{"title":"Identification of Flavonoid-based Hypoxia-inducible Factor-2 Alpha Inhibitors for the Treatment of Breast Cancer- In silico and In vitro Evidence.","authors":"Shahinaz, Mursaleen Baba, Ravi Gor, Chandrasudan Ramamurthy, Habeeb Shaik Mohideen, Satish Ramalingam, Thangavel Mahalingam Vijayakumar","doi":"10.2174/0118715206377378250414052656","DOIUrl":"https://doi.org/10.2174/0118715206377378250414052656","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer (BC) is a common malignancy that poses a serious threat to women's health. The hypoxic tumor microenvironment in BC promotes drug resistance, making hypoxia-targeted therapies crucial. Targeting hypoxia-inducible factors (HIFs), particularly HIF-2α, has emerged as a promising approach to inhibit tumor growth and improve response to chemotherapy and radiotherapy. However, further research is required to fully understand the role of HIF-2α to develop more effective treatments for BC.</p><p><strong>Aim: </strong>The aim of this study is to identify phytochemicals that target HIF-2α and evaluate their effects on the MCF-7 breast cancer cell line under hypoxic conditions.</p><p><strong>Methods: </strong>Molecular docking identified phytochemicals targeting HIF-2α, with high-affinity compounds undergoing stability evaluation via GROMACS molecular dynamics simulations. ADMET and toxicity assessments were performed using SwissADME and ProTox-3.0. In-vitro assays on hypoxic MCF-7 cells examined cell viability and gene expression. The expression of HIF-2α-regulated genes (VEGFA, CCND1, GLUT1) was analyzed by using qRT-PCR.</p><p><strong>Results: </strong>Molecular docking revealed that naringin (-8.2 Kcal/mol) and morin (-7.1 Kcal/mol) showed better binding affinity than the standard drug, belzutifan (-7.7 Kcal/mol). Dynamic simulations, including RMSD, RMSF, Hbond interactions, Rg, SASA, and PE, confirmed their strong binding potential. Morin, in particular, demonstrated more H-bond interactions and met Lipinski's Rule of Five, making it a promising candidate for in vitro studies. It reduced cell viability with an IC50 of 118 μM and significantly downregulated HIF-2α-associated genes.</p><p><strong>Conclusion: </strong>Morin demonstrated promising anti-cancer activity under hypoxic conditions by inhibiting HIF-2α in the hypoxia signaling pathway.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143956514","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}
Laura Sartori Assunção, Iara Patricia Kretzer, Jelver Alexander Sierra Restrepo, Leonidas João de Mello Junior, Flavio Augusto Rocha Barbosa, Misael Ferreira, Marcus Mandolesi Sá, Tânia Beatriz Creczynski-Pasa
{"title":"Methyl (Z)-2-(Isothioureidomethyl)-2-pentenoate Hydrobromide Induces Cell Cycle Arrest and Disrupts Mitosis in a Melanoma Cell Line.","authors":"Laura Sartori Assunção, Iara Patricia Kretzer, Jelver Alexander Sierra Restrepo, Leonidas João de Mello Junior, Flavio Augusto Rocha Barbosa, Misael Ferreira, Marcus Mandolesi Sá, Tânia Beatriz Creczynski-Pasa","doi":"10.2174/0118715206358941250413154017","DOIUrl":"https://doi.org/10.2174/0118715206358941250413154017","url":null,"abstract":"<p><strong>Introduction/objective: </strong>Cancer is a global health burden. Despite advances in early detection and therapeutics, cancer prevalence continues to increase, underscoring the need for innovative therapeutic strategies. Dysregulation of cell death mechanisms is a hallmark of cancer that can lead to apoptosis evasion, which strongly contributes to tumor progression and therapy resistance. Isothiouronium salts have attracted attention as promising antitumor agents. This study aimed to evaluate the in vitro antitumor effect of an isothiouronium salt (ISMF08) on the B16F10 melanoma cell line.</p><p><strong>Methods: </strong>The antitumor properties of IS-MF08 were investigated by incubating B16F10 cells with the compound at different concentrations. Cytotoxicity was determined by the (3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide) (MTT) assay, cell cycle arrest and cell death mechanisms by flow cytometry, and morphological alterations by transmission electron microscopy. Physicochemical parameters related to druglikeness were predicted in silico using the SwissADME tool.</p><p><strong>Results: </strong>IS-MF08 was cytotoxic to melanoma cells, triggering cell cycle arrest and disrupting mitosis. The mechanism of cell death was compatible with apoptosis, as indicated by annexin V-FITC experiments and the relevant morphological changes in cell structure observed by transmission electron microscopy. SwissADME predicted that IS-MF08 has good physicochemical properties related to absorption and permeation.</p><p><strong>Conclusion: </strong>The numerous mechanisms of cell death triggered by IS-MF08 and its drug-likeness make it an interesting molecule in the search for new antitumor compounds, contributing to therapies targeting the dysregulation of cellular mechanisms such as apoptosis.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143967524","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}
Zai Wang, Mengtian Tan, Junqi Zhang, Hang Ren, Xueshuai Ye
{"title":"BCL6 Inhibitors Exhibit Therapeutic Effects on Breast Cancer Cells through the BCL6-STAT4 Pathway.","authors":"Zai Wang, Mengtian Tan, Junqi Zhang, Hang Ren, Xueshuai Ye","doi":"10.2174/0118715206364308250410104338","DOIUrl":"https://doi.org/10.2174/0118715206364308250410104338","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to explore the roles of BCL6 and STAT4 in breast cancer, their biological functions, and their relationships with the prognosis of patients with breast cancer.</p><p><strong>Methods: </strong>Online databases were used to analyze the expression characteristics of BCL6 and STAT4 in breast cancer, as well as the correlation between STAT4 and both the prognosis of breast cancer patients and the biological function of breast cancer cells. BC cell lines, such as MCF7 and MDA-436 cells, were treated with the BCL6 inhibitor TP-021, and STAT4 and BCL6 mRNA expression levels were detected. Sh-RNAs were used to downregulate STAT4 in MCF7 and MDA-436 cells, and their proliferation ability was measured via a CCK-8 assay.</p><p><strong>Results: </strong>BCL6 expression was detected in BC cell lines and tissues, but the expression of STAT4 was downregulated in BC, and the expression level of STAT4 was negatively correlated with patient prognosis. Inhibition of BCL6 can increase the STAT4 level in BC cells and inhibit their proliferation ability in vitro. Poor prognosis may be related to the expression of STAT4 and the characteristics of immune cell infiltration in tumor tissues.</p><p><strong>Conclusion: </strong>BCL6 inhibitors demonstrated therapeutic effects on breast cancer cells through the BCL6-STAT4 pathway.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143973251","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}
Beyza Ecem Öz Bedir, Emine Terzi, Tuba Ozdemir Sanci, Francesco Melfi, Ecem Kaya-Sezginer, Betül Kaya, Ulviye Acar Çevik
{"title":"Design, Synthesis, Molecular Modeling and Biological Evaluation of Novel Benzothiazole-Based 1,3,4-Thiadiazole Derivatives as Potential Anticancer Agents.","authors":"Beyza Ecem Öz Bedir, Emine Terzi, Tuba Ozdemir Sanci, Francesco Melfi, Ecem Kaya-Sezginer, Betül Kaya, Ulviye Acar Çevik","doi":"10.2174/0118715206353584241018051852","DOIUrl":"https://doi.org/10.2174/0118715206353584241018051852","url":null,"abstract":"<p><strong>Objective: </strong>The present study aimed to design and synthesize a new series of benzothiazole analogues containing 1,3,4-thiadiazole, and assess their biological activities as potential anticancer agents.</p><p><strong>Methods: </strong>N-(5,6-dimethylbenzo[d]thiazol-2-yl)-2-((5-(substituted amino)-1,3,4-thiadiazol-2-yl)thio)acetamide derivatives (4a-4h) were synthesized via the reaction of thiadiazole derivatives (3a-3h) with 2-chloro-N-(5,6- dimethylbenzo[d]thiazol-2-yl)acetamide (1) in the presence of potassium carbonate. All the target compounds have been characterized by spectral analysis. The anticancer activities of compounds 4a-4h were tested against two human HT-1376 bladder and HT-29 colorectal carcinoma cells using the WST-1 assay. Flow cytometry was used for the determination of apoptosis, cell cycle, and caspase 3/7 activity. Moreover, wound-healing assay was utilized to evaluate cell migration. In silico physicochemical, pharmacokinetics, and toxicological properties of compound 4g were determined by pkCSM, SwissADME, and SwissTargetPrediction online web tools.</p><p><strong>Results: </strong>Among all synthesized derivatives, compound 4g (N-(5,6-dimethylbenzo[d]thiazol-2-yl)-2-((5-((3- methoxyphenyl)amino)-1,3,4-thiadiazol-2-yl)thio)acetamide) recorded the highest antiproliferative activity against HT-1376 cells with an IC50 as 26.51 μM at 24 h, which was less cytotoxic than cisplatin (IC50=14.85 μM). The combined treatment with compound 4g and cisplatin increased the cellular apoptosis with a higher impact compared with the cisplatin group. The higher accumulation of cells in the G2 phase, a significant increase of caspase 3/7 activity, and the inhibition of migration rate were also observed in HT-1376 following a combination of compound 4g and cisplatin treatment versus cisplatin alone, which might be involved in the apoptotic effects of compound 4g.</p><p><strong>Conclusion: </strong>The in vitro anticancer potential of compound 4g lays the foundation for future research to focus on its value as a novel and advanced cancer therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144061948","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}
{"title":"The Pivotal Role of Irradiation-induced Autophagy Mechanisms in Glioma Therapy.","authors":"Seidu A Richard","doi":"10.2174/0118715206371143250402175030","DOIUrl":"https://doi.org/10.2174/0118715206371143250402175030","url":null,"abstract":"<p><p>Glioma epitomizes exclusively primary brain cancer of glial cell or neuroepithelial derivation and irradiation (IR) is one of the key and standard treatment modalities for all kinds of gliomas. Patients with glioma often undergo IR, such as whole-brain radiotherapy, stereotactic radiosurgery, as well as intensity modulated radiation therapy. However, IR therapy for malignant glioma is still facing severe hindrances because gliomas have high resistance to the IR. Autophagy is a type II programmed cell death which has been implicated in IR to gliomas. Autophagy was able to protect cells under sublethal damage circumstances, and it differentially triggered cell death after lethal damage in glioma. Furthermore, IR induced cerebral vascular damage was associated with progressive endothelial cells loss. IR triggered the acceleration of autophagic flux in cerebral endothelial cells which was characterized with robust upregulation of autophagy genes. Thus, autophagy plays a pivotal role in modulating the sensitivity and resistance of glioma cells to IR therapy. However, the exact autophagic mechanisms underlying radiosensitivity and/or radioresistance is still a matter of debate, and the development of effective radiosensitizers are lacking. Specific conditions pointing to the capabilities of IR-induced autophagy augmentation or inhibition of IR-induced cell death mostly contribute to radiosensitivity or radioresistance. Thus, IRinduced autophagy mechanisms in gliomas therapy are multiplex and they either induce radiosensitivity or inhibit radioresistance leading to potential effective treatment strategies for glioma. The aim of this review is to elucidate the autophagic mechanisms associated with radiosensitivity and/or radioresistance in glioma at the bench level, and accordingly highlight the development of potentially effective and efficient radiosensitizers to argument the treatment of glioma.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143956582","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}
{"title":"Recent Approaches on Oncolytic Viruses, Aptamers, TTFields and Personalized Treatment used for the Management of Glioblastoma: A Comprehensive Review.","authors":"Archna Singh, Anmol Kanda, Jyoti Kanda","doi":"10.2174/0118715206364677250401190214","DOIUrl":"https://doi.org/10.2174/0118715206364677250401190214","url":null,"abstract":"<p><p>Glioblastoma (GB) remains a formidable challenge in oncology, with current treatment approaches providing only marginal improvements in patient outcomes. Despite significant advances in understanding its molecular and genetic characteristics, median survival for untreated patients remains distressingly low, emphasizing the urgent need for novel therapeutic strategies. This review comprehensively examines the standard first-line treatments for GB, including surgery, concomitant radio-chemotherapy, and maintenance chemotherapy, while highlighting the limitations of these approaches. Consequently, we explore emerging novel therapeutic modalities such as Oncolytic Viral Therapy with genetically modified oncolytic viruses that enhance the capabilities of antigen- presenting cells. These cells migrate to lymph nodes to recruit cytotoxic CD8+ T lymphocytes, directing them to the site of infection where they eradicate cells that promote tumour growth. Aptamer-based therapies, such as GMT-3, AS1411, GS24, GMT8, and Gint4.T, which exhibit specificity for their biological targets and can act as drug transporters by facilitating receptor-mediated transcytosis within the endothelial cells of the blood-brain barrier, thus improving drug delivery. Tumour-treating fields (TTFields) that have shown increased overall survival rates in patients. Personalized genomic medicine, driven by biomarkers, which provokes immune responses tailored to the tumour's specific antigens, thereby customizing patient-specific treatments to improve effectiveness. By synthesizing current evidence and recent breakthroughs, we underscore the potential use of advancing novel therapies to address the unmet clinical needs of GB patients and ultimately enhance their overall survival and quality of life.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143958976","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}
{"title":"Advanced Technologies for Cancer Immunotherapy: Focus on Gastrointestinal Cancers.","authors":"Shahrzad Ahangarzadeh, Elmira Mohammadi, Hajar Yaghoobi, Kiana Shahzamani, Armina Alagheband Bahrami, Roghaye Arezumand, Leila Beikmohammadi, Abbas Alibakhshi","doi":"10.2174/0118715206369319250402150638","DOIUrl":"https://doi.org/10.2174/0118715206369319250402150638","url":null,"abstract":"<p><p>Immunotherapy is becoming an alternative method for gastrointestinal cancers, such as colorectal, gastric, and liver cancers. This field of research focuses on utilizing the immune system to recognize and eliminate cancer cells. One important method is immune checkpoint inhibitors, which enable T cells to recognize and attack tumor cells by releasing the immune system's brakes. Chimeric antigen receptor (CAR) T-cell therapy is another approach that modifies a patient's T cells to express receptors specific to tumor-associated antigens. Some cancer vaccines have demonstrated positive results in clinical trials, particularly colorectal and gastric cancers. Despite progress, challenges exist in immunotherapy for gastrointestinal cancers, such as treatment resistance, limited biomarkers for patient selection, and identifying new targets. In this review, different immunotherapy methods for all types of gastrointestinal cancers will be studied, and the limitations and benefits of each will be discussed in detail. By delving into the various immunotherapy methods, their limitations, and benefits, this review offers valuable insights that could potentially shape the future of gastrointestinal cancer treatment. It not only sheds light on the promising advancements in immune checkpoint inhibitors, CAR T-cell therapy, and cancer vaccines but also highlights the existing challenges that demand further research and innovation.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143966325","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}
Shalaka R Patki, Shyam Sundar P, Arehelli S Manjappa, Alagarsamy Veerachamy, Murugesan Sankaranarayanan, Mohammed Muzaffar-Ur-Rehman, Ahmad Salawi, Sunil T Galatage, John I Disouza, Vinyas Mayasa
{"title":"Identification of Active Phytochemicals to Inhibit Signal Transducer and Activator of Transcription 5A (STAT5A) Dimerization for Prostate Cancer Therapy: An In Silico Approach.","authors":"Shalaka R Patki, Shyam Sundar P, Arehelli S Manjappa, Alagarsamy Veerachamy, Murugesan Sankaranarayanan, Mohammed Muzaffar-Ur-Rehman, Ahmad Salawi, Sunil T Galatage, John I Disouza, Vinyas Mayasa","doi":"10.2174/0118715206367609250329195533","DOIUrl":"https://doi.org/10.2174/0118715206367609250329195533","url":null,"abstract":"<p><strong>Background: </strong>The Src Homology 2 (SH2) domain, the most conserved region of STAT5a/b (aa 573- 712), is crucial for receptor-specific recruitment and STAT5 dimerization, making it a therapeutic target in prostate cancer (PCa).</p><p><strong>Objectives: </strong>This study explored the SH2 domain of STAT5a and carried out the identification of natural STAT5a inhibitors.</p><p><strong>Methods: </strong>Using template-based homology modeling, we constructed the structure of human STAT5a (VP1P) and compared it with its 3D crystal of the STAT5a protein obtained from the RCSB database and the model generated by the AlphaFold database. In this study, we carried out molecular docking studies using AutoDock Vina on the top 500 natural compounds identified through a pharmacophore search of the ZINC database using ZINCPharmer. Furthermore, the top ten compounds with the highest binding energies were evaluated for their drug-likeness and ADMET properties using SWISS ADME and ProTox-II, followed by 100 ns molecular dynamics (MD) simulations using the Desmond module of the Schrodinger suite.</p><p><strong>Results: </strong>Docking studies revealed Pedunculagin (-10.5 kcal/mol), Folic acid (-10.1 kcal/mol), Chebulinic acid (- 10.0 kcal/mol), Chebulagic acid (-9.8 kcal/mol), and Oleandrin (-9.8 kcal/mol) as the top candidates, compared to the STAT5 inhibitor (Phase-II Clinical Trial) (-8.5 kcal/mol). ADMET analysis confirmed their safety profiles. MD simulations showed stable protein-ligand complexes, with all compounds interacting with the conserved Arg638 residue at the active site, similar to the STAT5 inhibitor.</p><p><strong>Conclusion: </strong>Pedunculagin demonstrated the strongest binding energy and stability, making it a promising candidate for further development as a novel lead compound to disrupt STAT5a/b dimerization in PCa therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143952262","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}
Mukta Rani, Amit Kumar Sharma, Anuradha Nischal, Sanjay Khattri, Ganesh Chandra Sahoo, Rajesh K Singh
{"title":"Molecular Dynamics (MD) Simulation of GPR87-LPA Binding: Therapeutic Implications for Targeted Cancer Treatment.","authors":"Mukta Rani, Amit Kumar Sharma, Anuradha Nischal, Sanjay Khattri, Ganesh Chandra Sahoo, Rajesh K Singh","doi":"10.2174/0118715206374428250403103159","DOIUrl":"https://doi.org/10.2174/0118715206374428250403103159","url":null,"abstract":"<p><p>GPR87 is an orphan G-protein-coupled receptor (GPCR) that represents a potential molecular target for developing novel drugs aimed at treating Squamous Cell Carcinomas (SCCs) or adenocarcinomas of the lungs and bladder. The aim of the present research is to screen a variety of LPA analogues using computational methods to identify the most promising inhibitors of the GPR87 protein. Further, the objective of this study was to construct the structure of the human GPR87 protein using two template- based tools, Phyre2 and SWISS-MODEL, alongside the Iterative Threading Assembly Refinement (ITASSER) server. Additionally, neural network-based de novo modeling approaches, such as AlphaFold2, were utilized. The resulting GPR87 structure was validated by comparing it with the structural properties of the templates using the Verify-3D, ProSA, and ERRAT servers. We conducted a comprehensive structural and functional analysis of the target protein using various computational tools. Several computational techniques were employed to explore the structural and functional characteristics of the target, with LPA selected as the initial pharmacological candidate. A library of 2,605 LPA analogues was screened against orphan GPR87 through in-silico docking analysis to identify higher-affinity and more selective potential drugs. Molecular Dynamics (MD) simulations were performed to track structural changes and convergence during the simulations. Key metrics, including the root mean square fluctuation (RMSF) of Cα-atoms, the radius of gyration, and the RMSD of backbone atoms, were calculated for both the apo-form and the LPA-GPR87 complex structures. These studies on structure-based drug targeting could pave the way for the development of specific inhibitors for the treatment of squamous cell carcinomas, and the findings may contribute to the design and development of new therapeutic compounds targeting GPR87 for the treatment of SCC.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143962849","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}
{"title":"Clinical Characteristics and Prognostic Factors Associated with Herpes Zoster in Patients with Malignant Tumors: A Systematic Review and Meta-analysis.","authors":"Mingming Ding, Shantao Qiu, Guan Jiang","doi":"10.2174/0118715206350399250324183333","DOIUrl":"https://doi.org/10.2174/0118715206350399250324183333","url":null,"abstract":"<p><strong>Background: </strong>Herpes zoster (HZ) is a common complication in patients with malignant tumors (MT), impacting prognosis. Immunocompromised states due to malignancy or treatment increase HZ risk. However, comprehensive assessments of HZ's clinical features and its impact on prognosis in these patients are limited, general conclusions are challenging, prompting a systematic review and meta-analysis to better understand the relative risk of HZ in malignancy.</p><p><strong>Objective: </strong>To assess the clinical features and prognostic factors of HZ in cancer patients through systematic review and meta-analysis. The study aimed to calculate the relative risk of HZ in malignancy and analyze factors affecting prognosis, such as age, gender, tumor type, and treatment.</p><p><strong>Methods: </strong>A systematic search in PubMed (2016-2024) identified studies on HZ and malignancy. Two reviewers independently screened and selected studies, extracting data on study characteristics, population demographics, and outcomes. Statistical heterogeneity across the studies was addressed using random-effects models, while subgroup analyses were performed to identify potential sources of heterogeneity.</p><p><strong>Results: </strong>Out of the 633 records reviewed, 13 studies satisfied the eligibility criteria and were incorporated into the meta-analysis. The combined relative risk for any type of cancer was found to be 1.82(95% CI: 1.29,2.57). The combined relative risk for any solid tumors was 1.63(95% CI: 1.08,2.46). The combined relative risk for any haematological cancer was 3.43(95% CI: 1.33,8.86). The combined analysis of all treatment modalities (including Radiotherapy, Chemotherapy, Immunosuppression, HSCT) shows a significant overall effect with a risk ratio of 1.78(95%CI: 1.59,2.00).</p><p><strong>Conclusion: </strong>Cancer patients have increased HZ risk due to immunosuppression from the malignancy and its treatment, especially in hematological cancers and those undergoing stem cell transplantation.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802308","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}