33
|
McPhillie M, Zhou Y, El Bissati K, Dubey J, Lorenzi H, Capper M, Lukens AK, Hickman M, Muench S, Verma SK, Weber CR, Wheeler K, Gordon J, Sanders J, Moulton H, Wang K, Kim TK, He Y, Santos T, Woods S, Lee P, Donkin D, Kim E, Fraczek L, Lykins J, Esaa F, Alibana-Clouser F, Dovgin S, Weiss L, Brasseur G, Wirth D, Kent M, Hood L, Meunieur B, Roberts CW, Hasnain SS, Antonyuk SV, Fishwick C, McLeod R. New paradigms for understanding and step changes in treating active and chronic, persistent apicomplexan infections. Sci Rep 2016; 6:29179. [PMID: 27412848 PMCID: PMC4944145 DOI: 10.1038/srep29179] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/31/2016] [Indexed: 12/24/2022] Open
Abstract
Toxoplasma gondii, the most common parasitic infection of human brain and eye, persists across lifetimes, can progressively damage sight, and is currently incurable. New, curative medicines are needed urgently. Herein, we develop novel models to facilitate drug development: EGS strain T. gondii forms cysts in vitro that induce oocysts in cats, the gold standard criterion for cysts. These cysts highly express cytochrome b. Using these models, we envisioned, and then created, novel 4-(1H)-quinolone scaffolds that target the cytochrome bc1 complex Qi site, of which, a substituted 5,6,7,8-tetrahydroquinolin-4-one inhibits active infection (IC50, 30 nM) and cysts (IC50, 4 μM) in vitro, and in vivo (25 mg/kg), and drug resistant Plasmodium falciparum (IC50, <30 nM), with clinically relevant synergy. Mutant yeast and co-crystallographic studies demonstrate binding to the bc1 complex Qi site. Our results have direct impact on improving outcomes for those with toxoplasmosis, malaria, and ~2 billion persons chronically infected with encysted bradyzoites.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Amanda K Lukens
- Harvard School of Public Health, Boston, Massachusetts, USA
- The Broad Institute, Boston, Massachusetts, USA
| | - Mark Hickman
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | | | | | - Kai Wang
- Institute for Systems Biology, Seattle, Washington, USA
| | - Taek-Kyun Kim
- Institute for Systems Biology, Seattle, Washington, USA
| | - Yuqing He
- Institute for Systems Biology, Seattle, Washington, USA
| | - Tatiana Santos
- Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Patty Lee
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - David Donkin
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Eric Kim
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | - Louis Weiss
- Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Dyann Wirth
- Harvard School of Public Health, Boston, Massachusetts, USA
- The Broad Institute, Boston, Massachusetts, USA
| | | | - Leroy Hood
- Institute for Systems Biology, Seattle, Washington, USA
| | - Brigitte Meunieur
- Institute for Integrative Biology of the Cell (12BC), Gif-sur-Yvette, France
| | | | | | | | | | | |
Collapse
|
34
|
Moradin N, Torre S, Gauthier S, Tam M, Hawari J, Vandercruyssen K, De Spiegeleer B, Fortin A, Stevenson MM, Gros P. Cysteamine broadly improves the anti-plasmodial activity of artemisinins against murine blood stage and cerebral malaria. Malar J 2016; 15:260. [PMID: 27150250 PMCID: PMC4858922 DOI: 10.1186/s12936-016-1317-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The potential emergence and spread of resistance to artemisinins in the Plasmodium falciparum malaria parasite constitutes a major global health threat. Hence, improving the efficacy of artemisinins and of artemisinin-based combination therapy (ACT) represents a major short-term goal in the global fight against malaria. Mice defective in the enzyme pantetheinase (Vnn3) show increased susceptibility to blood-stage malaria (increased parasitaemia, reduced survival), and supplementation of Vnn3 mutants with the reaction product of pantetheinase, cysteamine, corrects in part the malaria-susceptibility phenotype of the mutants. Cysteamine (Cys) is a small, naturally occurring amino-thiol that has very low toxicity in vivo and is approved for clinical use in the life-long treatment of the kidney disorder nephropathic cystinosis. METHODS The ability of Cys to improve the anti-plasmodial activity of different clinically used artemisinins was tested. The effect of different CYS/ART combinations on malarial phenotypes (parasite blood-stage replication, overall and survival from lethal infection) was assessed in a series of in vivo experiments using Plasmodium strains that induce either blood-stage (Plasmodium chabaudi AS) or cerebral disease (Plasmodium berghei ANKA). This was also evaluated in an ex vivo experimental protocol that directly assesses the effect of such drug combinations on the viability of Plasmodium parasites, as measured by the ability of tested parasites to induce a productive infection in vivo in otherwise naïve animals. RESULTS Cys is found to potentiate the anti-plasmodial activity of artesunate, artemether, and arteether, towards the blood-stage malaria parasite P. chabaudi AS. Ex vivo experiments, indicate that potentiation of the anti-plasmodial activity of artemisinins by Cys is direct and does not require the presence of host factors. In addition, potentiation occurs at sub-optimal concentrations of artemisinins and Cys that on their own have little or no effect on parasite growth. Cys also dramatically enhances the efficacy and protective effect of artemisinins against cerebral malaria induced by infection with the P. berghei ANKA parasite. CONCLUSION These findings indicate that inclusion of Cys in current formulations of ACT, or its use as adjunct therapy could improve the anti-plasmodial activity of artemisinin, decrease mortality in cerebral malaria patients, and prevent or delay the development and spread of artemisinin resistance.
Collapse
Affiliation(s)
- Neda Moradin
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Sabrina Torre
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Susan Gauthier
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Mifong Tam
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Jalal Hawari
- Department of Civil, Geological and Mining Engineering, Ecole Polytechnique, Université de Montreal, Montreal, Canada
| | | | | | - Anny Fortin
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Mary M Stevenson
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
35
|
Goodman CD, Siregar JE, Mollard V, Vega-Rodríguez J, Syafruddin D, Matsuoka H, Matsuzaki M, Toyama T, Sturm A, Cozijnsen A, Jacobs-Lorena M, Kita K, Marzuki S, McFadden GI. Parasites resistant to the antimalarial atovaquone fail to transmit by mosquitoes. Science 2016; 352:349-53. [PMID: 27081071 DOI: 10.1126/science.aad9279] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/10/2016] [Indexed: 12/16/2022]
Abstract
Drug resistance compromises control of malaria. Here, we show that resistance to a commonly used antimalarial medication, atovaquone, is apparently unable to spread. Atovaquone pressure selects parasites with mutations in cytochrome b, a respiratory protein with low but essential activity in the mammalian blood phase of the parasite life cycle. Resistance mutations rescue parasites from the drug but later prove lethal in the mosquito phase, where parasites require full respiration. Unable to respire efficiently, resistant parasites fail to complete mosquito development, arresting their life cycle. Because cytochrome b is encoded by the maternally inherited parasite mitochondrion, even outcrossing with wild-type strains cannot facilitate spread of resistance. Lack of transmission suggests that resistance will be unable to spread in the field, greatly enhancing the utility of atovaquone in malaria control.
Collapse
Affiliation(s)
| | - Josephine E Siregar
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia. Eijkman Institute for Molecular Biology, JI Diponegoro no. 69, Jakarta, 10430, Indonesia. Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Vanessa Mollard
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Joel Vega-Rodríguez
- Johns Hopkins University Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, MD 21205, USA
| | - Din Syafruddin
- Eijkman Institute for Molecular Biology, JI Diponegoro no. 69, Jakarta, 10430, Indonesia. Department of Parasitology, Faculty of Medicine, Hasanuddin University, Jalan Perintis Kemerdekaan Km10, Makassar 90245, Indonesia
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Motomichi Matsuzaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoko Toyama
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Angelika Sturm
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anton Cozijnsen
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Marcelo Jacobs-Lorena
- Johns Hopkins University Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, MD 21205, USA
| | - Kiyoshi Kita
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan. School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
| | - Sangkot Marzuki
- Eijkman Institute for Molecular Biology, JI Diponegoro no. 69, Jakarta, 10430, Indonesia
| | - Geoffrey I McFadden
- School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
36
|
Discovery of Dual-Stage Malaria Inhibitors with New Targets. Antimicrob Agents Chemother 2015; 60:1430-7. [PMID: 26666931 DOI: 10.1128/aac.02110-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/08/2015] [Indexed: 11/20/2022] Open
Abstract
Malaria remains a major global health problem, with more than half of the world population at risk of contracting the disease and nearly a million deaths each year. Here, we report the discovery of inhibitors that target multiple stages of malaria parasite growth. To identify these inhibitors, we took advantage of the Tres Cantos Antimalarial Compound Set (TCAMS) small-molecule library, which is comprised of diverse and potent chemical scaffolds with activities against the blood stage of the malaria parasite, and investigated their effects against the elusive liver stage of the malaria parasite using a forward chemical screen. From a screen of nearly 14,000 compounds, we identified and confirmed 103 compounds as dual-stage malaria inhibitors. Interestingly, these compounds show preferential inhibition of parasite growth in liver- versus blood-stage malaria parasite assays, highlighting the drug susceptibility of this parasite form. Mode-of-action studies were completed using genetically modified and drug-resistant Plasmodium parasite strains. While we identified some compound targets as classical antimalarial pathways, such as the mitochondrial electron transport chain through cytochrome bc1 complex inhibition or the folate biosynthesis pathway, most compounds induced parasite death through as yet unknown mechanisms of action. Importantly, the identification of new chemotypes with different modes of action in killing Plasmodium parasites represents a promising opportunity for probing essential and novel molecular processes that remain to be discovered. The chemical scaffolds identified with activity against drug-resistant Plasmodium parasites represent starting points for dual-stage antimalarial development to surmount the threat of malaria parasite drug resistance.
Collapse
|