1
|
Chen M, Cheng S, Dai X, Yu J, Wang H, Xu B, Luo H, Xu G. Design, Synthesis, and Biological Evaluation of Novel Quinazoline Derivatives Possessing a Trifluoromethyl Moiety as Potential Antitumor Agents. Chem Biodivers 2024; 21:e202301776. [PMID: 38602834 DOI: 10.1002/cbdv.202301776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
A novel series of trifluoromethyl-containing quinazoline derivatives with a variety of functional groups was designed, synthesized, and tested for their antitumor activity by following a pharmacophore hybridization strategy. Most of the 20 compounds displayed moderate to excellent antiproliferative activity against five different cell lines (PC3, LNCaP, K562, HeLa, and A549). After three rounds of screening and structural optimization, compound 10 b was identified as the most potent one, with IC50 values of 3.02, 3.45, and 3.98 μM against PC3, LNCaP, and K562 cells, respectively, which were comparable to the effect of the positive control gefitinib. To further explore the mechanism of action of 10 b against cancer, experiments focusing on apoptosis induction, cell cycle arrest, and cell migration assay were conducted. The results showed that 10 b was able to induce apoptosis and prevent tumor cell migration, but had no effect on the cell cycle of tumor cells.
Collapse
Affiliation(s)
- Mingxiu Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xing Dai
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - HuiDi Wang
- The Affiliated Hospital of Wuhan Sports University, Wuhan, 430079, China
| | - BiXue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| |
Collapse
|
2
|
Pindjakova D, Mascaretti S, Hricoviniova J, Hosek J, Gregorova J, Kos J, Cizek A, Hricoviniova Z, Jampilek J. Critical view on antimicrobial, antibiofilm and cytotoxic activities of quinazolin-4(3 H)-one derived schiff bases and their Cu(II) complexes. Heliyon 2024; 10:e29051. [PMID: 38601653 PMCID: PMC11004567 DOI: 10.1016/j.heliyon.2024.e29051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
A series of nine 2,3-disubstituted-quinazolin-4(3H)-one derived Schiff bases and their three Cu(II) complexes was prepared and tested for their antimicrobial activities against reference strains Staphylococcus aureus ATCC 29213 and Enterococcus faecalis ATCC 29212 and resistant clinical isolates of methicillin-resistant S. aureus (MRSA) and vancomycin-resistant E. faecalis (VRE). All the substances were tested in vitro against Mycobacterium tuberculosis H37Ra ATCC 25177, M. kansasii DSM 44162 and M. smegmatis ATCC 700084. While anti-enterococcal and antimycobacterial activities were insignificant, 3-[(E)-(2-hydroxy-5-nitrobenzylidene)amino]-2-(2-hydroxy-5-nitrophenyl)-2,3-dihydroquinazolin-4(1H)-one (SB3) and its Cu(II) complex (SB3-Cu) demonstrated bacteriostatic antistaphylococcal activity. In addition, both compounds, as well as the other two prepared complexes, showed antibiofilm activity, which resulted in a reduction of biofilm formation and eradication of mature S. aureus biofilm by 80% even at concentrations lower than the values of their minimum inhibitory concentrations. In addition, the compounds were tested for their cytotoxic effect on the human monocytic leukemia cell line THP-1. The antileukemic efficiency was improved by the preparation of Cu(II) complexes from the corresponding non-chelated Schiff base ligands.
Collapse
Affiliation(s)
- Dominika Pindjakova
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Sarka Mascaretti
- Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho 1946/1, 612 42 Brno, Czech Republic
| | - Jana Hricoviniova
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, Odbojarov 10, 832 32 Bratislava, Slovakia
| | - Jan Hosek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - Jana Gregorova
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jiri Kos
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Alois Cizek
- Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho 1946/1, 612 42 Brno, Czech Republic
| | - Zuzana Hricoviniova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
| | - Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| |
Collapse
|
3
|
Monsen RC, Maguire JM, DeLeeuw LW, Chaires JB, Trent JO. Drug discovery of small molecules targeting the higher-order hTERT promoter G-quadruplex. PLoS One 2022; 17:e0270165. [PMID: 35709230 PMCID: PMC9202945 DOI: 10.1371/journal.pone.0270165] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
DNA G-quadruplexes (G4s) are now widely accepted as viable targets in the pursuit of anticancer therapeutics. To date, few small molecules have been identified that exhibit selectivity for G4s over alternative forms of DNA, such as the ubiquitous duplex. We posit that the lack of current ligand specificity arises for multiple reasons: G4 atomic models are often small, monomeric, single quadruplex structures with few or no druggable pockets; targeting G-tetrad faces frequently results in the enrichment of extended electron-deficient polyaromatic end-pasting scaffolds; and virtual drug discovery efforts often under-sample chemical search space. We show that by addressing these issues we can enrich for non-standard molecular templates that exhibit high selectivity towards G4s over other forms of DNA. We performed an extensive virtual screen against the higher-order hTERT core promoter G4 that we have previously characterized, targeting 12 of its unique loop and groove pockets using libraries containing 40 million drug-like compounds for each screen. Using our drug discovery funnel approach, which utilizes high-throughput fluorescence thermal shift assay (FTSA) screens, microscale thermophoresis (MST), and orthogonal biophysical methods, we have identified multiple unique G4 binding scaffolds. We subsequently used two rounds of catalogue-based SAR to increase the affinity of a disubstituted 2-aminoethyl-quinazoline that stabilizes the higher-order hTERT G-quadruplex by binding across its G4 junctional sites. We show selectivity of its binding affinity towards hTERT is virtually unaffected in the presence of near-physiological levels of duplex DNA, and that this molecule downregulates hTERT transcription in breast cancer cells.
Collapse
Affiliation(s)
- Robert C. Monsen
- UofL Health Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Jon M. Maguire
- UofL Health Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Lynn W. DeLeeuw
- UofL Health Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Jonathan B. Chaires
- UofL Health Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail: (JBC); (JOT)
| | - John O. Trent
- UofL Health Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail: (JBC); (JOT)
| |
Collapse
|
4
|
Pal S, Paul B, Bandopadhyay P, Preethy N, Sarkar D, Rahaman O, Goon S, Roy S, Ganguly D, Talukdar A. Synthesis and characterization of new potent TLR7 antagonists based on analysis of the binding mode using biomolecular simulations. Eur J Med Chem 2020; 210:112978. [PMID: 33189437 DOI: 10.1016/j.ejmech.2020.112978] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Aberrant activation of the endosomal Toll-like receptor 7 (TLR7) has been implicated in myriad autoimmune diseases and is an established therapeutic target in such conditions. Development of diverse TLR7 antagonists is mainly accomplished through random screening. To correlate human TLR7 (hTLR7) antagonistic activity with the structural features in different chemotypes, we derived a hypothetical binding model based on molecular docking analysis along with molecular dynamics (MD) simulations study. The binding hypothesis revealed different pockets, grooves and a central cavity where ligand-receptor interaction with specific residues through hydrophobic and hydrogen bond interactions take place, which correlate with TLR7 antagonistic activity thus paving the way for rational design using varied chemotypes. Based on the structural insight thus gained, TLR7 antagonists with quinazoline were designed to understand the effect of engagement of these pockets as well as boundaries of the chemical space associated with them. The newly synthesized most potent hTLR7 antagonist, i.e. compound 63, showed IC50 value of 1.03 ± 0.05 μM and was validated by performing primary assay in human plasmacytoid dendritic cells (pDC) (IC50pDC: 1.42 μM). The biological validation of the synthesized molecules was performed in TLR7-reporter HEK293 cells as well as in human plasmacytoid dendritic cells (pDCs). Our study provides a rational design approach thus facilitating further development of novel small molecule hTLR7 antagonists based on different chemical scaffolds.
Collapse
Affiliation(s)
- Sourav Pal
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Barnali Paul
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata, 700091, WB, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Nagothy Preethy
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Dipika Sarkar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Oindrila Rahaman
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata, 700091, WB, India
| | - Sunny Goon
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Swarnali Roy
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, Department of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology, CN6, Sector V, Salt Lake, Kolkata, 700091, WB, India
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India.
| |
Collapse
|
5
|
Turcano L, Battista T, De Haro ET, Missineo A, Alli C, Paonessa G, Colotti G, Harper S, Fiorillo A, Ilari A, Bresciani A. Spiro-containing derivatives show antiparasitic activity against Trypanosoma brucei through inhibition of the trypanothione reductase enzyme. PLoS Negl Trop Dis 2020; 14:e0008339. [PMID: 32437349 PMCID: PMC7269337 DOI: 10.1371/journal.pntd.0008339] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/03/2020] [Accepted: 04/30/2020] [Indexed: 11/19/2022] Open
Abstract
Trypanothione reductase (TR) is a key enzyme that catalyzes the reduction of trypanothione, an antioxidant dithiol that protects Trypanosomatid parasites from oxidative stress induced by mammalian host defense systems. TR is considered an attractive target for the development of novel anti-parasitic agents as it is essential for parasite survival but has no close homologue in humans. We report here the identification of spiro-containing derivatives as inhibitors of TR from Trypanosoma brucei (TbTR), the parasite responsible for Human African Trypanosomiasis. The hit series, identified by high throughput screening, was shown to bind TbTR reversibly and to compete with the trypanothione (TS2) substrate. The prototype compound 1 from this series was also found to impede the growth of Trypanosoma brucei parasites in vitro. The X-ray crystal structure of TbTR in complex with compound 1 solved at 1.98 Å allowed the identification of the hydrophobic pocket where the inhibitor binds, placed close to the catalytic histidine (His 461’) and lined by Trp21, Val53, Ile106, Tyr110 and Met113. This new inhibitor is specific for TbTR and no activity was detected against the structurally similar human glutathione reductase (hGR). The central spiro scaffold is known to be suitable for brain active compounds in humans thus representing an attractive starting point for the future treatment of the central nervous system stage of T. brucei infections. Trypanosoma brucei is a parasite responsible for neglected pathologies such as human African trypanosomiasis, also known as sleeping sickness. This disease is endemic in sub-Saharan Africa, with 70 million people at risk of infection. Current treatments for this type of disease are limited by their toxicity, administration in endemic countries and treatment resistance. Therapies against infectious diseases typically rely on targeting one or more components of the parasite that are not present in humans to ensure the best possible therapeutic window. In this case we aimed at targeting the Trypanosoma brucei trypanothione reductase (TR), one enzyme that synthesize the reduced trypanothione a key molecule for preserving the parasite redox balance. This enzyme does not exist in humans that have glutathione instead of trypanothione. Past attempts to identify novel inhibitors of this target has failed to generate drug-like molecules. To overcome this limitation we employed a recent, higher quality, TR activity assay to test a collection of compounds previously reported to be active against these parasites. This approach led to the identification and validation of a new chemotype with a unique mode of inhibition of TR. This chemical series is a drug-like starting point, in fact its core (spiro) is present in drugs approved for human use.
Collapse
Affiliation(s)
- Lorenzo Turcano
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Theo Battista
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | | | - Antonino Missineo
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Cristina Alli
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Giacomo Paonessa
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Gianni Colotti
- Istituto di Biologia e Patologia Molecolari del CNR c/o Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | | | - Annarita Fiorillo
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | - Andrea Ilari
- Istituto di Biologia e Patologia Molecolari del CNR c/o Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
- * E-mail: (AI); (AB)
| | - Alberto Bresciani
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
- * E-mail: (AI); (AB)
| |
Collapse
|
6
|
In vitro anticancer activity of pyrano[3, 2-c]chromene derivatives with both cell cycle arrest and apoptosis induction. Med Chem Res 2020. [DOI: 10.1007/s00044-019-02494-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
7
|
Soares FGN, Göethel G, Kagami LP, das Neves GM, Sauer E, Birriel E, Varela J, Gonçalves IL, Von Poser G, González M, Kawano DF, Paula FR, de Melo EB, Garcia SC, Cerecetto H, Eifler-Lima VL. Novel coumarins active against Trypanosoma cruzi and toxicity assessment using the animal model Caenorhabditis elegans. BMC Pharmacol Toxicol 2019; 20:76. [PMID: 31852548 PMCID: PMC6921407 DOI: 10.1186/s40360-019-0357-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chagas disease (CD) is a tropical parasitic disease. Although the number of people infected is very high, the only drugs available to treat CD, nifurtimox (Nfx) and benznidazole, are highly toxic, particularly in the chronic stage of the disease. Coumarins are a large class of compounds that display a wide range of interesting biological properties, such as antiparasitic. Hence, the aim of this work is to find a good antitrypanosomal drug with less toxicity. The use of simple organism models has become increasingly attractive for planning and simplifying efficient drug discovery. Within these models, Caenorhabditis elegans has emerged as a convenient and versatile tool with significant advantages for the toxicological potential identification for new compounds. METHODS Trypanocidal activity: Forty-two 4-methylamino-coumarins were assayed against the epimastigote form of Trypanosoma cruzi (Tulahuen 2 strain) by inhibitory concentration 50% (IC50). Toxicity assays: Lethal dose 50% (LD50) and Body Area were determined by Caenorhabditis elegans N2 strain (wild type) after acute exposure. Structure-activity relationship: A classificatory model was built using 3D descriptors. RESULTS Two of these coumarins demonstrated near equipotency to Nifurtimox (IC50 = 5.0 ± 1 μM), with values of: 11 h (LaSOM 266), (IC50 = 6.4 ± 1 μM) and 11 g (LaSOM 231), (IC50 = 8.2 ± 2.3 μM). In C. elegans it was possible to observe that Nfx showed greater toxicity in both the LD50 assay and the evaluation of the development of worms. It is possible to observe that the efficacy between Nfx and the synthesized compounds (11 h and 11 g) are similar. On the other hand, the toxicity of Nfx is approximately three times higher than that of the compounds. Results from the QSAR-3D study indicate that the volume and hydrophobicity of the substituents have a significant impact on the trypanocidal activities for derivatives that cause more than 50% of inhibition. These results show that the C. elegans model is efficient for screening potentially toxic compounds. CONCLUSION Two coumarins (11 h and 11 g) showed activity against T. cruzi epimastigote similar to Nifurtimox, however with lower toxicity in both LD50 and development of C. elegans assays. These two compounds may be a feasible starting point for the development of new trypanocidal drugs.
Collapse
Affiliation(s)
- Fabiana Gomes Nascimento Soares
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gabriela Göethel
- Laboratório Toxicologia/LATOX, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luciano Porto Kagami
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gustavo Machado das Neves
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Elisa Sauer
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Estefania Birriel
- Facultad de Ciencias-Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Javier Varela
- Facultad de Ciencias-Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Itamar Luís Gonçalves
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gilsane Von Poser
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mercedes González
- Facultad de Ciencias-Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Daniel Fábio Kawano
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, Campinas, SP, Brazil
- Departamento de Química Orgânica, Instituto de Química, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Fávero Reisdorfer Paula
- Universidade Estadual do Oeste do Paraná, Centro de Ciências Médicas e Farmacêuticas, Cascavel, PR, Brazil
| | - Eduardo Borges de Melo
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná, Cascavel, PR, Brazil
| | - Solange Cristina Garcia
- Laboratório Toxicologia/LATOX, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Hugo Cerecetto
- Facultad de Ciencias-Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Vera Lucia Eifler-Lima
- Laboratório de Síntese Orgânica Medicinal/LaSOM, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
8
|
Bollini M, Bruno AM, Niño ME, Casal JJ, Sasiambarrena LD, Valdez DAG, Battini L, Puente VR, Lombardo ME. Synthesis, 2D-QSAR Studies and Biological Evaluation of Quinazoline Derivatives as Potent Anti-Trypanosoma cruzi Agents. Med Chem 2019; 15:265-276. [PMID: 30295191 DOI: 10.2174/1573406414666181005145042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/24/2018] [Accepted: 08/23/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chagas disease affects about 7 million people worldwide. Only two drugs are currently available for the treatment for this parasite disease, namely, benznidazol (Bzn) and nifurtimox (Nfx). Both drugs have limited curative power in the chronic phase of the disease. Therefore, continuous research is an urgent need so as to discover novel therapeutic alternatives. OBJECTIVE The development of safer and more efficient therapeutic anti-T. cruzi drugs continues to be a major goal in trypanocidal chemotherapy. METHOD Synthesis, 2D-QSAR and drug-like physicochemical properties of a set of quinazolinone and quinazoline derivatives were studied as trypanocidal agents. All compounds were screened in vitro against Trypanosoma cruzi (Tulahuen strain, Tul 2 stock) epimastigotes and bloodstream trypomastigotes. RESULTS Out of 34 compounds synthesized and tested, six compounds (5a, 5b, 9b, 9h, 13f and 13p) displayed significant activity against both epimastigotes and tripomastigotes, without exerting toxicity on Vero cells. CONCLUSION The antiprotozoal activity of these quinazolinone and quinazoline derivatives represents an interesting starting point for a medicinal chemistry program aiming at the development of novel chemotherapies for Chagas disease.
Collapse
Affiliation(s)
- Mariela Bollini
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Ana M Bruno
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - María E Niño
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Juan J Casal
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Leandro D Sasiambarrena
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Damián A G Valdez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Leandro Battini
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Vanesa R Puente
- Centro de Investigaciones Sobre Porfirinas y Porfirias (CIPYP, UBA-CONICET), Hospital de Clinicas Jose de San Martin, Avenida Cordoba 2351, C1120AAR, Ciudad Autonoma de Buenos Aires, Argentina
| | - María E Lombardo
- Centro de Investigaciones Sobre Porfirinas y Porfirias (CIPYP, UBA-CONICET), Hospital de Clinicas Jose de San Martin, Avenida Cordoba 2351, C1120AAR, Ciudad Autonoma de Buenos Aires, Argentina
| |
Collapse
|
9
|
Matus-Meza AS, Velasco-Velázquez MA, Hernández-Luis F. Design, synthesis and cytotoxic evaluation of quinazoline-2,4,6-triamine and 2,6-diaminoquinazolin-4(3H)-one derivatives. Med Chem Res 2018; 27:1748-1756. [DOI: 10.1007/s00044-018-2188-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
|
10
|
Zulfiqar B, Jones AJ, Sykes ML, Shelper TB, Davis RA, Avery VM. Screening a Natural Product-Based Library against Kinetoplastid Parasites. Molecules 2017; 22:E1715. [PMID: 29023425 PMCID: PMC6151456 DOI: 10.3390/molecules22101715] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 01/06/2023] Open
Abstract
Kinetoplastid parasites cause vector-borne parasitic diseases including leishmaniasis, human African trypanosomiasis (HAT) and Chagas disease. These Neglected Tropical Diseases (NTDs) impact on some of the world's lowest socioeconomic communities. Current treatments for these diseases cause severe toxicity and have limited efficacy, highlighting the need to identify new treatments. In this study, the Davis open access natural product-based library was screened against kinetoplastids (Leishmania donovani DD8, Trypanosoma brucei brucei and Trypanosoma cruzi) using phenotypic assays. The aim of this study was to identify hit compounds, with a focus on improved efficacy, selectivity and potential to target several kinetoplastid parasites. The IC50 values of the natural products were obtained for L. donovani DD8, T. b. brucei and T. cruzi in addition to cytotoxicity against the mammalian cell lines, HEK-293, 3T3 and THP-1 cell lines were determined to ascertain parasite selectivity. Thirty-one compounds were identified with IC50 values of ≤ 10 µM against the kinetoplastid parasites tested. Lissoclinotoxin E (1) was the only compound identified with activity across all three investigated parasites, exhibiting IC50 values < 5 µM. In this study, natural products with the potential to be new chemical starting points for drug discovery efforts for kinetoplastid diseases were identified.
Collapse
Affiliation(s)
- Bilal Zulfiqar
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Amy J Jones
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Melissa L Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Todd B Shelper
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Rohan A Davis
- Natural Product Chemistry, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
11
|
Hamed MM, Darwish SS, Herrmann J, Abadi AH, Engel M. First Bispecific Inhibitors of the Epidermal Growth Factor Receptor Kinase and the NF-κB Activity As Novel Anticancer Agents. J Med Chem 2017; 60:2853-2868. [PMID: 28291344 DOI: 10.1021/acs.jmedchem.6b01774] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The activation of the NF-κB transcription factor is a major adaptive response induced upon treatment with EGFR kinase inhibitors, leading to the emergence of resistance in nonsmall cell lung cancer and other tumor types. To suppress this survival mechanism, we developed new thiourea quinazoline derivatives that are dual inhibitors of both EGFR kinase and the NF-κB activity. Optimization of the hit compound, identified in a NF-κB reporter gene assay, led to compound 9b, exhibiting a cellular IC50 for NF-κB inhibition of 0.3 μM while retaining a potent EGFR kinase inhibition (IC50 = 60 nM). The dual inhibitors showed a higher potency than gefitinib to inhibit cell growth of EGFR-overexpressing tumor cell lines in vitro and in a xenograft model in vivo, while no signs of toxicity were observed. An investigation of the molecular mechanism of NF-κB suppression revealed that the dual inhibitors depleted the transcriptional coactivator CREB-binding protein from the NF-κB complex in the nucleus.
Collapse
Affiliation(s)
- Mostafa M Hamed
- Pharmaceutical and Medicinal Chemistry, Campus C2.3, and Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarland University , Campus E8.1, D-66123 Saarbrücken, Germany.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Sarah S Darwish
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) , Campus E8.1, D-66123 Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo , Cairo 11835, Egypt
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University , Campus C2.3, D-66123 Saarbrücken, Germany
| |
Collapse
|
12
|
Kotha S, Deodhar D, Khedkar P. Diversity-oriented synthesis of medicinally important 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic) derivatives and higher analogs. Org Biomol Chem 2015; 12:9054-91. [PMID: 25299735 DOI: 10.1039/c4ob01446d] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
1,2,3,4-Tetrahydroisoquinoline-3-carboxylic acid (Tic) is a constrained analog of phenylalanine (Phe). The Tic unit has been identified as a core structural element present in several peptide-based drugs and forms an integral part of various biologically active compounds. This report covers the biological significance of the Tic core and provides a detailed account of various synthetic approaches available for the construction of Tic derivatives. Along with the traditional methods such as the Pictet-Spengler and Bischler-Nepieralski reactions, we cover various recent approaches such as enyne metathesis, [2 + 2 + 2] cycloaddition and the Diels-Alder reaction to generate Tic derivatives. In addition, syntheses of higher analogs of Tic are also discussed.
Collapse
Affiliation(s)
- Sambasivarao Kotha
- Department of Chemistry, Indian Institute of Technology-Bombay, Powai, Mumbai - 400 076, India.
| | | | | |
Collapse
|
13
|
A facile one-pot synthesis of functionalized fused benzochromene derivatives via intramolecular Wittig reactions. RESEARCH ON CHEMICAL INTERMEDIATES 2015. [DOI: 10.1007/s11164-013-1438-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
14
|
Staderini M, Bolognesi ML, Menéndez JC. Lewis Acid-Catalyzed Generation of CC and CN Bonds on π-Deficient Heterocyclic Substrates. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201400674] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
15
|
Nagle A, Khare S, Kumar AB, Supek F, Buchynskyy A, Mathison CJN, Chennamaneni N, Pendem N, Buckner FS, Gelb M, Molteni V. Recent developments in drug discovery for leishmaniasis and human African trypanosomiasis. Chem Rev 2014; 114:11305-47. [PMID: 25365529 PMCID: PMC4633805 DOI: 10.1021/cr500365f] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Indexed: 02/08/2023]
Affiliation(s)
- Advait
S. Nagle
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Shilpi Khare
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Arun Babu Kumar
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Frantisek Supek
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Andriy Buchynskyy
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Casey J. N. Mathison
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Naveen
Kumar Chennamaneni
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Nagendar Pendem
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Frederick S. Buckner
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Michael
H. Gelb
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Valentina Molteni
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| |
Collapse
|
16
|
Yolacan C, Mavis ME, Aydogan F. Evaluation of mono- and dipeptides as organocatalysts for enantioselective aldol reaction. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
17
|
Gupta S, Shivahare R, Korthikunta V, Singh R, Gupta S, Tadigoppula N. Synthesis and biological evaluation of chalcones as potential antileishmanial agents. Eur J Med Chem 2014; 81:359-66. [DOI: 10.1016/j.ejmech.2014.05.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 05/09/2014] [Accepted: 05/11/2014] [Indexed: 12/12/2022]
|
18
|
Shivahare R, Korthikunta V, Chandasana H, Suthar MK, Agnihotri P, Vishwakarma P, Chaitanya TK, Kancharla P, Khaliq T, Gupta S, Bhatta RS, Pratap JV, Saxena JK, Gupta S, Tadigoppula N. Synthesis, Structure–Activity Relationships, and Biological Studies of Chromenochalcones as Potential Antileishmanial Agents. J Med Chem 2014; 57:3342-57. [DOI: 10.1021/jm401893j] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Rahul Shivahare
- Division
of Parasitology, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Venkateswarlu Korthikunta
- Division
of Medicinal and Process Chemistry, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Hardik Chandasana
- Division
of Pharmacokinetics and Metabolism, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Manish K. Suthar
- Division
of Biochemistry, CSIR−Central Drug Research Institute, Lucknow-226 031, Uttar Pradesh, India
| | - Pragati Agnihotri
- Division
of Molecular and Structural Biology, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Preeti Vishwakarma
- Division
of Parasitology, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Telaprolu K. Chaitanya
- Division
of Pharmacokinetics and Metabolism, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Papireddy Kancharla
- Division
of Medicinal and Process Chemistry, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Tanvir Khaliq
- Division
of Medicinal and Process Chemistry, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Shweta Gupta
- Division
of Medicinal and Process Chemistry, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Division
of Pharmacokinetics and Metabolism, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - J. Venkatesh Pratap
- Division
of Molecular and Structural Biology, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Jitendra K. Saxena
- Division
of Biochemistry, CSIR−Central Drug Research Institute, Lucknow-226 031, Uttar Pradesh, India
| | - Suman Gupta
- Division
of Parasitology, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| | - Narender Tadigoppula
- Division
of Medicinal and Process Chemistry, CSIR−Central Drug Research Institute, Lucknow 226 031, Uttar Pradesh, India
| |
Collapse
|
19
|
Kalniņa A, Bizdēna Ē, Kiselovs G, Mishnev A, Turks M. Structural Proof of Tetrazolo[1,5-a]quinazoline Derivatives and their Application in the Synthesis of 4-Amino-2-(1,2,3-triazol-1-yl)quinazolines*. Chem Heterocycl Compd (N Y) 2014. [DOI: 10.1007/s10593-014-1418-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Toward the Development of Dual-Targeted Glyceraldehyde-3-phosphate Dehydrogenase/Trypanothione Reductase Inhibitors againstTrypanosoma bruceiandTrypanosoma cruzi. ChemMedChem 2014; 9:371-82. [DOI: 10.1002/cmdc.201300399] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/05/2013] [Indexed: 12/22/2022]
|
21
|
Lizzi F, Veronesi G, Belluti F, Bergamini C, López-Sánchez A, Kaiser M, Brun R, Krauth-Siegel RL, Hall DG, Rivas L, Bolognesi ML. Conjugation of Quinones with Natural Polyamines: Toward an Expanded Antitrypanosomatid Profile. J Med Chem 2012; 55:10490-500. [DOI: 10.1021/jm301112z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Federica Lizzi
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126 Bologna,
Italy
| | - Giacomo Veronesi
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126 Bologna,
Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126 Bologna,
Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126 Bologna,
Italy
| | - Almudena López-Sánchez
- Physico-Chemical
Biology, Centro
de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland
- University of Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Reto Brun
- Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland
- University of Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - R. Luise Krauth-Siegel
- Biochemistry Center (BZH), Heidelberg University, Im, Neuenheimer Feld 328, 69120
Heidelberg, Germany
| | - Dennis G. Hall
- Department
of Chemistry, University of Alberta, Edmonton,
Alberta, T6G 2G2,
Canada
| | - Luis Rivas
- Physico-Chemical
Biology, Centro
de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126 Bologna,
Italy
| |
Collapse
|
22
|
|
23
|
Synthesis and biological evaluation of new 3-(6-hydroxyindol-2-yl)-5-(Phenyl) pyridine or pyrazine V-Shaped molecules as kinase inhibitors and cytotoxic agents. Eur J Med Chem 2011; 46:5416-34. [DOI: 10.1016/j.ejmech.2011.08.048] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/29/2011] [Accepted: 08/31/2011] [Indexed: 12/22/2022]
|
24
|
Patterson S, Alphey MS, Jones DC, Shanks EJ, Street IP, Frearson JA, Wyatt PG, Gilbert IH, Fairlamb AH. Dihydroquinazolines as a novel class of Trypanosoma brucei trypanothione reductase inhibitors: discovery, synthesis, and characterization of their binding mode by protein crystallography. J Med Chem 2011; 54:6514-30. [PMID: 21851087 PMCID: PMC3188286 DOI: 10.1021/jm200312v] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Trypanothione reductase (TryR) is a genetically validated drug target in the parasite Trypanosoma brucei , the causative agent of human African trypanosomiasis. Here we report the discovery, synthesis, and development of a novel series of TryR inhibitors based on a 3,4-dihydroquinazoline scaffold. In addition, a high resolution crystal structure of TryR, alone and in complex with substrates and inhibitors from this series, is presented. This represents the first report of a high resolution complex between a noncovalent ligand and this enzyme. Structural studies revealed that upon ligand binding the enzyme undergoes a conformational change to create a new subpocket which is occupied by an aryl group on the ligand. Therefore, the inhibitor, in effect, creates its own small binding pocket within the otherwise large, solvent exposed active site. The TryR-ligand structure was subsequently used to guide the synthesis of inhibitors, including analogues that challenged the induced subpocket. This resulted in the development of inhibitors with improved potency against both TryR and T. brucei parasites in a whole cell assay.
Collapse
Affiliation(s)
- Stephen Patterson
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee , Dow Street, Dundee DD1 5EH, U.K
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bello ML, Chiaradia LD, Dias LRS, Pacheco LK, Stumpf TR, Mascarello A, Steindel M, Yunes RA, Castro HC, Nunes RJ, Rodrigues CR. Trimethoxy-chalcone derivatives inhibit growth of Leishmania braziliensis: synthesis, biological evaluation, molecular modeling and structure-activity relationship (SAR). Bioorg Med Chem 2011; 19:5046-52. [PMID: 21757358 DOI: 10.1016/j.bmc.2011.06.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 06/03/2011] [Accepted: 06/08/2011] [Indexed: 01/08/2023]
Abstract
In this work we described the synthesis, the antileishmanial activity and the molecular modeling and structure-activity relationship (SAR) evaluations of a series of chalcone derivatives. Among these compounds, the methoxychalcones 2h, 2i, 2j, 2k and 2l showed significant antileishmanial activity (IC(50)<10 μM). Interestingly 2i (IC(50)=2.7 μM), 2j (IC(50)=3.9 μM) and 2k (IC(50)=4.6 μM) derivatives presented better antileishmanial activity than the control drug pentamidine (IC(50)=6.0 μM). Our SAR study showed the importance of methoxy di-ortho substitution at phenyl ring A and the relationship between the frontier orbital HOMO coefficients distribution of these molecules and their activity. The most active compounds 2h, 2i, 2j, 2k, and 2l fulfilled the Lipinski rule-of-five which theoretically is important for good drug absorption and permeation through biological membranes. The potential profile of 2j (IC(50)=3.9 μM and CC(50)=216 μM) pointed this chalcone derivative as a hit compound to be further explored in antileishmanial drug design.
Collapse
Affiliation(s)
- Murilo Lamim Bello
- Laboratório de Modelagem Molecular e QSAR (ModMolQSAR-3D), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bongarzone S, Bolognesi ML. The concept of privileged structures in rational drug design: focus on acridine and quinoline scaffolds in neurodegenerative and protozoan diseases. Expert Opin Drug Discov 2011; 6:251-68. [PMID: 22647203 DOI: 10.1517/17460441.2011.550914] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION For nearly 20 years, privileged structures have offered an optimal source of core scaffolds and capping fragments for the design of combinatorial libraries directed at a broad spectrum of targets. From describing structures promiscuous within a given target family, the concept has evolved to include frameworks that can modulate proteins lacking a strict target class relation. AREAS COVERED Based on a literature search from 2000 to 2010, we discuss how two privileged motifs, quinolines and acridines, are particularly recurrent in compounds active against two quite different pathologies, neurodegenerative and protozoan diseases. EXPERT OPINION As privileged structures, quinolines and acridines could improve the productivity of drug discovery projects in the field of neurodegenerative and protozoan diseases. They could be particularly relevant for protozoan diseases because of the importance of cost-effective strategies and less stringent intellectual property concerns. Furthermore, because of their inherent affinity for various targets, privileged structures could offer a viable starting point in the search for novel multi-target ligands. Finally, from a broader perspective, they can serve as effective probes for investigating unknown but interrelated mechanisms of action.
Collapse
Affiliation(s)
- Salvatore Bongarzone
- Statistical and Biological Physics Sector, Scuola Internazionale Superiore di Studi Avanzati - International School for Advanced Studies, (SISSA-ISAS), Italian Institute of Technology, SISSA-ISAS Unit, 34151 Trieste, Italy
| | | |
Collapse
|
27
|
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: 7.4] [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
|
28
|
Jacobs RT, Ding C. Recent Advances in Drug Discovery for Neglected Tropical Diseases Caused by Infective Kinetoplastid Parasites. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2010. [DOI: 10.1016/s0065-7743(10)45017-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
Abstract
The protozoan parasitesTrypanosoma bruceiandTrypanosoma cruziare the causative agents of African trypanosomiasis and Chagas disease, respectively. These are debilitating infections that exert a considerable health burden on some of the poorest people on the planet. Treatment of trypanosome infections is dependent on a small number of drugs that have limited efficacy and can cause severe side effects. Here, we review the properties of these drugs and describe new findings on their modes of action and the mechanisms by which resistance can arise. We further outline how a greater understanding of parasite biology is being exploited in the search for novel chemotherapeutic agents. This effort is being facilitated by new research networks that involve academic and biotechnology/pharmaceutical organisations, supported by public–private partnerships, and are bringing a new dynamism and purpose to the search for trypanocidal agents.
Collapse
|
30
|
Cavalli A, Lizzi F, Bongarzone S, Belluti F, Piazzi L, Bolognesi ML. Complementary medicinal chemistry-driven strategies toward new antitrypanosomal and antileishmanial lead drug candidates. ACTA ACUST UNITED AC 2009; 58:51-60. [PMID: 19845762 DOI: 10.1111/j.1574-695x.2009.00615.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Trypanosomiases and Leishmaniases are neglected tropical diseases that affect the less developed countries. For this reason, they did not and still do not have high visibility in Western societies. The name neglected diseases also refers to the fact that they often received little interest at the level of public investment, research and development. The drug discovery scenario, however, is changing dramatically. After a period in which different socioeconomic factors have prevented massive research efforts in this field, such efforts have increased considerably in the very recent years, with significant scientific advancements. In this context, we have embarked on a new drug discovery project devoted to identification of new small molecules for the treatment of trypanosomal and leishmanial diseases. Two complementary approaches have been pursued and are reported here. The first deals with a structure-based drug design, and a privileged structure-guided synthesis of quinazoline compounds able to modulate trypanothione reductase activity was accomplished. In the second, a combinatorial library, built on a natural product-based strategy, was synthesized. Using whole parasite assays, different quinones have been identified as promising lead compounds. A combination of both approaches to hopefully overcome some of the challenges of anti-trypanosomatid drug discovery has eventually been proposed.
Collapse
Affiliation(s)
- Andrea Cavalli
- Department of Pharmaceutical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | | | | | | | | | | |
Collapse
|