1
|
Sit SY, Chen Y, Chen J, Venables BL, Swidorski JJ, Xu L, Sin N, Hartz RA, Lin Z, Zhang S, Li Z, Wu DR, Li P, Kempson J, Hou X, Shanmugam Y, Parker D, Jenkins S, Simmermacher J, Falk P, McAuliffe B, Cockett M, Hanumegowda U, Dicker I, Krystal M, Meanwell NA, Regueiro-Ren A. Invention of VH-937, a Potent HIV-1 Maturation Inhibitor with the Potential for Infrequent Oral Dosing in Humans. ACS Med Chem Lett 2024; 15:1997-2004. [PMID: 39563829 PMCID: PMC11571082 DOI: 10.1021/acsmedchemlett.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Newer generation HIV-1 maturation inhibitors have proven to be viable antiretroviral agents in the clinic. VH3739937, (VH-937, 24) is an advanced HIV-1 maturation inhibitor (MI) with a 4-cyanopyridyl ether replacing the fluorine present in the previous lead MI GSK3640254 (GSK254, 3). The introduction of aromatic methylene ethers α to the carboxylic acid moiety significantly enhanced the antiviral profile, with additional inhibitory effects observed toward the A364V mutation, the primary resistance mutation emerging in response to selective pressure by MIs. Structure-activity optimization led to the invention of VH-937, which combined the best overall antiviral profile with pharmacokinetic properties in animal models. These properties indicate the potential for infrequent dosing, a finding confirmed in initial clinical studies in humans that suggests its potential as a once-weekly dosing agent.
Collapse
Affiliation(s)
- Sing-Yuen Sit
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Yan Chen
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Jie Chen
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Brian L Venables
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Jacob J Swidorski
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Li Xu
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Ny Sin
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Richard A Hartz
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Zeyu Lin
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Sharon Zhang
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Zhufang Li
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Dauh-Rurng Wu
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Peng Li
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - James Kempson
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Xiaoping Hou
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Yoganand Shanmugam
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Dawn Parker
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Susan Jenkins
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Jean Simmermacher
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Paul Falk
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Brian McAuliffe
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Mark Cockett
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Umesh Hanumegowda
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Ira Dicker
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Mark Krystal
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- ViiV Healthcare, 36 East Industrial Road, Branford, Connecticut 06405, United States
| | - Nicholas A Meanwell
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Alicia Regueiro-Ren
- Departments of Discovery Chemistry, Virology, Pharmaceutical Candidate Optimization, and Discovery Synthesis, Bristol Myers Squibb Research and Early Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
2
|
Dupouy B, Donzel M, Roignant M, Charital S, Keumoe R, Yamaryo-Botté Y, Feckler A, Bundschuh M, Bordat Y, Rottmann M, Mäser P, Botté CY, Blandin SA, Besteiro S, Davioud-Charvet E. 3-Benzylmenadiones and their Heteroaromatic Analogues Target the Apicoplast of Apicomplexa Parasites: Synthesis and Bioimaging Studies. ACS Infect Dis 2024; 10:3553-3576. [PMID: 39327729 DOI: 10.1021/acsinfecdis.4c00304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
The apicoplast is an essential organelle for the viability of apicomplexan parasites Plasmodium falciparum or Toxoplasma gondii, which has been proposed as a suitable drug target for the development of new antiplasmodial drug-candidates. Plasmodione, an antimalarial redox-active lead drug is active at low nM concentrations on several blood stages of Plasmodiumsuch as early rings and gametocytes. Nevertheless, its precise biological targets remain unknown. Here, we described the synthesis and the evaluation of new heteroaromatic analogues of plasmodione, active on asexual blood P. falciparum stages and T. gondii tachyzoites. Using a bioimaging-based analysis, we followed the morphological alterations of T. gondii tachyzoites and revealed a specific loss of the apicoplast upon drug treatment. Lipidomic and fluxomic analyses determined that drug treatment severely impacts apicoplast-hosted FASII activity in T. gondii tachyzoites, further supporting that the apicoplast is a primary target of plasmodione analogues. To follow the drug localization, "clickable" analogues of plasmodione were designed as tools for fluorescence imaging through a Cu(I)-catalyzed azide-alkyne cycloaddition reaction. Short-time incubation of two probes with P. falciparum trophozoites and T. gondii tachyzoites showed that the clicked products localize within, or in the vicinity of, the apicoplast of both Apicomplexa parasites. In P. falciparum, the fluorescence signal was also associated with the mitochondrion, suggesting that bioactivation and activity of plasmodione and related analogues are potentially associated with these two organelles in malaria parasites.
Collapse
Affiliation(s)
- Baptiste Dupouy
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Bio(in)organic & Medicinal Chemistry Team, European School of Chemistry, Polymers and Materials (ECPM), 25, Rue Becquerel, Strasbourg F-67087, France
| | - Maxime Donzel
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Bio(in)organic & Medicinal Chemistry Team, European School of Chemistry, Polymers and Materials (ECPM), 25, Rue Becquerel, Strasbourg F-67087, France
| | - Matthieu Roignant
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Bio(in)organic & Medicinal Chemistry Team, European School of Chemistry, Polymers and Materials (ECPM), 25, Rue Becquerel, Strasbourg F-67087, France
| | - Sarah Charital
- Apicolipid Team, Institut pour l'Avancée des Biosciences, CNRS UMR5309,INSERM U1209, Université Grenoble Alpes, Bat. Jean Roget, Domaine de la Merci, La Tronche F-38700, France
| | - Rodrigue Keumoe
- INSERM, CNRS, Université de Strasbourg, U1257/UPR9022, Mosquito Immune Responses IBMC, 2 Allée Konrad Roentgen, Strasbourg F-67000, France
| | - Yoshiki Yamaryo-Botté
- Apicolipid Team, Institut pour l'Avancée des Biosciences, CNRS UMR5309,INSERM U1209, Université Grenoble Alpes, Bat. Jean Roget, Domaine de la Merci, La Tronche F-38700, France
| | - Alexander Feckler
- Functional Aquatic Ecotoxicology, Institute for Environmental Sciences (iES), RPTU Kaiserslautern-Landau, Fortstrasse 7, Landau D-76829, Germany
| | - Mirco Bundschuh
- Functional Aquatic Ecotoxicology, Institute for Environmental Sciences (iES), RPTU Kaiserslautern-Landau, Fortstrasse 7, Landau D-76829, Germany
| | - Yann Bordat
- UMR5294 CNRS-Université de Montpellier, Laboratory of Pathogens and Host Immunity (LPHI), Place Eugène Bataillon, Bâtiment 24, CC 107, Montpellier cedex 5 F-34095, France
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil CH-4123, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil CH-4123, Switzerland
- University of Basel, Petersgraben 1, Basel CH-4001, Switzerland
| | - Cyrille Y Botté
- Apicolipid Team, Institut pour l'Avancée des Biosciences, CNRS UMR5309,INSERM U1209, Université Grenoble Alpes, Bat. Jean Roget, Domaine de la Merci, La Tronche F-38700, France
| | - Stéphanie A Blandin
- INSERM, CNRS, Université de Strasbourg, U1257/UPR9022, Mosquito Immune Responses IBMC, 2 Allée Konrad Roentgen, Strasbourg F-67000, France
| | - Sébastien Besteiro
- UMR5294 CNRS-Université de Montpellier, Laboratory of Pathogens and Host Immunity (LPHI), Place Eugène Bataillon, Bâtiment 24, CC 107, Montpellier cedex 5 F-34095, France
| | - Elisabeth Davioud-Charvet
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), Bio(in)organic & Medicinal Chemistry Team, European School of Chemistry, Polymers and Materials (ECPM), 25, Rue Becquerel, Strasbourg F-67087, France
| |
Collapse
|
3
|
Mohamed AM, Abou-Ghadir OMF, Mostafa YA, Dahlous KA, Bräse S, Youssif BGM. Design and synthesis of new 1,2,4-oxadiazole/quinazoline-4-one hybrids with antiproliferative activity as multitargeted inhibitors. Front Chem 2024; 12:1447618. [PMID: 39281035 PMCID: PMC11393688 DOI: 10.3389/fchem.2024.1447618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction The combination of BRAF and tyrosine kinase (TK) inhibitors has been demonstrated to be highly effective in inhibiting tumor development and is an approach for overcoming resistance in clinical trials. Accordingly, a novel series of 1,2,4-oxadiazole/quinazoline-4-one hybrids was developed as antiproliferative multitargeted inhibitors. Methods The structures of the newly synthesized compounds 9a-o were validated using IR, NMR, MS, and elemental techniques. 9a-o were tested as antiproliferative agents. Results and Discussion The results showed that the majority of the tested compounds showed significant antiproliferative action with 9b, 9c, 9h, 9k, and 9l being the most potent. Compounds 9b, 9c, 9h, 9k, and 9l were tested as EGFR and BRAFV600E inhibitors. These in vitro tests revealed that compounds 9b, 9c, and 9h are strong antiproliferative agents that may act as dual EGFR/BRAFV600E inhibitors. 9b, 9c, and 9h were further investigated for their inhibitory effect on mutant EGFR (EGFRT790M), and the results showed that the tested compounds had considerable inhibitory action. Cell cycle study and apoptosis detection demonstrated that compound 9b exhibits cell cycle arrest at the G2/M transition. Molecular docking simulations reveal the binding mechanism of the most active antiproliferative agents.
Collapse
Affiliation(s)
- Amira M Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Ola M F Abou-Ghadir
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Yaser A Mostafa
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Kholood A Dahlous
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, IBCS-FMS, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
4
|
Ungureanu D, Tiperciuc B, Nastasă C, Ionuț I, Marc G, Oniga I, Oniga O. An Overview of the Structure-Activity Relationship in Novel Antimicrobial Thiazoles Clubbed with Various Heterocycles (2017-2023). Pharmaceutics 2024; 16:89. [PMID: 38258100 PMCID: PMC10820536 DOI: 10.3390/pharmaceutics16010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/31/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Antimicrobial resistance is an increasing problem for global public health. One of the strategies to combat this issue is the synthesis of novel antimicrobials through rational drug design based on extensive structure-activity relationship studies. The thiazole nucleus is a prominent feature in the structure of many authorized antimicrobials, being clubbed with different heterocycles. The purpose of this review is to study the structure-activity relationship in antimicrobial thiazoles clubbed with various heterocycles, as reported in the literature between 2017 and 2023, in order to offer an overview of the last years in terms of antimicrobial research and provide a helpful instrument for future research in the field.
Collapse
Affiliation(s)
- Daniel Ungureanu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
- “Prof. Dr. Ion Chiricuță” Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Brîndușa Tiperciuc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
| | - Cristina Nastasă
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
| | - Ioana Ionuț
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
| | - Gabriel Marc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
| | - Ilioara Oniga
- Department of Pharmacognosy, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 12 Ion Creangă Street, 400010 Cluj-Napoca, Romania;
| | - Ovidiu Oniga
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (D.U.); (C.N.); (I.I.); (G.M.); (O.O.)
| |
Collapse
|
5
|
Shivers GN, Pigge FC. Palladium-catalyzed allylation of 2- and 4-alkylpyridines via N-allyl alkylidene dihydropyridine intermediates. Tetrahedron Lett 2023; 128:154701. [PMID: 37841749 PMCID: PMC10569290 DOI: 10.1016/j.tetlet.2023.154701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
A method to introduce allyl or cinnamyl groups to the picolyl positions of 2- or 4-alkylpyridines is described. Substituted N-allyl pyridinium salts are first treated with base (KOtBu) followed by catalytic [(η3-allyl)PdCl]2 and PPh3 to result in formal Pd-catalyzed transfer of N-allyl groups to the pyridine periphery. The reaction is believed to proceed through initial formation of nucleophilic alkylidene dihydropyridine intermediates that react with (π-allyl)Pd(II) electrophiles, thereby regenerating N-allyl pyridinium cations. Catalytic turnover and liberation of pyridine products is then achieved by oxidative addition of Pd(0) to these activated allyl groups.
Collapse
Affiliation(s)
- Grant N Shivers
- Department of Chemistry, University of Iowa Iowa City, Iowa, 52242, USA
| | | |
Collapse
|
6
|
Tun SL, Shivers GN, Pigge FC. C-Sulfonylation of 4-Alkylpyridines: Formal Picolyl C-H Activation via Alkylidene Dihydropyridine Intermediates. J Org Chem 2023; 88:3998-4002. [PMID: 36848377 PMCID: PMC10028608 DOI: 10.1021/acs.joc.3c00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
4-Picoline derivatives are converted to the corresponding aryl picolyl sulfones upon treatment with aryl sulfonyl chlorides and Et3N in the presence of catalytic DMAP. The reaction proceeds smoothly for a variety of alkyl and aryl picolines using a range of aryl sulfonyl chlorides. The reaction is believed to involve N-sulfonyl 4-alkylidene dihydropyridine intermediates and results in formal sulfonylation of unactivated picolyl C-H bonds.
Collapse
Affiliation(s)
- Soe L Tun
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242, United States
| | - Grant N Shivers
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242, United States
| | - F Christopher Pigge
- Department of Chemistry, University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
7
|
Meanwell NA. The pyridazine heterocycle in molecular recognition and drug discovery. Med Chem Res 2023; 32:1-69. [PMID: 37362319 PMCID: PMC10015555 DOI: 10.1007/s00044-023-03035-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/06/2023] [Indexed: 03/17/2023]
Abstract
The pyridazine ring is endowed with unique physicochemical properties, characterized by weak basicity, a high dipole moment that subtends π-π stacking interactions and robust, dual hydrogen-bonding capacity that can be of importance in drug-target interactions. These properties contribute to unique applications in molecular recognition while the inherent polarity, low cytochrome P450 inhibitory effects and potential to reduce interaction of a molecule with the cardiac hERG potassium channel add additional value in drug discovery and development. The recent approvals of the gonadotropin-releasing hormone receptor antagonist relugolix (24) and the allosteric tyrosine kinase 2 inhibitor deucravacitinib (25) represent the first examples of FDA-approved drugs that incorporate a pyridazine ring. In this review, the properties of the pyridazine ring are summarized in comparison to the other azines and its potential in drug discovery is illustrated through vignettes that explore applications that take advantage of the inherent physicochemical properties as an approach to solving challenges associated with candidate optimization. Graphical Abstract
Collapse
|
8
|
Akinnusi PA, Olubode SO, Alade AA, Ashimi AA, Onawola OL, Agbolade AO, Emeka AP, Shodehinde SA, Adeniran OY. Potential Inhibitory Biomolecular Interactions of Natural Compounds With Different Molecular Targets of Diabetes. Bioinform Biol Insights 2023; 17:11779322231167970. [PMID: 37124131 PMCID: PMC10134171 DOI: 10.1177/11779322231167970] [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: 04/23/2022] [Accepted: 03/17/2023] [Indexed: 05/02/2023] Open
Abstract
Type II diabetes is an endemic disease and is responsible for approximately 90% to 95% of diabetes cases. The pathophysiological distortions are majorly β-cell dysfunction, insulin resistance, and long-term inflammation, which all progressively unsettle the control of blood glucose levels and trigger microvascular and macrovascular complications. The diverse pathological disruptions which patients with type II diabetes mellitus exhibit precipitate the opinion that different antidiabetic agents, administered in combination, might be required to curb this menace and maintain normal blood glucose. To this end, natural compounds were screened to identify small molecular weight compounds with inhibitory effects on protein tyrosine phosphatase 1B (PTP1B), dipeptidyl-peptidase-4 (DPP-4), and α-amylase. From the result, the top 5 anthocyanins with the highest binding affinity are reported herein. Further ADMET profiling showed moderate pharmacokinetic profiles for these compounds as well as insignificant toxicity. Cyanidin 3-(p-coumaroyl)-diglucoside-5-glucoside (-15.272 kcal/mol), cyanidin 3-O-(6"-malonyl-3"-glucosyl-glucoside) (-9.691 kcal/mol), and delphinidin 3,5-O-diglucoside (-12.36 kcal/mol) had the highest binding affinities to PTP1B, DPP-4, and α-amylase, respectively, and can be used in combination to control glucose fluctuations. However, validations must be carried out through further in vitro and in vivo tests.
Collapse
Affiliation(s)
- Precious A Akinnusi
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
- Department of Biochemistry, Federal University of Oye-Ekiti, Oye-Ekiti, Nigeria
- Precious A Akinnusi, Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko 342111, Ondo, Nigeria.
| | - Samuel O Olubode
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Adebowale A Alade
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Aderemi A Ashimi
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Olamide L Onawola
- Department of Microbiology, Lagos State University, Ojo, Nigeria
- Molecular Biology Research Laboratory, Department of Microbiology, Covenant University, Ota, Nigeria
| | - Abigail O Agbolade
- Department of Biochemistry, Federal University of Oye-Ekiti, Oye-Ekiti, Nigeria
- Department of Biochemistry, Osun State University, Osogbo, Nigeria
| | - Adaobi P Emeka
- Department of Biological Sciences, Godfrey Okoye University, Enugu, Nigeria
| | | | - Olawole Y Adeniran
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| |
Collapse
|
9
|
Humphreys PG, Anderson NA, Bamborough P, Baxter A, Chung CW, Cookson R, Craggs PD, Dalton T, Fournier JCL, Gordon LJ, Gray HF, Gray MW, Gregory R, Hirst DJ, Jamieson C, Jones KL, Kessedjian H, Lugo D, McGonagle G, Patel VK, Patten C, Poole DL, Prinjha RK, Ramirez-Molina C, Rioja I, Seal G, Stafford KAJ, Shah RR, Tape D, Theodoulou NH, Tomlinson L, Ukuser S, Wall ID, Wellaway N, White G. Identification and Optimization of a Ligand-Efficient Benzoazepinone Bromodomain and Extra Terminal (BET) Family Acetyl-Lysine Mimetic into the Oral Candidate Quality Molecule I-BET432. J Med Chem 2022; 65:15174-15207. [DOI: 10.1021/acs.jmedchem.2c01102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | - Niall A. Anderson
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Paul Bamborough
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Andrew Baxter
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Chun-wa Chung
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Rosa Cookson
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Peter D. Craggs
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Toryn Dalton
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Laurie J. Gordon
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Heather F. Gray
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Matthew W. Gray
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Richard Gregory
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - David J. Hirst
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Craig Jamieson
- WestCHEM, Department of Pure and Applied Chemistry, Thomas Graham Building, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | | | | | - David Lugo
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Grant McGonagle
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | | | - Darren L. Poole
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Rab K. Prinjha
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Inmaculada Rioja
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Gail Seal
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Rishi R. Shah
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Daniel Tape
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Laura Tomlinson
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Sabri Ukuser
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D. Wall
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Natalie Wellaway
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Gemma White
- GSK, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
10
|
Ding Y, Zhu Z, Chen M, Yu C, Zhou Y. Rhodium‐Catalyzed Asymmetric Hydrogenation of All‐Carbon Aromatic Rings. Angew Chem Int Ed Engl 2022; 61:e202205623. [DOI: 10.1002/anie.202205623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Yi‐Xuan Ding
- State Key Laboratory of Catalysis Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Zhou‐Hao Zhu
- State Key Laboratory of Catalysis Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Mu‐Wang Chen
- State Key Laboratory of Catalysis Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 P. R. China
| | - Chang‐Bin Yu
- State Key Laboratory of Catalysis Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 P. R. China
| | - Yong‐Gui Zhou
- State Key Laboratory of Catalysis Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 P. R. China
- Zhang Dayu School of Chemistry Dalian University of Technology Dalian 116023 P. R. China
| |
Collapse
|
11
|
Gomaa HAM. A Comprehensive Review of Recent Advances in the Biological Activities of Quinazolines. Chem Biol Drug Des 2022; 100:639-655. [PMID: 35920244 DOI: 10.1111/cbdd.14129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/17/2022] [Accepted: 07/30/2022] [Indexed: 11/25/2022]
Abstract
Quinazoline heterocycles are critical in the development of medications. Quinazoline derivatives have been intensively researched, providing a wide range of compounds with diverse biological roles. The quinazoline nucleus has garnered a lot of attention in medical chemistry in recent years. It was assumed to be a pharmacophore component in the development of physiologically interesting drugs. This review is an attempt to increase the potential of quinazoline by highlighting a wide range of advancements demonstrated by numerous derivatives of the quinazoline moiety, as well as focusing on diverse pharmacological actions of the quinazoline moiety. This review compiles recent studies on the quinazoline moiety described in the literature by researchers.
Collapse
Affiliation(s)
- Hesham A M Gomaa
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| |
Collapse
|
12
|
Serkov SA, Sigai NV, Kostikova NN, Fedorov AE, Gazieva GA. Synthesis and evaluation of cytotoxicity of S-substituted 5-sulfanylmethyl(ethyl)-1,3,4-thiadiazol-2-amines. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3592-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
13
|
Novel cyclohepta[b]thiophene derivative incorporating pyrimidine, pyridine, and chromene moiety as potential antimicrobial agents targeting DNA gyrase. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Zhou YG, Ding YX, Zhu ZH, Chen MW, Yu CB. Rhodium‐Catalyzed Asymmetric Hydrogenation of All‐Carbon Aromatic Rings. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202205623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yong-Gui Zhou
- Dalian Institute of Chemical Physics Department of Fine Chemicals 457 Zhongshan Road 116023 Dalian CHINA
| | - Yi-Xuan Ding
- Dalian Institute of Chemical Physics State Key Laboratory of Catalysis State Key Laboratory of Catalysis CHINA
| | - Zhou-Hao Zhu
- Dalian Institute of Chemical Physics State Key Laboratory of Catalysis State Key Laboratory of Catalysis Dalian CHINA
| | - Mu-Wang Chen
- Dalian Institute of Chemical Physics State Key Laboratory of Catalysis State Key Laboratory of Catalysis Dalian CHINA
| | - Chang-Bin Yu
- Dalian Institute of Chemical Physics State Key Laboratory of Catalysis State Key Laboratory of Catalysis Dalian CHINA
| |
Collapse
|
15
|
Subbaiah MAM, Meanwell NA. Bioisosteres of the Phenyl Ring: Recent Strategic Applications in Lead Optimization and Drug Design. J Med Chem 2021; 64:14046-14128. [PMID: 34591488 DOI: 10.1021/acs.jmedchem.1c01215] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The benzene moiety is the most prevalent ring system in marketed drugs, underscoring its historic popularity in drug design either as a pharmacophore or as a scaffold that projects pharmacophoric elements. However, introspective analyses of medicinal chemistry practices at the beginning of the 21st century highlighted the indiscriminate deployment of phenyl rings as an important contributor to the poor physicochemical properties of advanced molecules, which limited their prospects of being developed into effective drugs. This Perspective deliberates on the design and applications of bioisosteric replacements for a phenyl ring that have provided practical solutions to a range of developability problems frequently encountered in lead optimization campaigns. While the effect of phenyl ring replacements on compound properties is contextual in nature, bioisosteric substitution can lead to enhanced potency, solubility, and metabolic stability while reducing lipophilicity, plasma protein binding, phospholipidosis potential, and inhibition of cytochrome P450 enzymes and the hERG channel.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon-Bristol Myers Squibb Research and Development Centre, Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bangalore, Karnataka 560099, India
| | - Nicholas A Meanwell
- Department of Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
16
|
Wang T, Kadow JF, Meanwell NA. Innovation in the discovery of the HIV-1 attachment inhibitor temsavir and its phosphonooxymethyl prodrug fostemsavir. Med Chem Res 2021; 30:1955-1980. [PMID: 34602806 PMCID: PMC8476988 DOI: 10.1007/s00044-021-02787-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 11/25/2022]
Abstract
The discovery and development of fostemsavir (2), the tromethamine salt of the phosphonooxymethyl prodrug of temsavir (1), encountered significant challenges at many points in the preclinical and clinical development program that, in many cases, stimulated the implementation of innovative solutions in order to enable further progression. In the preclinical program, a range of novel chemistry methodologies were developed during the course of the discovery effort that enabled a thorough examination and definition of the HIV-1 attachment inhibitor (AI) pharmacophore. These discoveries helped to address the challenges associated with realizing a molecule with all of the properties necessary to successfully advance through development and this aspect of the program is the major focus of this retrospective. Although challenges and innovation are not unusual in drug discovery and development programs, the HIV-1 AI program is noteworthy not only because of the serial nature of the challenges encountered along the development path, but also because it resulted in a compound that remains the first and only example of a mechanistically novel class of HIV-1 inhibitor that is proving to be very beneficial for controlling virus levels in highly treatment-experienced HIV-1 infected patients. ![]()
Collapse
Affiliation(s)
- Tao Wang
- Beijing Kawin Technology Share-Holdiing Co., 6 Rongjing East Street, BDA, Beijing, PR China
| | - John F Kadow
- ViiV Healthcare, 36 East Industrial Road, Branford, CT 06405 USA
| | - Nicholas A Meanwell
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ 08543-4000 USA
| |
Collapse
|
17
|
Gu X, Ma S. Recent Advances in the Development of Pyrazolopyridines as Anticancer Agents. Anticancer Agents Med Chem 2021; 22:1643-1657. [PMID: 34488593 DOI: 10.2174/1871520621666210901102832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/12/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022]
Abstract
Cancer, especially malignant tumor, is a serious threat to people's life and health. It is recognized as an enormous challenge in the 21st century. Continuous efforts are needed to overcome this problem. Pyrazolopyridine nucleus, similar in structure to purine, shows a variety of biological activities, which is mainly attributed to the antagonistic nature towards the natural purines in many biological processes. This has aroused enormous attention for many researchers. At present, a large number of new chemical entities containing pyrazolopyridine nucleus have been found as anticancer agents. In this review we summarize novel pyrazolopyridine-containing derivatives with biological activities. Furthermore, we outline the relationships between the structures of variously modified pyrazolopyridines and their anticancer activity.
Collapse
Affiliation(s)
- Xiaotong Gu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012. China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012. China
| |
Collapse
|
18
|
Vinogradova EE, Gazieva GA, Izmest'ev AN, Karnoukhova VA, Kravchenko AN. Dimroth-type N/S-interchange of N-aminothioglycolurils in the synthesis of 2-hydrazonoimidazo[4,5- d]thiazolones. RSC Adv 2021; 11:28395-28400. [PMID: 35480725 PMCID: PMC9038022 DOI: 10.1039/d1ra05568b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/14/2021] [Indexed: 11/21/2022] Open
Abstract
An original method for the synthesis of 2-hydrazonoimidazo[4,5-d]thiazolone derivatives has been developed based on a one-pot acid-induced sequence of hydrazone formation from 3-thioxoperhydroimidazo[4,5-e]-1,2,4-triazinones and aromatic aldehydes, triazine ring contraction to imidazolidine one, and Dimroth-type N/S-interchange of N-aminothioglycolurils formed in situ into 2-hydrazonoimidazo[4,5-d]thiazolones. 3-Phenylacroleine derivatives are also suitable substrates for the reaction with thioxoperhydroimidazotriazinones. An original method for the synthesis of 2-hydrazonoimidazo[4,5-d]thiazolone derivatives has been developed based on an acid-induced Dimroth-type N/S-interchange of N-aminothioglycolurils formed in situ into 2-hydrazonoimidazo[4,5-d]thiazolones.![]()
Collapse
Affiliation(s)
- Ekaterina E Vinogradova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Leninsky Prosp., 47 Moscow 119991 Russian Federation
| | - Galina A Gazieva
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Leninsky Prosp., 47 Moscow 119991 Russian Federation
| | - Alexei N Izmest'ev
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Leninsky Prosp., 47 Moscow 119991 Russian Federation
| | - Valentina A Karnoukhova
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences Vavilova Str., 28 Moscow 119991 Russian Federation
| | - Angelina N Kravchenko
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Leninsky Prosp., 47 Moscow 119991 Russian Federation .,Plekhanov Russian University of Economics Stremyanny Lane, 36 Moscow 117997 Russian Federation
| |
Collapse
|
19
|
Modelling of an autonomous Nav1.5 channel system as a part of in silico pharmacology study. J Mol Model 2021; 27:182. [PMID: 34031769 DOI: 10.1007/s00894-021-04799-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 05/17/2021] [Indexed: 12/22/2022]
Abstract
A homology model of Nav1.5, based mainly on the crystal structures of Nav1.2/1.5 was built, optimized and successfully inserted into the membrane bilayer. We applied steered and free MD simulation protocols for the visualization of the mechanism of Nav1.5 activation. We constrained dihedrals of S4 trigger to introduce a structural tension with further rearrangement and movement of secondary structure elements. From these, we observed an intracellular gate opening and movement of the Lys1419 residue caused by a gradual displacement of the distal S6 α-helix with the extended S4 3-10 helix of voltage-sensing domains (VSD). A construction containing the Lys1419 residue in P-loop also changed its position due to the extension of this helix and subsequent induction of the pore-forming helixes motion. From this point, a double membrane system was generated, implying a free of ligand Nav1.5 protein and on the opposite side its copy containing a docked bupivacaine molecule inside the pore channel. The system can be used for the design of selective inhibitors against the Nav1.7 channel, instead of mixed effect on both channels.
Collapse
|
20
|
Murai T, Krisna Puji Pamungkas K, Hattori S, Maruyama T, Ebihara M. Synthesis of 5-H Thiazoles via Thioamide Dianions with Thioformamides: Pyridylmethyl Group on the Nitrogen Atom of Thiazole Promotes the Formation of 5-H Thiazoles. HETEROCYCLES 2021. [DOI: 10.3987/com-20-s(k)7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
21
|
Synthesis, biological evaluation and QSAR studies of new thieno[2,3-d]pyrimidin-4(3H)-one derivatives as antimicrobial and antifungal agents. Bioorg Chem 2020; 106:104509. [PMID: 33288321 DOI: 10.1016/j.bioorg.2020.104509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 11/24/2022]
Abstract
A series of new thieno[2,3-d]pyrimidin-4(3H)-one derivatives were synthesized and evaluated for their activity against four gram-positive and four gram-negative bacterial and eight fungal species. The majority of the compounds exhibited excellent antimicrobial and antifungal activity, being more potent than the control compounds. Compound 22, bearing a m-methoxyphenyl group and an ethylenediamine side chain anchored at C-2 of the thienopyrimidinone core, is the most potent antibacterial compound with broad antimicrobial activity with MIC values in the range of 0.05-0.13 mM, being 6 to 15 fold more potent than the controls, streptomycin and ampicillin. Furthermore, compounds 14 and 15 which bear a p-chlorophenyl and m-methoxyphenyl group, respectively, and share a 2-(2-mercaptoethoxy)ethan-1-ol side chain showed the best antifungal activity, being 10-15 times more potent than ketoconazole or bifonazole with MIC values 0.013-0.026 and 0.027 mM, respectively. Especially in the case of compound 15 the low MIC values were accompanied by excellent MFC values ranging from 0.056 to 0.058 mM. Evaluation of toxicity in vitro on HFL-1 human embryonic primary cells and in vivo in the nematode C. elegans revealed no toxic effects for both compounds 15 and 22 tested at the MIC concentrations. Ligand-based similarity search and molecular docking predicted that the antibacterial activity of analogue 22 is related to inhibition of the topoisomerase II DNA gyrase enzyme and the antifungal activity of compound 15 to CYP51 lanosterol demethylase enzyme. R-Group analysis as a means of computational structure activity relationship tool, highlighted the compounds' crucial pharmacophore features and their impact on the antibacterial and antifungal activity. The presence of a N-methyl piperidine ring fused to the thienopyrimidinone core plays an important role in both activities.
Collapse
|
22
|
Bansal R, Malhotra A. Therapeutic progression of quinazolines as targeted chemotherapeutic agents. Eur J Med Chem 2020; 211:113016. [PMID: 33243532 DOI: 10.1016/j.ejmech.2020.113016] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/16/2020] [Accepted: 11/08/2020] [Indexed: 02/08/2023]
Abstract
Presently cancer is a grave health issue with predominance beyond restrictions. It can affect any organ of the body. Most of the available chemotherapeutic drugs are highly toxic, not much selective and eventually lead to the development of resistance. Therefore, a target specific palliative approach for the treatment of cancer is required. Remarkable advancements in science have illuminated various molecular pathways responsible for cancer. This has resulted in abundant opportunities to develop targeted anticancer agents. Quinazoline nucleus is a privileged scaffold with significant diversified pharmacological activities. Numerous established anticancer quinazoline derivatives constitute a new class of chemotherapeutic agents which are found to act by inhibiting various protein kinases as well as other molecular targets. A recent update on various quinazoline derivatives acting on different types of molecular targets for the treatment of cancer has been compiled in this review. Brief SAR studies of quinazoline derivatives acting through different mechanisms of action have been highlighted. The comprehensive medicinal chemistry aspects of these agents in this review provide a panoramic view to the biologists as well as medicinal chemists working in this area and would assist them in their efforts to design and synthesize novel quinazoline based anticancer compounds.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Sector-14, Panjab University, Chandigarh, 160014, India.
| | - Anjleena Malhotra
- University Institute of Pharmaceutical Sciences, Sector-14, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
23
|
Kim AN, Stoltz BM. Recent Advances in Homogeneous Catalysts for the Asymmetric Hydrogenation of Heteroarenes. ACS Catal 2020; 10:13834-13851. [PMID: 34567830 PMCID: PMC8460131 DOI: 10.1021/acscatal.0c03958] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The asymmetric hydrogenation of heteroarenes has recently emerged as an effective strategy for the direct access to enantioenriched, saturated heterocycles. Although several homogeneous catalyst systems have been extensively developed for the hydrogenation of heteroarenes with high levels of chemo- and stereoselectivity, the development of mild conditions that allow for efficient and stereoselective hydrogenation of a broad range of substrates remains a challenge. This Perspective highlights recent advances in homogeneous catalysis of heteroarene hydrogenation as inspiration for the further development of asymmetric hydrogenation catalysts, and addresses underdeveloped areas and limitations of the current technology.
Collapse
Affiliation(s)
- Alexia N. Kim
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Brian M. Stoltz
- The Warren and Katharine Schlinger Laboratory for Chemistry and Chemical Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
24
|
Cascioferro S, Parrino B, Carbone D, Schillaci D, Giovannetti E, Cirrincione G, Diana P. Thiazoles, Their Benzofused Systems, and Thiazolidinone Derivatives: Versatile and Promising Tools to Combat Antibiotic Resistance. J Med Chem 2020; 63:7923-7956. [PMID: 32208685 PMCID: PMC7997583 DOI: 10.1021/acs.jmedchem.9b01245] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Thiazoles,
their benzofused systems, and thiazolidinone derivatives
are widely recognized as nuclei of great value for obtaining molecules
with various biological activities, including analgesic, anti-inflammatory,
anti-HIV, antidiabetic, antitumor, and antimicrobial. In particular,
in the past decade, many compounds bearing these heterocycles have
been studied for their promising antibacterial properties due to their
action on different microbial targets. Here we assess the recent development
of this class of compounds to address mechanisms underlying antibiotic
resistance at both bacterial-cell and community levels (biofilms).
We also explore the SAR and the prospective clinical application of
thiazole and its benzofused derivatives, which act as inhibitors of
mechanisms underlying antibiotic resistance in the treatment of severe
drug-resistant infections. In addition, we examined all bacterial
targets involved in their antimicrobial activity reporting, when described,
their spontaneous frequencies of resistance.
Collapse
Affiliation(s)
- Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Daniela Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Domenico Schillaci
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, DeBoelelaan 1117, 1081HV, Amsterdam, The Netherlands.,Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, via Giovannini 13, 56017 San Giuliano Terme, Pisa, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
25
|
Belen’kii LI, Evdokimenkova YB. The literature of heterocyclic chemistry, part XVII, 2017. ADVANCES IN HETEROCYCLIC CHEMISTRY 2019:337-418. [DOI: 10.1016/bs.aihch.2019.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Nakagawa Y, Sawaki Y, Kimura T, Tomura T, Igarashi Y, Ojika M. Quinocidin, a Cytotoxic Antibiotic with an Unusual 3,4-Dihydroquinolizinium Ring and Michael Acceptor Reactivity toward Thiols. Chemistry 2017; 23:17894-17897. [DOI: 10.1002/chem.201704729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Yu Nakagawa
- Department of Applied Molecular Biosciences; Graduate School of Bioagricultural Sciences; Nagoya University; Furo-cho Chikusa-ku Nagoya 464-8601 Japan
| | - Yuki Sawaki
- Department of Applied Molecular Biosciences; Graduate School of Bioagricultural Sciences; Nagoya University; Furo-cho Chikusa-ku Nagoya 464-8601 Japan
| | - Takahiro Kimura
- Department of Applied Molecular Biosciences; Graduate School of Bioagricultural Sciences; Nagoya University; Furo-cho Chikusa-ku Nagoya 464-8601 Japan
| | - Tomohiko Tomura
- Department of Applied Molecular Biosciences; Graduate School of Bioagricultural Sciences; Nagoya University; Furo-cho Chikusa-ku Nagoya 464-8601 Japan
| | - Yasuhiro Igarashi
- Biotechnology Research Center; Toyama Prefectural University; 5180 Kurokawa Imazu Toyama 939-0398 Japan
| | - Makoto Ojika
- Department of Applied Molecular Biosciences; Graduate School of Bioagricultural Sciences; Nagoya University; Furo-cho Chikusa-ku Nagoya 464-8601 Japan
| |
Collapse
|
27
|
Azad CS, Narula AK. Substituted, Fused, Tricyclic 6,7-Dihydro-1H
,5H
-pyrido[1,2,3-de
]quinoxaline-3-amines by Isocyanide-Abetted Cycloaddition Reaction. European J Org Chem 2017. [DOI: 10.1002/ejoc.201701118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Chandra S. Azad
- “Hygeia”, Centre of Excellence in Pharmaceutical Sciences; Guru Gobind Singh Indraprastha University; Sector 16-C, Dwarka 110078 New Delhi India
| | - Anudeep K. Narula
- “Hygeia”, Centre of Excellence in Pharmaceutical Sciences; Guru Gobind Singh Indraprastha University; Sector 16-C, Dwarka 110078 New Delhi India
| |
Collapse
|
28
|
Barone V, Cacelli I, Ferretti A, Prampolini G. Noncovalent Interactions in the Catechol Dimer. Biomimetics (Basel) 2017; 2:E18. [PMID: 31105180 PMCID: PMC6352673 DOI: 10.3390/biomimetics2030018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 12/02/2022] Open
Abstract
Noncovalent interactions play a significant role in a wide variety of biological processes and bio-inspired species. It is, therefore, important to have at hand suitable computational methods for their investigation. In this paper, we report on the contribution of dispersion and hydrogen bonds in both stacked and T-shaped catechol dimers, with the aim of delineating the respective role of these classes of interactions in determining the most stable structure. By using second-order Møller⁻Plesset (MP2) calculations with a small basis set, specifically optimized for these species, we have explored a number of significant sections of the interaction potential energy surface and found the most stable structures for the dimer, in good agreement with the highly accurate, but computationally more expensive coupled cluster single and double excitation and the perturbative triples (CCSD(T))/CBS) method.
Collapse
Affiliation(s)
- Vincenzo Barone
- Scuola Normale Superiore di Pisa, Piazza dei Cavalieri, I-56126 Pisa, Italy.
| | - Ivo Cacelli
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy.
- Istituto di Chimica dei Composti OrganoMetallici (ICCOM-CNR), Area della Ricerca, Via G. Moruzzi 1, I-56124 Pisa, Italy.
| | - Alessandro Ferretti
- Istituto di Chimica dei Composti OrganoMetallici (ICCOM-CNR), Area della Ricerca, Via G. Moruzzi 1, I-56124 Pisa, Italy.
| | - Giacomo Prampolini
- Istituto di Chimica dei Composti OrganoMetallici (ICCOM-CNR), Area della Ricerca, Via G. Moruzzi 1, I-56124 Pisa, Italy.
| |
Collapse
|
29
|
Yeung KS, Beno BR, Parcella K, Bender JA, Grant-Young KA, Nickel A, Gunaga P, Anjanappa P, Bora RO, Selvakumar K, Rigat K, Wang YK, Liu M, Lemm J, Mosure K, Sheriff S, Wan C, Witmer M, Kish K, Hanumegowda U, Zhuo X, Shu YZ, Parker D, Haskell R, Ng A, Gao Q, Colston E, Raybon J, Grasela DM, Santone K, Gao M, Meanwell NA, Sinz M, Soars MG, Knipe JO, Roberts SB, Kadow JF. Discovery of a Hepatitis C Virus NS5B Replicase Palm Site Allosteric Inhibitor (BMS-929075) Advanced to Phase 1 Clinical Studies. J Med Chem 2017; 60:4369-4385. [PMID: 28430437 DOI: 10.1021/acs.jmedchem.7b00328] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The hepatitis C virus (HCV) NS5B replicase is a prime target for the development of direct-acting antiviral drugs for the treatment of chronic HCV infection. Inspired by the overlay of bound structures of three structurally distinct NS5B palm site allosteric inhibitors, the high-throughput screening hit anthranilic acid 4, the known benzofuran analogue 5, and the benzothiadiazine derivative 6, an optimization process utilizing the simple benzofuran template 7 as a starting point for a fragment growing approach was pursued. A delicate balance of molecular properties achieved via disciplined lipophilicity changes was essential to achieve both high affinity binding and a stringent targeted absorption, distribution, metabolism, and excretion profile. These efforts led to the discovery of BMS-929075 (37), which maintained ligand efficiency relative to early leads, demonstrated efficacy in a triple combination regimen in HCV replicon cells, and exhibited consistently high oral bioavailability and pharmacokinetic parameters across preclinical animal species. The human PK properties from the Phase I clinical studies of 37 were better than anticipated and suggest promising potential for QD administration.
Collapse
Affiliation(s)
- Kap-Sun Yeung
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Brett R Beno
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Kyle Parcella
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - John A Bender
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Katherine A Grant-Young
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Andrew Nickel
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Prashantha Gunaga
- Department of Discovery Chemistry, Biocon Bristol-Myers Squibb Research and Development Center , Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Prakash Anjanappa
- Department of Discovery Chemistry, Biocon Bristol-Myers Squibb Research and Development Center , Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Rajesh Onkardas Bora
- Department of Discovery Chemistry, Biocon Bristol-Myers Squibb Research and Development Center , Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Kumaravel Selvakumar
- Department of Discovery Chemistry, Biocon Bristol-Myers Squibb Research and Development Center , Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Karen Rigat
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Ying-Kai Wang
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Mengping Liu
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Julie Lemm
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Kathy Mosure
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Steven Sheriff
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Changhong Wan
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Mark Witmer
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kevin Kish
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Umesh Hanumegowda
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Xiaoliang Zhuo
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Yue-Zhong Shu
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dawn Parker
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Roy Haskell
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Alicia Ng
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Qi Gao
- Bristol-Myers Squibb Research and Development , 1 Squibb Drive, New Brunswick, New Jersey 08901, United States
| | - Elizabeth Colston
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joseph Raybon
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dennis M Grasela
- Bristol-Myers Squibb Research and Development , P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kenneth Santone
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Min Gao
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Nicholas A Meanwell
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Michael Sinz
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Matthew G Soars
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Jay O Knipe
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Susan B Roberts
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - John F Kadow
- Bristol-Myers Squibb Research and Development , P.O. Box 5100, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| |
Collapse
|