Future medicinal chemistry最新文献

筛选
英文 中文
Hybrid chalcone-pyrazoline derivatives: synthesis and in silico evaluation for anticancer potential. 杂化查尔酮-吡唑啉衍生物:合成及其抗癌潜力的硅评价。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-15 DOI: 10.1080/17568919.2025.2561464
Purabi Saha, Azra Yasmin, Ritesh Jha, Aarti Passi, Vikramdeep Monga, Shammy Jindal, Kamya Goyal
{"title":"Hybrid chalcone-pyrazoline derivatives: synthesis and in silico evaluation for anticancer potential.","authors":"Purabi Saha, Azra Yasmin, Ritesh Jha, Aarti Passi, Vikramdeep Monga, Shammy Jindal, Kamya Goyal","doi":"10.1080/17568919.2025.2561464","DOIUrl":"https://doi.org/10.1080/17568919.2025.2561464","url":null,"abstract":"<p><strong>Aim: </strong>This study aimed to design, synthesize, and evaluate a hybrid chalcone-pyrazoline derivatives as potential anticancer agents targeting B-Raf kinase in lung cancer.</p><p><strong>Material & methods: </strong>Chalcone-pyrazoline derivatives (<b>PY1</b>-<b>PY10</b>) were synthesized via Claisen-Schmidt condensation followed by cyclization, characterized using FT-IR, NMR, and LC-MS. In vitro cytotoxic activity was assessed against A549 human lung cancer cells using the MTT assay. Molecular docking studies were performed with B-Raf kinase (PDB ID: 2FB8) using Schrödinger software. ADME properties were predicted using SwissADME.</p><p><strong>Result: </strong>Compound <b>PY7</b> exhibited the most potent cytotoxicity (IC₅₀ = 6.45 µM) and the highest docking score (-8.89 kcal/mol), showing strong binding interactions with GLN530 in B-Raf kinase. Structure Activity Relationship analysis revealed that electron-withdrawing para-nitro substituents enhanced potency, while electron-donating groups generally reduced activity. ADME profiling confirmed all compounds complied with Lipinski's Rule of Five, had high gastrointestinal absorption, and displayed favorable drug-likeness.</p><p><strong>Conclusion: </strong>The findings identify <b>PY7</b> as a promising lead candidate with potent anticancer activity, strong B-Raf binding affinity, and favorable pharmacokinetics. This work supports chalcone-pyrazole scaffolds as viable templates for the development of novel targeted lung cancer therapeutics.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-13"},"PeriodicalIF":3.4,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145069461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design, development, and therapeutic applications of PARP-1 selective inhibitors. PARP-1选择性抑制剂的设计、开发和治疗应用。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-15 DOI: 10.1080/17568919.2025.2561467
Yue Xu, Xiangqian Li, Yuanyuan Zhao, Chao Xie, Jiashu Chen, Yuxi Lin, Pan Xing, Jiqiang Zhu, Bokan Wang, Dayong Shi
{"title":"Design, development, and therapeutic applications of PARP-1 selective inhibitors.","authors":"Yue Xu, Xiangqian Li, Yuanyuan Zhao, Chao Xie, Jiashu Chen, Yuxi Lin, Pan Xing, Jiqiang Zhu, Bokan Wang, Dayong Shi","doi":"10.1080/17568919.2025.2561467","DOIUrl":"https://doi.org/10.1080/17568919.2025.2561467","url":null,"abstract":"<p><p>Poly(ADP-ribose) polymerase (PARP) plays a key role in DNA damage repair and has become a critical target for tumor therapy. In recent years, several PARP inhibitors, such as Olaparib and Niraparib, have achieved clinical success in breast cancer susceptibility genes (BRCA) mutant tumors by exploiting the synthetic lethality of homologous recombination-deficient cancers. However, problems have emerged in clinical application, such as hematologic toxicity, which may be related to the lack of subtype selectivity of PARP-1/-2. Selective inhibitors of PARP-1 that can overcome toxicity have emerged as a new strategy for PARP inhibitor development. In this review, we first reveal the conformational heterogeneity of the PARP-1/-2 active region through homology comparison and systematically explain the spatial topological characteristics of its selective binding pockets. Then, the structure-activity relationships of 14 reported selective inhibitors of PARP-1 are analyzed to reveal the key pharmacophores occupying the active region, as well as to characterize the specific groups bound to the selective binding domain. Finally, we discuss the structural requirements of selective PARP-1 inhibitors and propose the \"secondary site contact\" design strategy for the development of new PARP inhibitors.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-15"},"PeriodicalIF":3.4,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145063896","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of KRAS mutants (G12C, G12D, and G12V) inhibitors. KRAS突变体(G12C、G12D和G12V)抑制剂的鉴定。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-09-17 DOI: 10.1080/17568919.2025.2557177
Vikas Yadav, Mohammad Kashif, Zenab Kamalia, Vikas, Santhipriya P R, Samudrala Gourinath, Neelima Mondal, Naidu Subbarao
{"title":"Identification of KRAS mutants (G12C, G12D, and G12V) inhibitors.","authors":"Vikas Yadav, Mohammad Kashif, Zenab Kamalia, Vikas, Santhipriya P R, Samudrala Gourinath, Neelima Mondal, Naidu Subbarao","doi":"10.1080/17568919.2025.2557177","DOIUrl":"10.1080/17568919.2025.2557177","url":null,"abstract":"<p><strong>Aim: </strong>To identify and validate novel small-molecule inhibitors targeting KRAS G12C, G12D, and G12V mutants through a structure-based drug design and experimental approach.</p><p><strong>Methods: </strong>We employed molecular docking, molecular dynamics (MD) simulations, MM-PBSA binding free energy calculations, and principal component analysis (PCA) to screen and evaluate potential inhibitors targeting the Switch-II pocket of KRAS mutants. Top-ranking compounds were experimentally validated using Bio-Layer Interferometry (BLI) for binding affinity and MTT assays to assess anticancer activity in breast and lung cancer cell lines.</p><p><strong>Results: </strong>Compound C797-1505 showed strong binding to KRAS G12V (Dissociation constant (KD) = 141 µM), outperforming the reference Sotorasib (KD = 345 µM). C190-0346 displayed weak affinity toward KRAS G12C. MTT assays revealed that C797-1505 reduced breast cancer cell viability (Half-maximal Inhibitory Concentration (IC50) = 43.51 µM), while both compounds demonstrated significant cytotoxicity against lung cancer cells (IC50 = 18.78 µM and 22.93 µM, respectively).</p><p><strong>Conclusion: </strong>Our integrated computational and experimental strategies successfully identified selective KRAS mutant inhibitors with promising anticancer activity, particularly against G12V and G12C driven tumors. These findings support further development and preclinical evaluation of these compounds as targeted therapeutics.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2221-2234"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12456861/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145074864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metallacarboranes: abiotic scaffolds for advanced drug discovery. 金属碳硼烷:用于高级药物发现的非生物支架。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-09-10 DOI: 10.1080/17568919.2025.2557181
Tomasz M Goszczyński, Krzysztof Fink
{"title":"Metallacarboranes: abiotic scaffolds for advanced drug discovery.","authors":"Tomasz M Goszczyński, Krzysztof Fink","doi":"10.1080/17568919.2025.2557181","DOIUrl":"10.1080/17568919.2025.2557181","url":null,"abstract":"","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2193-2196"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12452483/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145029236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Structural insights into Arylidenehydrazinyl Benzenesulfonamides as potent mycobacterial carbonic anhydrase inhibitors. 芳基肼基苯磺酰胺作为分枝杆菌碳酸酐酶抑制剂的结构见解。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-09-02 DOI: 10.1080/17568919.2025.2552642
Pardeep Kumar, Anuradha Singampalli, Rani Bandela, Sri Mounika Bellapukonda, Sarvan Maddipatla, Aman Dalal, Ankita Devi, Srinivas Nanduri, Nitin Pal Kalia, Niccolo Paoletti, Claudiu T Supuran, Yaddanapudi Venkata Madhavi
{"title":"Structural insights into Arylidenehydrazinyl Benzenesulfonamides as potent mycobacterial carbonic anhydrase inhibitors.","authors":"Pardeep Kumar, Anuradha Singampalli, Rani Bandela, Sri Mounika Bellapukonda, Sarvan Maddipatla, Aman Dalal, Ankita Devi, Srinivas Nanduri, Nitin Pal Kalia, Niccolo Paoletti, Claudiu T Supuran, Yaddanapudi Venkata Madhavi","doi":"10.1080/17568919.2025.2552642","DOIUrl":"10.1080/17568919.2025.2552642","url":null,"abstract":"<p><strong>Aims: </strong>To design, synthesize, and assess novel sulfonamide hydrazone derivatives as selective inhibitors of <i>Mycobacterium tuberculosis</i> carbonic anhydrase.</p><p><strong>Materials and methods: </strong>Two series of 4-(arylidenehydrazinyl)benzenesulfonamides (<b>5a-r</b>) and N-arylidene-4-methylbenzenesulfonohydrazides (<b>6a-h</b>) were synthesized and evaluated against recombinant MtCA isoforms 1 and 3, and human carbonic anhydrase isoforms I and II by enzyme inhibition assays. Molecular docking and molecular dynamics simulations assessed the binding stability and coordination with the active-site zinc ion. Anti-mycobacterial activity was determined by minimum inhibitory concentrations (MICs) against M. tuberculosis. Time-kill kinetics and cytotoxicity assays evaluated the bactericidal potential and selectivity of the compound toward mammalian cells.</p><p><strong>Results: </strong>The compounds showed potent inhibition of MtCA 3 and hCA II, with moderate activity against MtCA 1 and hCA I. Notably, compounds <b>3e</b> and <b>3f</b> exhibited K<sub>i</sub> values of 0.0931 µM and 0.0984 µM, respectively, surpassing acetazolamide (K<sub>i</sub> = 0.104 µM). Docking and simulations confirmed stable zinc coordination. MIC values ranged from 4 to 128 µg/mL. Time-kill and cytotoxicity studies confirmed rapid bactericidal activity and low mammalian toxicity.</p><p><strong>Conclusion: </strong>These sulfonamide hydrazone derivatives demonstrate potent, selective MtCA inhibition, robust antimycobacterial efficacy, and favorable safety profiles, representing promising scaffolds for novel tuberculosis therapies with a novel mode of action.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2131-2144"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144949242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancing oxacillin efficacy against planktonic and biofilms of foodborne Enterobacteriaceae by hylin-a1-derived peptide. 利用海林-a1衍生肽增强oxacillin对食源性肠杆菌科浮游生物和生物膜的抑制作用。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-08-29 DOI: 10.1080/17568919.2025.2552632
Victor Alves Carneiro, Francisco Figueredo de Lima Filho, Benise Ferreira da Silva, Rafaela Mesquita Bastos Cavalcante, Renata Albuquerque Costa, Esteban Nicolás Lorenzón, Eduardo Maffud Cilli
{"title":"Enhancing oxacillin efficacy against planktonic and biofilms of foodborne Enterobacteriaceae by hylin-a1-derived peptide.","authors":"Victor Alves Carneiro, Francisco Figueredo de Lima Filho, Benise Ferreira da Silva, Rafaela Mesquita Bastos Cavalcante, Renata Albuquerque Costa, Esteban Nicolás Lorenzón, Eduardo Maffud Cilli","doi":"10.1080/17568919.2025.2552632","DOIUrl":"10.1080/17568919.2025.2552632","url":null,"abstract":"<p><strong>Aims: </strong>This study aims to evaluate the antibacterial and antibiofilm activity of Lys-a1, particularly in combination with oxacillin (OXA), against foodborne <i>E. coli</i> and <i>Klebsiella</i> spp.</p><p><strong>Materials and methods: </strong>The antibacterial activity was determined by minimum inhibitory/bactericidal concentration (MIC/MBC) and time-kill curves. Additionally, the potential synergistic effects between Lys-a1 and OXA were assessed using a checkerboard assay, and the ability of this combination to disrupt pre-formed biofilms was also investigated.</p><p><strong>Results: </strong>All tested strains were sensitive to Lys-a1, exhibiting MIC values ranging from 25 to 100 µg/mL and MBC typically one or twice the MIC values. The time-kill curve indicated the bactericidal effect within 180-240 minutes, achieving a 99.99% reduction in viable cells. Despite the detectable additive action, a notable synergistic effect was observed when Lys-a1 was combined with OXA for most strains (5/8), with an FIC index between 0.31 and 0.50. The Lys-a1/OXA combination significantly reduced biomass by around 50% and 90% of pre-formed biofilms compared with control and OXA-treated and untreated cells, respectively.</p><p><strong>Conclusions: </strong>The results suggest that Lys-a1/OXA combination could be a promising approach for sensitizing Gram-negative bacteria and expanding treatment options using OXA.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2079-2086"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12427473/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144949374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Analysis of antimicrobial activity and biofilm inhibition of Ag-NHC complexes by in-vitro and molecular docking method. Ag-NHC配合物的体外抑菌活性及生物膜抑制作用分析。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-08-28 DOI: 10.1080/17568919.2025.2552635
Neslihan Şahin, Elvan Üstün, Uğur Tutar, Cem Çelik
{"title":"Analysis of antimicrobial activity and biofilm inhibition of Ag-NHC complexes by in-vitro and molecular docking method.","authors":"Neslihan Şahin, Elvan Üstün, Uğur Tutar, Cem Çelik","doi":"10.1080/17568919.2025.2552635","DOIUrl":"10.1080/17568919.2025.2552635","url":null,"abstract":"<p><strong>Aims: </strong>Metal-N-heterocyclic carben (NHC) complexes have garnered significant attention from synthesis chemistry. Silver is well known for its broad-spectrum antimicrobial activity, and it exhibits their activities with different mechanisms. In this study, we combined these two important scaffolds, analyzed for possible antimicrobial and antibiofilm activity, and evaluated the interactions against DNA Gyrase, SarA, Human Serum Albumin, and DNA for getting insight into the antimicrobial and antibiofilm details.</p><p><strong>Materials & methods: </strong>Four new Ag-NHC complexes (<b>2a-d</b>) were prepared from corresponding benzimidazolium salts (<b>1a-d</b>) and revealed by elemental analysis, FT-IR, NMR, LC-MS, and HRMS. The antimicrobial and antibiofilm properties of both ligands and complexes were evaluated with in-vitro and molecular docking methods which were performed against DNA Gyrase, SarA, Human Serum Albumin, and DNA.</p><p><strong>Results and conclusions: </strong><b>1d</b> showed superior activity while <b>2a</b> and <b>2d</b> were effective against <i>C. albicans</i>, with activity comparable to fluconazole in the range of 8.6-8.7 µM. The highest binding affinity was recorded for 2a as -7.93 kcal/mol against DNA Gyrase, while <b>2b</b> has the best interactions with -5.49 kcal/mol against SarA. and -7.74 kcal/mol binding affinity was determined for <b>2a</b> with molecular docking. All the molecules interacted with the same grove of DNA.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2087-2100"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144949377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Research status of small molecule inhibitors, probes, and degraders of NSDs: a comprehensive review. NSDs小分子抑制剂、探针和降解剂的研究现状综述
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-09-10 DOI: 10.1080/17568919.2025.2557180
Zunyun Jiang, Hongyi Chen, Lianhua Piao, Shan Chang, Yingguang Zhu, Ren Kong
{"title":"Research status of small molecule inhibitors, probes, and degraders of NSDs: a comprehensive review.","authors":"Zunyun Jiang, Hongyi Chen, Lianhua Piao, Shan Chang, Yingguang Zhu, Ren Kong","doi":"10.1080/17568919.2025.2557180","DOIUrl":"10.1080/17568919.2025.2557180","url":null,"abstract":"<p><p>The nuclear receptor binding SET domain (NSD) family of histone methyltransferases, which comprised NSD1, NSD2, and NSD3. They play a pivotal role in catalyzing mono- and dimethylation of histone H3 at lysine 36 (H3K36me1/2), a modification critical for maintaining chromatin structure and transcriptional fidelity. Dysregulation of NSD enzymes, often through overexpression, mutation, or chromosomal translocation, has been implicated in a broad spectrum of malignancies and various diseases. Due to their critical role in disease pathogenesis and recent technological advances, NSD proteins have become attractive targets for therapeutic intervention. This review highlights recent progress in developing small molecule inhibitors and chemical probes targeting NSD family members, focusing on the catalytic SET domain, the PWWP domain, and other functional motifs. Among these, several chemical classes have been investigated, including quinoline-5,8-dione, 2-aminobenzothiazole, 5-aminonaphthalene, quinazoline, purine, benzoxazinone-cyclopropylamide, and imidazole derivatives. In addition, novel strategies such as protein degradation via PROTACs and dual-target inhibitors are discussed. By systematically summarizing recent advances, this review seeks to facilitate and accelerate the development of effective NSD modulators, ultimately advancing therapeutic options for diseases driven by NSD dysregulation.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2295-2310"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12452435/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145029400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Indole-selenide derivatives: recent advances in design, synthesis and future prospects for therapeutic applications. 吲哚硒化物衍生物:设计、合成的最新进展和治疗应用的未来前景。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-09-14 DOI: 10.1080/17568919.2025.2557185
Yunjun Chen, Hongtao Xu, Wei Hou
{"title":"Indole-selenide derivatives: recent advances in design, synthesis and future prospects for therapeutic applications.","authors":"Yunjun Chen, Hongtao Xu, Wei Hou","doi":"10.1080/17568919.2025.2557185","DOIUrl":"10.1080/17568919.2025.2557185","url":null,"abstract":"<p><p>In recent years, Selenium (Se) has become an emerging versatile player in medicinal chemistry. Indole scaffolds, known as a \"privileged structure,\" are widely used in drug design due to their unique physicochemical properties and broad biological activities. Indole selenide compounds, which integrate the two important elements of selenium and indole, represent a new and significant structural type and have been reported to exhibit various biological activities, including antitumor, antioxidant and P-glycoprotein inhibitory activities. In this review, we summarize the recent advances in the synthesis, design and biological activity studies of indole-selenide compounds. We also provide perspectives regarding the challenges in indole-selenide-based medicinal chemistry and future research directions.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2281-2293"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12456862/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145063906","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synthesis, crystal structure, anticancer activity and computational studies of novel Pyrazolo[4',3':5,6]pyrano[2,3-d]pyrimidine derivatives as potential anticancer agents. 新型吡唑[4',3':5,6]吡喃[2,3-d]嘧啶衍生物的合成、晶体结构、抗癌活性及计算研究。
IF 3.4 4区 医学
Future medicinal chemistry Pub Date : 2025-09-01 Epub Date: 2025-08-29 DOI: 10.1080/17568919.2025.2552641
Younesse Ait Elmachkouri, Ezaddine Irrou, Imane Yamari, Samir Chtita, Yahya Toubi, Mohamed F AlAjmi, Ali Oubella, Joel T Mague, Nada Kheira Sebbar, Mohamed Labd Taha
{"title":"Synthesis, crystal structure, anticancer activity and computational studies of novel Pyrazolo[4',3':5,6]pyrano[2,3-<i>d</i>]pyrimidine derivatives as potential anticancer agents.","authors":"Younesse Ait Elmachkouri, Ezaddine Irrou, Imane Yamari, Samir Chtita, Yahya Toubi, Mohamed F AlAjmi, Ali Oubella, Joel T Mague, Nada Kheira Sebbar, Mohamed Labd Taha","doi":"10.1080/17568919.2025.2552641","DOIUrl":"10.1080/17568919.2025.2552641","url":null,"abstract":"<p><strong>Aims: </strong>The search for effective anticancer agents remains a major goal in medicinal chemistry. This study aims to synthesize and evaluate new pyrazolo[4',3':5,6]pyrano[2,3-<i>d</i>]pyrimidine derivatives as potential anticancer agents.</p><p><strong>Materials & methods: </strong>Compounds <b>3-12</b> were synthesized <i>via N</i>2 or <i>N</i>1 alkylation reactions and substitution of chlorine at the <i>C</i>5 position of the pyrimidine ring under mild conditions. The structures were characterized by NMR (<sup>1</sup>H and <sup>13</sup>C), DEPT-135, HRMS, and single-crystal X-ray diffraction. Derivatives <b>6</b>, <b>7</b>, <b>8</b>, <b>11</b> and <b>12</b> were evaluated for their <i>in vitro</i> anticancer activity against 60 human cancer cell lines. Computational studies included molecular docking, binding affinity analysis against cancer-related protein targets, and ADME-T predictions.</p><p><strong>Results: </strong>Compounds <b>7</b> and <b>8</b> exhibited broad-spectrum anticancer activity against several tumor cell lines. Molecular docking and ADME-T analysis supported their potential as drug candidates.</p><p><strong>Conclusions: </strong>The synthesized pyrazolopyranopyrimidine derivatives, particularly <b>7</b> and <b>8</b>, show promise as anticancer agents and warrant further investigation.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"2101-2118"},"PeriodicalIF":3.4,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144949200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信