{"title":"Natural scaffold, modern strategy: Coumarin hybrids as potential carbohydrate-digesting enzyme inhibitors-A decade of medicinal chemistry exploration (2015–2025)","authors":"Ananya Pradhan, Priyabrata Pattanayak","doi":"10.1016/j.bmc.2025.118386","DOIUrl":"10.1016/j.bmc.2025.118386","url":null,"abstract":"<div><div>Over the past decade (2015–2025), coumarin and its hybrid derivatives have emerged as promising scaffolds in the search for potent inhibitors of carbohydrate-digesting enzymes, particularly α-amylase (AMY) and α-glucosidase (AG), which are key pharmacological targets in controlling postprandial hyperglycemia in type-2 diabetic patients. This review compiles and critically evaluates the medicinal chemistry efforts focused on coumarin-based hybrids, highlighting their development strategies, synthesis, enzyme inhibitory potential, and structure–activity relationships (SAR). Various pharmacophoric conjugates, including thiazole, thiazolidinedione, triazole, chalcone, isatin, sulfonamide, oxadiazole, hydrazone, and cinnamic acid, have been effectively linked to the coumarin nucleus to enhance enzyme inhibitory potential. Several derivatives have exhibited superior enzyme inhibition compared to the standard drug acarbose, with favorable <em>in vitro</em> and <em>in vivo</em> profiles, low cytotoxicity, and enhanced binding interactions as supported by docking analysis. The SAR insights reveal that substitution patterns, electronic effects, and linker modifications significantly influence biological activity. The findings underscore the versatility of coumarin as a lead scaffold and deliver a robust foundation for the rational design of next-generation lead molecules targeting the two carbohydrate-digesting enzymes.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"131 ","pages":"Article 118386"},"PeriodicalIF":3.0,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145156525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neha V. Rathod, Parmeswar Dalai, Reena Agrawal-Rajput, Satyendra Mishra
{"title":"Design, synthesis and molecular modelling studies of bile acid-curcumin conjugates as potential antiproliferative agents for breast cancer","authors":"Neha V. Rathod, Parmeswar Dalai, Reena Agrawal-Rajput, Satyendra Mishra","doi":"10.1016/j.bmc.2025.118387","DOIUrl":"10.1016/j.bmc.2025.118387","url":null,"abstract":"<div><div>In this study, novel bile acid–curcumin conjugates (conjugates <strong>3</strong> and <strong>4</strong>) were synthesized and structurally characterized using FT-IR, <sup>1</sup>H NMR, and <sup>13</sup>C NMR spectroscopy. The cytotoxic potential of these conjugates was evaluated against two triple negative breast cancer cell lines (TNBC), 4T1, and MDA-MB-231. Conjugate 3 displayed exceptional cytotoxic activity, with IC₅₀ values of 3.3 ± 0.06 μM for 4 T1 and 0.7 ± 0.01 μM for MDA-MB-231, significantly surpassing the activity of curcumin (IC₅₀ = 29.7 ± 0.9 μM and10.04 μM, respectively). Conjugate 4 also demonstrated enhanced efficacy, with IC₅₀ values of 5.3 ± 0.5 μM (4 T1) and 2.5 ± 0.03 μM (MDA-MB-231). Cell cycle analysis revealed a substantial reduction in the G0/G1 phase population from 89.90 % in untreated cells to 75.10 % for conjugate <strong>3</strong>, which induced apoptosis of 37.10 % than curcumin 20.39 % and 75.90 % for conjugate <strong>4</strong>, indicating disruption of cell cycle progression and enhanced antiproliferative effects. Both Conjugates <strong>3</strong> and <strong>4</strong> demonstrated excellent biocompatibility, exhibiting no toxicity toward normal macrophages. These findings confirm that bile acid conjugation significantly enhances the antiproliferative potential of curcumin.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118387"},"PeriodicalIF":3.0,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145045306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exploring the anticancer potential of β-amyrin: Scalable process for isolation, semi synthesis, and molecular docking studies","authors":"Telukuntla Sai Priya , Sonam Swain , Banoth Venkateswara Rao , Rajitha Rajeshwar Tatikonda , Malaya Kumar Nanda , Nishant Jain , Srinivasa Rao Mutheneni , K. Suresh Babu","doi":"10.1016/j.bmc.2025.118388","DOIUrl":"10.1016/j.bmc.2025.118388","url":null,"abstract":"<div><div>β-Amyrin, a pentacyclic triterpenoid with diverse biological activities, was efficiently isolated from <em>Alstonia scholaris</em> using a scalable and simplified process. To enhance its therapeutic potential, a series of C-3 modified semisynthetic derivatives bearing heterocyclic moieties were synthesized (<strong>3a-3o</strong>, <strong>4a-4k</strong>, <strong>6a-6h</strong> and <strong>7a-7n</strong>). Several derivatives exhibited promising anticancer activity, notably through Bax-mediated apoptotic pathways. Molecular docking and simulation studies confirmed strong interactions with the Bax protein trigger site, supporting their proposed mechanism of action. These findings highlight the potential of β-amyrin derivatives as lead candidates for further development in anticancer drug discovery.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118388"},"PeriodicalIF":3.0,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145060018","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Phytoestrogens and their synthetic analogues as substrate mimic inhibitors of CYP1B1 – An update (2020–2025)","authors":"Abanish Biswas, Venkatesan Jayaprakash","doi":"10.1016/j.bmc.2025.118385","DOIUrl":"10.1016/j.bmc.2025.118385","url":null,"abstract":"<div><div>Cytochrome P450 enzymes (CYPs) play crucial roles in metabolizing diverse substances, including drugs, pollutants, hormones, and steroids. Among them, CYP1B1 has gained significant attention due to its involvement in cancer initiation, progression, and drug resistance. Overexpression of CYP1B1 is frequently observed in multiple cancers, including breast, lung, colorectal, and brain cancers, making it an important therapeutic target. This review is an update covering the last six years (2019–2025) to our earlier publication proposing linker-based classification. Articles published over this period broadly covers efforts taken to increase the aqueous solubility <span><math><mi>α</mi></math></span>-naphthoflavone derivatives and ability to reverse the resistance to main-stream anticancer agents. We also covered PET tracers and PROTACs designed to hit the CYP1B1 by conjugating appropriate elements to the potent CYP1B1 inhibitors reported earlier. Overall, this review highlights significant recent innovations and strategic advancements in CYP1B1 inhibitor research, offering promising directions for cancer diagnosis and treatment.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118385"},"PeriodicalIF":3.0,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145045303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Céline Martin-Ghoteimi , Kyle A. Davis , Rayane Ghoteimi , Meareg Amare , Meghan V. Diefenbacher , Christianna Kutz , Jessica L. Smith , Laura J. Stevens , Egor Tchesnokov , Simon M. Walker , Mark Denison , Matthias Götte , Peter Halfmann , Alec J. Hirsch , Timothy P. Sheahan , Katherine L. Seley-Radtke
{"title":"Novel tricycle expanded purine nucleosides with pan-viral activity","authors":"Céline Martin-Ghoteimi , Kyle A. Davis , Rayane Ghoteimi , Meareg Amare , Meghan V. Diefenbacher , Christianna Kutz , Jessica L. Smith , Laura J. Stevens , Egor Tchesnokov , Simon M. Walker , Mark Denison , Matthias Götte , Peter Halfmann , Alec J. Hirsch , Timothy P. Sheahan , Katherine L. Seley-Radtke","doi":"10.1016/j.bmc.2025.118384","DOIUrl":"10.1016/j.bmc.2025.118384","url":null,"abstract":"<div><div>A series of thiophene-expanded tricyclic nucleosides featuring modifications not previously investigated, including sugar and nucleobase modifications, were synthesized as potential antiviral therapeutics. In addition, their corresponding prodrugs were also pursued to probe their potential antiviral activity, as typically prodrugs increase solubility and delivery. Initial interest was focused on ribose analogues as we had previously only synthesized a few compounds, and none had been tested against new viruses, particularly those of pandemic concern. In that regard, CEM-007, showed promising antiviral activity against several viral families. Given this promising lead, the 2′-α-fluoro-2′-β-methyl-modified analogues of CEM-007 were also synthesized for comparison. While 2′-modified analogue CEM-024 showed little to no antiviral activity, several of its prodrugs including CEM-042, CEM-052, CEM-053, CEM-054 and CEM-055, exhibited potent, broad-spectrum antiviral activity across several viral families including flaviviruses, filoviruses, and coronaviruses, with EC<sub>50</sub> values in the single digit low micromolar range, with minimal toxicity. Additional studies are underway to elucidate the mechanism of action of these analogues. The design, synthesis, and biological testing of these structurally novel analogues are reported herein.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118384"},"PeriodicalIF":3.0,"publicationDate":"2025-09-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145045305","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaoman Zhao , Yueshan Ji , Yue Kong , Aixia Yan , Changyuan Yu
{"title":"Discovery of novel tropomyosin receptor kinase A inhibitors by virtual screening merging ligand-based and structure-based methods","authors":"Xiaoman Zhao , Yueshan Ji , Yue Kong , Aixia Yan , Changyuan Yu","doi":"10.1016/j.bmc.2025.118381","DOIUrl":"10.1016/j.bmc.2025.118381","url":null,"abstract":"<div><div>Tropomyosin receptor kinase (TRK) is a crucial broad-spectrum anticancer target. Its inhibitors are among the first “tumor agnostic” drugs approved for pan-cancer therapy. TRKA, a subtype of tropomyosin receptor kinase, is one of the most frequently detected in human cancers and has emerged as a key target for anticancer drug development. Our work employed a virtual screening approach integrating ligand- and structure-based strategies to identify novel inhibitors targeting TRKA. A large-scale virtual screening system was constructed for a screening database of 9.87 million commercial compounds containing a broad chemical space. A funnel-shaped multistage combinatorial screening process was adopted, with the combined time-consuming and precision of the methods in the following order: screening based on pharmacophore and shape, screening based on the QSAR model obtained from the prequel study, screening based on molecular docking, prediction of ADME properties, and patent searching. The above virtual screening system received sixteen potentially active novel inhibitors. Subsequent enzymatic activity assays revealed one hit compound ZA16 with nanomolar-level inhibitory potency, validating the efficacy of the large-scale virtual screening system. This approach can be a robust framework for guiding drug discovery and design across diverse compound libraries. Furthermore, molecular dynamics simulations revealed the interaction between this novel inhibitor and the TRKA protein domain, emphasizing the crucial role of the hydrogen bond at MET592. Through the analysis of MM/GBSA binding free energy, this study offered significant insights for drug discovery and design targeting the same protein.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118381"},"PeriodicalIF":3.0,"publicationDate":"2025-09-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145045307","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Redox-cleavable disulfide linker enhances siRNA delivery by prodrug-type bifunctional cell-penetrating peptide","authors":"Keita Wakamori, Yuzuki Hashikawa, Hidehito Urata, Shun-ichi Wada","doi":"10.1016/j.bmc.2025.118383","DOIUrl":"10.1016/j.bmc.2025.118383","url":null,"abstract":"<div><div>In prodrug strategies, introducing a cleavable linker enables activation within target tissues. We developed a novel prodrug-type cell-penetrating peptide (CPP), <strong>PI-linker-cRGD</strong>, incorporating a sterically refined redox-cleavable linker for intracellular siRNA delivery. Disulfide bonds are valuable in CPP-based prodrug designs because they are cleavable in the reductive environment of cancer cells. We previously designed a bifunctional CPP, <strong>PI-cRGD</strong>, by conjugating the membrane-permeable α-aminoisobutyric acid (Aib)-containing amphipathic helical peptide (<strong>PI</strong>) to the α<sub>v</sub>β<sub>3</sub> integrin-targeting ligand cRGD via a disulfide bond. The <strong>PI-cRGD</strong>/siRNA complex exhibited low cytotoxicity and significant cellular uptake via endocytosis. However, it failed to achieve efficient cytosolic siRNA delivery, likely due to steric hindrance that prevented the disulfide cleavage in the reductive cell surface and intracellular environments, which in turn inhibited the release of membrane-permeable <strong>PI</strong>. To address this issue, we demonstrate that inserting a sterically refined redox-cleavable linker between <strong>PI</strong> and cRGD sustains low toxicity and targeting ability while enabling cleavage of the disulfide bond through thiol–disulfide exchange at the cell surface, thereby activating the CPP, promoting membrane translocation, and ultimately achieving efficient cytosolic siRNA delivery. This simple cleavable-linker strategy holds broad applicability for diverse therapeutic agents.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118383"},"PeriodicalIF":3.0,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145045304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nathália R.C. Martins , Ruan C.B. Ribeiro , Mariana O.L. Desmarais , André Luiz Lourenço , Aldo R. da Silva , Thaís D.C. Almeida , Sarah Christina G. Gonçalves , Norman A. Ratcliffe , Isadora M.G. Andrade , Laís M. Marins , Sandy P. Valle , Luana da S.M. Forezi , Vitor F. Ferreira , Vanessa Nascimento , Helena Carla Castro , Fernando de C. da Silva
{"title":"Antifungal profile of menadione tethered to 1H-1,2,3-triazolyl-selenoester: synthesis, in vitro and in silico analyses","authors":"Nathália R.C. Martins , Ruan C.B. Ribeiro , Mariana O.L. Desmarais , André Luiz Lourenço , Aldo R. da Silva , Thaís D.C. Almeida , Sarah Christina G. Gonçalves , Norman A. Ratcliffe , Isadora M.G. Andrade , Laís M. Marins , Sandy P. Valle , Luana da S.M. Forezi , Vitor F. Ferreira , Vanessa Nascimento , Helena Carla Castro , Fernando de C. da Silva","doi":"10.1016/j.bmc.2025.118380","DOIUrl":"10.1016/j.bmc.2025.118380","url":null,"abstract":"<div><div>Invasive Candidiasis infections are a clinical challenge, with limited effective therapeutic agents and increasing resistance. The discovery of new antifungal agents is urgently required. Here, we developed a new series of 2-methyl-1,4-naphthoquinone (Menadione) Tethered to 1<em>H</em>-1,2,3-triazolyl-selenoester in good yields, which exhibit antifungal potential activity against <em>Candida</em> species. Antifungal and antibiofilm activities for the twenty-one compounds were performed <em>in vitro</em> against <em>Candida spp</em> and exhibited inhibitory effects at different concentrations for several <em>Candida</em> species. Two compounds showed strong inhibitory effects, with fungicidal activity at 16 and 32 μg/mL, respectively, against planktonic cells and biofilms of <em>Candida glabrata</em>. <em>In silico</em> studies were conducted to determine the binding energies against BonFIRE spectral images of antifungal protein targets, and provided stereo electronic, pharmacokinetic and toxicological profiles. The analysis through molecular docking with CYP51 and Hsp90 highlighted the inhibitory potential of the compounds in comparison to itraconazole and radicicol, and further underscored the favorable pharmacokinetic profile for all molecules, characterized by a low toxicity profile. We therefore concluded that two compounds especially exhibited potent antifungal activity against <em>C. glabrata</em>.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118380"},"PeriodicalIF":3.0,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145019877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chang-Jiang Yang , Qi-Hang Cui , Hui-Juan Lyu , Zhao-Yang Wang , Qing-Shan Wang , Rong Xiang , Shi-Wu Chen
{"title":"Identification and biological evaluation of novel indole-2-one derivatives as BRD4-BD2 inhibitors","authors":"Chang-Jiang Yang , Qi-Hang Cui , Hui-Juan Lyu , Zhao-Yang Wang , Qing-Shan Wang , Rong Xiang , Shi-Wu Chen","doi":"10.1016/j.bmc.2025.118379","DOIUrl":"10.1016/j.bmc.2025.118379","url":null,"abstract":"<div><div>Pan-inhibitions of Bromodomain and Extra Terminal Domain (BET) proteins have shown great potential in anti-tumor therapy but exhibited clinical toxicities, while selective inhibition of BRD4-BD2 could improve specificity and have better safety. Herein, a series of indole-2-one derivatives were designed and synthesized as novel BD2-selective inhibitors. The representative compound <strong>47</strong> showed good inhibitory effect on BRD4-BD2 with the IC<sub>50</sub> of 27 nM and displayed 102-fold selectivity over BRD4-BD1, and exhibited extensive anti-tumor proliferation activities <em>in vitro</em>, especially against tumor cells, such as K562 and HGC-27 (IC<sub>50</sub> = 0.15 and 3 μM), and it was less toxic to normal cell in GES-1 (IC<sub>50</sub> = 71 μM). Further biological studies revealed that <strong>47</strong> could down-regulate c-Myc and up-regulate p21, arrest cell cycle at G<sub>0</sub>/G<sub>1</sub> phase and induce apoptosis in HGC-27 cells. More importantly, <strong>47</strong> showed moderate pharmacokinetic properties and low toxicity <em>in vivo</em>. These results demonstrated that <strong>47</strong> might serve as a potent lead compound with potential for the treatment of cancers such as gastric cancer.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118379"},"PeriodicalIF":3.0,"publicationDate":"2025-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145019878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Miaomiao Shi , Jiale Yan , Jiangyu Zhao , Khurshed Bozorov , Lifei Nie , Haji Akber Aisa , Chao Niu
{"title":"Aurone and its analogues as targeted drugs for the anticancer treatment","authors":"Miaomiao Shi , Jiale Yan , Jiangyu Zhao , Khurshed Bozorov , Lifei Nie , Haji Akber Aisa , Chao Niu","doi":"10.1016/j.bmc.2025.118375","DOIUrl":"10.1016/j.bmc.2025.118375","url":null,"abstract":"<div><div>Targeted therapy has become a crucial strategy in modern anticancer treatment. Among various pharmacophores explored, aurones (2-benzylidenebenzofuran-3(2H)-ones) and their analogues have garnered increasing attention as promising scaffolds for targeted cancer therapy due to their ability to interact with multiple oncological targets. This review comprehensively examines the medicinal chemistry development of aurone-based lead compounds over the past decade, with particular emphasis on their derivatization strategies, pharmacological potency, and structure-activity relationships against key cancer targets. Accumulated evidence demonstrates that aurone derivatives and their analogues show promising potential as candidate drugs for next-generation targeted anticancer agents. The present review not only summarizes current research progress but also provides valuable perspectives for future directions in aurone-based anticancer drug discovery.</div></div>","PeriodicalId":255,"journal":{"name":"Bioorganic & Medicinal Chemistry","volume":"130 ","pages":"Article 118375"},"PeriodicalIF":3.0,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145038813","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}