51
|
Huang YN, Li YL, Li J, Deng J. Beyond a Protecting Reagent: DMAP-Catalyzed Cyclization of Boc-Anhydride with 2-Alkenylanilines. J Org Chem 2016; 81:4645-53. [DOI: 10.1021/acs.joc.6b00519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ya-Nan Huang
- School of Pharmaceutical Science and Technology, Key Laboratory for Modern Drug Delivery & High-Efficiency, Tianjin University, Tianjin, 300072 P. R. China
| | - Yin-Long Li
- School of Pharmaceutical Science and Technology, Key Laboratory for Modern Drug Delivery & High-Efficiency, Tianjin University, Tianjin, 300072 P. R. China
| | - Jian Li
- School of Pharmaceutical Science and Technology, Key Laboratory for Modern Drug Delivery & High-Efficiency, Tianjin University, Tianjin, 300072 P. R. China
| | - Jun Deng
- School of Pharmaceutical Science and Technology, Key Laboratory for Modern Drug Delivery & High-Efficiency, Tianjin University, Tianjin, 300072 P. R. China
| |
Collapse
|
52
|
Chen X, Cui X, Wu Y. “One-Pot” Approach to 8-Acylated 2-Quinolinones via Palladium-Catalyzed Regioselective Acylation of Quinoline N-Oxides. Org Lett 2016; 18:2411-4. [DOI: 10.1021/acs.orglett.6b00923] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaopei Chen
- Department
of Chemistry, Henan Key Laboratory of Chemical Biology and Organic
Chemistry, Key Laboratory of Applied Chemistry of Henan Universities, Zhengzhou University, Zhengzhou 450052, P. R. China
| | - Xiuling Cui
- Department
of Chemistry, Henan Key Laboratory of Chemical Biology and Organic
Chemistry, Key Laboratory of Applied Chemistry of Henan Universities, Zhengzhou University, Zhengzhou 450052, P. R. China
- Xiamen Key Laboratory of Ocean and Gene Drugs, School of Biomedical Sciences, Institute of Molecular Medicine of Huaqiao University & Engineering Research Centre of Molecular Medicine of Chinese Education Ministry, Xiamen, Fujian 361021, P. R. China
| | - Yangjie Wu
- Department
of Chemistry, Henan Key Laboratory of Chemical Biology and Organic
Chemistry, Key Laboratory of Applied Chemistry of Henan Universities, Zhengzhou University, Zhengzhou 450052, P. R. China
| |
Collapse
|
53
|
Dzhons DY, Budruev AV. Synthesis of 2,1-benzisoxazole-3(1H)-ones by base-mediated photochemical N-O bond-forming cyclization of 2-azidobenzoic acids. Beilstein J Org Chem 2016; 12:874-81. [PMID: 27340478 PMCID: PMC4902054 DOI: 10.3762/bjoc.12.86] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/18/2016] [Indexed: 01/03/2023] Open
Abstract
The base-mediated photochemical cyclization of 2-azidobenzoic acids with the formation of 2,1-benzisoxazole-3(1H)-ones is reported. The optimization and scope of this cyclization reaction is discussed. It is shown that an essential step of the ring closure of 2-azidobenzoic acids is the formation and photolysis of 2-azidobenzoate anions.
Collapse
Affiliation(s)
- Daria Yu Dzhons
- Chemistry Department, Lobachevsky State University of Nizhny Novgorod, Gagarina pr. 23, Building 5, 603950, Nizhny Novgorod, Russian Federation
| | - Andrei V Budruev
- Chemistry Department, Lobachevsky State University of Nizhny Novgorod, Gagarina pr. 23, Building 5, 603950, Nizhny Novgorod, Russian Federation
| |
Collapse
|
54
|
Recent advances in the application of group-10 transition metal based catalysts in C–H activation and functionalization. J Organomet Chem 2015. [DOI: 10.1016/j.jorganchem.2015.03.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
55
|
Ragusa G, Gómez-Cañas M, Morales P, Hurst DP, Deligia F, Pazos R, Pinna GA, Fernández-Ruiz J, Goya P, Reggio PH, Jagerovic N, García-Arencibia M, Murineddu G. Synthesis, pharmacological evaluation and docking studies of pyrrole structure-based CB2 receptor antagonists. Eur J Med Chem 2015. [PMID: 26209834 DOI: 10.1016/j.ejmech.2015.06.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
During the last years, there has been a continuous interest in the development of cannabinoid receptor ligands that may serve as therapeutic agents and/or as experimental tools. This prompted us to design and synthesize analogues of the CB2 receptor antagonist N-fenchyl-5-(4-chloro-3-methyl-phenyl)-1-(4-methyl-benzyl)-1H-pyrazole-3-carboxamide (SR144528). The structural modifications involved the bioisosteric replacement of the pyrazole ring by a pyrrole ring and variations on the amine carbamoyl substituents. Two of these compounds, the fenchyl pyrrole analogue 6 and the myrtanyl derivative 10, showed high affinity (Ki in the low nM range) and selectivity for the CB2 receptor and both resulted to be antagonists/inverse agonists in [(35)S]-GTPγS binding analysis and in an in vitro CB2 receptor bioassay. Cannabinoid receptor binding data of the series allowed identifying steric constraints within the CB2 binding pocket using a study of Van der Waals' volume maps. Glide docking studies revealed that all docked compounds bind in the same region of the CB2 receptor inactive state model.
Collapse
Affiliation(s)
- Giulio Ragusa
- Dipartimento di Chimica e Farmacia, Università degli Studi di Sassari, via F. Muroni 23/A, 07100 Sassari, Italy
| | - María Gómez-Cañas
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigación en Neuroquímica, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; Campus de Excelencia Internacional (CEI-Moncloa), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Paula Morales
- Instituto de Química Médica, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Dow P Hurst
- Center for Drug Discovery, Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Francesco Deligia
- Dipartimento di Chimica e Farmacia, Università degli Studi di Sassari, via F. Muroni 23/A, 07100 Sassari, Italy
| | - Ruth Pazos
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigación en Neuroquímica, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; Campus de Excelencia Internacional (CEI-Moncloa), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Gerard A Pinna
- Dipartimento di Chimica e Farmacia, Università degli Studi di Sassari, via F. Muroni 23/A, 07100 Sassari, Italy
| | - Javier Fernández-Ruiz
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigación en Neuroquímica, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; Campus de Excelencia Internacional (CEI-Moncloa), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pilar Goya
- Instituto de Química Médica, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Patricia H Reggio
- Center for Drug Discovery, Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Nadine Jagerovic
- Instituto de Química Médica, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Moisés García-Arencibia
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigación en Neuroquímica, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; Campus de Excelencia Internacional (CEI-Moncloa), Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Gabriele Murineddu
- Dipartimento di Chimica e Farmacia, Università degli Studi di Sassari, via F. Muroni 23/A, 07100 Sassari, Italy.
| |
Collapse
|
56
|
Kancherla R, Naveen T, Maiti D. Palladium-Catalyzed [3+3] Annulation between Diarylamines and α,β-Unsaturated Acids through CH Activation: Direct Access to 4-Substituted 2-Quinolinones. Chemistry 2015; 21:8360-4. [DOI: 10.1002/chem.201500774] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 11/07/2022]
|
57
|
Wang W, Peng X, Qin X, Zhao X, Ma C, Tung CH, Xu Z. Synthesis of quinolinones with palladium-catalyzed oxidative annulation between acrylamides and arynes. J Org Chem 2015; 80:2835-41. [PMID: 25686292 DOI: 10.1021/jo5027673] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
An unprecedented palladium-catalyzed oxidative annulation of acrylamides with benzyne precursors has been successfully developed. By using this mild "N-H activation/Heck reaction" method, a wide variety of quinolinones were conveniently prepared in one step with high efficiency.
Collapse
Affiliation(s)
- Weiguo Wang
- Key Lab of Colloid and Interface Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University , No. 27 South Shanda Road, Jinan, Shandong 250100, China
| | | | | | | | | | | | | |
Collapse
|
58
|
Wu J, Xiang S, Zeng J, Leow M, Liu XW. Practical route to 2-quinolinones via a Pd-catalyzed C-H bond activation/C-C bond formation/cyclization cascade reaction. Org Lett 2014; 17:222-5. [PMID: 25545799 DOI: 10.1021/ol503292p] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Quinolinone derivatives were constructed via a Pd-catalyzed C-H bond activation/C-C bond formation/cyclization cascade process with simple anilines as the substrates. This finding provides a practical procedure for the synthesis of quinolinone-containing alkaloids and drug molecules. The utility of this method was demonstrated by a formal synthesis of Tipifarnib.
Collapse
Affiliation(s)
- Junliang Wu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, Singapore 637371
| | | | | | | | | |
Collapse
|
59
|
Choi JY, Podust LM, Roush WR. Drug strategies targeting CYP51 in neglected tropical diseases. Chem Rev 2014; 114:11242-71. [PMID: 25337991 PMCID: PMC4254036 DOI: 10.1021/cr5003134] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Jun Yong Choi
- Department
of Chemistry, Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Larissa M. Podust
- Center for Discovery and Innovation in Parasitic Diseases, and Department of
Pathology, University of California—San
Francisco, San Francisco, California 94158, United States
| | - William R. Roush
- Department
of Chemistry, Scripps Florida, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
60
|
Vieira DF, Choi JY, Calvet CM, Siqueira-Neto JL, Johnston JB, Kellar D, Gut J, Cameron MD, McKerrow JH, Roush WR, Podust LM. Binding mode and potency of N-indolyloxopyridinyl-4-aminopropanyl-based inhibitors targeting Trypanosoma cruzi CYP51. J Med Chem 2014; 57:10162-75. [PMID: 25393646 PMCID: PMC4266343 DOI: 10.1021/jm501568b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Chagas disease is a chronic infection
in humans caused by Trypanosoma cruzi and manifested
in progressive cardiomyopathy
and/or gastrointestinal dysfunction. Limited therapeutic options to
prevent and treat Chagas disease put 8 million people infected with T. cruzi worldwide at risk. CYP51, involved in the biosynthesis
of the membrane sterol component in eukaryotes, is a promising drug
target in T. cruzi. We report the structure–activity
relationships (SAR) of an N-arylpiperazine series
of N-indolyloxopyridinyl-4-aminopropanyl-based inhibitors
designed to probe the impact of substituents in the terminal N-phenyl
ring on binding mode, selectivity and potency. Depending on the substituents
at C-4, two distinct ring binding modes, buried and solvent-exposed,
have been observed by X-ray structure analysis (resolution of 1.95–2.48
Å). The 5-chloro-substituted analogs 9 and 10 with no substituent at C-4 demonstrated improved selectivity
and potency, suppressing ≥99.8% parasitemia in mice when administered
orally at 25 mg/kg, b.i.d., for 4 days.
Collapse
Affiliation(s)
- Debora F Vieira
- Center for Discovery and Innovation in Parasitic Diseases, ‡Department of Pathology, and §Department of Pharmaceutical Chemistry, University of California-San Francisco , San Francisco, California 94158, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Wen X, Chen J, Peng Y. Synthesis of Multifunctional 3-Amino-4-phosphono-2-quinolinonesviaRegioselective Ring Enlargement of Imino Isatins. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201400652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
62
|
Chen J, Natte K, Spannenberg A, Neumann H, Beller M, Wu XF. Palladium-Catalyzed Carbonylative [3+2+1] Annulation ofN-Aryl-Pyridine-2-Amines with Internal Alkynes by CH Activation: Facile Synthesis of 2-Quinolinones. Chemistry 2014; 20:14189-93. [DOI: 10.1002/chem.201404462] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Indexed: 01/02/2023]
|
63
|
Abstract
We report here a new, atom economical annulation to 2,1-benzisoxazole scaffolds via the BF3·Et2O-catalyzed reaction of glyoxylate esters and nitrosoarenes. The developed method represents a convergent route to this compound class from previously unexplored inputs and provides a range of 2,1-benzisoxazoles in moderate to high yields under convenient conditions. Along with exploration of substrate scope, initial mechanistic investigation through (18)O labeling and the synthesis of a reaction intermediate provides evidence for an unusual umpolung addition of glyoxylates to nitrosobenzenes with high O-selectivity, followed by a new type of Friedel-Crafts cyclization.
Collapse
Affiliation(s)
- Kate D. Otley
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| | - Jonathan A. Ellman
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
64
|
Calvet C, Vieira D, Choi JY, Kellar D, Cameron MD, Siqueira-Neto JL, Gut J, Johnston JB, Lin L, Khan S, McKerrow JH, Roush WR, Podust LM. 4-Aminopyridyl-based CYP51 inhibitors as anti-Trypanosoma cruzi drug leads with improved pharmacokinetic profile and in vivo potency. J Med Chem 2014; 57:6989-7005. [PMID: 25101801 PMCID: PMC4148169 DOI: 10.1021/jm500448u] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Indexed: 12/26/2022]
Abstract
CYP51 is a P450 enzyme involved in the biosynthesis of the sterol components of eukaryotic cell membranes. CYP51 inhibitors have been developed to treat infections caused by fungi, and more recently the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease. To specifically optimize drug candidates for T. cruzi CYP51 (TcCYP51), we explored the structure-activity relationship (SAR) of a N-indolyl-oxopyridinyl-4-aminopropanyl-based scaffold originally identified in a target-based screen. This scaffold evolved via medicinal chemistry to yield orally bioavailable leads with potent anti-T. cruzi activity in vivo. Using an animal model of infection with a transgenic T. cruzi Y luc strain expressing firefly luciferase, we prioritized the biaryl and N-arylpiperazine analogues by oral bioavailability and potency. The drug-target complexes for both scaffold variants were characterized by X-ray structure analysis. Optimization of both binding mode and pharmacokinetic properties of these compounds led to potent inhibitors against experimental T. cruzi infection.
Collapse
Affiliation(s)
- Claudia
M. Calvet
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
- Cellular
Ultra-Structure Laboratory, Oswaldo Cruz
Institute (IOC), FIOCRUZ, Rio de
Janeiro, Re de Janeiro 21040-362, Brazil
| | - Debora
F. Vieira
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - Jun Yong Choi
- Department
of Chemistry, Department of Molecular Therapeutics, Scripps
Florida, Jupiter, Florida 33458, United
States
| | - Danielle Kellar
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - Michael D. Cameron
- Department
of Chemistry, Department of Molecular Therapeutics, Scripps
Florida, Jupiter, Florida 33458, United
States
| | - Jair Lage Siqueira-Neto
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - Jiri Gut
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - Jonathan B. Johnston
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - Li Lin
- Department
of Chemistry, Department of Molecular Therapeutics, Scripps
Florida, Jupiter, Florida 33458, United
States
| | - Susan Khan
- Department
of Chemistry, Department of Molecular Therapeutics, Scripps
Florida, Jupiter, Florida 33458, United
States
| | - James H. McKerrow
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| | - William R. Roush
- Department
of Chemistry, Department of Molecular Therapeutics, Scripps
Florida, Jupiter, Florida 33458, United
States
| | - Larissa M. Podust
- Center for Discovery and Innovation in Parasitic
Diseases, Department of Pathology and Department of Medicine, Department of Pharmaceutical
Chemistry, University of California San
Francisco, San Francisco, California 94158, United States
| |
Collapse
|
65
|
Planer JD, Hulverson MA, Arif JA, Ranade RM, Don R, Buckner FS. Synergy testing of FDA-approved drugs identifies potent drug combinations against Trypanosoma cruzi. PLoS Negl Trop Dis 2014; 8:e2977. [PMID: 25033456 PMCID: PMC4102417 DOI: 10.1371/journal.pntd.0002977] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/12/2014] [Indexed: 12/14/2022] Open
Abstract
An estimated 8 million persons, mainly in Latin America, are infected with Trypanosoma cruzi, the etiologic agent of Chagas disease. Existing antiparasitic drugs for Chagas disease have significant toxicities and suboptimal effectiveness, hence new therapeutic strategies need to be devised to address this neglected tropical disease. Due to the high research and development costs of bringing new chemical entities to the clinic, we and others have investigated the strategy of repurposing existing drugs for Chagas disease. Screens of FDA-approved drugs (described in this paper) have revealed a variety of chemical classes that have growth inhibitory activity against mammalian stage Trypanosoma cruzi parasites. Aside from azole antifungal drugs that have low or sub-nanomolar activity, most of the active compounds revealed in these screens have effective concentrations causing 50% inhibition (EC50's) in the low micromolar or high nanomolar range. For example, we have identified an antihistamine (clemastine, EC50 of 0.4 µM), a selective serotonin reuptake inhibitor (fluoxetine, EC50 of 4.4 µM), and an antifolate drug (pyrimethamine, EC50 of 3.8 µM) and others. When tested alone in the murine model of Trypanosoma cruzi infection, most compounds had insufficient efficacy to lower parasitemia thus we investigated using combinations of compounds for additive or synergistic activity. Twenty-four active compounds were screened in vitro in all possible combinations. Follow up isobologram studies showed at least 8 drug pairs to have synergistic activity on T. cruzi growth. The combination of the calcium channel blocker, amlodipine, plus the antifungal drug, posaconazole, was found to be more effective at lowering parasitemia in mice than either drug alone, as was the combination of clemastine and posaconazole. Using combinations of FDA-approved drugs is a promising strategy for developing new treatments for Chagas disease. Chronic infection with Trypanosoma cruzi causes progressive damage to the heart and other organs that is fatal in about 30% of cases. Known as Chagas disease, this is a major public health problem in Latin America. The existing medicines were developed over forty years ago and are not widely used because of toxicity and unreliable effectiveness. To discover better treatments, we screened a collection of existing drugs for growth inhibitory activity on Trypanosoma cruzi. Several dozen orally administered drugs were discovered, but when used by themselves they were not strong enough to cure the infection in an animal model. We tested a set of 24 of these drugs in every two-way combination and identified eight synergistic partners. At least two of these combinations were able to substantially lower parasite levels in the mouse model of Trypanosoma cruzi infection. Thus, finding pairs of FDA-approved drugs that can be used in combination may be a pragmatic and effective strategy for designing new therapies for Chagas disease.
Collapse
Affiliation(s)
- Joseph D. Planer
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Matthew A. Hulverson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jennifer A. Arif
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ranae M. Ranade
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Robert Don
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Frederick S. Buckner
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
66
|
Manikandan R, Jeganmohan M. Ruthenium-catalyzed cyclization of anilides with substituted propiolates or acrylates: an efficient route to 2-quinolinones. Org Lett 2014; 16:3568-71. [PMID: 24956409 DOI: 10.1021/ol501548e] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A Ru-catalyzed cyclization of anilides with propiolates or acrylates affording 2-quinolinones having diverse functional groups in good to excellent yields is described. Later, 2-quinolinones were converted into 3-halo-2-quinolinones and 2-chloroquinolines. The proposed mechanism was strongly supported by experimental evidence and deuterium labeling studies.
Collapse
Affiliation(s)
- Rajendran Manikandan
- Department of Chemistry, Indian Institute of Science Education and Research , Pune 411021, India
| | | |
Collapse
|
67
|
Deng Y, Gong W, He J, Yu JQ. Ligand-enabled triple C-H activation reactions: one-pot synthesis of diverse 4-aryl-2-quinolinones from propionamides. Angew Chem Int Ed Engl 2014; 53:6692-5. [PMID: 24828691 PMCID: PMC4114037 DOI: 10.1002/anie.201403878] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Indexed: 11/06/2022]
Abstract
Diverse 4-aryl-2-quinolinones are prepared from propionamides in one pot by ligand-promoted triple sequential C-H activation reactions and a stereospecific Heck reaction. In these cascade reactions, three new C-C bonds and one C-N bond are formed to rapidly build molecular complexity from propionic acid.
Collapse
Affiliation(s)
| | | | - Jian He
- Department of Chemistry, The Scripps Research Institute (TSRI), 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA)
| | - Jin-Quan Yu
- Department of Chemistry, The Scripps Research Institute (TSRI), 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA)
| |
Collapse
|
68
|
Deng Y, Gong W, He J, Yu JQ. Ligand-Enabled Triple CH Activation Reactions: One-Pot Synthesis of Diverse 4-Aryl-2-quinolinones from Propionamides. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403878] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
69
|
Tatipaka HB, Gillespie JR, Chatterjee AK, Norcross NR, Hulverson MA, Ranade RM, Nagendar P, Creason SA, McQueen J, Duster NA, Nagle A, Supek F, Molteni V, Wenzler T, Brun R, Glynne R, Buckner FS, Gelb MH. Substituted 2-phenylimidazopyridines: a new class of drug leads for human African trypanosomiasis. J Med Chem 2014; 57:828-35. [PMID: 24354316 DOI: 10.1021/jm401178t] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A phenotypic screen of a compound library for antiparasitic activity on Trypanosoma brucei, the causative agent of human African trypanosomiasis, led to the identification of substituted 2-(3-aminophenyl)oxazolopyridines as a starting point for hit-to-lead medicinal chemistry. A total of 110 analogues were prepared, which led to the identification of 64, a substituted 2-(3-aminophenyl)imidazopyridine. This compound showed antiparasitic activity in vitro with an EC50 of 2 nM and displayed reasonable druglike properties when tested in a number of in vitro assays. The compound was orally bioavailable and displayed good plasma and brain exposure in mice. Compound 64 cured mice infected with Trypanosoma brucei when dosed orally down to 2.5 mg/kg. Given its potent antiparasitic properties and its ease of synthesis, compound 64 represents a new lead for the development of drugs to treat human African trypanosomiasis.
Collapse
Affiliation(s)
- Hari Babu Tatipaka
- Departments of †Chemistry, ‡Medicine, and §Biochemistry, University of Washington , Seattle, Washington 98195, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Gupta S, Ganguly B, Das S. A straight forward synthesis of 4-aryl substituted 2-quinolones via Heck reaction. RSC Adv 2014. [DOI: 10.1039/c4ra06284a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pd–NHC catalyzed one pot synthesis of 4-aryl-2-quinolones through the Heck reaction followed by cyclization. Additionally an efficient methodology has been developed for Heck reaction with a wide range of arylhalides and olefins.
Collapse
Affiliation(s)
- Sumanta Gupta
- Department of Chemistry
- North Bengal University
- Darjeeling, India
| | - Bhaskar Ganguly
- Department of Chemistry
- North Bengal University
- Darjeeling, India
| | - Sajal Das
- Department of Chemistry
- North Bengal University
- Darjeeling, India
| |
Collapse
|
71
|
Choi JY, Calvet CM, Gunatilleke SS, Ruiz C, Cameron MD, McKerrow JH, Podust LM, Roush WR. Rational development of 4-aminopyridyl-based inhibitors targeting Trypanosoma cruzi CYP51 as anti-chagas agents. J Med Chem 2013; 56:7651-68. [PMID: 24079662 PMCID: PMC3864028 DOI: 10.1021/jm401067s] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A new series of 4-aminopyridyl-based lead inhibitors targeting Trypanosoma cruzi CYP51 (TcCYP51) has been developed using structure-based drug design as well as structure-property relationship (SPR) analyses. The screening hit starting point, LP10 (KD ≤ 42 nM; EC50 = 0.65 μM), has been optimized to give the potential leads 14t, 27i, 27q, 27r, and 27t, which have low-nanomolar binding affinity to TcCYP51 and significant activity against T. cruzi amastigotes cultured in human myoblasts (EC50 = 14-18 nM for 27i and 27r). Many of the optimized compounds have improved microsome stability, and most are selective against human CYPs 1A2, 2D6, and 3A4 (<50% inhibition at 1 μM). A rationale for the improvement in microsome stability and selectivity of inhibitors against human metabolic CYP enzymes is presented. In addition, the binding mode of 14t with the Trypanosoma brucei CYP51 (TbCYP51) orthologue has been characterized by X-ray structure analysis.
Collapse
Affiliation(s)
- Jun Yong Choi
- Department of Chemistry, Scripps Florida, Jupiter, Florida 33458, United States
| | - Claudia M. Calvet
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California 94158, United States
- Department of Pathology, University of California San Francisco, San Francisco, California 94158, United States
| | - Shamila S. Gunatilleke
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California 94158, United States
- Department of Pathology, University of California San Francisco, San Francisco, California 94158, United States
| | - Claudia Ruiz
- Department of Molecular Therapeutics, Scripps Florida, Jupiter, Florida 33458, United States
| | - Michael D. Cameron
- Department of Molecular Therapeutics, Scripps Florida, Jupiter, Florida 33458, United States
| | - James H. McKerrow
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California 94158, United States
- Department of Pathology, University of California San Francisco, San Francisco, California 94158, United States
| | - Larissa M. Podust
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California 94158, United States
- Department of Pathology, University of California San Francisco, San Francisco, California 94158, United States
| | - William R. Roush
- Department of Chemistry, Scripps Florida, Jupiter, Florida 33458, United States
| |
Collapse
|
72
|
Paterna R, André V, Duarte MT, Veiros LF, Candeias NR, Gois PMP. Ring-Expansion Reaction of Isatins with Ethyl Diazoacetate Catalyzed by Dirhodium(II)/DBU Metal-Organic System: En Route to Viridicatin Alkaloids. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300796] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
73
|
Sykes ML, Avery VM. Approaches to Protozoan Drug Discovery: Phenotypic Screening. J Med Chem 2013; 56:7727-40. [DOI: 10.1021/jm4004279] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Melissa L. Sykes
- Discovery Biology, Eskitis Institute
for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Vicky M. Avery
- Discovery Biology, Eskitis Institute
for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
74
|
Xie P, Wang ZQ, Deng GB, Song RJ, Xia JD, Hu M, Li JH. Copper-Catalyzed Aerobic Oxidative Carbocyclization- Ketonization Cascade: Selective Synthesis of Quinolinones. Adv Synth Catal 2013. [DOI: 10.1002/adsc.201300160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
75
|
Upadhayaya RS, Dixit SS, Földesi A, Chattopadhyaya J. New antiprotozoal agents: Their synthesis and biological evaluations. Bioorg Med Chem Lett 2013; 23:2750-8. [DOI: 10.1016/j.bmcl.2013.02.054] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 02/08/2013] [Accepted: 02/12/2013] [Indexed: 11/26/2022]
|
76
|
Ferguson J, Zeng F, Alwis N, Alper H. Synthesis of 2(1H)-quinolinones via Pd-catalyzed oxidative cyclocarbonylation of 2-vinylanilines. Org Lett 2013; 15:1998-2001. [PMID: 23550614 DOI: 10.1021/ol4006739] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Palladium-catalyzed oxidative cyclocarbonylation of N-monosubstituted-2-vinylanilines constitutes a simple, direct, and selective method for the synthesis of 2(1H)-quinolinones. The reaction conditions are attractive in terms of environmental considerations and operational simplicity. 2(1H)-Quinolinones with a variety of functional groups were prepared in up to 97% yield.
Collapse
Affiliation(s)
- Jamie Ferguson
- Centre for Catalysis Research and Innovation, Department of Chemistry, University of Ottawa, 10 Marie Curie, Ottawa, Ontario, Canada, K1N 6N5
| | | | | | | |
Collapse
|
77
|
Andriani G, Amata E, Beatty J, Clements Z, Coffey BJ, Courtemanche G, Devine W, Erath J, Juda CE, Wawrzak Z, Wood JT, Lepesheva GI, Rodriguez A, Pollastri MP. Antitrypanosomal lead discovery: identification of a ligand-efficient inhibitor of Trypanosoma cruzi CYP51 and parasite growth. J Med Chem 2013; 56:2556-67. [PMID: 23448316 DOI: 10.1021/jm400012e] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chagas disease is caused by the intracellular protozoan parasite Trypanosomal cruzi , and current drugs are lacking in terms of desired safety and efficacy profiles. Following on a recently reported high-throughput screening campaign, we have explored initial structure-activity relationships around a class of imidazole-based compounds. This profiling has uncovered compounds 4c (NEU321) and 4j (NEU704), which are potent against in vitro cultures of T. cruzi and are greater than 160-fold selective over host cells. We report in vitro drug metabolism and properties profiling of 4c and show that this chemotype inhibits the T. cruzi CYP51 enzyme, an observation confirmed by X-ray crystallographic analysis. We compare the binding orientation of 4c to that of other, previously reported inhibitors. We show that 4c displays a significantly better ligand efficiency and a shorter synthetic route over previously disclosed CYP51 inhibitors, and should therefore be considered a promising lead compound for further optimization.
Collapse
Affiliation(s)
- Grasiella Andriani
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Keenan M, Alexander PW, Diao H, Best WM, Khong A, Kerfoot M, Thompson RCA, White KL, Shackleford DM, Ryan E, Gregg AD, Charman SA, von Geldern TW, Scandale I, Chatelain E. Design, structure-activity relationship and in vivo efficacy of piperazine analogues of fenarimol as inhibitors of Trypanosoma cruzi. Bioorg Med Chem 2013; 21:1756-63. [PMID: 23462713 DOI: 10.1016/j.bmc.2013.01.050] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/16/2013] [Accepted: 01/23/2013] [Indexed: 10/27/2022]
Abstract
A scaffold hopping exercise undertaken to expand the structural diversity of the fenarimol series of anti-Trypanosoma cruzi (T. cruzi) compounds led to preparation of simple 1-[phenyl(pyridin-3-yl)methyl]piperazinyl analogues of fenarimol which were investigated for their ability to inhibit T. cruzi in vitro in a whole organism assay. A range of compounds bearing amide, sulfonamide, carbamate/carbonate and aryl moieties exhibited low nM activities and two analogues were further studied for in vivo efficacy in a mouse model of T. cruzi infection. One compound, the citrate salt of 37, was efficacious in a mouse model of acute T. cruzi infection after once daily oral dosing at 20, 50 and 100 mg/kg for 5 days.
Collapse
Affiliation(s)
- Martine Keenan
- Epichem Pty Ltd, Murdoch University Campus, South Street, Murdoch, Western Australia 6150, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Villalta F, Dobish MC, Nde PN, Kleshchenko YY, Hargrove TY, Johnson CA, Waterman MR, Johnston JN, Lepesheva GI. VNI cures acute and chronic experimental Chagas disease. J Infect Dis 2013; 208:504-11. [PMID: 23372180 DOI: 10.1093/infdis/jit042] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chagas disease is a deadly infection caused by the protozoan parasite Trypanosoma cruzi. Afflicting approximately 8 million people in Latin America, Chagas disease is now becoming a serious global health problem proliferating beyond the traditional geographical borders, mainly because of human and vector migration. Because the disease is endemic in low-resource areas, industrial drug development has been lethargic. The chronic form remains incurable, there are no vaccines, and 2 existing drugs for the acute form are toxic and have low efficacy. Here we report the efficacy of a small molecule, VNI, including evidence of its effectiveness against chronic Chagas disease. VNI is a potent experimental inhibitor of T. cruzi sterol 14α-demethylase. Nontoxic and highly selective, VNI displays promising pharmacokinetics and administered orally to mice at 25 mg/kg for 30 days cures, with 100% cure rate and 100% survival, the acute and chronic T. cruzi infection.
Collapse
Affiliation(s)
- Fernando Villalta
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Nakai K, Kurahashi T, Matsubara S. Synthesis of Quinolones by Nickel-Catalyzed Cycloaddition via Elimination of Nitrile. Org Lett 2013; 15:856-9. [DOI: 10.1021/ol303546p] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kenichiro Nakai
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan, and JST, ACT-C, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Takuya Kurahashi
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan, and JST, ACT-C, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Seijiro Matsubara
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan, and JST, ACT-C, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| |
Collapse
|
81
|
Choy JW, Bryant C, Calvet CM, Doyle PS, Gunatilleke SS, Leung SSF, Ang KKH, Chen S, Gut J, Oses-Prieto JA, Johnston JB, Arkin MR, Burlingame AL, Taunton J, Jacobson MP, McKerrow JM, Podust LM, Renslo AR. Chemical-biological characterization of a cruzain inhibitor reveals a second target and a mammalian off-target. Beilstein J Org Chem 2013; 9:15-25. [PMID: 23400640 PMCID: PMC3566858 DOI: 10.3762/bjoc.9.3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/27/2012] [Indexed: 12/17/2022] Open
Abstract
Inhibition of the Trypanosoma cruzi cysteine protease cruzain has been proposed as a therapeutic approach for the treatment of Chagas’ disease. Among the best-studied cruzain inhibitors to date is the vinylsulfone K777 (1), which has proven effective in animal models of Chagas’ disease. Recent structure–activity studies aimed at addressing potential liabilities of 1 have now produced analogues such as N-[(2S)-1-[[(E,3S)-1-(benzenesulfonyl)-5-phenylpent-1-en-3-yl]amino]-3-(4-methylphenyl)-1-oxopropan-2-yl]pyridine-4-carboxamide (4), which is trypanocidal at ten-fold lower concentrations than for 1. We now find that the trypanocidal activity of 4 derives primarily from the inhibition of T. cruzi 14-α-demethylase (TcCYP51), a cytochrome P450 enzyme involved in the biosynthesis of ergosterol in the parasite. Compound 4 also inhibits mammalian CYP isoforms but is trypanocidal at concentrations below those required to significantly inhibit mammalian CYPs in vitro. A chemical-proteomics approach employing an activity-based probe derived from 1 was used to identify mammalian cathepsin B as a potentially important off-target of 1 and 4. Computational docking studies and the evaluation of truncated analogues of 4 reveal structural determinants for TcCYP51 binding, information that will be useful in further optimization of this new class of inhibitors.
Collapse
Affiliation(s)
- Jonathan W Choy
- Small Molecule Discovery Center, University of California San Francisco, 1700 4th Street, San Francisco, CA, 94158, USA ; Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th Street, San Francisco, CA, 94158, USA ; Department of Cellular and Molecular Pharmacology, University of California San Francisco, 1700 4th Street, San Francisco, CA, 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Recent Developments in Sterol 14-demethylase Inhibitors for Chagas Disease. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2012; 2:236-242. [PMID: 23277882 DOI: 10.1016/j.ijpddr.2011.12.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The protozoan parasite, Trypanosoma cruzi, causes the most prevalent parasitic infection in the American continent. It gives rise to life-long infection in humans and results in severe cardiomyopathy or other life-threatening manifestations (Chagas disease) in ~30% of those infected. Animal models and clinical studies indicate that etiological treatment of the infection reduces the risk of developing the disease manifestations. Unfortunately, the existing chemotherapeutics have suboptimal antiparasitic activity and cause significant side effects in many patients, thus better anti-trypanosomal drugs are greatly needed. The sterol biosynthesis pathway has received attention as a target for the development of new drugs for Chagas disease. In particular, inhibitors of sterol 14-demethylase (CYP51) are shown to be extremely active on Trypanosoma cruzi in vitro and in animal models. Antifungal drugs (i.e. azoles) in clinical use or in clinical studies have been extensively tested preclinically on Trypanosoma cruzi with posaconazole and ravuconazole demonstrating the most promising activity. As a result, posaconazole and a pro-drug of ravuconazole (E1224) are currently being evaluated in Phase II studies for Chagas disease. Additional CYP51 inhibitors that are specifically optimized for anti-Trypanosoma cruzi activity are in development by academia. These represent an alternative to proprietary antifungal drugs if the latter fall short in clinical trials or are too expensive for widespread clinical use in disease endemic countries. The research over the next few years will help define the role of CYP51 inhibitors, alone or in combination with other drugs, for managing patients with Chagas disease.
Collapse
|
83
|
Gunatilleke SS, Calvet CM, Johnston JB, Chen CK, Erenburg G, Gut J, Engel JC, Ang KKH, Mulvaney J, Chen S, Arkin MR, McKerrow JH, Podust LM. Diverse inhibitor chemotypes targeting Trypanosoma cruzi CYP51. PLoS Negl Trop Dis 2012; 6:e1736. [PMID: 22860142 PMCID: PMC3409115 DOI: 10.1371/journal.pntd.0001736] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 06/04/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chagas Disease, a WHO- and NIH-designated neglected tropical disease, is endemic in Latin America and an emerging infection in North America and Europe as a result of population moves. Although a major cause of morbidity and mortality due to heart failure, as well as inflicting a heavy economic burden in affected regions, Chagas Disease elicits scant notice from the pharmaceutical industry because of adverse economic incentives. The discovery and development of new routes to chemotherapy for Chagas Disease is a clear priority. METHODOLOGY/PRINCIPAL FINDINGS The similarity between the membrane sterol requirements of pathogenic fungi and those of the parasitic protozoon Trypanosoma cruzi, the causative agent of Chagas human cardiopathy, has led to repurposing anti-fungal azole inhibitors of sterol 14α-demethylase (CYP51) for the treatment of Chagas Disease. To diversify the therapeutic pipeline of anti-Chagasic drug candidates we exploited an approach that included directly probing the T. cruzi CYP51 active site with a library of synthetic small molecules. Target-based high-throughput screening reduced the library of ∼104,000 small molecules to 185 hits with estimated nanomolar K(D) values, while cross-validation against T. cruzi-infected skeletal myoblast cells yielded 57 active hits with EC(50) <10 µM. Two pools of hits partially overlapped. The top hit inhibited T. cruzi with EC(50) of 17 nM and was trypanocidal at 40 nM. CONCLUSIONS/SIGNIFICANCE The hits are structurally diverse, demonstrating that CYP51 is a rather permissive enzyme target for small molecules. Cheminformatic analysis of the hits suggests that CYP51 pharmacology is similar to that of other cytochromes P450 therapeutic targets, including thromboxane synthase (CYP5), fatty acid ω-hydroxylases (CYP4), 17α-hydroxylase/17,20-lyase (CYP17) and aromatase (CYP19). Surprisingly, strong similarity is suggested to glutaminyl-peptide cyclotransferase, which is unrelated to CYP51 by sequence or structure. Lead compounds developed by pharmaceutical companies against these targets could also be explored for efficacy against T. cruzi.
Collapse
Affiliation(s)
- Shamila S. Gunatilleke
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Claudia M. Calvet
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Cellular Ultra-Structure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonathan B. Johnston
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Chiung-Kuang Chen
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Grigori Erenburg
- King's University College at the University of Western Ontario, London, Ontario, Canada
| | - Jiri Gut
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Juan C. Engel
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Kenny K. H. Ang
- Small Molecule Discovery Center, University of California San Francisco, San Francisco, California, United States of America
| | - Joseph Mulvaney
- Small Molecule Discovery Center, University of California San Francisco, San Francisco, California, United States of America
| | - Steven Chen
- Small Molecule Discovery Center, University of California San Francisco, San Francisco, California, United States of America
| | - Michelle R. Arkin
- Small Molecule Discovery Center, University of California San Francisco, San Francisco, California, United States of America
| | - James H. McKerrow
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Larissa M. Podust
- Sandler Center for Drug Discovery, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
84
|
Pharmacological characterization, structural studies, and in vivo activities of anti-Chagas disease lead compounds derived from tipifarnib. Antimicrob Agents Chemother 2012; 56:4914-21. [PMID: 22777048 DOI: 10.1128/aac.06244-11] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chagas disease, caused by the protozoan pathogen Trypanosoma cruzi, remains a challenging infection due to the unavailability of safe and efficacious drugs. Inhibitors of the trypanosome sterol 14α-demethylase enzyme (CYP51), including azole antifungal drugs, are promising candidates for development as anti-Chagas disease drugs. Posaconazole is under clinical investigation for Chagas disease, although the high cost of this drug may limit its widespread use. We have previously reported that the human protein farnesyltransferase (PFT) inhibitor tipifarnib has potent anti-T. cruzi activity by inhibiting the CYP51 enzyme. Furthermore, we have developed analogs that minimize the PFT-inhibitory activity and enhance the CYP51 inhibition. In this paper, we describe the efficacy of the lead tipifarnib analog compared to that of posaconazole in a murine model of T. cruzi infection. The plasma exposure profiles for each compound following a single oral dose in mice and estimated exposure parameters after repeated twice-daily dosing for 20 days are also presented. The lead tipifarnib analog had potent suppressive activity on parasitemia in mice but was unsuccessful at curing mice, whereas posaconazole as well as benznidazole cured 3 of 5 and 4 of 6 mice, respectively. The efficacy results are consistent with posaconazole having substantially higher predicted exposure than that of the tipifarnib analog after repeat twice-daily administration. Further changes to the tipifarnib analogs to reduce plasma clearance are therefore likely to be important. A crystal structure of a trypanosomal CYP51 bound to a tipifarnib analog is reported here and provides new insights to guide structure-based drug design for further optimized compounds.
Collapse
|
85
|
Alcaide B, Almendros P, Aragoncillo C, Gómez-Campillos G, Arnó M, Domingo LR. Scandium-Catalyzed Preparation of Cytotoxic 3-Functionalized Quinolin-2-ones: Regioselective Ring Enlargement of Isatins or Imino Isatins. Chempluschem 2012. [DOI: 10.1002/cplu.201200090] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
86
|
Berrino R, Cacchi S, Fabrizi G, Goggiamani A. 4-Aryl-2-quinolones from 3,3-Diarylacrylamides through Intramolecular Copper-Catalyzed C–H Functionalization/C–N Bond Formation. J Org Chem 2012; 77:2537-42. [DOI: 10.1021/jo202427m] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Roberta Berrino
- Dipartimento
di Chimica e Tecnologie del Farmaco, La
Sapienza, Università di Roma, P.
le A. Moro 5, 00185 Rome, Italy
| | - Sandro Cacchi
- Dipartimento
di Chimica e Tecnologie del Farmaco, La
Sapienza, Università di Roma, P.
le A. Moro 5, 00185 Rome, Italy
| | - Giancarlo Fabrizi
- Dipartimento
di Chimica e Tecnologie del Farmaco, La
Sapienza, Università di Roma, P.
le A. Moro 5, 00185 Rome, Italy
| | - Antonella Goggiamani
- Dipartimento
di Chimica e Tecnologie del Farmaco, La
Sapienza, Università di Roma, P.
le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
87
|
Experimental chemotherapy and approaches to drug discovery for Trypanosoma cruzi infection. ADVANCES IN PARASITOLOGY 2011; 75:89-119. [PMID: 21820553 DOI: 10.1016/b978-0-12-385863-4.00005-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the 100 years since the discovery of Chagas disease, only two drugs have been developed and introduced into clinical practice, and these drugs were introduced over 40 years ago. The tools of drug discovery have improved dramatically in the interim; however, this has not translated into new drugs for Chagas disease. This has been largely because the main practitioners of drug discovery are pharmaceutical companies who are not financially motivated to invest in Chagas disease and other "orphan" diseases. As a result, it has largely been up to academic groups to bring drug candidates through the discovery pipeline and to clinical trials. The difficulty with drug discovery in academia has been the challenge of bringing together the diverse expertise in biology, chemistry, and pharmacology in concerted efforts towards a common goal of developing therapeutics. Funding is often inadequate, but lack of coordination amongst academic investigators with different expertise has also contributed to the slow progress. The purpose of this chapter is to provide an overview of approaches that can be accomplished in academic settings for preclinical drug discovery for Chagas disease. The chapter addresses methods of drug screening against Trypanosoma cruzi cultures and in animal models and includes general topics on compound selection, testing for drug-like properties (including oral bioavailability), investigating the pharmacokinetics and toxicity of compounds, and finally providing parameters to help with triaging compounds.
Collapse
|
88
|
Lepesheva GI, Villalta F, Waterman MR. Targeting Trypanosoma cruzi sterol 14α-demethylase (CYP51). ADVANCES IN PARASITOLOGY 2011; 75:65-87. [PMID: 21820552 DOI: 10.1016/b978-0-12-385863-4.00004-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There are at least two obvious features that must be considered upon targeting specific metabolic pathways/enzymes for drug development: the pathway must be essential and the enzyme must allow the design of pharmacologically useful inhibitors. Here, we describe Trypanosoma cruzi sterol 14α-demethylase as a promising target for anti-Chagasic chemotherapy. The use of anti-fungal azoles, which block sterol biosynthesis and therefore membrane formation in fungi, against the protozoan parasite has turned out to be highly successful: a broad spectrum anti-fungal drug, the triazole compound posaconazole, is now entering phase II clinical trials for treatment of Chagas disease. This review summarizes comparative information on anti-fungal azoles and novel inhibitory scaffolds selective for Trypanosomatidae sterol 14α-demethylase through the lens of recent structure/functional characterization of the target enzyme. We believe our studies open wide opportunities for rational design of novel, pathogen-specific and therefore more potent and efficient anti-trypanosomal drugs.
Collapse
Affiliation(s)
- Galina I Lepesheva
- Department of Biochemistry School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | | | | |
Collapse
|
89
|
Muscia GC, Cazorla SI, Frank FM, Borosky GL, Buldain GY, Asís SE, Malchiodi EL. Synthesis, trypanocidal activity and molecular modeling studies of 2-alkylaminomethylquinoline derivatives. Eur J Med Chem 2011; 46:3696-703. [PMID: 21664012 DOI: 10.1016/j.ejmech.2011.05.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 04/19/2011] [Accepted: 05/13/2011] [Indexed: 01/15/2023]
Abstract
Research and development of new drugs effective in the treatment of Trypanosoma cruzi infections are a real need for the 16 million people infected in the Americas. In a previous work, a quinoline derivative substituted by a 2-piperidylmethyl moiety showed to be active against Chagas disease and was considered a lead compound for further optimization. A series of ten analogous derivatives were tested against epimastigotes as a first approach. In view of their promising results, six of them were evaluated against the blood and intracellular replicative forms of the parasite in humans. Among them, compound 12 which possesses a 6-acetamidohexylamino substituent showed remarkable improvement in activity against epimastigotes, trypomastigotes and amastigotes compared with the structure lead, as well as a good selectivity index for the two parasite stages present in humans. In addition, treatment of infected mice with compound 12 induced a significant reduction in parasitemia compared with non-treated mice. Molecular modeling studies were performed by computational methods in order to elucidate the factors determining these experimental bioactivities.
Collapse
Affiliation(s)
- Gisela C Muscia
- Departamento de Química Orgánica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
90
|
Alcaide B, Almendros P, Luna A, Cembellín S, Arnó M, Domingo LR. Controlled Rearrangement of Lactam-Tethered Allenols with Brominating Reagents: A Combined Experimental and Theoretical Study on α- versus β-Keto Lactam Formation. Chemistry 2011; 17:11559-66. [DOI: 10.1002/chem.201101160] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Indexed: 11/06/2022]
|
91
|
Shi XL, Ge JF, Liu BQ, Kaiser M, Wittlin S, Brun R, Ihara M. Synthesis and in vitro antiprotozoal activities of 5-phenyliminobenzo[a]phenoxazine derivatives. Bioorg Med Chem Lett 2011; 21:5804-7. [PMID: 21868222 DOI: 10.1016/j.bmcl.2011.07.112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 07/27/2011] [Accepted: 07/29/2011] [Indexed: 11/28/2022]
Abstract
A series of 5-phenyliminobenzo[a]phenoxazine derivatives were synthesized. The in vitro antiprotozoal activities were evaluated against Plasmodium falciparum K1, Trypanosoma cruzi, Leishmania donovani and Trypanosoma brucei rhodesiense. N,N-Diethyl-5-((4-methoxyphenyl)imino)-5H-benzo[a]phenoxazin-9-amine shows IC(50)=0.040 μmol L(-1) with a selective index of 1425 against Plasmodium falciparum K1.
Collapse
Affiliation(s)
- Xue-Liang Shi
- Key Laboratory of Organic Synthesis of Jiangsu Province, Soochow University, 199 Ren'Ai Road, Suzhou 215123, China
| | | | | | | | | | | | | |
Collapse
|
92
|
Henderson WR, Oslund RC, Bollinger JG, Ye X, Tien YT, Xue J, Gelb MH. Blockade of human group X secreted phospholipase A2 (GX-sPLA2)-induced airway inflammation and hyperresponsiveness in a mouse asthma model by a selective GX-sPLA2 inhibitor. J Biol Chem 2011; 286:28049-55. [PMID: 21652694 DOI: 10.1074/jbc.m111.235812] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Group X (GX) phospholipase A(2), a member of a large group of secreted phospholipases A(2) (sPLA(2)s), has recently been demonstrated to play an important in vivo role in the release of arachidonic acid and subsequent formation of eicosanoids. In a Th2 cytokine-driven mouse asthma model, deficiency of mouse GX (mGX)-sPLA(2) significantly impairs development of the asthma phenotype. In this study, we generated mGX-sPLA(2)(-/-) mice with knock-in of human GX (hGX)-sPLA(2) (i.e. hGX-sPLA(2)(+/+) knock-in mice) to understand more fully the role of GX-sPLA(2) in these allergic pulmonary responses and to assess the effect of pharmacological blockade of the GX-sPLA(2)-mediated responses. Knock-in of hGX-sPLA(2) in mGX-sPLA(2)(-/-) mice restored the allergen-induced airway infiltration by inflammatory cells, including eosinophils, goblet cell metaplasia, and hyperresponsiveness to methacholine in the mGX-sPLA(2)-deficient mice. This knock-in mouse model enabled the use of a highly potent indole-based inhibitor of hGX-sPLA(2), RO061606 (which is ineffective against mGX-sPLA(2)), to assess the potential utility of GX-sPLA(2) blockade as a therapeutic intervention in asthma. Delivery of RO061606 via mini-osmotic pumps enabled the maintenance in vivo in the mouse asthma model of plasma inhibitor concentrations near 10 μm, markedly higher than the IC(50) for inhibition of hGX-sPLA(2) in vitro. RO061606 significantly decreased allergen-induced airway inflammation, mucus hypersecretion, and hyperresponsiveness in the hGX-sPLA(2)(+/+) knock-in mouse. Thus, development of specific hGX-sPLA(2) inhibitors may provide a new pharmacological opportunity for the treatment of patients with asthma.
Collapse
Affiliation(s)
- William R Henderson
- Center for Allergy and Inflammation, Department of Medicine, University of Washington, Seattle, Washington 98195-1700, USA
| | | | | | | | | | | | | |
Collapse
|
93
|
Psilostachyin C: a natural compound with trypanocidal activity. Int J Antimicrob Agents 2011; 37:536-43. [DOI: 10.1016/j.ijantimicag.2011.02.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/27/2011] [Accepted: 02/02/2011] [Indexed: 12/27/2022]
|
94
|
Abstract
PURPOSE OF REVIEW The need for better drugs to treat patients with Chagas disease remains urgent. This review summarizes the advancements in drug development over the past 2 years. RECENT FINDINGS Drug development efforts are almost exclusively occurring as preclinical research, although phase II studies for the antifungal drug, posaconazole, and a prodrug of ravuconazole are being planned. Several recent laboratory investigations demonstrate anti-Trypanosoma cruzi activity of novel small molecules in animal models. These include nonpeptidic cruzain inhibitors, novel inhibitors of the sterol 14α-demethylase enzyme, new compounds (arylimidamides) related to pentamidine, derivatives of nifurtimox, compounds using ruthenium complexes, and several natural products. The recent implementation of a high-throughput screen of more than 300 000 compounds against intracellular T. cruzi amastigotes done at the Broad Institute is an important development, yielding approximately 300 selective inhibitors, many of which may serve as leads for medicinal chemistry efforts. SUMMARY Progress is slow, but recent advancements in both drug development and advocacy for research on neglected diseases are encouraging. Efforts to define a target product profile and to harmonize methodologies for testing drugs for Chagas disease are described herein.
Collapse
|
95
|
Zucca M, Savoia D. Current developments in the therapy of protozoan infections. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2011; 5:4-10. [PMID: 21629507 PMCID: PMC3103884 DOI: 10.2174/1874104501105010004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 05/25/2010] [Accepted: 06/20/2010] [Indexed: 12/03/2022]
Abstract
Protozoan parasites cause serious human and zoonotic infections, including life-threatening diseases such as malaria, African and American trypanosomiasis, and leishmaniasis. These diseases are no more common in the developed world, but together they still threaten about 40% of the world population (WHO estimates). Mortality and morbidity are high in developing countries, and the lack of vaccines makes chemotherapy the only suitable option. However, available antiparasitic drugs are hampered by more or less marked toxic side effects and by the emergence of drug resistance. As the main prevalence of parasitic diseases occurs in the poorest areas of the world, the interest of the pharmaceutical companies in the development of new drugs has been traditionally scarce. The establishment of public-private partnerships focused on tropical diseases is changing this situation, allowing the exploitation of the technological advances that took place during the past decade related to genomics, proteomics, and in silico drug discovery approaches. These techniques allowed the identification of new molecular targets that in some cases are shared by different parasites. In this review we outline the recent developments in the fields of protease and topoisomerase inhibitors, antimicrobial and cell-penetrating peptides, and RNA interference. We also report on the rapidly developing field of new vectors (micro and nano particles, mesoporous materials) that in some cases can cross host or parasite natural barriers and, by selectively delivering new or already in use drugs to the target site, minimize dosage and side effects.
Collapse
Affiliation(s)
- Mario Zucca
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | | |
Collapse
|
96
|
Tang BX, Song RJ, Wu CY, Wang ZQ, Liu Y, Huang XC, Xie YX, Li JH. Ruthenium-catalyzed intramolecular carbocyclization of alkynes with an sp3 carbon involving an oxidative deprotonation process. Chem Sci 2011. [DOI: 10.1039/c1sc00423a] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
97
|
|
98
|
Jacobs RT, Nare B, Phillips MA. State of the art in African trypanosome drug discovery. Curr Top Med Chem 2011; 11:1255-74. [PMID: 21401507 PMCID: PMC3101707 DOI: 10.2174/156802611795429167] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 11/25/2010] [Indexed: 11/22/2022]
Abstract
African sleeping sickness is endemic in sub-Saharan Africa where the WHO estimates that 60 million people are at risk for the disease. Human African trypanosomiasis (HAT) is 100% fatal if untreated and the current drug therapies have significant limitations due to toxicity and difficult treatment regimes. No new chemical agents have been approved since eflornithine in 1990. The pentamidine analog DB289, which was in late stage clinical trials for the treatment of early stage HAT recently failed due to toxicity issues. A new protocol for the treatment of late-stage T. brucei gambiense that uses combination nifurtomox/eflornithine (NECT) was recently shown to have better safety and efficacy than eflornithine alone, while being easier to administer. This breakthrough represents the only new therapy for HAT since the approval of eflornithine. A number of research programs are on going to exploit the unusual biochemical pathways in the parasite to identify new targets for target based drug discovery programs. HTS efforts are also underway to discover new chemical entities through whole organism screening approaches. A number of inhibitors with anti-trypanosomal activity have been identified by both approaches, but none of the programs are yet at the stage of identifying a preclinical candidate. This dire situation underscores the need for continued effort to identify new chemical agents for the treatment of HAT.
Collapse
Affiliation(s)
- Robert T. Jacobs
- SCYNEXIS, Inc., Research Triangle Park, North Carolina 27709-2878
| | - Bakela Nare
- SCYNEXIS, Inc., Research Triangle Park, North Carolina 27709-2878
| | - Margaret A. Phillips
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park Rd, Dallas, Texas 75390-9041
| |
Collapse
|
99
|
Mansfield BE, Oltean HN, Oliver BG, Hoot SJ, Leyde SE, Hedstrom L, White TC. Azole drugs are imported by facilitated diffusion in Candida albicans and other pathogenic fungi. PLoS Pathog 2010; 6:e1001126. [PMID: 20941354 PMCID: PMC2947996 DOI: 10.1371/journal.ppat.1001126] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 09/01/2010] [Indexed: 12/21/2022] Open
Abstract
Despite the wealth of knowledge regarding the mechanisms of action and the mechanisms of resistance to azole antifungals, very little is known about how the azoles are imported into pathogenic fungal cells. Here the in-vitro accumulation and import of Fluconazole (FLC) was examined in the pathogenic fungus, Candida albicans. In energized cells, FLC accumulation correlates inversely with expression of ATP-dependent efflux pumps. In de-energized cells, all strains accumulate FLC, suggesting that FLC import is not ATP-dependent. The kinetics of import in de-energized cells displays saturation kinetics with a Km of 0.64 uM and Vmax of 0.0056 pmol/min/108 cells, demonstrating that FLC import proceeds via facilitated diffusion through a transporter rather than passive diffusion. Other azoles inhibit FLC import on a mole/mole basis, suggesting that all azoles utilize the same facilitated diffusion mechanism. An analysis of related compounds indicates that competition for azole import depends on an aromatic ring and an imidazole or triazole ring together in one molecule. Import of FLC by facilitated diffusion is observed in other fungi, including Cryptococcus neoformans, Saccharomyces cerevisiae, and Candida krusei, indicating that the mechanism of transport is conserved among fungal species. FLC import was shown to vary among Candida albicans resistant clinical isolates, suggesting that altered facilitated diffusion may be a previously uncharacterized mechanism of resistance to azole drugs. Azole antifungals are used to treat a wide variety of fungal infections of humans, animals and plants. A great deal is known about how the azoles interact with their target enzyme within fungal cells and how the azoles are exported from the fungal cell through various efflux pumps. Altered interactions with the target enzyme and altered efflux pump expression are common mechanisms of azole resistance in fungi. However, the mechanism by which azoles enter a fungal cell is not clear—many have assumed that azoles passively diffuse into the cell. This study demonstrates that azoles are not passively diffused, or actively pumped, into the cell. Instead, azoles are imported by facilitated diffusion, mediated by a transporter. Facilitated diffusion of azoles is saturable. All clinically important azoles, and many structurally related compounds, compete for FLC import, while structurally unrelated drugs do not compete. Azole import by facilitated diffusion is shown in four species of fungi, suggesting that it is common for most if not all fungi. Altered facilitated diffusion is observed in a collection of clinical isolates, suggesting that altered import is a previously uncharacterized mechanism of resistance.
Collapse
Affiliation(s)
- Bryce E. Mansfield
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Hanna N. Oltean
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Brian G. Oliver
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Samantha J. Hoot
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Sarah E. Leyde
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Lizbeth Hedstrom
- Brandeis University Department of Biology and Chemistry, Waltham, Massachusetts, United States of America
| | - Theodore C. White
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- University of Washington Program in Pathobiology, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
100
|
Nohara LL, Lema C, Bader JO, Aguilera RJ, Almeida IC. High-content imaging for automated determination of host-cell infection rate by the intracellular parasite Trypanosoma cruzi. Parasitol Int 2010; 59:565-70. [PMID: 20688189 DOI: 10.1016/j.parint.2010.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Revised: 07/17/2010] [Accepted: 07/24/2010] [Indexed: 10/19/2022]
Abstract
Chagas disease affects 8-11 million people, mostly in Latin America. Sequelae include cardiac, peripheral nervous and/or gastrointestinal disorders, thus placing a large economic and social burden on endemic countries. The pathogenesis and the evolutive pattern of the disease are not fully clarified. Moreover, available drugs are partially effective and toxic, and there is no vaccine. Therefore, there is an urgent need to speed up basic and translational research in the field. Here, we applied automated high-content imaging to generate multiparametric data on a cell-by-cell basis to precisely and quickly determine several parameters associated with in vitro infection of host cell by Trypanosoma cruzi, the causative agent of Chagas disease. Automated and manual quantifications were used to determine the percentage of T. cruzi-infected cells in a 96-well microplate format and the data generated was statistically evaluated. Most importantly, this automated approach can be widely applied for discovery of potential drugs as well as molecular pathway elucidation not only in T. cruzi but also in other human intracellular pathogens.
Collapse
Affiliation(s)
- L L Nohara
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | | | | | | | | |
Collapse
|