首页 > 最新文献

Molecular Diversity最新文献

英文 中文
Synthesis of porphyrin-formononetin derivatives and their anti-tumor activity studies.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-09 DOI: 10.1007/s11030-025-11183-w
Lingyan Yang, Shan He, Chen Tang, Lei Shi, Zhenhua Liu, Zejie Tian, Hui Li, Xufeng He, Jiding Liao, Yunmei Liu

Photodynamic therapy (PDT) has received much attention in cancer treatment because of its low toxicity and side effects. In this study, we successfully synthesized 14 novel porphyrin-formononetin derivatives. In reactive oxygen species detection experiments, the target compounds 4a-6d caused a significant decrease in the fluorescence intensity of DPBF compared with the porphyrin parent and formononetin feedstock after illumination, and it was found that the target compound had a higher ROS quantum yield, among which the quantum yield of compound 6c was higher. In the in vitro anti-tumor activity assay, the target compounds 4a-6d exhibited a certain degree of growth inhibition against six cancer cells (A549, MDA-MB-231, HCT-116, HGC-27, DU145, and TCCSUP) under light conditions, whereas the cytotoxicity of the target compounds against the normal cells H9c2 was less. The results of the scratch assay showed that 6c could inhibit the growth of tumor cells by inhibiting the migration of DU145 cells. The experimental results indicate that the target compounds achieve the synergistic effect of PDT and chemotherapy.

{"title":"Synthesis of porphyrin-formononetin derivatives and their anti-tumor activity studies.","authors":"Lingyan Yang, Shan He, Chen Tang, Lei Shi, Zhenhua Liu, Zejie Tian, Hui Li, Xufeng He, Jiding Liao, Yunmei Liu","doi":"10.1007/s11030-025-11183-w","DOIUrl":"https://doi.org/10.1007/s11030-025-11183-w","url":null,"abstract":"<p><p>Photodynamic therapy (PDT) has received much attention in cancer treatment because of its low toxicity and side effects. In this study, we successfully synthesized 14 novel porphyrin-formononetin derivatives. In reactive oxygen species detection experiments, the target compounds 4a-6d caused a significant decrease in the fluorescence intensity of DPBF compared with the porphyrin parent and formononetin feedstock after illumination, and it was found that the target compound had a higher ROS quantum yield, among which the quantum yield of compound 6c was higher. In the in vitro anti-tumor activity assay, the target compounds 4a-6d exhibited a certain degree of growth inhibition against six cancer cells (A549, MDA-MB-231, HCT-116, HGC-27, DU145, and TCCSUP) under light conditions, whereas the cytotoxicity of the target compounds against the normal cells H9c2 was less. The results of the scratch assay showed that 6c could inhibit the growth of tumor cells by inhibiting the migration of DU145 cells. The experimental results indicate that the target compounds achieve the synergistic effect of PDT and chemotherapy.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810162","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A scaffold repositioning approach: dihydroBenzoImidazoTriazineDione (BITD) derivatives as selective ALDH1A1 inhibitors. 支架重新定位方法:二氢苯并咪唑三嗪二酮 (BITD) 衍生物作为 ALDH1A1 的选择性抑制剂。
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-04 DOI: 10.1007/s11030-025-11179-6
Bianca Laura Bernardoni, Ilaria D'Agostino, Sonia Siragusa, Mattia Mori, Silvia Garavaglia, Concettina La Motta

The overexpression of the Aldehyde Dehydrogenases 1A subfamily (ALDH1As) in various diseases, particularly in cancer, has made it an important target for therapeutic applications. Interestingly, the 1A1 isoenzyme plays a role in tumor initiation and progression, being identified as a biomarker for cancer stem cells. However, although promising, current ALDH1A1 inhibitors suffer from a lack of isoform selectivity and off-target toxicity. This study aims to address these limitations by developing a new class of ALDH1A1-selective inhibitors. By leveraging structural analogies with Isatin-based ALDH1A1 inhibitors, we designed compounds containing a dihydrobenzo[4,5]imidazo[2,1-c][1,2,4]triazine-3,4-dione (BITD) core, that emerged from a repositioning approach. Using a microwave-assisted protocol, a small library of derivatives was synthesized, and enzymatic assays highlighted a promising isoform specificity for ALDH1A1 among ALDH1As, with the best-in-class compound 5, showing an inhibition of the enzyme activity of 86% for ALDH1A1 and no inhibition for 1A2 and 1A3 isoenzymes. In silico studies further elucidated the binding mode of 5, providing a rational basis for the observed selectivity. These findings represent a promising strategy for the development of more selective ALDH1A1 inhibitors, laying the foundation for further optimization processes.

{"title":"A scaffold repositioning approach: dihydroBenzoImidazoTriazineDione (BITD) derivatives as selective ALDH1A1 inhibitors.","authors":"Bianca Laura Bernardoni, Ilaria D'Agostino, Sonia Siragusa, Mattia Mori, Silvia Garavaglia, Concettina La Motta","doi":"10.1007/s11030-025-11179-6","DOIUrl":"https://doi.org/10.1007/s11030-025-11179-6","url":null,"abstract":"<p><p>The overexpression of the Aldehyde Dehydrogenases 1A subfamily (ALDH1As) in various diseases, particularly in cancer, has made it an important target for therapeutic applications. Interestingly, the 1A1 isoenzyme plays a role in tumor initiation and progression, being identified as a biomarker for cancer stem cells. However, although promising, current ALDH1A1 inhibitors suffer from a lack of isoform selectivity and off-target toxicity. This study aims to address these limitations by developing a new class of ALDH1A1-selective inhibitors. By leveraging structural analogies with Isatin-based ALDH1A1 inhibitors, we designed compounds containing a dihydrobenzo[4,5]imidazo[2,1-c][1,2,4]triazine-3,4-dione (BITD) core, that emerged from a repositioning approach. Using a microwave-assisted protocol, a small library of derivatives was synthesized, and enzymatic assays highlighted a promising isoform specificity for ALDH1A1 among ALDH1As, with the best-in-class compound 5, showing an inhibition of the enzyme activity of 86% for ALDH1A1 and no inhibition for 1A2 and 1A3 isoenzymes. In silico studies further elucidated the binding mode of 5, providing a rational basis for the observed selectivity. These findings represent a promising strategy for the development of more selective ALDH1A1 inhibitors, laying the foundation for further optimization processes.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrating deep learning and molecular dynamics simulations for FXR antagonist discovery.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-02 DOI: 10.1007/s11030-025-11145-2
Yueying Yang, Yuxin Huang, Hanxiao Shen, Ding Wang, Zhen Liu, Wei Zhu, Qing Liu

Farnesoid X receptor (FXR) is a key regulator of bile acid, lipid, and glucose homeostasis, making it a promising target for treating metabolic diseases. FXR antagonists have shown therapeutic potential in cholestasis, metabolic disorders, and certain cancers, while clinically approved FXR antagonists remain unavailable and underrepresented in current treatment strategies. To address this, we developed deep learning models for predicting FXR antagonistic activity (ANTCL) and toxicity (TOXCL). Screening 217,345 compounds from the HMDB database identified eleven human metabolite candidates with significant FXR binding potential. Molecular dynamics simulations and binding free energy calculations revealed five more stable complexes compared to the reference compound Gly-MCA, with HMDB0253354 (Fulvestrant) and HMDB0242367 (ZM 189154) standing out for their binding free energies. Hydrophobic interactions, particularly involving residues MET328, PHE329, and ALA291, contributed to their stability. These results demonstrate the effectiveness of deep learning in FXR antagonist discovery and highlight the potential of HMDB0253354 and HMDB0242367 as promising candidates for metabolic disease treatment.

法尼类固醇 X 受体(FXR)是胆汁酸、脂质和葡萄糖稳态的关键调节因子,使其成为治疗代谢性疾病的有望靶点。FXR拮抗剂在胆汁淤积症、代谢紊乱和某些癌症中显示出治疗潜力,而临床批准的FXR拮抗剂在目前的治疗策略中仍然不可用且代表性不足。为了解决这个问题,我们开发了用于预测 FXR 拮抗活性(ANTCL)和毒性(TOXCL)的深度学习模型。从 HMDB 数据库中筛选出 217,345 种化合物,发现了 11 种具有显著 FXR 结合潜力的候选人类代谢物。分子动力学模拟和结合自由能计算显示,与参考化合物 Gly-MCA 相比,五种复合物更为稳定,其中 HMDB0253354(氟维司群)和 HMDB0242367(ZM 189154)的结合自由能更为突出。疏水相互作用,尤其是涉及残基 MET328、PHE329 和 ALA291 的疏水相互作用,有助于提高它们的稳定性。这些结果证明了深度学习在发现 FXR 拮抗剂方面的有效性,并凸显了 HMDB0253354 和 HMDB0242367 作为代谢疾病治疗候选药物的潜力。
{"title":"Integrating deep learning and molecular dynamics simulations for FXR antagonist discovery.","authors":"Yueying Yang, Yuxin Huang, Hanxiao Shen, Ding Wang, Zhen Liu, Wei Zhu, Qing Liu","doi":"10.1007/s11030-025-11145-2","DOIUrl":"https://doi.org/10.1007/s11030-025-11145-2","url":null,"abstract":"<p><p>Farnesoid X receptor (FXR) is a key regulator of bile acid, lipid, and glucose homeostasis, making it a promising target for treating metabolic diseases. FXR antagonists have shown therapeutic potential in cholestasis, metabolic disorders, and certain cancers, while clinically approved FXR antagonists remain unavailable and underrepresented in current treatment strategies. To address this, we developed deep learning models for predicting FXR antagonistic activity (ANTCL) and toxicity (TOXCL). Screening 217,345 compounds from the HMDB database identified eleven human metabolite candidates with significant FXR binding potential. Molecular dynamics simulations and binding free energy calculations revealed five more stable complexes compared to the reference compound Gly-MCA, with HMDB0253354 (Fulvestrant) and HMDB0242367 (ZM 189154) standing out for their binding free energies. Hydrophobic interactions, particularly involving residues MET328, PHE329, and ALA291, contributed to their stability. These results demonstrate the effectiveness of deep learning in FXR antagonist discovery and highlight the potential of HMDB0253354 and HMDB0242367 as promising candidates for metabolic disease treatment.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143762606","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Strategic 1,n-migration of boronate complexes: a novel platform for remote C-C bond construction.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-02 DOI: 10.1007/s11030-025-11169-8
Yi-Ming Chen, Xue Li, Zhi-Gang Xu

Organoboron compounds play a pivotal role in diverse scientific disciplines, including chemistry, materials science, energy research, and medicinal chemistry. In recent years, research efforts have predominantly focused on 1,2-metallate migrations of tetracoordinate boronate complexes, while remote migrations, particularly 1,n-metallate migrations (n > 2), remain challenging due to their inherent complexity. This comprehensive review systematically examines seminal contributions to the field of 1,n-metallate migration reactions (n > 2). Our critical analysis reveals that progress in this domain has been fundamentally driven by the strategic design and synthesis of novel tetracoordinate boron complexes, with a notable evolution from conventional O-B coordination motifs to more sophisticated C-B-bonded architectures. Recent methodological advancements have further expanded the structural diversity and mechanistic understanding of these transformations. Although the number of reported cases remains limited and the research landscape is somewhat fragmented, the existing systems underscore the significance of these migration reactions, drawing considerable attention to this area and inspiring further exploration.

{"title":"Strategic 1,n-migration of boronate complexes: a novel platform for remote C-C bond construction.","authors":"Yi-Ming Chen, Xue Li, Zhi-Gang Xu","doi":"10.1007/s11030-025-11169-8","DOIUrl":"https://doi.org/10.1007/s11030-025-11169-8","url":null,"abstract":"<p><p>Organoboron compounds play a pivotal role in diverse scientific disciplines, including chemistry, materials science, energy research, and medicinal chemistry. In recent years, research efforts have predominantly focused on 1,2-metallate migrations of tetracoordinate boronate complexes, while remote migrations, particularly 1,n-metallate migrations (n > 2), remain challenging due to their inherent complexity. This comprehensive review systematically examines seminal contributions to the field of 1,n-metallate migration reactions (n > 2). Our critical analysis reveals that progress in this domain has been fundamentally driven by the strategic design and synthesis of novel tetracoordinate boron complexes, with a notable evolution from conventional O-B coordination motifs to more sophisticated C-B-bonded architectures. Recent methodological advancements have further expanded the structural diversity and mechanistic understanding of these transformations. Although the number of reported cases remains limited and the research landscape is somewhat fragmented, the existing systems underscore the significance of these migration reactions, drawing considerable attention to this area and inspiring further exploration.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143770917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Searching potential GSK-3β inhibitors from marine sources using atomistic simulations.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-02 DOI: 10.1007/s11030-025-11174-x
Tran Thi Hoai Van, Minh Quan Pham, Truong Thi Thu Huong, Bui Nguyen Thanh Long, Pham Quoc Long, Le Thi Thuy Huong, George Binh Lenon, Nguyen Thi Thanh Uyen, Son Tung Ngo

Glycogen synthase kinase-3 beta, GSK-3β, is one of the most common targets for cancer treatment. Inhibiting the biological activity of the enzyme can lead to the prevention of cancer development. Especially, estimating a new inhibitor for preventing GSK-3β by using natural compounds is of great interest. In this context, the marine compounds were investigated for their ligand-binding affinity to GSK-3β via atomistic simulations. The compounds, including xanalteric acid I, chaunolidone A, macrolactin V, and aspergiolide A, were suggested that can inhibit GSK-3β via molecular docking and steered-MD simulations. Moreover, the potency of these compounds was also confirmed via the perturbation simulations. Furthermore, the toxicity prediction also indicates that these compounds would adopt less toxicity. Therefore, it may be argued that four compounds can play as potential inhibitors preventing GSK-3β. In addition, the residues including Ile62, Val135, Pro136, Arg141, Lys183, Gln185, Asn186, and Asp200 play a crucial role in the GSK-3β binding process.

{"title":"Searching potential GSK-3β inhibitors from marine sources using atomistic simulations.","authors":"Tran Thi Hoai Van, Minh Quan Pham, Truong Thi Thu Huong, Bui Nguyen Thanh Long, Pham Quoc Long, Le Thi Thuy Huong, George Binh Lenon, Nguyen Thi Thanh Uyen, Son Tung Ngo","doi":"10.1007/s11030-025-11174-x","DOIUrl":"https://doi.org/10.1007/s11030-025-11174-x","url":null,"abstract":"<p><p>Glycogen synthase kinase-3 beta, GSK-3β, is one of the most common targets for cancer treatment. Inhibiting the biological activity of the enzyme can lead to the prevention of cancer development. Especially, estimating a new inhibitor for preventing GSK-3β by using natural compounds is of great interest. In this context, the marine compounds were investigated for their ligand-binding affinity to GSK-3β via atomistic simulations. The compounds, including xanalteric acid I, chaunolidone A, macrolactin V, and aspergiolide A, were suggested that can inhibit GSK-3β via molecular docking and steered-MD simulations. Moreover, the potency of these compounds was also confirmed via the perturbation simulations. Furthermore, the toxicity prediction also indicates that these compounds would adopt less toxicity. Therefore, it may be argued that four compounds can play as potential inhibitors preventing GSK-3β. In addition, the residues including Ile62, Val135, Pro136, Arg141, Lys183, Gln185, Asn186, and Asp200 play a crucial role in the GSK-3β binding process.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143762612","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery and optimization of AAK1 inhibitors based on 1H-indazole scaffold for the potential treatment of SARS-CoV-2 infection.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-04-02 DOI: 10.1007/s11030-025-11135-4
Zi Hui, Haowen Deng, Yueying Xu, Yuan Gao, Chenfeng Zhai, Nian-Dong Mao, Hao Che, Zhen Li, Yuting Zhang, Hang Zhang, Tian Xie, Xiang-Yang Ye

The process of various virus entry into host cells, including SARS-CoV-2, is mediated by clathrin-mediated endocytosis (CME). AP-2 plays a crucial role in this process by recognizing membrane receptors and binding with clathrin, facilitating the formation of clathrin-coated vesicles and promoting CME. AAK1 catalyzes the phosphorylation of AP2M1 subunit at Thr156. Therefore, suppressing AAK1 activity can hinder virus invasion by blocking CME. indicating that AAK1 could be a potential target for developing novel antiviral drugs against SARS-CoV-2. In this study, we present a series of novel AAK1 inhibitors based on previously reported AAK1 inhibitors. Drug design was carried out by fusing the 1H-indazole scaffold of SGC-AAK1-1 with pharmacophore groups of compound 6, and further optimized with the assistance of molecular docking. Among the 42 compounds novelly synthesized, compounds 9i, 9s, 11f and 11l exhibited comparable antiviral activity against SARS-CoV-2 infection compared to reference compound 6 at the concentration of 3 μM. Particularly, 11f showed almost no cytotoxicity at all tested concentrations. Additionally, 11f exhibited favorable predictive pharmacokinetic properties. These findings support the potential of 11f as a lead compound for developing antiviral drugs targeting SARS-CoV-2 infection, as well as potentially other viruses which are dependent on the CME process to enter host cells. In summary, we have expanded the structural types of AAK1 inhibitors and successfully obtained effective AAK1 inhibitors with antiviral capabilities.

{"title":"Discovery and optimization of AAK1 inhibitors based on 1H-indazole scaffold for the potential treatment of SARS-CoV-2 infection.","authors":"Zi Hui, Haowen Deng, Yueying Xu, Yuan Gao, Chenfeng Zhai, Nian-Dong Mao, Hao Che, Zhen Li, Yuting Zhang, Hang Zhang, Tian Xie, Xiang-Yang Ye","doi":"10.1007/s11030-025-11135-4","DOIUrl":"https://doi.org/10.1007/s11030-025-11135-4","url":null,"abstract":"<p><p>The process of various virus entry into host cells, including SARS-CoV-2, is mediated by clathrin-mediated endocytosis (CME). AP-2 plays a crucial role in this process by recognizing membrane receptors and binding with clathrin, facilitating the formation of clathrin-coated vesicles and promoting CME. AAK1 catalyzes the phosphorylation of AP2M1 subunit at Thr156. Therefore, suppressing AAK1 activity can hinder virus invasion by blocking CME. indicating that AAK1 could be a potential target for developing novel antiviral drugs against SARS-CoV-2. In this study, we present a series of novel AAK1 inhibitors based on previously reported AAK1 inhibitors. Drug design was carried out by fusing the 1H-indazole scaffold of SGC-AAK1-1 with pharmacophore groups of compound 6, and further optimized with the assistance of molecular docking. Among the 42 compounds novelly synthesized, compounds 9i, 9s, 11f and 11l exhibited comparable antiviral activity against SARS-CoV-2 infection compared to reference compound 6 at the concentration of 3 μM. Particularly, 11f showed almost no cytotoxicity at all tested concentrations. Additionally, 11f exhibited favorable predictive pharmacokinetic properties. These findings support the potential of 11f as a lead compound for developing antiviral drugs targeting SARS-CoV-2 infection, as well as potentially other viruses which are dependent on the CME process to enter host cells. In summary, we have expanded the structural types of AAK1 inhibitors and successfully obtained effective AAK1 inhibitors with antiviral capabilities.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143770913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computational screening for natural compounds as potential immune checkpoint inhibitors against TIGIT, a new avenue in cancer immunotherapy.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-03-31 DOI: 10.1007/s11030-025-11172-z
Aritra Chakraborty, Amit Kumar

The TIGIT-PVR signalling pathway is a key mechanism of tumour immune evasion, making it an attractive target for cancer immunotherapy. Despite the recent advances in anti-TIGIT antibodies, monoclonal antibody-based therapeutics present significant challenges because of their immunogenicity and immune-related side effects. This study presents a new path involving natural compounds as potential small molecule inhibitors of TIGIT, providing a possible alternative to antibodies in cancer immunotherapy. Through a comprehensive in silico workflow combining structure-based virtual screening, ADMET analysis, Molecular docking and molecular dynamics simulations, six promising candidates, mostly of bacterial origin, were identified: Neomycin K, 4'-Deoxybutirosin A, 5-Glucosyl-neamine, S-11-A, 12-carbamoylstreptothricin E acid, and Zwittermicin A. These candidates demonstrated favourable binding energies, stable interactions, and the capacity to block TIGIT-PVR signalling. The compounds can potentially compete with PVR to bind to TIGIT, limiting the formation of the TIGIT-PVR complex, which typically activates an inhibitory cascade in T cells and NK cells, reducing their anti-tumour activity. By disrupting this interaction, the identified compounds have the potential to stimulate T cell and NK cell responses against cancer cells. Such natural compounds potentially provide better tissue penetration and reduced immunogenicity compared to conventional antibody therapies. The discovery of bacterial-derived compounds as TIGIT inhibitors presents a new direction in the investigation of microbial metabolites for cancer immunotherapy. This strategy not only identifies a new class of TIGIT inhibitors but also provides a robust computational framework for discovering and characterizing small molecule immune checkpoint inhibitors, paving the way for subsequent experimental validation to explore their efficacy in restoring anti-tumour immune responses and improving clinical outcomes for cancer patients.

{"title":"Computational screening for natural compounds as potential immune checkpoint inhibitors against TIGIT, a new avenue in cancer immunotherapy.","authors":"Aritra Chakraborty, Amit Kumar","doi":"10.1007/s11030-025-11172-z","DOIUrl":"https://doi.org/10.1007/s11030-025-11172-z","url":null,"abstract":"<p><p>The TIGIT-PVR signalling pathway is a key mechanism of tumour immune evasion, making it an attractive target for cancer immunotherapy. Despite the recent advances in anti-TIGIT antibodies, monoclonal antibody-based therapeutics present significant challenges because of their immunogenicity and immune-related side effects. This study presents a new path involving natural compounds as potential small molecule inhibitors of TIGIT, providing a possible alternative to antibodies in cancer immunotherapy. Through a comprehensive in silico workflow combining structure-based virtual screening, ADMET analysis, Molecular docking and molecular dynamics simulations, six promising candidates, mostly of bacterial origin, were identified: Neomycin K, 4'-Deoxybutirosin A, 5-Glucosyl-neamine, S-11-A, 12-carbamoylstreptothricin E acid, and Zwittermicin A. These candidates demonstrated favourable binding energies, stable interactions, and the capacity to block TIGIT-PVR signalling. The compounds can potentially compete with PVR to bind to TIGIT, limiting the formation of the TIGIT-PVR complex, which typically activates an inhibitory cascade in T cells and NK cells, reducing their anti-tumour activity. By disrupting this interaction, the identified compounds have the potential to stimulate T cell and NK cell responses against cancer cells. Such natural compounds potentially provide better tissue penetration and reduced immunogenicity compared to conventional antibody therapies. The discovery of bacterial-derived compounds as TIGIT inhibitors presents a new direction in the investigation of microbial metabolites for cancer immunotherapy. This strategy not only identifies a new class of TIGIT inhibitors but also provides a robust computational framework for discovering and characterizing small molecule immune checkpoint inhibitors, paving the way for subsequent experimental validation to explore their efficacy in restoring anti-tumour immune responses and improving clinical outcomes for cancer patients.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143750639","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of novel quinazoline derivatives containing trifluoromethyl against cell proliferation by targeting werner helicase.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-03-28 DOI: 10.1007/s11030-025-11175-w
Gang Yu, Jia Yu, Yunyun Zhou, Kun Liu, Xiaolin Peng, Guangcan Xu, Chao Chen, Xueling Meng, Xiaoping Zeng, Hui Wu, Ningning Zan, Heng Luo, Bixue Xu

A series of novel 2-trifluoromethyl-4-aminoquinazoline derivatives were designed and synthesized, and their antitumor activities were evaluated. Among them, several target compounds exhibited nanomolar inhibitory activities against K562 and LNCaP. Meanwhile, the results of in vitro and in vivo activity evaluation showed that compound 9 had the significant selective anticancer activity and the lower toxicity. The target prediction and pathway analysis showed that the mechanism of compound 9 on the proliferation inhibitory activity of K562 and PC3 cells may be via inhibiting werner helicase (WRN) activity and affecting DNA damage repair. As expected, biological evaluation showed that compound 9 bind to WRN, significantly downregulated the expression of WRN, inhibited the MDM2/p53 pathway, to render the damaged DNA unrepaired, eventually causing mitotic arrest and cell death. Our findings provide a foundation for further research of trifluoromethyl-quinazoline-4-amines as WRN-dependent anticancer agents that targeting DNA damage repair pathway.

{"title":"Discovery of novel quinazoline derivatives containing trifluoromethyl against cell proliferation by targeting werner helicase.","authors":"Gang Yu, Jia Yu, Yunyun Zhou, Kun Liu, Xiaolin Peng, Guangcan Xu, Chao Chen, Xueling Meng, Xiaoping Zeng, Hui Wu, Ningning Zan, Heng Luo, Bixue Xu","doi":"10.1007/s11030-025-11175-w","DOIUrl":"https://doi.org/10.1007/s11030-025-11175-w","url":null,"abstract":"<p><p>A series of novel 2-trifluoromethyl-4-aminoquinazoline derivatives were designed and synthesized, and their antitumor activities were evaluated. Among them, several target compounds exhibited nanomolar inhibitory activities against K562 and LNCaP. Meanwhile, the results of in vitro and in vivo activity evaluation showed that compound 9 had the significant selective anticancer activity and the lower toxicity. The target prediction and pathway analysis showed that the mechanism of compound 9 on the proliferation inhibitory activity of K562 and PC3 cells may be via inhibiting werner helicase (WRN) activity and affecting DNA damage repair. As expected, biological evaluation showed that compound 9 bind to WRN, significantly downregulated the expression of WRN, inhibited the MDM2/p53 pathway, to render the damaged DNA unrepaired, eventually causing mitotic arrest and cell death. Our findings provide a foundation for further research of trifluoromethyl-quinazoline-4-amines as WRN-dependent anticancer agents that targeting DNA damage repair pathway.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143727154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An updated survey on the recent advancement of antimalarial molecules: synthetic methodologies and SAR studies.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-03-28 DOI: 10.1007/s11030-025-11143-4
Hamna Nawaz, Ayesha Malik, Nasir Rasool, Aqsa Kanwal, Tahira Khalid, Iffat Almas, Samreen Gul Khan

An ongoing escalation in malaria cases is a critical global health issue and also a leading source of increased mortality rates. Infected mosquitos spread the Plasmodium parasite-based disease,. Recent advancements in antimalarial drug discovery have focused on developing novel molecules with unique mechanisms of action to combat this growing resistance. This review focuses on the latest findings in synthesizing hybrid antimalarial compounds, which combine different pharmacophores to enhance therapeutic efficacy, resulting in compounds that exhibit potent action against both resistant/sensitive strains of chloroquine, quinine, and artemisinin. Notably, the incorporation of diverse scaffolds such as 1,2,4-trioxanes, halogenated quinolines, pyrazoline, and sulfonamides has led to promising candidates that not only demonstrate high potency but also favorable pharmacokinetic profiles for drug development, the exploration of novel targets. This review highlights the progress (2019-2024) made in identifying and synthesizing hybrid antimalarial molecules while addressing existing challenges in the field.

{"title":"An updated survey on the recent advancement of antimalarial molecules: synthetic methodologies and SAR studies.","authors":"Hamna Nawaz, Ayesha Malik, Nasir Rasool, Aqsa Kanwal, Tahira Khalid, Iffat Almas, Samreen Gul Khan","doi":"10.1007/s11030-025-11143-4","DOIUrl":"https://doi.org/10.1007/s11030-025-11143-4","url":null,"abstract":"<p><p>An ongoing escalation in malaria cases is a critical global health issue and also a leading source of increased mortality rates. Infected mosquitos spread the Plasmodium parasite-based disease,. Recent advancements in antimalarial drug discovery have focused on developing novel molecules with unique mechanisms of action to combat this growing resistance. This review focuses on the latest findings in synthesizing hybrid antimalarial compounds, which combine different pharmacophores to enhance therapeutic efficacy, resulting in compounds that exhibit potent action against both resistant/sensitive strains of chloroquine, quinine, and artemisinin. Notably, the incorporation of diverse scaffolds such as 1,2,4-trioxanes, halogenated quinolines, pyrazoline, and sulfonamides has led to promising candidates that not only demonstrate high potency but also favorable pharmacokinetic profiles for drug development, the exploration of novel targets. This review highlights the progress (2019-2024) made in identifying and synthesizing hybrid antimalarial molecules while addressing existing challenges in the field.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143735604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deep learning in the discovery of antiviral peptides and peptidomimetics: databases and prediction tools.
IF 3.9 2区 化学 Q2 CHEMISTRY, APPLIED Pub Date : 2025-03-28 DOI: 10.1007/s11030-025-11173-y
Maryam Nawaz, Yao Huiyuan, Fahad Akhtar, Ma Tianyue, Heng Zheng

Antiviral peptides (AVPs) represent a novel and promising therapeutic alternative to conventional antiviral treatments, due to their broad-spectrum activity, high specificity, and low toxicity. The emergence of zoonotic viruses such as Zika, Ebola, and SARS-CoV-2 have accelerated AVP research, driven by advancements in data availability and artificial intelligence (AI). This review focuses on the development of AVP databases, their physicochemical properties, and predictive tools utilizing machine learning for AVP discovery. Machine learning plays a pivotal role in advancing and developing antiviral peptides and peptidomimetics, particularly through the development of specialized databases such as DRAVP, AVPdb, and DBAASP. These resources facilitate AVP characterization but face limitations, including small datasets, incomplete annotations, and inadequate integration with multi-omics data.The antiviral efficacy of AVPs is closely linked to their physicochemical properties, such as hydrophobicity and amphipathic α-helical structures, which enable viral membrane disruption and specific target interactions. Computational prediction tools employing machine learning and deep learning have significantly advanced AVP discovery. However, challenges like overfitting, limited experimental validation, and a lack of mechanistic insights hinder clinical translation.Future advancements should focus on improved validation frameworks, integration of in vivo data, and the development of interpretable models to elucidate AVP mechanisms. Expanding predictive models to address multi-target interactions and incorporating complex biological environments will be crucial for translating AVPs into effective clinical therapies.

{"title":"Deep learning in the discovery of antiviral peptides and peptidomimetics: databases and prediction tools.","authors":"Maryam Nawaz, Yao Huiyuan, Fahad Akhtar, Ma Tianyue, Heng Zheng","doi":"10.1007/s11030-025-11173-y","DOIUrl":"https://doi.org/10.1007/s11030-025-11173-y","url":null,"abstract":"<p><p>Antiviral peptides (AVPs) represent a novel and promising therapeutic alternative to conventional antiviral treatments, due to their broad-spectrum activity, high specificity, and low toxicity. The emergence of zoonotic viruses such as Zika, Ebola, and SARS-CoV-2 have accelerated AVP research, driven by advancements in data availability and artificial intelligence (AI). This review focuses on the development of AVP databases, their physicochemical properties, and predictive tools utilizing machine learning for AVP discovery. Machine learning plays a pivotal role in advancing and developing antiviral peptides and peptidomimetics, particularly through the development of specialized databases such as DRAVP, AVPdb, and DBAASP. These resources facilitate AVP characterization but face limitations, including small datasets, incomplete annotations, and inadequate integration with multi-omics data.The antiviral efficacy of AVPs is closely linked to their physicochemical properties, such as hydrophobicity and amphipathic α-helical structures, which enable viral membrane disruption and specific target interactions. Computational prediction tools employing machine learning and deep learning have significantly advanced AVP discovery. However, challenges like overfitting, limited experimental validation, and a lack of mechanistic insights hinder clinical translation.Future advancements should focus on improved validation frameworks, integration of in vivo data, and the development of interpretable models to elucidate AVP mechanisms. Expanding predictive models to address multi-target interactions and incorporating complex biological environments will be crucial for translating AVPs into effective clinical therapies.</p>","PeriodicalId":708,"journal":{"name":"Molecular Diversity","volume":" ","pages":""},"PeriodicalIF":3.9,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143735606","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Diversity
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:481959085
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