1
|
Ong HW, Adderley J, Tobin AB, Drewry DH, Doerig C. Parasite and host kinases as targets for antimalarials. Expert Opin Ther Targets 2023; 27:151-169. [PMID: 36942408 DOI: 10.1080/14728222.2023.2185511] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION The deployment of Artemisinin-based combination therapies and transmission control measures led to a decrease in the global malaria burden over the recent decades. Unfortunately, this trend is now reversing, in part due to resistance against available treatments, calling for the development of new drugs against untapped targets to prevent cross-resistance. AREAS COVERED In view of their demonstrated druggability in noninfectious diseases, protein kinases represent attractive targets. Kinase-focussed antimalarial drug discovery is facilitated by the availability of kinase-targeting scaffolds and large libraries of inhibitors, as well as high-throughput phenotypic and biochemical assays. We present an overview of validated Plasmodium kinase targets and their inhibitors, and briefly discuss the potential of host cell kinases as targets for host-directed therapy. EXPERT OPINION We propose priority research areas, including (i) diversification of Plasmodium kinase targets (at present most efforts focus on a very small number of targets); (ii) polypharmacology as an avenue to limit resistance (kinase inhibitors are highly suitable in this respect); and (iii) preemptive limitation of resistance through host-directed therapy (targeting host cell kinases that are required for parasite survival) and transmission-blocking through targeting sexual stage-specific kinases as a strategy to protect curative drugs from the spread of resistance.
Collapse
Affiliation(s)
- Han Wee Ong
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC USA
| | - Jack Adderley
- Department of Laboratory Medicine, School of Health and Biomedical Sciences, Rmit University, Bundoora VIC Australia
| | - Andrew B Tobin
- Advanced Research Centre, University of Glasgow, Glasgow, UK
| | - David H Drewry
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC USA
| | - Christian Doerig
- Department of Laboratory Medicine, School of Health and Biomedical Sciences, Rmit University, Bundoora VIC Australia
| |
Collapse
|
2
|
Pal K, Raza MK, Legac J, Rahman A, Manzoor S, Bhattacharjee S, Rosenthal PJ, Hoda N. Identification, in-vitro anti-plasmodial assessment and docking studies of series of tetrahydrobenzothieno[2,3-d]pyrimidine-acetamide molecular hybrids as potential antimalarial agents. Eur J Med Chem 2023; 248:115055. [PMID: 36621136 DOI: 10.1016/j.ejmech.2022.115055] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022]
Abstract
Malaria is the most lethal parasitic infections in the world. To address the emergence of drug resistance to current antimalarials, here we report the design and synthesis of new series of tetrahydrobenzothieno[2,3-d]pyrimidine-acetamide hybrids by using multicomponent Petasis reaction as the key step and evaluated in vitro for their antimalarial effectiveness. The structure of all the compounds were confirmed by NMR Spectroscopy and mass spectrometry. Most of the compounds showed potent antimalarial activity against both CQ-sensitive (3D7) and CQ-resistant (W2) strains. A8, A5, and A4 are the most potent compounds that showed excellent anti-plasmodial activity against CQ-resistant strain in the nanomolar range with IC50 values 55.7 nM, 60.8 nM, and 68.0 nM respectively. To assess the parasite selectivity, the in vitro cytotoxicity of selected compounds (A3-A6, A8) was tested against HPL1D cells, demonstrating low cytotoxicity with high selectivity indices. Furthermore, these compounds were also evaluated on two additional human cancerous cell lines (A549 and MDA-MB-231), confirming their anticancer effectiveness. The in vitro hemolysis assay also showed the non-toxicity of these compounds on normal uninfected human RBCs. The interaction of these hybrids was also investigated by the molecular docking studies in the binding site of wild type Pf-DHFR-TS and quadruple mutant Pf-DHFR-TS. The in silico ADMET profiling also revealed promising physicochemical and pharmacokinetic parameters for the most active hybrids, which provide strong vision for further development of potential antimalarials.
Collapse
Affiliation(s)
- Kavita Pal
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Jenny Legac
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Abdur Rahman
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Souvik Bhattacharjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
3
|
Lagardère P, Fersing C, Masurier N, Lisowski V. Thienopyrimidine: A Promising Scaffold to Access Anti-Infective Agents. Pharmaceuticals (Basel) 2021; 15:35. [PMID: 35056092 PMCID: PMC8780093 DOI: 10.3390/ph15010035] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/16/2022] Open
Abstract
Thienopyrimidines are widely represented in the literature, mainly due to their structural relationship with purine base such as adenine and guanine. This current review presents three isomers-thieno[2,3-d]pyrimidines, thieno[3,2-d]pyrimidines and thieno[3,4-d]pyrimidines-and their anti-infective properties. Broad-spectrum thienopyrimidines with biological properties such as antibacterial, antifungal, antiparasitic and antiviral inspired us to analyze and compile their structure-activity relationship (SAR) and classify their synthetic pathways. This review explains the main access route to synthesize thienopyrimidines from thiophene derivatives or from pyrimidine analogs. In addition, SAR study and promising anti-infective activity of these scaffolds are summarized in figures and explanatory diagrams. Ligand-receptor interactions were modeled when the biological target was identified and the crystal structure was solved.
Collapse
Affiliation(s)
- Prisca Lagardère
- IBMM, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (P.L.); (C.F.); (V.L.)
| | - Cyril Fersing
- IBMM, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (P.L.); (C.F.); (V.L.)
- Nuclear Medicine Department, Montpellier Cancer Institute (ICM), University of Montpellier, 208 Avenue des Apothicaires, CEDEX 5, 34298 Montpellier, France
| | - Nicolas Masurier
- IBMM, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (P.L.); (C.F.); (V.L.)
| | - Vincent Lisowski
- IBMM, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (P.L.); (C.F.); (V.L.)
- Department of Pharmacy, Lapeyronie Hospital, CHU Montpellier, 191 Av. du Doyen Gaston Giraud, 34295 Montpellier, France
| |
Collapse
|
4
|
Pal K, Raza MK, Legac J, Ataur Rahman M, Manzoor S, Rosenthal PJ, Hoda N. Design, synthesis, crystal structure and anti-plasmodial evaluation of tetrahydrobenzo[4,5]thieno[2,3- d]pyrimidine derivatives. RSC Med Chem 2021; 12:970-981. [PMID: 34223162 DOI: 10.1039/d1md00038a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/26/2021] [Indexed: 12/26/2022] Open
Abstract
Effective chemotherapy is essential for controlling malaria. However, resistance of Plasmodium falciparum to existing antimalarial drugs has undermined attempts to control and eventually eradicate the disease. In this study, a series of 2-((substituted)(4-(5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4-yl)piperazin-1-yl)methyl)-6-substitutedphenol derivatives were prepared using Petasis reaction with a view to evaluate their activities against P. falciparum. The development of synthesized compounds (F1-F16) was justified through the study of H1 NMR, C13 NMR, mass spectra. Compound F1 and F2 were also structurally validated by single crystal X-ray diffraction analysis. All the compounds were evaluated for their in vitro antiplasmodial assessment against the W2 strain (chloroquine-resistant) of P. falciparum IC50 values ranging from 0.74-6.4 μM. Two compounds, F4 and F16 exhibited significant activity against W2 strain of P. falciparum with 0.75 and 0.74 μM. The compounds (F3-F6 and F16) were also evaluated for in vitro cytotoxicity against two cancer cell lines, human lung (A549) and cervical (HeLa) cells, which demonstrated non-cytotoxicity with significant selectivity indices. In addition, in silico ADME profiling and physiochemical properties predicts drug-like properties with a very low toxic effect. Thus, all these results indicate that tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidine scaffolds may serve as models for the development of antimalarial agents.
Collapse
Affiliation(s)
- Kavita Pal
- Drug Design and Synthesis Laboratory, Department of chemistry, Jamia Millia Islamia New Delhi 110025 India +91 11 26985507 +91 9910200655
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| | - Jenny Legac
- Department of Medicine, University of California San Francisco CA USA
| | - Md Ataur Rahman
- Department of Chemistry and Chemical Biology, Harvard University Cambridge Massachusetts 02138 USA
| | - Shoaib Manzoor
- Drug Design and Synthesis Laboratory, Department of chemistry, Jamia Millia Islamia New Delhi 110025 India +91 11 26985507 +91 9910200655
| | | | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of chemistry, Jamia Millia Islamia New Delhi 110025 India +91 11 26985507 +91 9910200655
| |
Collapse
|
5
|
Yan W, Zhang L, Lv F, Moccia M, Carlomagno F, Landry C, Santoro M, Gosselet F, Frett B, Li HY. Discovery of pyrazolo-thieno[3,2-d]pyrimidinylamino-phenyl acetamides as type-II pan-tropomyosin receptor kinase (TRK) inhibitors: Design, synthesis, and biological evaluation. Eur J Med Chem 2021; 216:113265. [PMID: 33652352 DOI: 10.1016/j.ejmech.2021.113265] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/21/2022]
Abstract
Tropomyosin receptor kinase (TRK) represents an attractive oncology target for cancer therapy related to its critical role in cancer formation and progression. NTRK fusions are found to occur in 3.3% of lung cancers, 2.2% of colorectal cancers, 16.7% of thyroid cancers, 2.5% of glioblastomas, and 7.1% of pediatric gliomas. In this paper, we described the discovery of the type-II pan-TRK inhibitor 4c through the structure-based drug design strategy from the original hits 1b and 2b. Compound 4c exhibited excellent in vitro TRKA, TRKB, and TRKC kinase inhibitory activity and anti-proliferative activity against human colorectal carcinoma derived cell line KM12. In the NCI-60 human cancer cell lines screen, compound 4g demonstrated nearly 80% of growth inhibition for KM12, while only minimal inhibitory activity was observed for the remaining 59 cancer cell lines. Western blot analysis demonstrated that 4c and its urea cousin 4k suppressed the TPM3-TRKA autophosphorylation at the concentrations of 100 nM and 10 nM, respectively. The work presented that 2-(4-(thieno[3,2-d]pyrimidin-4-ylamino)phenyl)acetamides could serve as a novel scaffold for the discovery and development of type-II pan-TRK inhibitors for the treatment of TRK driven cancers.
Collapse
Affiliation(s)
- Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Fengping Lv
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy; Istituto di Endocrinologia e Oncologia Sperimentale Del CNR, Via S Pansini 5, 80131, Naples, Italy
| | - Christophe Landry
- Blood Brain Barrier Laboratory (LBHE), University of Artois, UR2465, F-62300, Lens, France
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131, Naples, Italy
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory (LBHE), University of Artois, UR2465, F-62300, Lens, France
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
6
|
Borba JVVB, Silva AC, Lima MNN, Mendonca SS, Furnham N, Costa FTM, Andrade CH. Chemogenomics and bioinformatics approaches for prioritizing kinases as drug targets for neglected tropical diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 124:187-223. [PMID: 33632465 DOI: 10.1016/bs.apcsb.2020.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neglected tropical diseases (NTDs) are a group of twenty-one diseases classified by the World Health Organization that prevail in regions with tropical and subtropical climate and affect more than one billion people. There is an urgent need to develop new and safer drugs for these diseases. Protein kinases are a potential class of targets for developing new drugs against NTDs, since they play crucial role in many biological processes, such as signaling pathways, regulating cellular communication, division, metabolism and death. Bioinformatics is a field that aims to organize large amounts of biological data as well as develop and use tools for understanding and analyze them in order to produce meaningful information in a biological manner. In combination with chemogenomics, which analyzes chemical-biological interactions to screen ligands against selected targets families, these approaches can be used to stablish a rational strategy for prioritizing new drug targets for NTDs. Here, we describe how bioinformatics and chemogenomics tools can help to identify protein kinases and their potential inhibitors for the development of new drugs for NTDs. We present a review of bioinformatics tools and techniques that can be used to define an organisms kinome for drug prioritization, drug and target repurposing, multi-quinase inhibition approachs and selectivity profiling. We also present some successful examples of the application of such approaches in recent case studies.
Collapse
Affiliation(s)
- Joyce Villa Verde Bastos Borba
- LabMol-Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil; Laboratory of Tropical Diseases-Prof. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, University of Campinas, Campinas, SP, Brazil
| | - Arthur Carvalho Silva
- LabMol-Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Marilia Nunes Nascimento Lima
- LabMol-Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Sabrina Silva Mendonca
- LabMol-Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Nicholas Furnham
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fabio Trindade Maranhão Costa
- Laboratory of Tropical Diseases-Prof. Luiz Jacintho da Silva, Department of Genetics, Evolution and Bioagents, University of Campinas, Campinas, SP, Brazil
| | - Carolina Horta Andrade
- LabMol-Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil; Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
7
|
Ahmed M, Younis O, Orabi EA, Sayed AM, Kamal El-Dean AM, Hassanien R, Davis RL, Tsutsumi O, Tolba MS. Synthesis of Novel Biocompatible Thienopyrimidine Chromophores with Aggregation-Induced Emission Sensitive to Molecular Aggregation. ACS OMEGA 2020; 5:29988-30000. [PMID: 33251435 PMCID: PMC7689934 DOI: 10.1021/acsomega.0c04358] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 05/08/2023]
Abstract
Biocompatible luminogens with aggregation-induced emission (AIE) have several applications in the biology field, such as in detecting biomacromolecules bioprobes and in bio-imaging. Due to their bioactivities and light-emitting properties, many heterocyclic compounds are good candidates for such applications. However, heterocyclic π-conjugated systems with AIE behavior remain rare as strong intermolecular π-π interactions usually quench their emission. In this work, new thienopyrimidine heterocyclic compounds were synthesized and their structures were verified by elemental analysis and Fourier transform infrared (FT-IR), 1H nuclear magnetic resonance (NMR), and 13C NMR spectra. The photophysical properties of some compounds were investigated in the solution and solid states. Density functional theory calculations were also performed to confirm the observed photophysical properties of the compounds. The studied dyes displayed AIE properties with spectral shapes related to the aggregate structure and a quantum yield up to 10.8%. The emission efficiency of the powder is attributed to the incorporation of multiply rotatable and twisted aryl groups to the fused heterocyclic moieties. The dyes also showed high thermal stability and potent antimicrobial activities against numerous bacterial and fungal strains. Additionally, the cytotoxicity of the new compounds was evaluated against the Caco-2 cell line, and molecular docking was used to investigate the binding conformation of the most effective compound with the MNK2 enzyme. Therefore, the presented structures may potentially be used for bioapplications.
Collapse
Affiliation(s)
- Mostafa Ahmed
- Chemistry Department,
Faculty of Science, New Valley University, El-Kharja 72511, Egypt
| | - Osama Younis
- Chemistry Department,
Faculty of Science, New Valley University, El-Kharja 72511, Egypt
- Department of Applied Chemistry, College
of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Esam A. Orabi
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
- Department of Chemistry, University of
Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ahmed M. Sayed
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | | | - Reda Hassanien
- Chemistry Department,
Faculty of Science, New Valley University, El-Kharja 72511, Egypt
| | - Rebecca L. Davis
- Department of Chemistry, University of
Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Osamu Tsutsumi
- Department of Applied Chemistry, College
of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Mahmoud S. Tolba
- Chemistry Department,
Faculty of Science, New Valley University, El-Kharja 72511, Egypt
| |
Collapse
|
8
|
Kesely K, Noomuna P, Vieth M, Hipskind P, Haldar K, Pantaleo A, Turrini F, Low PS. Identification of tyrosine kinase inhibitors that halt Plasmodium falciparum parasitemia. PLoS One 2020; 15:e0242372. [PMID: 33180822 PMCID: PMC7660480 DOI: 10.1371/journal.pone.0242372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/31/2020] [Indexed: 01/23/2023] Open
Abstract
Although current malaria therapies inhibit pathways encoded in the parasite’s genome, we have looked for anti-malaria drugs that can target an erythrocyte component because development of drug resistance might be suppressed if the parasite cannot mutate the drug’s target. In search for such erythrocyte targets, we noted that human erythrocytes express tyrosine kinases, whereas the Plasmodium falciparum genome encodes no obvious tyrosine kinases. We therefore screened a library of tyrosine kinase inhibitors from Eli Lilly and Co. in a search for inhibitors with possible antimalarial activity. We report that although most tyrosine kinase inhibitors exerted no effect on parasite survival, a subset of tyrosine kinase inhibitors displayed potent anti-malarial activity. Moreover, all inhibitors found to block tyrosine phosphorylation of band 3 specifically suppressed P. falciparum survival at the parasite egress stage of its intra-erythrocyte life cycle. Conversely, tyrosine kinase inhibitors that failed to block band 3 tyrosine phosphorylation but still terminated the parasitemia were observed to halt parasite proliferation at other stages of the parasite’s life cycle. Taken together these results suggest that certain erythrocyte tyrosine kinases may be important to P. falciparum maturation and that inhibitors that block these kinases may contribute to novel therapies for P. falciparum malaria.
Collapse
Affiliation(s)
- Kristina Kesely
- Department of Chemistry, Purdue University, West Lafayette, IN, United States of America.,Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States of America
| | - Panae Noomuna
- Department of Chemistry, Purdue University, West Lafayette, IN, United States of America.,Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States of America
| | - Michal Vieth
- Eli Lilly and Company, San Diego, CA, United States of America
| | - Philip Hipskind
- School of Medicine, Indiana University, Bloomington, IN, United States of America.,Clinical Pharmacology R2 402 MDEP, Indianapolis, IN, United States of America
| | - Kasturi Haldar
- Galvin Life Science Center, University of Notre Dame, Notre Dame, IN, United States of America
| | | | | | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, United States of America.,Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
9
|
Younis O, Tolba MS, Orabi EA, Kamal AM, Hassanien R, Tsutsumi O, Ahmed M. Biologically-Active Heterocyclic Molecules with Aggregation-Induced Blue-Shifted Emission and Efficient Luminescence both in Solution and Solid States. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2020.112642] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Abstract
Parasitic infections are responsible for significant morbidity and mortality throughout the world. Management strategies rely primarily on antiparasitic drugs that have side effects and risk of drug resistance. Therefore, novel strategies are needed for treatment of parasitic infections. Host-directed therapy (HDT) is a viable alternative, which targets host pathways responsible for parasite invasion/survival/pathogenicity. Recent innovative combinations of genomics, proteomics and computational biology approaches have led to discovery of several host pathways that could be promising targets for HDT for treating parasitic infections. Herein, we review major advances in HDT for parasitic disease with regard to core regulatory pathways and their interactions.
Collapse
|
11
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
12
|
Senerovic L, Opsenica D, Moric I, Aleksic I, Spasić M, Vasiljevic B. Quinolines and Quinolones as Antibacterial, Antifungal, Anti-virulence, Antiviral and Anti-parasitic Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1282:37-69. [PMID: 31515709 DOI: 10.1007/5584_2019_428] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Infective diseases have become health threat of a global proportion due to appearance and spread of microorganisms resistant to majority of therapeutics currently used for their treatment. Therefore, there is a constant need for development of new antimicrobial agents, as well as novel therapeutic strategies. Quinolines and quinolones, isolated from plants, animals, and microorganisms, have demonstrated numerous biological activities such as antimicrobial, insecticidal, anti-inflammatory, antiplatelet, and antitumor. For more than two centuries quinoline/quinolone moiety has been used as a scaffold for drug development and even today it represents an inexhaustible inspiration for design and development of novel semi-synthetic or synthetic agents exhibiting broad spectrum of bioactivities. The structural diversity of synthetized compounds provides high and selective activity attained through different mechanisms of action, as well as low toxicity on human cells. This review describes quinoline and quinolone derivatives with antibacterial, antifungal, anti-virulent, antiviral, and anti-parasitic activities with the focus on the last 10 years literature.
Collapse
Affiliation(s)
- Lidija Senerovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.
| | - Dejan Opsenica
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Center of excellence in Environmental Chemistry and Engineering, ICTM - University of Belgrade, Belgrade, Serbia
| | - Ivana Moric
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Aleksic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marta Spasić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Branka Vasiljevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
13
|
Varikuti S, Jha BK, Volpedo G, Ryan NM, Halsey G, Hamza OM, McGwire BS, Satoskar AR. Host-Directed Drug Therapies for Neglected Tropical Diseases Caused by Protozoan Parasites. Front Microbiol 2018; 9:2655. [PMID: 30555425 PMCID: PMC6284052 DOI: 10.3389/fmicb.2018.02655] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
The neglected tropical diseases (NTDs) caused by protozoan parasites are responsible for significant morbidity and mortality worldwide. Current treatments using anti-parasitic drugs are toxic and prolonged with poor patient compliance. In addition, emergence of drug-resistant parasites is increasing worldwide. Hence, there is a need for safer and better therapeutics for these infections. Host-directed therapy using drugs that target host pathways required for pathogen survival or its clearance is a promising approach for treating infections. This review will give a summary of the current status and advances of host-targeted therapies for treating NTDs caused by protozoa.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Greta Volpedo
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Nathan M Ryan
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Gregory Halsey
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Omar M Hamza
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Bradford S McGwire
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Abhay R Satoskar
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
14
|
Synthesis, antitrypanosomal activity and molecular docking studies of pyrimidine derivatives. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2253-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
15
|
Woodring J, Behera R, Sharma A, Wiedeman J, Patel G, Singh B, Guyett P, Amata E, Erath J, Roncal N, Penn E, Leed SE, Rodriguez A, Sciotti RJ, Mensa-Wilmot K, Pollastri MP. Series of Alkynyl-Substituted Thienopyrimidines as Inhibitors of Protozoan Parasite Proliferation. ACS Med Chem Lett 2018; 9:996-1001. [PMID: 30344906 PMCID: PMC6187419 DOI: 10.1021/acsmedchemlett.8b00245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/04/2018] [Indexed: 12/23/2022] Open
Abstract
Discovery of new chemotherapeutic lead agents can be accelerated by optimizing chemotypes proven to be effective in other diseases to act against parasites. One such medicinal chemistry campaign has focused on optimizing the anilinoquinazoline drug lapatinib (1) and the alkynyl thieno[3,2-d]pyrimidine hit GW837016X (NEU-391, 3) into leads for antitrypanosome drugs. We now report the structure-activity relationship studies of 3 and its analogs against Trypanosoma brucei, which causes human African trypanosomiasis (HAT). The series was also tested against Trypanosoma cruzi, Leishmania major, and Plasmodium falciparum. In each case, potent antiparasitic hits with acceptable toxicity margins over mammalian HepG2 and NIH3T3 cell lines were identified. In a mouse model of HAT, 3 extended life of treated mice by 50%, compared to untreated controls. At the cellular level, 3 inhibited mitosis and cytokinesis in T. brucei. Thus, the alkynylthieno[3,2-d]pyrimidine chemotype is an advanced hit worthy of further optimization as a potential chemotherapeutic agent for HAT.
Collapse
Affiliation(s)
- Jennifer
L. Woodring
- Department
of Chemistry & Chemical Biology, Northeastern
University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Ranjan Behera
- Department
of Cellular Biology, Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Amrita Sharma
- Department
of Cellular Biology, Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Justin Wiedeman
- Department
of Cellular Biology, Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Gautam Patel
- Department
of Chemistry & Chemical Biology, Northeastern
University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Baljinder Singh
- Department
of Chemistry & Chemical Biology, Northeastern
University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Paul Guyett
- Department
of Cellular Biology, Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Emanuele Amata
- Department
of Chemistry & Chemical Biology, Northeastern
University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Jessey Erath
- Department
of Microbiology, New York University School
of Medicine, 430 E. 29th Street New York, New York 10010, United
States
- Anti-Infectives
Screening Core, New York University School
of Medicine, New York, New York 10010, United
States
| | - Norma Roncal
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United
States
| | - Erica Penn
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United
States
| | - Susan E. Leed
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United
States
| | - Ana Rodriguez
- Department
of Microbiology, New York University School
of Medicine, 430 E. 29th Street New York, New York 10010, United
States
- Anti-Infectives
Screening Core, New York University School
of Medicine, New York, New York 10010, United
States
| | - Richard J. Sciotti
- Experimental
Therapeutics, Walter Reed Army Institute
of Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United
States
| | - Kojo Mensa-Wilmot
- Department
of Cellular Biology, Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Michael P. Pollastri
- Department
of Chemistry & Chemical Biology, Northeastern
University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
The First Catalytic Direct C–H Arylation on C2 and C3 of Thiophene Ring Applied to Thieno-Pyridines, -Pyrimidines and -Pyrazines. Catalysts 2018. [DOI: 10.3390/catal8040137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
17
|
Triloknadh S, Venkata Rao C, Nagaraju K, Hari Krishna N, Venkata Ramaiah C, Rajendra W, Trinath D, Suneetha Y. Design, synthesis, neuroprotective, antibacterial activities and docking studies of novel thieno[2,3-d]pyrimidine-alkyne Mannich base and oxadiazole hybrids. Bioorg Med Chem Lett 2018; 28:1663-1669. [PMID: 29602681 DOI: 10.1016/j.bmcl.2018.03.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 03/05/2018] [Accepted: 03/12/2018] [Indexed: 01/07/2023]
Abstract
A series of thieno[2,3-d]pyrimidine alkyne Mannich base derivatives (7a-e, 8a-e) and thieno[2,3-d]pyrimidine 1,3,4-oxadiazole derivatives (9a-e, 10a-e) have been synthesized and evaluated for their neuroprotective and neurotoxicity activities where 9a, 10d displayed good neuroprotection 10.6 and 11.88 µg/mL respectively against the H2O2 induced cell death at the EC50 values and 9b, 9d showed respective toxic effects on PC12 cells at CC50 86.12 and 94.16 µg/mL. Compounds 9a, 9e, 10a and 10b showed strong antibacterial activity against two gram positive (S. aureus, B. subtilis) and two gram-negative strains (E. coli, P. aeruginosa) and showed good binding affinities with C(30) carotenoid dehydrosqualene synthase, Gyrase A and LpxC. This is the first report for the demonstration of thieno[2,3-d] pyrimidine derivatives as promising neuroprotective agents against H2O2 induced neurotoxicity on PC12 cells.
Collapse
Affiliation(s)
- Settypalli Triloknadh
- Department of Chemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Chunduri Venkata Rao
- Department of Chemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India.
| | - Kerru Nagaraju
- Department of Chemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | | | | | - Wudayagiri Rajendra
- Department of Zoology, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Daggupati Trinath
- Department of Zoology, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Yeguvapalli Suneetha
- Department of Zoology, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| |
Collapse
|
18
|
Borsari C, Quotadamo A, Ferrari S, Venturelli A, Cordeiro-da-Silva A, Santarem N, Costi MP. Scaffolds and Biological Targets Avenue to Fight Against Drug Resistance in Leishmaniasis. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2018. [DOI: 10.1016/bs.armc.2018.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
19
|
Optimization of physicochemical properties for 4-anilinoquinazoline inhibitors of trypanosome proliferation. Eur J Med Chem 2017; 141:446-459. [PMID: 29049963 DOI: 10.1016/j.ejmech.2017.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 01/15/2023]
Abstract
Human African trypanosomiasis (HAT) is a deadly disease in need of new chemotherapeutics that can cross into the central nervous system. We previously reported the discovery of 2 (NEU-617), a small molecule with activity against T. brucei bloodstream proliferation. Further optimization of 2 to improve the physicochemical properties (LogP, LLE, [1], and MPO score) [2] have led us to twelve sub-micromolar compounds, most importantly the headgroup variants 9i and 9j, and the linker variant 18. Although these 3 compounds had reduced potency compared to 2, they all had improved LogP, LLE and MPO scores. Cross-screening these analogs against other protozoan parasites uncovered 9o with potent activity towards T. brucei, T. cruzi and L. major, while four others compounds (17, 18, 21, 26) showed activity towards P. falciparum D6. This reinforces the effectiveness of lead repurposing for the discovery of new protozoan disease therapeutics.
Collapse
|
20
|
Berninger M, Schmidt I, Ponte-Sucre A, Holzgrabe U. Novel lead compounds in pre-clinical development against African sleeping sickness. MEDCHEMCOMM 2017; 8:1872-1890. [PMID: 30108710 PMCID: PMC6072528 DOI: 10.1039/c7md00280g] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/29/2017] [Indexed: 01/21/2023]
Abstract
Human African trypanosomiasis (HAT), also known as African sleeping sickness, is caused by parasitic protozoa of the genus Trypanosoma. As the disease progresses, the parasites cross the blood brain barrier and are lethal for the patients if the disease is left untreated. Current therapies suffer from several drawbacks due to e.g. toxicity of the respective compounds or resistance to approved antitrypanosomal drugs. In this review, the different strategies of drug development against HAT are considered, namely the target-based approach, the phenotypic high throughput screening and the drug repurposing strategy. The most promising compounds emerging from these approaches entering an in vivo evaluation are mentioned herein. Of note, it may turn out to be difficult to confirm in vitro activity in an animal model of infection; however, possible reasons for the missing efficacy in unsuccessful in vivo studies are discussed.
Collapse
Affiliation(s)
- Michael Berninger
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| | - Ines Schmidt
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology , Institute of Experimental Medicine , Luis Razetti School of Medicine , Faculty of Medicine , Universidad Central de Venezuela Caracas , Venezuela . Tel: +0931 31 85461
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry , University of Würzburg , Am Hubland , 97074 Würzburg , Germany .
| |
Collapse
|
21
|
Dichiara M, Marrazzo A, Prezzavento O, Collina S, Rescifina A, Amata E. Repurposing of Human Kinase Inhibitors in Neglected Protozoan Diseases. ChemMedChem 2017; 12:1235-1253. [PMID: 28590590 DOI: 10.1002/cmdc.201700259] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Indexed: 12/11/2022]
Abstract
Human African trypanosomiasis (HAT), Chagas disease, and leishmaniasis belong to a group of infectious diseases known as neglected tropical diseases and are induced by infection with protozoan parasites named trypanosomatids. Drugs in current use have several limitations, and therefore new candidate drugs are required. The majority of current therapeutic trypanosomatid targets are enzymes or cell-surface receptors. Among these, eukaryotic protein kinases are a major group of protein targets whose modulation may be beneficial for the treatment of neglected tropical protozoan diseases. This review summarizes the finding of new hit compounds for neglected tropical protozoan diseases, by repurposing known human kinase inhibitors on trypanosomatids. Kinase inhibitors are grouped by human kinase family and discussed according to the screening (target-based or phenotypic) reported for these compounds on trypanosomatids. This collection aims to provide insight into repurposed human kinase inhibitors and their importance in the development of new chemical entities with potential beneficial effects on the diseases caused by trypanosomatids.
Collapse
Affiliation(s)
- Maria Dichiara
- Department of Drug Sciences, University of Catania, V.le A. Doria, 6, 95100, Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania, V.le A. Doria, 6, 95100, Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, University of Catania, V.le A. Doria, 6, 95100, Catania, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, V.le Taramelli, 12, 27100, Pavia, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania, V.le A. Doria, 6, 95100, Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania, V.le A. Doria, 6, 95100, Catania, Italy
| |
Collapse
|
22
|
Wilding B, Klempier N. Newest Developments in the Preparation of Thieno[2,3-d]pyrimidines. ORG PREP PROCED INT 2017. [DOI: 10.1080/00304948.2017.1320513] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- B. Wilding
- Institute of Organic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| | - N. Klempier
- Institute of Organic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz, Austria
| |
Collapse
|
23
|
Park J, Leung CY, Matralis AN, Lacbay CM, Tsakos M, Fernandez De Troconiz G, Berghuis AM, Tsantrizos YS. Pharmacophore Mapping of Thienopyrimidine-Based Monophosphonate (ThP-MP) Inhibitors of the Human Farnesyl Pyrophosphate Synthase. J Med Chem 2017; 60:2119-2134. [PMID: 28208018 DOI: 10.1021/acs.jmedchem.6b01888] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The human farnesyl pyrophosphate synthase (hFPPS), a key regulatory enzyme in the mevalonate pathway, catalyzes the biosynthesis of the C-15 isoprenoid farnesyl pyrophosphate (FPP). FPP plays a crucial role in the post-translational prenylation of small GTPases that perform a plethora of cellular functions. Although hFPPS is a well-established therapeutic target for lytic bone diseases, the currently available bisphosphonate drugs exhibit poor cellular uptake and distribution into nonskeletal tissues. Recent drug discovery efforts have focused primarily on allosteric inhibition of hFPPS and the discovery of non-bisphosphonate drugs for potentially treating nonskeletal diseases. Hit-to-lead optimization of a new series of thienopyrimidine-based monosphosphonates (ThP-MPs) led to the identification of analogs with nanomolar potency in inhibiting hFPPS. Their interactions with the allosteric pocket of the enzyme were characterized by crystallography, and the results provide further insight into the pharmacophore requirements for allosteric inhibition.
Collapse
Affiliation(s)
- Jaeok Park
- Department of Biochemistry, McGill University , 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1, Canada
| | - Chun Yuen Leung
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| | - Alexios N Matralis
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| | - Cyrus M Lacbay
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| | - Michail Tsakos
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| | | | - Albert M Berghuis
- Department of Biochemistry, McGill University , 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1, Canada.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University , 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1, Canada
| | - Youla S Tsantrizos
- Department of Chemistry, McGill University , 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada.,Department of Biochemistry, McGill University , 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1, Canada.,Groupe de Recherche Axé sur la Structure des Protéines, McGill University , 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1, Canada
| |
Collapse
|
24
|
Novel Effects of Lapatinib Revealed in the African Trypanosome by Using Hypothesis-Generating Proteomics and Chemical Biology Strategies. Antimicrob Agents Chemother 2017; 61:AAC.01865-16. [PMID: 27872081 DOI: 10.1128/aac.01865-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 01/07/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the protozoan parasite Trypanosoma brucei Lapatinib, a human epidermal growth factor receptor (EGFR) inhibitor, can cure 25% of trypanosome-infected mice, although the parasite lacks EGFR-like tyrosine kinases. Four trypanosome protein kinases associate with lapatinib, suggesting that the drug may be a multitargeted inhibitor of phosphoprotein signaling in the bloodstream trypanosome. Phosphoprotein signaling pathways in T. brucei have diverged significantly from those in humans. As a first step in the evaluation of the polypharmacology of lapatinib in T. brucei, we performed a proteome-wide phosphopeptide analysis before and after drug addition to cells. Lapatinib caused dephosphorylation of Ser/Thr sites on proteins predicted to be involved in scaffolding, gene expression, and intracellular vesicle trafficking. To explore the perturbation of phosphotyrosine (pTyr)-dependent signaling by lapatinib, proteins in lapatinib-susceptible pTyr complexes were identified by affinity chromatography; they included BILBO-1, MORN, and paraflagellar rod (PFR) proteins PFR1 and PFR2. These data led us to hypothesize that lapatinib disrupts PFR functions and/or endocytosis in the trypanosome. In direct chemical biology tests of these speculations, lapatinib-treated trypanosomes (i) lost segments of the PFR inside the flagellum, (ii) were inhibited in the endocytosis of transferrin, and (iii) changed morphology from long and slender to rounded. Thus, our hypothesis-generating phosphoproteomics strategy predicted novel physiological pathways perturbed by lapatinib, which were verified experimentally. General implications of this workflow for identifying signaling pathways perturbed by drug hits discovered in phenotypic screens are discussed.
Collapse
|
25
|
Zhang Q, Zhang L, Yu J, Li H, He S, Tang W, Zuo J, Lu W. Discovery of new BTK inhibitors with B cell suppression activity bearing a 4,6-substituted thieno[3,2-d]pyrimidine scaffold. RSC Adv 2017. [DOI: 10.1039/c7ra04261b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Seventeen compounds with 4,6-substituted thieno[3,2-d]pyrimidine scaffold were prepared as new Bruton's tyrosine kinase inhibitors. Compound 8 exhibits anti-BTK activity, immunosuppressive activity, enzymatic selectivity and low toxicity.
Collapse
Affiliation(s)
- Qiumeng Zhang
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
| | - Luyao Zhang
- Laboratory of Immunopharmacology
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- P. R. China
| | - Jie Yu
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
| | - Heng Li
- Laboratory of Immunopharmacology
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- P. R. China
| | - Shijun He
- Laboratory of Immunopharmacology
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- P. R. China
| | - Wei Tang
- Laboratory of Immunopharmacology
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- P. R. China
| | - Jianping Zuo
- Laboratory of Immunopharmacology
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- P. R. China
| | - Wei Lu
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
| |
Collapse
|
26
|
Russell S, Rahmani R, Jones AJ, Newson HL, Neilde K, Cotillo I, Rahmani Khajouei M, Ferrins L, Qureishi S, Nguyen N, Martinez-Martinez MS, Weaver DF, Kaiser M, Riley J, Thomas J, De Rycker M, Read KD, Flematti GR, Ryan E, Tanghe S, Rodriguez A, Charman SA, Kessler A, Avery VM, Baell JB, Piggott MJ. Hit-to-Lead Optimization of a Novel Class of Potent, Broad-Spectrum Trypanosomacides. J Med Chem 2016; 59:9686-9720. [DOI: 10.1021/acs.jmedchem.6b00442] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Stephanie Russell
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| | - Raphaël Rahmani
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Amy J. Jones
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane Innovation Park, Don Young
Road, Nathan, Queensland 4111, Australia
| | - Harriet L. Newson
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| | - Kevin Neilde
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- GlaxoSmithKline, 28760 Tres Cantos, Spain
| | | | - Marzieh Rahmani Khajouei
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| | - Lori Ferrins
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sana Qureishi
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| | - Nghi Nguyen
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - Donald F. Weaver
- Department
of Chemistry, Dalhousie University, Halifax Nova Scotia B3H 4R2, Canada
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Socinstrasse, 4051 Basel, Switzerland
- University of Basel, Petesplatz
1, 4003 Basel, Switzerland
| | - Jennifer Riley
- Drug
Discovery Unit, Division of Biological Chemistry and Drug Discovery,
School of Life Sciences, University of Dundee, DD1 5EH Dundee, U.K
| | - John Thomas
- Drug
Discovery Unit, Division of Biological Chemistry and Drug Discovery,
School of Life Sciences, University of Dundee, DD1 5EH Dundee, U.K
| | - Manu De Rycker
- Drug
Discovery Unit, Division of Biological Chemistry and Drug Discovery,
School of Life Sciences, University of Dundee, DD1 5EH Dundee, U.K
| | - Kevin D. Read
- Drug
Discovery Unit, Division of Biological Chemistry and Drug Discovery,
School of Life Sciences, University of Dundee, DD1 5EH Dundee, U.K
| | - Gavin R. Flematti
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| | - Eileen Ryan
- Centre
for Drug Candidate Optimisation, Monash University, Parkville, Victoria 3052, Australia
| | - Scott Tanghe
- Anti-Infectives
Screening Core, New York University School of Medicine, New York, New York 10010, United States
| | - Ana Rodriguez
- Anti-Infectives
Screening Core, New York University School of Medicine, New York, New York 10010, United States
| | - Susan A. Charman
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Centre
for Drug Candidate Optimisation, Monash University, Parkville, Victoria 3052, Australia
| | | | - Vicky M. Avery
- Eskitis
Institute for Drug Discovery, Griffith University, Brisbane Innovation Park, Don Young
Road, Nathan, Queensland 4111, Australia
| | - Jonathan B. Baell
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Matthew J. Piggott
- School
of Chemistry and Biochemistry, The University of Western Australia, 35 Stirling Highway, Crawley, Perth 6009, Western Australia, Australia
| |
Collapse
|
27
|
Devine W, Woodring JL, Swaminathan U, Amata E, Patel G, Erath J, Roncal NE, Lee PJ, Leed SE, Rodriguez A, Mensa-Wilmot K, Sciotti RJ, Pollastri MP. Protozoan Parasite Growth Inhibitors Discovered by Cross-Screening Yield Potent Scaffolds for Lead Discovery. J Med Chem 2015; 58:5522-37. [PMID: 26087257 PMCID: PMC4515785 DOI: 10.1021/acs.jmedchem.5b00515] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
![]()
Tropical protozoal infections are
a significant cause of morbidity
and mortality worldwide; four in particular (human African trypanosomiasis
(HAT), Chagas disease, cutaneous leishmaniasis, and malaria) have
an estimated combined burden of over 87 million disability-adjusted
life years. New drugs are needed for each of these diseases. Building
on the previous identification of NEU-617 (1) as a potent
and nontoxic inhibitor of proliferation for the HAT pathogen (Trypanosoma brucei), we have now tested this class of analogs
against other protozoal species: T. cruzi (Chagas
disease), Leishmania major (cutaneous leishmaniasis),
and Plasmodium falciparum (malaria). Based on hits
identified in this screening campaign, we describe the preparation
of several replacements for the quinazoline scaffold and report these
inhibitors’ biological activities against these parasites.
In doing this, we have identified several potent proliferation inhibitors
for each pathogen, such as 4-((3-chloro-4-((3-fluorobenzyl)oxy)phenyl)amino)-6-(4-((4-methyl-1,4-diazepan-1-yl)sulfonyl)phenyl)quinoline-3-carbonitrile
(NEU-924, 83) for T. cruzi and N-(3-chloro-4-((3-fluorobenzyl)oxy)phenyl)-7-(4-((4-methyl-1,4-diazepan-1-yl)sulfonyl)phenyl)cinnolin-4-amine
(NEU-1017, 68) for L. major and P. falciparum.
Collapse
Affiliation(s)
| | | | | | | | | | - Jessey Erath
- ‡Division of Parasitology, Department of Microbiology, New York University School of Medicine, 341 E. 25th St., New York, New York 10010, United States
| | - Norma E Roncal
- §Experimental Therapeutics, Walter Reed Army Institute for Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United States
| | - Patricia J Lee
- §Experimental Therapeutics, Walter Reed Army Institute for Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United States
| | - Susan E Leed
- §Experimental Therapeutics, Walter Reed Army Institute for Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United States
| | - Ana Rodriguez
- ‡Division of Parasitology, Department of Microbiology, New York University School of Medicine, 341 E. 25th St., New York, New York 10010, United States.,⊥Anti-Infectives Screening Core, New York University School of Medicine, New York, New York 10010, United States
| | - Kojo Mensa-Wilmot
- ∥Department of Cellular Biology, University of Georgia, Athens, Georgia 30602, United States
| | - Richard J Sciotti
- §Experimental Therapeutics, Walter Reed Army Institute for Research, 2460 Linden Lane, Silver Spring, Maryland 20910, United States
| | | |
Collapse
|