首页 > 最新文献

Future medicinal chemistry最新文献

英文 中文
Autophagosome degraders: a novel therapeutic strategy with broad application potential in human diseases. 自噬体降解剂:一种具有广泛应用潜力的治疗人类疾病的新策略。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-19 DOI: 10.1080/17568919.2025.2605014
Wei-Fang Zuo, Xinyi Huang, Cheng Peng, Bo Han, Qian Zhao

Autophagy-mediated targeted protein degradation, exemplified by technologies such as autophagosome-tethering compounds (ATTECs), AUTOphagy-TArgeting chimeras (AUTOTACs), and autophagy-targeting chimeras (AUTACs), leverages the autophagy-lysosome pathway for the clearance of challenging substrates that often exceed proteasomal capacity. These substrates include large protein aggregates, multi-protein complexes, and even entire organelles. This review synthesizes key advances in the development of autophagy-based degraders since 2022, highlighting their therapeutic potential through exemplar applications. We discuss their utility in oncology, neurodegenerative disorders, and inflammatory/cardiometabolic diseases. These novel modalities have demonstrated potent, selective, and durable substrate elimination in vivo, successfully overcoming resistance mechanisms associated with traditional occupancy-driven inhibition. Finally, we summarize the general workflow for developing autophagy-based degraders, outline the current challenges and future directions in this field, and aim to promote fundamental mechanistic studies and innovative medicinal chemistry research, thereby accelerating the clinical translation of autophagy-targeting degraders for the treatment of various human diseases.

自噬介导的靶向蛋白降解,以自噬体系栓化合物(attec)、自噬靶向嵌合体(AUTOTACs)和自噬靶向嵌合体(AUTACs)等技术为例,利用自噬-溶酶体途径清除通常超过蛋白酶体容量的挑战性底物。这些底物包括大的蛋白质聚集体,多蛋白质复合物,甚至整个细胞器。本文综述了自2022年以来基于自噬的降解剂发展的关键进展,通过范例应用强调了它们的治疗潜力。我们讨论了它们在肿瘤学、神经退行性疾病和炎症/心脏代谢疾病中的应用。这些新模式已经在体内证明了有效的、选择性的和持久的底物消除,成功克服了与传统占领驱动抑制相关的耐药机制。最后,我们总结了基于自噬的降解物开发的一般工作流程,概述了该领域目前面临的挑战和未来的发展方向,旨在促进基础机制研究和创新药物化学研究,从而加快自噬靶向降解物的临床转化,治疗各种人类疾病。
{"title":"Autophagosome degraders: a novel therapeutic strategy with broad application potential in human diseases.","authors":"Wei-Fang Zuo, Xinyi Huang, Cheng Peng, Bo Han, Qian Zhao","doi":"10.1080/17568919.2025.2605014","DOIUrl":"https://doi.org/10.1080/17568919.2025.2605014","url":null,"abstract":"<p><p>Autophagy-mediated targeted protein degradation, exemplified by technologies such as autophagosome-tethering compounds (ATTECs), AUTOphagy-TArgeting chimeras (AUTOTACs), and autophagy-targeting chimeras (AUTACs), leverages the autophagy-lysosome pathway for the clearance of challenging substrates that often exceed proteasomal capacity. These substrates include large protein aggregates, multi-protein complexes, and even entire organelles. This review synthesizes key advances in the development of autophagy-based degraders since 2022, highlighting their therapeutic potential through exemplar applications. We discuss their utility in oncology, neurodegenerative disorders, and inflammatory/cardiometabolic diseases. These novel modalities have demonstrated potent, selective, and durable substrate elimination <i>in vivo</i>, successfully overcoming resistance mechanisms associated with traditional occupancy-driven inhibition. Finally, we summarize the general workflow for developing autophagy-based degraders, outline the current challenges and future directions in this field, and aim to promote fundamental mechanistic studies and innovative medicinal chemistry research, thereby accelerating the clinical translation of autophagy-targeting degraders for the treatment of various human diseases.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-12"},"PeriodicalIF":3.4,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793796","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
Current scenario of indole-azole hybrids with anticancer potential: part I. triazole and pyrazole hybrids. 具有抗癌潜力的吲哚-唑类杂交种的现状:第一部分:三唑和吡唑类杂交种。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-16 DOI: 10.1080/17568919.2025.2602420
Dong Gao-Li, Wang Jun-Jian, Yang Jia

Cancer therapy is still hampered by key challenges, including drug resistance, poor target selectivity, and narrow therapeutic spectra, driving the pursuit of novel anticancer agents with enhanced efficacy and safety. Indole-triazole/pyrazole hybrids, formed by fusing indole scaffolds with triazole/pyrazole, confer inherent structural diversity and high modifiability. Structurally, rational modification of indole/triazole/pyrazole moieties allows optimization of pharmacokinetic properties and improves cancer cell selectivity, minimizing toxicity to normal cells. Functionally, indole-triazole/pyrazole hybrids exhibit multitargeted activity to simultaneously inhibit key oncogenic pathways, addressing the heterogeneity of cancer pathogenesis, while their hybrid structure enhances anticancer potency. This multitargeted mode also aids in overcoming drug resistance, a major bottleneck in clinical therapy. Accordingly, indole-triazole/pyrazole hybrids have emerged as a promising class of anticancer candidates. This review summarizes recent advances in indole-triazole/pyrazole hybrids with anticancer potential, covering articles published from 2021 to the present. To delineate the key molecular features governing anticancer potency, this review further presents a detailed analysis of structure-activity relationships (SARs) and conducts an in-depth exploration of the underlying mechanisms of action.

癌症治疗仍然受到一些关键挑战的阻碍,包括耐药、低靶点选择性和狭窄的治疗谱,这促使人们追求具有更高疗效和安全性的新型抗癌药物。吲哚-三唑/吡唑杂合体是由吲哚支架与三唑/吡唑融合而成的,具有固有的结构多样性和高可修饰性。在结构上,合理修饰吲哚/三唑/吡唑基团可以优化药代动力学特性,提高癌细胞选择性,最大限度地减少对正常细胞的毒性。在功能上,吲哚-三唑/吡唑杂交种表现出多靶点活性,同时抑制关键的致癌途径,解决癌症发病机制的异质性,而它们的杂交结构增强了抗癌效力。这种多靶点模式也有助于克服耐药性,这是临床治疗的主要瓶颈。因此,吲哚-三唑/吡唑复合物已成为一类有前途的抗癌候选药物。本文综述了具有抗癌潜力的吲哚-三唑/吡唑杂交体的最新进展,涵盖了从2021年到现在发表的文章。为了描述控制抗癌能力的关键分子特征,本文进一步详细分析了结构-活性关系(SARs),并深入探讨了潜在的作用机制。
{"title":"Current scenario of indole-azole hybrids with anticancer potential: part I. triazole and pyrazole hybrids.","authors":"Dong Gao-Li, Wang Jun-Jian, Yang Jia","doi":"10.1080/17568919.2025.2602420","DOIUrl":"https://doi.org/10.1080/17568919.2025.2602420","url":null,"abstract":"<p><p>Cancer therapy is still hampered by key challenges, including drug resistance, poor target selectivity, and narrow therapeutic spectra, driving the pursuit of novel anticancer agents with enhanced efficacy and safety. Indole-triazole/pyrazole hybrids, formed by fusing indole scaffolds with triazole/pyrazole, confer inherent structural diversity and high modifiability. Structurally, rational modification of indole/triazole/pyrazole moieties allows optimization of pharmacokinetic properties and improves cancer cell selectivity, minimizing toxicity to normal cells. Functionally, indole-triazole/pyrazole hybrids exhibit multitargeted activity to simultaneously inhibit key oncogenic pathways, addressing the heterogeneity of cancer pathogenesis, while their hybrid structure enhances anticancer potency. This multitargeted mode also aids in overcoming drug resistance, a major bottleneck in clinical therapy. Accordingly, indole-triazole/pyrazole hybrids have emerged as a promising class of anticancer candidates. This review summarizes recent advances in indole-triazole/pyrazole hybrids with anticancer potential, covering articles published from 2021 to the present. To delineate the key molecular features governing anticancer potency, this review further presents a detailed analysis of structure-activity relationships (SARs) and conducts an in-depth exploration of the underlying mechanisms of action.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-13"},"PeriodicalIF":3.4,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145767694","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
Advances in hydroxamic acid hybrids for liver cancer therapy: a decade of progress (2016-2025). 混合羟基肟酸治疗肝癌的进展:十年进展(2016-2025)。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-16 DOI: 10.1080/17568919.2025.2594964
Yafei Zhuang, Yanjing Cheng, Kesong Zhu, Chenchen Song, Mengjie Zhao, Donghong Wang, Xia Cao, Aimei Liu

Liver cancer, which originates from hepatocytes, ranks among the most commonly diagnosed cancers and stands as a leading cause of cancer-related deaths, primarily due to late diagnosis and its rapid progression. Liver cancer, especially metastatic liver tumors, often relies on chemotherapy. Still, drug resistance driven by the overexpression of efflux pumps, reduced systemic drug exposure due to hepatic metabolism, low efficacy, and high toxicity creates an urgent need to explore novel chemotherapeutic agents. Hydroxamic acid serves as the zinc-binding group (ZBG) in most histone deacetylase (HDAC) inhibitors and is an important anti-liver cancer pharmacophore. Hydroxamic acid hybrids harness the epigenetic potency of hydroxamic acid through modular pharmacophore integration, providing multitarget efficacy, resistance overcoming, and therapeutic versatility, and thus represent promising candidates for next-generation liver cancer therapies.

肝癌起源于肝细胞,是最常诊断的癌症之一,也是癌症相关死亡的主要原因,主要原因是诊断较晚且进展迅速。肝癌,尤其是转移性肝肿瘤,往往依赖于化疗。然而,由于外排泵的过度表达、肝脏代谢导致的全身药物暴露减少、低疗效和高毒性,导致迫切需要探索新的化疗药物。在大多数组蛋白去乙酰化酶(HDAC)抑制剂中,羟肟酸作为锌结合基团(ZBG),是一种重要的抗肝癌药效团。羟肟酸杂交体通过模块化药效团整合利用羟肟酸的表观遗传效力,提供多靶点疗效,克服耐药性和治疗多功能性,因此代表了下一代肝癌治疗的有希望的候选者。
{"title":"Advances in hydroxamic acid hybrids for liver cancer therapy: a decade of progress (2016-2025).","authors":"Yafei Zhuang, Yanjing Cheng, Kesong Zhu, Chenchen Song, Mengjie Zhao, Donghong Wang, Xia Cao, Aimei Liu","doi":"10.1080/17568919.2025.2594964","DOIUrl":"https://doi.org/10.1080/17568919.2025.2594964","url":null,"abstract":"<p><p>Liver cancer, which originates from hepatocytes, ranks among the most commonly diagnosed cancers and stands as a leading cause of cancer-related deaths, primarily due to late diagnosis and its rapid progression. Liver cancer, especially metastatic liver tumors, often relies on chemotherapy. Still, drug resistance driven by the overexpression of efflux pumps, reduced systemic drug exposure due to hepatic metabolism, low efficacy, and high toxicity creates an urgent need to explore novel chemotherapeutic agents. Hydroxamic acid serves as the zinc-binding group (ZBG) in most histone deacetylase (HDAC) inhibitors and is an important anti-liver cancer pharmacophore. Hydroxamic acid hybrids harness the epigenetic potency of hydroxamic acid through modular pharmacophore integration, providing multitarget efficacy, resistance overcoming, and therapeutic versatility, and thus represent promising candidates for next-generation liver cancer therapies.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-14"},"PeriodicalIF":3.4,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145762750","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
Targeting autophagy kinases: from mechanisms to therapy with novel small molecules. 靶向自噬激酶:从机制到新小分子治疗。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-16 DOI: 10.1080/17568919.2025.2602421
Linlin Deng, Linwei Li, Yue Li, Ting Ma, Shaojie Liang, Erkang Tian

Autophagy is an evolutionarily conserved process in eukaryotic cells that degrades and recycles intracellular macromolecules and damaged organelles. It is closely related to a variety of physiological and pathological processes. Research on autophagy has become a current hotspot, with protein kinases regarded as crucial components that play essential roles throughout this process. During autophagy, diverse autophagy-related protein kinases continuously regulate different stages. Protein kinases are critical in signal transduction and the regulation of most cellular processes. Therefore, autophagy-associated protein kinases represent potential therapeutic targets for human diseases, and corresponding small-molecule compounds may provide promising therapeutic strategies. This review summarizes the current progress in autophagy research, with a focus on small-molecule drugs that influence autophagy-related kinases and their association with diseases.

自噬是真核细胞中降解和循环细胞内大分子和受损细胞器的进化保守过程。它与多种生理和病理过程密切相关。自噬的研究已成为当前的热点,蛋白激酶被认为是自噬过程中起重要作用的关键成分。在自噬过程中,多种自噬相关蛋白激酶不断调控不同阶段。蛋白激酶在信号转导和大多数细胞过程的调节中至关重要。因此,自噬相关蛋白激酶是人类疾病的潜在治疗靶点,相应的小分子化合物可能提供有前景的治疗策略。本文综述了自噬的研究进展,重点介绍了影响自噬相关激酶的小分子药物及其与疾病的关系。
{"title":"Targeting autophagy kinases: from mechanisms to therapy with novel small molecules.","authors":"Linlin Deng, Linwei Li, Yue Li, Ting Ma, Shaojie Liang, Erkang Tian","doi":"10.1080/17568919.2025.2602421","DOIUrl":"https://doi.org/10.1080/17568919.2025.2602421","url":null,"abstract":"<p><p>Autophagy is an evolutionarily conserved process in eukaryotic cells that degrades and recycles intracellular macromolecules and damaged organelles. It is closely related to a variety of physiological and pathological processes. Research on autophagy has become a current hotspot, with protein kinases regarded as crucial components that play essential roles throughout this process. During autophagy, diverse autophagy-related protein kinases continuously regulate different stages. Protein kinases are critical in signal transduction and the regulation of most cellular processes. Therefore, autophagy-associated protein kinases represent potential therapeutic targets for human diseases, and corresponding small-molecule compounds may provide promising therapeutic strategies. This review summarizes the current progress in autophagy research, with a focus on small-molecule drugs that influence autophagy-related kinases and their association with diseases.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-17"},"PeriodicalIF":3.4,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145767633","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
Integrative lipophilicity assessment and pharmacokinetic correlation of pyrimidine precursors and artesunate-pyrimidine hybrids: development of QSAR models for anticancer activity and interaction with P-glycoprotein. 嘧啶前体和青蒿琥酯-嘧啶杂交体的综合亲脂性评估和药动学相关性:抗癌活性及其与p糖蛋白相互作用的QSAR模型的建立。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-13 DOI: 10.1080/17568919.2025.2602963
Mirjana Mosić, Ljiljana Koračak, Marija Grozdanić, Nataša Terzić Jovanović, Milica Pešić, Sandra Šegan

Aims: To evaluate chromatographic lipophilicity of novel artesunate-pyrimidine hybrids and precursors using reversed-phase thin-layer chromatography (RP-TLC) and assess plasma protein binding (PPB). The impact of measured and predicted lipophilicity on pharmacokinetic descriptors was evaluated. Principal component analysis (PCA) explored relationships among lipophilicity, PPB, and physicochemical descriptors. Quantitative structure-activity relationship (QSAR) and partial least squares (PLS) models linked molecular descriptors to cytotoxicity and resistance modulation in nonsmall cell lung cancer (NSCLC) cells.

Materials and methods: Lipophilicity was measured by RP-TLC. PPB was determined using human serum albumin (HSA)-modified high-performance liquid chromatography (HPLC). PCA characterized physicochemical-pharmacokinetic correlations. Cytotoxicity in sensitive NCI-H460 and multidrug-resistant (MDR) NCI-H460/R cells was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. QSAR and PLS models identified key descriptors.

Results: Lipophilicity strongly influenced adsorption, distribution, and protein binding. Highly lipophilic hybrids showed near-complete HSA binding. Compound 2k lost cytotoxicity in the presence of albumin, whereas 4k retained potency. Models indicated steric and electronic features, alongside lipophilicity, dictate efficacy and P-glycoprotein (P-gp) interactions, particularly in resistant cells.

Conclusions: Lipophilicity and steric/electronic descriptors govern distribution, protein binding, and anticancer activity. Integrating these features enables design of hybrids overcoming P-gp-mediated multidrug resistance, with hybrid 4k emerging as a promising candidate.

目的:利用反相薄层色谱法(RP-TLC)评价新型青蒿琥酯-嘧啶杂合体和前体的层析亲脂性,并评价血浆蛋白结合(PPB)。评估测定和预测的亲脂性对药代动力学描述符的影响。主成分分析(PCA)探讨了亲脂性、PPB和理化描述符之间的关系。定量构效关系(QSAR)和偏最小二乘(PLS)模型将分子描述符与非小细胞肺癌(NSCLC)细胞毒性和耐药调节联系起来。材料与方法:采用反相薄层色谱法测定亲脂性。采用人血清白蛋白(HSA)-高效液相色谱法测定PPB。PCA表征了物理化学-药代动力学相关性。采用3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑(MTT)测定法,对NCI-H460敏感细胞和多药耐药(MDR) NCI-H460/R细胞的细胞毒性进行了评估。QSAR和PLS模型确定了关键描述符。结果:亲脂性强烈影响吸附、分布和蛋白质结合。高亲脂性杂交种显示出接近完全的HSA结合。化合物2k在白蛋白存在下失去细胞毒性,而4k保留效力。模型显示,立体和电子特征以及亲脂性决定了药效和p -糖蛋白(P-gp)的相互作用,特别是在耐药细胞中。结论:亲脂性和立体/电子描述子控制着分布、蛋白质结合和抗癌活性。整合这些特性可以设计出克服p- gp介导的多药耐药的杂交体,其中杂交体4k成为有希望的候选物。
{"title":"Integrative lipophilicity assessment and pharmacokinetic correlation of pyrimidine precursors and artesunate-pyrimidine hybrids: development of QSAR models for anticancer activity and interaction with <i>P</i>-glycoprotein.","authors":"Mirjana Mosić, Ljiljana Koračak, Marija Grozdanić, Nataša Terzić Jovanović, Milica Pešić, Sandra Šegan","doi":"10.1080/17568919.2025.2602963","DOIUrl":"https://doi.org/10.1080/17568919.2025.2602963","url":null,"abstract":"<p><strong>Aims: </strong>To evaluate chromatographic lipophilicity of novel artesunate-pyrimidine hybrids and precursors using reversed-phase thin-layer chromatography (RP-TLC) and assess plasma protein binding (PPB). The impact of measured and predicted lipophilicity on pharmacokinetic descriptors was evaluated. Principal component analysis (PCA) explored relationships among lipophilicity, PPB, and physicochemical descriptors. Quantitative structure-activity relationship (QSAR) and partial least squares (PLS) models linked molecular descriptors to cytotoxicity and resistance modulation in nonsmall cell lung cancer (NSCLC) cells.</p><p><strong>Materials and methods: </strong>Lipophilicity was measured by RP-TLC. PPB was determined using human serum albumin (HSA)-modified high-performance liquid chromatography (HPLC). PCA characterized physicochemical-pharmacokinetic correlations. Cytotoxicity in sensitive NCI-H460 and multidrug-resistant (MDR) NCI-H460/R cells was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. QSAR and PLS models identified key descriptors.</p><p><strong>Results: </strong>Lipophilicity strongly influenced adsorption, distribution, and protein binding. Highly lipophilic hybrids showed near-complete HSA binding. Compound 2k lost cytotoxicity in the presence of albumin, whereas 4k retained potency. Models indicated steric and electronic features, alongside lipophilicity, dictate efficacy and <i>P</i>-glycoprotein (<i>P</i>-gp) interactions, particularly in resistant cells.</p><p><strong>Conclusions: </strong>Lipophilicity and steric/electronic descriptors govern distribution, protein binding, and anticancer activity. Integrating these features enables design of hybrids overcoming <i>P</i>-gp-mediated multidrug resistance, with hybrid 4k emerging as a promising candidate.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-12"},"PeriodicalIF":3.4,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741947","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
Multi-target design of fused cyclic pyrimidine-2-thione candidates as DNA intercalators and topoisomerase I/II inhibitors. 融合环嘧啶-2-硫酮候选物作为DNA插入物和拓扑异构酶I/II抑制剂的多靶点设计。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-12 DOI: 10.1080/17568919.2025.2602424
Mahmoud S Khalil, Ahmed A Al-Karmalawy, Ayman Abo Elmaaty, Galal A Elsayed, A M A Hassan

Aim: We aimed to design and synthesize novel pyrimidine-2-thione derivatives (1-13) as Topoisomerase I/II (Topo I/II) inhibitors with DNA intercalation potential for cancer treatment.

Materials & methods: Inhibitory concentration 50 (IC50) against mammary gland breast cancer, hepatocellular carcinoma, and colorectal carcinoma was determined for all compounds. The frontier candidates (2, 6, 9, 10, and 11) were evaluated for their DNA-binding ability, Topo I, and Topo II inhibiting potential. Moreover, cell cycle and apoptosis analysis were carried out.

Results: Compound 2 displayed the best DNA-binding affinity with an IC50 value of 37.24 µM in comparison to doxorubicin (Dox). Both compounds 2 and 9 showed superior nanomolar Topo I inhibitory potential, compared to Dox. Similarly, compounds 2 and 9 achieved better Topo II inhibition, exceeding that of Dox. It was revealed that compound 9 halted the cell cycle at both the P1 and G2 phases. In addition, compound 9 was able to boost the apoptosis at both the early and late apoptotic phases.

Conclusion: Consequently, the compounds afforded can be regarded as prominent lead anticancer compounds for further optimization and investigation.

目的:设计并合成新型嘧啶-2-硫酮衍生物(1-13),作为具有DNA嵌入潜力的拓扑异构酶I/II (Topo I/II)抑制剂,用于癌症治疗。材料与方法:测定各化合物对乳腺、乳腺癌、肝细胞癌和结直肠癌的抑制浓度50 (IC50)。对前沿候选基因(2、6、9、10和11)的dna结合能力、Topo I和Topo II抑制潜力进行了评估。同时进行细胞周期和凋亡分析。结果:与阿霉素(Dox)相比,化合物2的dna结合亲和力最高,IC50值为37.24µM。与Dox相比,化合物2和9均表现出优异的纳米级Topo I抑制电位。同样,化合物2和9对Topo II的抑制效果更好,超过了Dox。结果表明,化合物9在P1期和G2期都能使细胞周期停止。此外,化合物9在凋亡早期和晚期都能促进细胞凋亡。结论:上述化合物可作为重要的抗癌先导化合物进行进一步的优化和研究。
{"title":"Multi-target design of fused cyclic pyrimidine-2-thione candidates as DNA intercalators and topoisomerase I/II inhibitors.","authors":"Mahmoud S Khalil, Ahmed A Al-Karmalawy, Ayman Abo Elmaaty, Galal A Elsayed, A M A Hassan","doi":"10.1080/17568919.2025.2602424","DOIUrl":"https://doi.org/10.1080/17568919.2025.2602424","url":null,"abstract":"<p><strong>Aim: </strong>We aimed to design and synthesize novel pyrimidine-2-thione derivatives (<b>1</b>-<b>13</b>) as Topoisomerase I/II (Topo I/II) inhibitors with DNA intercalation potential for cancer treatment.</p><p><strong>Materials & methods: </strong>Inhibitory concentration 50 (IC<sub>50</sub>) against mammary gland breast cancer, hepatocellular carcinoma, and colorectal carcinoma was determined for all compounds. The frontier candidates (<b>2</b>, <b>6</b>, <b>9</b>, <b>10</b>, and <b>11</b>) were evaluated for their DNA-binding ability, Topo I, and Topo II inhibiting potential. Moreover, cell cycle and apoptosis analysis were carried out.</p><p><strong>Results: </strong>Compound <b>2</b> displayed the best DNA-binding affinity with an IC<sub>50</sub> value of 37.24 µM in comparison to doxorubicin (Dox). Both compounds <b>2</b> and <b>9</b> showed superior nanomolar Topo I inhibitory potential, compared to Dox. Similarly, compounds <b>2</b> and <b>9</b> achieved better Topo II inhibition, exceeding that of Dox. It was revealed that compound <b>9</b> halted the cell cycle at both the P1 and G2 phases. In addition, compound <b>9</b> was able to boost the apoptosis at both the early and late apoptotic phases.</p><p><strong>Conclusion: </strong>Consequently, the compounds afforded can be regarded as prominent lead anticancer compounds for further optimization and investigation.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-14"},"PeriodicalIF":3.4,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741897","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
Small molecule inhibitors of HG-induced VEC ferroptosis: synthetic advances and therapeutic applications. hg诱导VEC铁下垂的小分子抑制剂:合成进展及治疗应用。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-12 DOI: 10.1080/17568919.2025.2602422
Yu-Xi Zheng, Ning Meng, Cheng-Shi Jiang

Ferroptosis is a form of Regulated Cell Death (RCD) found in recent years. Its typical characteristics are the abnormal accumulation of intracellular iron ions and the accumulation of lipid peroxidation products. Since its first systematic elucidation in 2012, a large number of studies have shown that ferroptosis is involved in a variety of pathophysiological processes. It has been reported that the pathological process of diabetic cardiovascular diseases (DVD) is closely associated with the activation of ferroptosis. Due to the long-term hyperglycemic environment in diabetic patients, vascular endothelial cells (VECs) are susceptible to ferroptosis, ultimately contributing to vascular dysfunction. Therefore, the development of inhibitors targeting ferroptosis is of great significance for the prevention and treatment of diabetic vascular complications. This review systematically expounds the latest research progress of the molecular mechanism of ferroptosis, and discusses its role in DVD. In addition, this review also comprehensively summarizes the latest advances in the synthesis and application of drugs and specific inhibitors targeting the ferroptosis pathway for disease treatment, thereby providing new therapeutic strategies for DVD.

铁下垂是近年来发现的一种细胞死亡形式。其典型特征是细胞内铁离子的异常积聚和脂质过氧化产物的积聚。自2012年首次系统阐明以来,大量研究表明,铁下垂参与多种病理生理过程。有报道称糖尿病性心血管疾病(DVD)的病理过程与铁下垂的激活密切相关。由于糖尿病患者长期处于高血糖环境,血管内皮细胞(VECs)易发生铁下垂,最终导致血管功能障碍。因此,开发针对铁下垂的抑制剂对于预防和治疗糖尿病血管并发症具有重要意义。本文系统阐述了铁下垂分子机制的最新研究进展,并讨论了其在DVD中的作用。此外,本文还全面综述了针对铁下垂通路的药物和特异性抑制剂的合成与应用的最新进展,从而为DVD提供新的治疗策略。
{"title":"Small molecule inhibitors of HG-induced VEC ferroptosis: synthetic advances and therapeutic applications.","authors":"Yu-Xi Zheng, Ning Meng, Cheng-Shi Jiang","doi":"10.1080/17568919.2025.2602422","DOIUrl":"https://doi.org/10.1080/17568919.2025.2602422","url":null,"abstract":"<p><p>Ferroptosis is a form of Regulated Cell Death (RCD) found in recent years. Its typical characteristics are the abnormal accumulation of intracellular iron ions and the accumulation of lipid peroxidation products. Since its first systematic elucidation in 2012, a large number of studies have shown that ferroptosis is involved in a variety of pathophysiological processes. It has been reported that the pathological process of diabetic cardiovascular diseases (DVD) is closely associated with the activation of ferroptosis. Due to the long-term hyperglycemic environment in diabetic patients, vascular endothelial cells (VECs) are susceptible to ferroptosis, ultimately contributing to vascular dysfunction. Therefore, the development of inhibitors targeting ferroptosis is of great significance for the prevention and treatment of diabetic vascular complications. This review systematically expounds the latest research progress of the molecular mechanism of ferroptosis, and discusses its role in DVD. In addition, this review also comprehensively summarizes the latest advances in the synthesis and application of drugs and specific inhibitors targeting the ferroptosis pathway for disease treatment, thereby providing new therapeutic strategies for DVD.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-14"},"PeriodicalIF":3.4,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741855","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
Purine derivatives as potent anticancer agents: a comprehensive review. 嘌呤衍生物作为强效抗癌剂的研究综述。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-09 DOI: 10.1080/17568919.2025.2594966
Fiza Farooqui, Abdul Rahman Khan, Mohd Arsh Khan, Malik Nasibullah, Jamal Akhtar Ansari

Cancer remains an incessantly rising cause of mortality worldwide, tempting millions of lives each year and posing a significant global health challenge. Available treatment modalities, including chemotherapy, have been associated with limited scope with severe side effects and complexities, underscoring the imperative need for more efficient and safe curative strategies. In this context, the rational design of multitargeted anticancer agents has gained momentum, aiming to enhance therapeutic outcomes while reducing systemic toxicity. The purine scaffold, a core structural motif found in essential biomolecules, such as deoxyribonucleic acid (DNA), ribonucleic acid (RNA), adenosine triphosphate (ATP), and nicotinamide adenine dinucleotide (NAD), has emerged as a promising pharmacophore in anticancer drug discovery. Notably, several synthetic purine analogues have received clinical approval owing to their potent anticancer activity, particularly when integrated with diverse heterocyclic frameworks. This review comprehensively summarizes the advances made over the past decade in the development of purine-based hybrid molecules, highlighting their mechanistic roles in overcoming drug resistance and targeting multiple oncogenic pathways. The insights presented herein underscore the versatility and therapeutic relevance of purine-based scaffolds and aim to guide future efforts in the rational design and development of drug-resistant and safer anticancer agents.

癌症仍然是世界范围内不断上升的死亡原因,每年夺去数百万人的生命,对全球健康构成重大挑战。现有的治疗方式,包括化疗,范围有限,副作用严重且复杂,强调迫切需要更有效和安全的治疗策略。在此背景下,合理设计多靶点抗癌药物已获得动力,旨在提高治疗效果,同时降低全身毒性。嘌呤支架是在脱氧核糖核酸(DNA)、核糖核酸(RNA)、三磷酸腺苷(ATP)和烟酰胺腺嘌呤二核苷酸(NAD)等生物分子中发现的核心结构基序,在抗癌药物研究中具有重要的应用前景。值得注意的是,一些合成嘌呤类似物由于其有效的抗癌活性而获得了临床批准,特别是当与各种杂环框架结合时。本文综述了近十年来嘌呤类杂化分子的研究进展,重点介绍了嘌呤类杂化分子在克服耐药和靶向多种致癌途径中的机制作用。本文提出的见解强调了嘌呤支架的多功能性和治疗相关性,旨在指导未来合理设计和开发耐药和更安全的抗癌药物。
{"title":"Purine derivatives as potent anticancer agents: a comprehensive review.","authors":"Fiza Farooqui, Abdul Rahman Khan, Mohd Arsh Khan, Malik Nasibullah, Jamal Akhtar Ansari","doi":"10.1080/17568919.2025.2594966","DOIUrl":"https://doi.org/10.1080/17568919.2025.2594966","url":null,"abstract":"<p><p>Cancer remains an incessantly rising cause of mortality worldwide, tempting millions of lives each year and posing a significant global health challenge. Available treatment modalities, including chemotherapy, have been associated with limited scope with severe side effects and complexities, underscoring the imperative need for more efficient and safe curative strategies. In this context, the rational design of multitargeted anticancer agents has gained momentum, aiming to enhance therapeutic outcomes while reducing systemic toxicity. The purine scaffold, a core structural motif found in essential biomolecules, such as deoxyribonucleic acid (DNA), ribonucleic acid (RNA), adenosine triphosphate (ATP), and nicotinamide adenine dinucleotide (NAD), has emerged as a promising pharmacophore in anticancer drug discovery. Notably, several synthetic purine analogues have received clinical approval owing to their potent anticancer activity, particularly when integrated with diverse heterocyclic frameworks. This review comprehensively summarizes the advances made over the past decade in the development of purine-based hybrid molecules, highlighting their mechanistic roles in overcoming drug resistance and targeting multiple oncogenic pathways. The insights presented herein underscore the versatility and therapeutic relevance of purine-based scaffolds and aim to guide future efforts in the rational design and development of drug-resistant and safer anticancer agents.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1-11"},"PeriodicalIF":3.4,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707914","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
Aurone-based α-glucosidase inhibitors for antidiabetic therapy: in vitro, in vivo, and in silico studies. 以aurone为基础的α-葡萄糖苷酶抑制剂用于降糖治疗:体外,体内和计算机研究。
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 Epub Date: 2025-11-12 DOI: 10.1080/17568919.2025.2587565
Ehsan Ullah Mughal, Nafeesa Naeem, Bushra Shakoor, Gehan Ahmed Othman, Amina Sadiq, Muhammad Naveed Zafar

Aims: This study aimed to evaluate the α-glucosidase inhibitory potential of newly synthesized aurone derivatives (1-14) using an integrated experimental and computational strategy, with emphasis on their antidiabetic potential.

Materials and methods: The compounds were evaluated through in vitro α-glucosidase inhibition and enzyme kinetic assays, along with in vivo studies to assess postprandial glucose control. Molecular docking, MM-GBSA calculations, and molecular dynamics (MD) simulations were performed to analyze interactions with diabetic targets (PDB IDs: 5NN4 and 6KK1). Furthermore, in silico ADME profiling and density functional theory (DFT) analyses were conducted to predict pharmacokinetic properties, drug-likeness, and electronic behavior.

Results: Several aurone derivatives exhibited strong α-glucosidase inhibition, surpassing standard drugs. Kinetic studies revealed a competitive inhibition mechanism, and in vivo evaluations confirmed their glucose-lowering effects - the first such report for aurones. Computational analyses indicated stable enzyme - ligand complexes with favorable binding affinities and ADME features. DFT results supported the observed structure - activity relationships and highlighted key electronic attributes influencing activity.

Conclusions: This comprehensive study identifies aurones as potent α-glucosidase inhibitors with significant therapeutic potential, providing a strong foundation for further development of aurone-based antidiabetic agents.

目的:采用实验与计算相结合的方法,研究新合成的金酮衍生物(1-14)对α-葡萄糖苷酶的抑制作用,重点研究其抗糖尿病作用。材料和方法:通过体外α-葡萄糖苷酶抑制和酶动力学分析,以及体内餐后血糖控制研究对化合物进行评价。通过分子对接、MM-GBSA计算和分子动力学(MD)模拟来分析与糖尿病靶点(PDB id: 5NN4和6KK1)的相互作用。此外,还进行了ADME分析和密度泛函理论(DFT)分析来预测药代动力学性质、药物相似性和电子行为。结果:几种aurone衍生物表现出较强的α-葡萄糖苷酶抑制作用,优于标准药物。动力学研究揭示了一种竞争性抑制机制,体内评价证实了它们的降血糖作用——这是对aurones的首次报道。计算分析表明,稳定的酶配体复合物具有良好的结合亲和力和ADME特征。DFT结果支持观察到的结构-活性关系,并突出了影响活性的关键电子属性。结论:本综合研究确定了aurone是有效的α-葡萄糖苷酶抑制剂,具有显著的治疗潜力,为进一步开发基于aurone的降糖药提供了坚实的基础。
{"title":"Aurone-based α-glucosidase inhibitors for antidiabetic therapy: <i>in vitro</i>, <i>in vivo</i>, and <i>in silico</i> studies.","authors":"Ehsan Ullah Mughal, Nafeesa Naeem, Bushra Shakoor, Gehan Ahmed Othman, Amina Sadiq, Muhammad Naveed Zafar","doi":"10.1080/17568919.2025.2587565","DOIUrl":"10.1080/17568919.2025.2587565","url":null,"abstract":"<p><strong>Aims: </strong>This study aimed to evaluate the α-glucosidase inhibitory potential of newly synthesized aurone derivatives (1-14) using an integrated experimental and computational strategy, with emphasis on their antidiabetic potential.</p><p><strong>Materials and methods: </strong>The compounds were evaluated through in vitro α-glucosidase inhibition and enzyme kinetic assays, along with <i>in</i> <i>vivo</i> studies to assess postprandial glucose control. Molecular docking, MM-GBSA calculations, and molecular dynamics (MD) simulations were performed to analyze interactions with diabetic targets (PDB IDs: 5NN4 and 6KK1). Furthermore, <i>in silico</i> ADME profiling and density functional theory (DFT) analyses were conducted to predict pharmacokinetic properties, drug-likeness, and electronic behavior.</p><p><strong>Results: </strong>Several aurone derivatives exhibited strong α-glucosidase inhibition, surpassing standard drugs. Kinetic studies revealed a competitive inhibition mechanism, and <i>in</i> <i>vivo</i> evaluations confirmed their glucose-lowering effects - the first such report for aurones. Computational analyses indicated stable enzyme - ligand complexes with favorable binding affinities and ADME features. DFT results supported the observed structure - activity relationships and highlighted key electronic attributes influencing activity.</p><p><strong>Conclusions: </strong>This comprehensive study identifies aurones as potent α-glucosidase inhibitors with significant therapeutic potential, providing a strong foundation for further development of aurone-based antidiabetic agents.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"3013-3024"},"PeriodicalIF":3.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698046/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145495109","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
α-Glucosidase-targeting 1,2,4-triazole antidiabetic candidates: comparative analysis and future perspectives. α-葡萄糖苷酶靶向1,2,4-三唑抗糖尿病候选物:比较分析及未来展望
IF 3.4 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 Epub Date: 2025-11-18 DOI: 10.1080/17568919.2025.2587561
Hamada S Abulkhair

Diabetes mellitus develops because of the disturbance in carbohydrate metabolism. The therapeutic goal for antidiabetic medications is to manage blood glucose level and to prevent hyperglycemia-associated complications. α-Glucosidase inhibitors represent one of the widely used oral hypoglycemics. This review highlights the potential of 1,2,4-triazole-containing synthetic molecules as antidiabetic agents, particularly focusing on their α-glucosidase inhibitory activity. It argues the significance of targeting α-glucosidase in managing type 2 diabetes and presents recent synthetic approaches for synthesizing 1,2,4-triazole derivatives. The mechanisms of action, SAR analysis, and docking insights are summarized for various reported 1,2,4-triazoles between 2020 and 2025. A comparative analysis was conducted to identify the most effective methodology and the best starting material for the synthesis of this class. Relative potencies and drug likeness characteristics of the reviewed candidates were also evaluated to identify whether one deserves forwarding to pre-clinical and clinical assessments. Many of these derivatives exhibited potent α-glucosidase enzyme inhibition, often outperforming standard marketed drugs like Acarbose. The review paves the way for medicinal chemists to develop new 1,2,4-triazole-incorporating molecular entities to build safe and effective agents for diabetes treatment.

糖尿病的发生是由于碳水化合物代谢紊乱。降糖药物的治疗目标是控制血糖水平和预防高血糖相关并发症。α-葡萄糖苷酶抑制剂是广泛应用的口服降糖药之一。本文综述了含1,2,4-三唑的合成分子作为抗糖尿病药物的潜力,特别是它们的α-葡萄糖苷酶抑制活性。本文讨论了靶向α-葡萄糖苷酶在治疗2型糖尿病中的意义,并介绍了合成1,2,4-三唑衍生物的最新合成方法。本文总结了2020年至2025年间各种1,2,4-三唑类化合物的作用机制、SAR分析和对接见解。进行了比较分析,以确定最有效的方法和最佳的起始材料,这类合成。还评估了审查的候选药物的相对效力和药物相似性特征,以确定是否值得转发到临床前和临床评估。许多这些衍生物表现出强有力的α-葡萄糖苷酶抑制作用,通常优于标准的上市药物,如阿卡波糖。该综述为药物化学家开发新的1,2,4-三唑类分子实体以构建安全有效的糖尿病治疗药物铺平了道路。
{"title":"α-Glucosidase-targeting 1,2,4-triazole antidiabetic candidates: comparative analysis and future perspectives.","authors":"Hamada S Abulkhair","doi":"10.1080/17568919.2025.2587561","DOIUrl":"10.1080/17568919.2025.2587561","url":null,"abstract":"<p><p>Diabetes mellitus develops because of the disturbance in carbohydrate metabolism. The therapeutic goal for antidiabetic medications is to manage blood glucose level and to prevent hyperglycemia-associated complications. α-Glucosidase inhibitors represent one of the widely used oral hypoglycemics. This review highlights the potential of 1,2,4-triazole-containing synthetic molecules as antidiabetic agents, particularly focusing on their α-glucosidase inhibitory activity. It argues the significance of targeting α-glucosidase in managing type 2 diabetes and presents recent synthetic approaches for synthesizing 1,2,4-triazole derivatives. The mechanisms of action, SAR analysis, and docking insights are summarized for various reported 1,2,4-triazoles between 2020 and 2025. A comparative analysis was conducted to identify the most effective methodology and the best starting material for the synthesis of this class. Relative potencies and drug likeness characteristics of the reviewed candidates were also evaluated to identify whether one deserves forwarding to pre-clinical and clinical assessments. Many of these derivatives exhibited potent α-glucosidase enzyme inhibition, often outperforming standard marketed drugs like Acarbose. The review paves the way for medicinal chemists to develop new 1,2,4-triazole-incorporating molecular entities to build safe and effective agents for diabetes treatment.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"3025-3040"},"PeriodicalIF":3.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698052/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145539946","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
期刊
Future medicinal chemistry
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1