首页 > 最新文献

Recent patents on anti-cancer drug discovery最新文献

英文 中文
How Oncovirus Affects Drug Resistance in Cancer Cells.
Pub Date : 2025-03-27 DOI: 10.2174/0115748928324072250317050917
Yuping Yang, Yuwen Wang, Zhengdong Zhou, Junlong Zhuang, Wei Zhao

Cancer drug resistance has emerged as a formidable challenge in the field of clinical oncology, significantly hampering the success of treatment strategies and leading to suboptimal outcomes for patients. In a broad array of therapeutic settings, the emergence of resistance has become a primary source of concern, ranging from conventional chemotherapy to modern immunotherapy and targeted therapies. The complexity of cancer drug resistance is further exacerbated by the involvement of oncoviruses, such as human papillomavirus (HPV), Epstein-Barr virus (EBV), and hepatitis B virus (HBV), which play pivotal roles in the initiation, progression, and response to treatment of various cancers. The intricate interactions between these oncoviruses and cancer cells have been found to significantly influence drug efficacy. These viruses can alter critical cellular pathways, including drug metabolism, DNA repair mechanisms, and the tumor microenvironment, thus promoting drug resistance. A profound understanding of these virus-cancer-drug interactions is crucial for the development of novel treatment approaches that can effectively overcome drug resistance. This review aimed to contribute to a broader awareness of the multifaceted nature of cancer drug resistance, particularly in the context of oncovirus involvement. By highlighting the critical role of oncoviruses in cancer development and treatment response, this review hopes to stimulate further research and the development of novel treatment strategies that can effectively overcome drug resistance and ultimately improve patient outcomes. As we advance toward precision oncology, a more holistic understanding of the complex interplay among cancer, its associated viruses, and therapeutic drugs is crucial for achieving optimal therapeutic responses.

{"title":"How Oncovirus Affects Drug Resistance in Cancer Cells.","authors":"Yuping Yang, Yuwen Wang, Zhengdong Zhou, Junlong Zhuang, Wei Zhao","doi":"10.2174/0115748928324072250317050917","DOIUrl":"https://doi.org/10.2174/0115748928324072250317050917","url":null,"abstract":"<p><p>Cancer drug resistance has emerged as a formidable challenge in the field of clinical oncology, significantly hampering the success of treatment strategies and leading to suboptimal outcomes for patients. In a broad array of therapeutic settings, the emergence of resistance has become a primary source of concern, ranging from conventional chemotherapy to modern immunotherapy and targeted therapies. The complexity of cancer drug resistance is further exacerbated by the involvement of oncoviruses, such as human papillomavirus (HPV), Epstein-Barr virus (EBV), and hepatitis B virus (HBV), which play pivotal roles in the initiation, progression, and response to treatment of various cancers. The intricate interactions between these oncoviruses and cancer cells have been found to significantly influence drug efficacy. These viruses can alter critical cellular pathways, including drug metabolism, DNA repair mechanisms, and the tumor microenvironment, thus promoting drug resistance. A profound understanding of these virus-cancer-drug interactions is crucial for the development of novel treatment approaches that can effectively overcome drug resistance. This review aimed to contribute to a broader awareness of the multifaceted nature of cancer drug resistance, particularly in the context of oncovirus involvement. By highlighting the critical role of oncoviruses in cancer development and treatment response, this review hopes to stimulate further research and the development of novel treatment strategies that can effectively overcome drug resistance and ultimately improve patient outcomes. As we advance toward precision oncology, a more holistic understanding of the complex interplay among cancer, its associated viruses, and therapeutic drugs is crucial for achieving optimal therapeutic responses.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143733153","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Resibufogenin and Oxaliplatin Synergistically Inhibit Diffuse Gastric Cancer by Inactivating the FAK/AKT/GSK3β/β-Catenin Signaling Pathway.
Pub Date : 2025-03-24 DOI: 10.2174/0115748928320605241112094549
Hui-Hui Hu, Hai-Li Shang, Yongjing Ren, Hui-Fang Lv, Shuiping Tu, Jian-Zheng Wang, Ya-Nan Gong, Zhi-Fei Li, He Zhang, Tiepeng Li, Yuhang Wang, Bei-Bei Chen, Sai-Qi Wang, Xiao-Bing Chen

Background: Diffuse Gastric Cancer (DGC) is a highly aggressive form of gastric cancer with a poor prognosis. Oxaliplatin (OX) is one of the first-line chemotherapeutic agents for the treatment of gastric cancer. However, some patients with DGC do not benefit from OX therapy. Resibufogenin (RBF), one of the main active components of the Chinese medicine Huachansu, has demonstrated significant anti-cancer effects. Nevertheless, the potential of RBF to enhance the sensitivity of OX treatment in DGC and its underlying mechanisms have not been reported.

Objective: The aim of this study is to investigate the sensitizing effect of RBF on OX therapy for DGC, as well as to elucidate the potential targets and mechanisms of action. This exploration is of significant importance for the development of sensitizers that can improve the therapeutic efficacy of OX and for the advancement of patentable innovations in this field.

Methods: MTT assay, flow cytometry, Western blotting, and immunofluorescence assays were employed to assess the inhibitory effects of Resibufogenin (RBF) in combination with OX on DGC in vitro. Human DGC cell xenografts were established in a mouse model to evaluate the efficacy and safety of RBF and OX for treating DGC in vivo.

Results: It was found that RBF inhibited the proliferation of DGC cells in a time- and dose-dependent manner. When RBF was used in combination with OX, the sensitivity of DGC cells to OX was improved. Significantly, the combination of OX and RBF acts synergistically to induce apoptosis and autophagy while inhibiting migration and invasion of DGC cells in vitro. In vivo, the combination of OX and RBF dramatically inhibited the progression of DGC in the subcutaneous xenograft model without observable toxicity. Mechanistically, RBF significantly inhibited the expression and activation of FAK. OX and RBF synergistically inhibited the phosphorylation of FAK, AKT, and GSK3β to abrogate the entry of β-catenin into the cell nucleus.

Conclusion: RBF exhibits a pronounced suppressive effect on FAK, and its combination with OX synergistically blocks the FAK/AKT/GSK3β/β-catenin signaling cascade, thereby inhibiting the growth and metastasis of DGC. This study provides a novel avenue for future research and patent development of FAK inhibitors, with the potential to enhance the therapeutic efficacy of DGC treatment and overcome drug resistance.

背景:弥漫性胃癌(DGC)是一种侵袭性很强的胃癌,预后较差。奥沙利铂(OX)是治疗胃癌的一线化疗药物之一。然而,一些 DGC 患者并不能从奥沙利铂治疗中获益。中药华蟾素的主要活性成分之一雷西布苷元(RBF)具有显著的抗癌作用。然而,RBF提高OX治疗对DGC敏感性的潜力及其内在机制尚未见报道:本研究旨在探讨 RBF 对 OX 治疗 DGC 的增敏作用,并阐明其潜在靶点和作用机制。这一探索对于开发可提高 OX 疗效的增敏剂以及推进该领域的专利创新具有重要意义:方法:采用 MTT 法、流式细胞术、Western 印迹法和免疫荧光法评估瑞布呋辛(RBF)与 OX 联用在体外对 DGC 的抑制作用。在小鼠模型中建立了人类 DGC 细胞异种移植,以评估 RBF 和 OX 在体内治疗 DGC 的有效性和安全性:结果:研究发现,RBF以时间和剂量依赖的方式抑制DGC细胞的增殖。当 RBF 与 OX 联用时,DGC 细胞对 OX 的敏感性有所提高。值得注意的是,在体外,OX 和 RBF 的组合能协同诱导细胞凋亡和自噬,同时抑制 DGC 细胞的迁移和侵袭。在体内,OX 和 RBF 的组合能显著抑制皮下异种移植模型中 DGC 的恶化,且无明显毒性。从机理上讲,RBF 能明显抑制 FAK 的表达和活化。OX和RBF可协同抑制FAK、AKT和GSK3β的磷酸化,从而抑制β-catenin进入细胞核:结论:RBF对FAK有明显的抑制作用,其与OX联合使用可协同阻断FAK/AKT/GSK3β/β-catenin信号级联,从而抑制DGC的生长和转移。这项研究为FAK抑制剂的未来研究和专利开发提供了一条新途径,有望提高DGC治疗的疗效并克服耐药性。
{"title":"Resibufogenin and Oxaliplatin Synergistically Inhibit Diffuse Gastric Cancer by Inactivating the FAK/AKT/GSK3β/β-Catenin Signaling Pathway.","authors":"Hui-Hui Hu, Hai-Li Shang, Yongjing Ren, Hui-Fang Lv, Shuiping Tu, Jian-Zheng Wang, Ya-Nan Gong, Zhi-Fei Li, He Zhang, Tiepeng Li, Yuhang Wang, Bei-Bei Chen, Sai-Qi Wang, Xiao-Bing Chen","doi":"10.2174/0115748928320605241112094549","DOIUrl":"https://doi.org/10.2174/0115748928320605241112094549","url":null,"abstract":"<p><strong>Background: </strong>Diffuse Gastric Cancer (DGC) is a highly aggressive form of gastric cancer with a poor prognosis. Oxaliplatin (OX) is one of the first-line chemotherapeutic agents for the treatment of gastric cancer. However, some patients with DGC do not benefit from OX therapy. Resibufogenin (RBF), one of the main active components of the Chinese medicine Huachansu, has demonstrated significant anti-cancer effects. Nevertheless, the potential of RBF to enhance the sensitivity of OX treatment in DGC and its underlying mechanisms have not been reported.</p><p><strong>Objective: </strong>The aim of this study is to investigate the sensitizing effect of RBF on OX therapy for DGC, as well as to elucidate the potential targets and mechanisms of action. This exploration is of significant importance for the development of sensitizers that can improve the therapeutic efficacy of OX and for the advancement of patentable innovations in this field.</p><p><strong>Methods: </strong>MTT assay, flow cytometry, Western blotting, and immunofluorescence assays were employed to assess the inhibitory effects of Resibufogenin (RBF) in combination with OX on DGC in vitro. Human DGC cell xenografts were established in a mouse model to evaluate the efficacy and safety of RBF and OX for treating DGC in vivo.</p><p><strong>Results: </strong>It was found that RBF inhibited the proliferation of DGC cells in a time- and dose-dependent manner. When RBF was used in combination with OX, the sensitivity of DGC cells to OX was improved. Significantly, the combination of OX and RBF acts synergistically to induce apoptosis and autophagy while inhibiting migration and invasion of DGC cells in vitro. In vivo, the combination of OX and RBF dramatically inhibited the progression of DGC in the subcutaneous xenograft model without observable toxicity. Mechanistically, RBF significantly inhibited the expression and activation of FAK. OX and RBF synergistically inhibited the phosphorylation of FAK, AKT, and GSK3β to abrogate the entry of β-catenin into the cell nucleus.</p><p><strong>Conclusion: </strong>RBF exhibits a pronounced suppressive effect on FAK, and its combination with OX synergistically blocks the FAK/AKT/GSK3β/β-catenin signaling cascade, thereby inhibiting the growth and metastasis of DGC. This study provides a novel avenue for future research and patent development of FAK inhibitors, with the potential to enhance the therapeutic efficacy of DGC treatment and overcome drug resistance.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143702608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vitexin's Role in Colon Cancer Apoptosis: AMPK/mTOR Pathway Modulation Explored Through Experimental and Computational Approaches.
Pub Date : 2025-03-21 DOI: 10.2174/0115748928361989250226083146
Mohammed Abdalla Hussein, Gamila A Farouk, Haneen Kh Abdelkader, Mina A Daniel, Mohamed A Youssef, Gaber E Eldesoky, Seikh Mafiz Alam, Mohammad Shahidul Islam, Yasser O Mosaad

Background: Colorectal cancer is a significant global public health challenge, contributing substantially to cancer-related mortality worldwide. Vitexin has been shown to promote the polarization of macrophages towards the M1 phenotype, a process dependent on the Vitamin D receptor. This polarization is crucial in the tumor microenvironment, as it helps mitigate the progression from chronic colitis to colorectal cancer. Despite its potential, the mechanisms of vitexin's action and its impact on colon cancer remain unclear.

Objective: This study aims to evaluate the inhibitory effects of vitexin on cell proliferation and apoptosis in the Caco-2 colon cancer cell line, with a specific focus on its modulation of antioxidant enzyme activities, pro-apoptotic factors, and key signaling pathways involved in cell survival and proliferation.

Methods: The IC50 of vitexin against Caco-2 cells was determined. Cell viability and necrosis rates were assessed after 48 hours of incubation with vitexin at concentrations of 19.01, 38.01, and 76.02 μg/mL. Additionally, levels of superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), P53, Bax, TSC2, Sestrin 2, and PUMA, as well as the expression of AMPK, PI3K, Akt, and mTOR genes and proteins, were measured using q-PCR and Western blotting techniques in Caco-2 cells post-incubation.

Results: Vitexin exhibited an IC50 of 38.01 ± 0.64 μg/mL against Caco-2 cells. Treatment with vitexin at the specified concentrations for 48 hours resulted in a significant decrease in cell viability by 28.40%, with inhibitory rates reaching 71.6%. Apoptosis rates increased to 93.81%, 171.41%, and 294.12%, respectively, with a corresponding rise in necrosis rates by 194.19%, 400.22%, and 811.44%. Pharmacological analysis revealed that vitexin significantly inhibited SOD and CAT activities while enhancing MDA production. Furthermore, vitexin treatment upregulated the expression of key apoptotic markers (P53, Bax, TSC2, Sestrin 2, and PUMA) and the expression of AMPK, PI3K, and Akt, while downregulating mTOR genes and proteins, implicating various signaling pathways.

Conclusion: This study demonstrates that vitexin induces apoptosis in Caco-2 colon cancer cells through multiple mechanisms, including modulation of antioxidant enzymes, upregulation of pro-apoptotic factors, and regulation of key signaling pathways involved in cell survival and proliferation. These findings suggest that vitexin's mechanisms of action involve complex interactions with various cellular pathways, making it a promising candidate for further research and potential therapeutic applications in colorectal cancer.

{"title":"Vitexin's Role in Colon Cancer Apoptosis: AMPK/mTOR Pathway Modulation Explored Through Experimental and Computational Approaches.","authors":"Mohammed Abdalla Hussein, Gamila A Farouk, Haneen Kh Abdelkader, Mina A Daniel, Mohamed A Youssef, Gaber E Eldesoky, Seikh Mafiz Alam, Mohammad Shahidul Islam, Yasser O Mosaad","doi":"10.2174/0115748928361989250226083146","DOIUrl":"https://doi.org/10.2174/0115748928361989250226083146","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer is a significant global public health challenge, contributing substantially to cancer-related mortality worldwide. Vitexin has been shown to promote the polarization of macrophages towards the M1 phenotype, a process dependent on the Vitamin D receptor. This polarization is crucial in the tumor microenvironment, as it helps mitigate the progression from chronic colitis to colorectal cancer. Despite its potential, the mechanisms of vitexin's action and its impact on colon cancer remain unclear.</p><p><strong>Objective: </strong>This study aims to evaluate the inhibitory effects of vitexin on cell proliferation and apoptosis in the Caco-2 colon cancer cell line, with a specific focus on its modulation of antioxidant enzyme activities, pro-apoptotic factors, and key signaling pathways involved in cell survival and proliferation.</p><p><strong>Methods: </strong>The IC50 of vitexin against Caco-2 cells was determined. Cell viability and necrosis rates were assessed after 48 hours of incubation with vitexin at concentrations of 19.01, 38.01, and 76.02 μg/mL. Additionally, levels of superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), P53, Bax, TSC2, Sestrin 2, and PUMA, as well as the expression of AMPK, PI3K, Akt, and mTOR genes and proteins, were measured using q-PCR and Western blotting techniques in Caco-2 cells post-incubation.</p><p><strong>Results: </strong>Vitexin exhibited an IC50 of 38.01 ± 0.64 μg/mL against Caco-2 cells. Treatment with vitexin at the specified concentrations for 48 hours resulted in a significant decrease in cell viability by 28.40%, with inhibitory rates reaching 71.6%. Apoptosis rates increased to 93.81%, 171.41%, and 294.12%, respectively, with a corresponding rise in necrosis rates by 194.19%, 400.22%, and 811.44%. Pharmacological analysis revealed that vitexin significantly inhibited SOD and CAT activities while enhancing MDA production. Furthermore, vitexin treatment upregulated the expression of key apoptotic markers (P53, Bax, TSC2, Sestrin 2, and PUMA) and the expression of AMPK, PI3K, and Akt, while downregulating mTOR genes and proteins, implicating various signaling pathways.</p><p><strong>Conclusion: </strong>This study demonstrates that vitexin induces apoptosis in Caco-2 colon cancer cells through multiple mechanisms, including modulation of antioxidant enzymes, upregulation of pro-apoptotic factors, and regulation of key signaling pathways involved in cell survival and proliferation. These findings suggest that vitexin's mechanisms of action involve complex interactions with various cellular pathways, making it a promising candidate for further research and potential therapeutic applications in colorectal cancer.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143702610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bioinformatics Analysis of Key Genes Associated with Resistance to the Combination of Bevacizumab and Pemetrexed Chemotherapy in Non-small Cell Lung Cancer.
Pub Date : 2025-02-12 DOI: 10.2174/0115748928376905250210055408
Chenling Hu, Shenjie Xu, Siwen Chen, Qian Sun, Xudong Pan

Objective: This study aimed to identify key genes linked to resistance to a combination treatment regimen of bevacizumab and pemetrexed in non-small cell lung cancer (NSCLC) through bioinformatics analysis and analysis of their associated pathways.

Methods: Expression data from the Gene Expression Omnibus (GEO) database (GSE154286) were analyzed. The differentially expressed genes (DEGs) between tissues sensitive and resistant to combined bevacizumab and pemetrexed treatment in NSCLC were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment was investigated, and protein-protein interaction (PPI) networks, as well as transcription factor (TFs)- DEGs-miRNA networks, were created using the STRING tool. Key genes were identified with the help of the MCODE plugin. Additionally, gene set enrichment analysis (GSEA) was utilized to identify pathways linked to the key genes. A retrospective analysis was conducted on clinical data from 80 NSCLC patients. Patients were categorized into drug-resistant and non-resistant groups based on RECIST1.1 criteria. The expression of the key gene TNFSF4 was analyzed using quantitative real-time PCR (qRT-PCR).

Results: In the GSE154286 dataset, 35 downregulated DEGs were discovered. KEGG pathway enrichment analysis revealed that these DEGs were primarily associated with immunity and inflammation-related pathways. The PPI network construction highlighted a significant module and led to the identification of 8 candidate genes: TNFRSF18, TNFSF4, LGALS9, FAS, LAG3, CD86, CD80, and FOXP3. The TFs-DEGs-miRNA network analysis pinpointed TNFSF4 as a key gene, potentially regulated by 7 transcription factors and interacting with 9 miRNAs. GSEA analysis suggested that TNFSF4 may influence NSCLC's pathological processes through involvement in pathways involved in chemokine, JAK/STAT, NOD-like receptor, T cell receptor, toll-like receptor, and PPAR signaling. qRT-PCR detection displayed significantly lower expression of TNFSF4 in the peripheral blood of the patients in the resistant group relative to the non-resistant group (p < 0.0001). Logistic regression analysis showed that low TNFSF4 levels were independently linked to a raised risk of resistance to bevacizumab combined with pemetrexed therapy in lung adenocarcinoma patients.

Conclusion: The identification of key genes, such as TNFSF4, and resistance-related signaling pathways through bioinformatics analysis offers valuable insights into potential mechanisms of chemotherapy resistance in NSCLC when treated with the combination of bevacizumab and pemetrexed. These findings provide a theoretical foundation for advancing clinical research on diagnosis and treatment.

{"title":"Bioinformatics Analysis of Key Genes Associated with Resistance to the Combination of Bevacizumab and Pemetrexed Chemotherapy in Non-small Cell Lung Cancer.","authors":"Chenling Hu, Shenjie Xu, Siwen Chen, Qian Sun, Xudong Pan","doi":"10.2174/0115748928376905250210055408","DOIUrl":"https://doi.org/10.2174/0115748928376905250210055408","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to identify key genes linked to resistance to a combination treatment regimen of bevacizumab and pemetrexed in non-small cell lung cancer (NSCLC) through bioinformatics analysis and analysis of their associated pathways.</p><p><strong>Methods: </strong>Expression data from the Gene Expression Omnibus (GEO) database (GSE154286) were analyzed. The differentially expressed genes (DEGs) between tissues sensitive and resistant to combined bevacizumab and pemetrexed treatment in NSCLC were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment was investigated, and protein-protein interaction (PPI) networks, as well as transcription factor (TFs)- DEGs-miRNA networks, were created using the STRING tool. Key genes were identified with the help of the MCODE plugin. Additionally, gene set enrichment analysis (GSEA) was utilized to identify pathways linked to the key genes. A retrospective analysis was conducted on clinical data from 80 NSCLC patients. Patients were categorized into drug-resistant and non-resistant groups based on RECIST1.1 criteria. The expression of the key gene TNFSF4 was analyzed using quantitative real-time PCR (qRT-PCR).</p><p><strong>Results: </strong>In the GSE154286 dataset, 35 downregulated DEGs were discovered. KEGG pathway enrichment analysis revealed that these DEGs were primarily associated with immunity and inflammation-related pathways. The PPI network construction highlighted a significant module and led to the identification of 8 candidate genes: TNFRSF18, TNFSF4, LGALS9, FAS, LAG3, CD86, CD80, and FOXP3. The TFs-DEGs-miRNA network analysis pinpointed TNFSF4 as a key gene, potentially regulated by 7 transcription factors and interacting with 9 miRNAs. GSEA analysis suggested that TNFSF4 may influence NSCLC's pathological processes through involvement in pathways involved in chemokine, JAK/STAT, NOD-like receptor, T cell receptor, toll-like receptor, and PPAR signaling. qRT-PCR detection displayed significantly lower expression of TNFSF4 in the peripheral blood of the patients in the resistant group relative to the non-resistant group (p < 0.0001). Logistic regression analysis showed that low TNFSF4 levels were independently linked to a raised risk of resistance to bevacizumab combined with pemetrexed therapy in lung adenocarcinoma patients.</p><p><strong>Conclusion: </strong>The identification of key genes, such as TNFSF4, and resistance-related signaling pathways through bioinformatics analysis offers valuable insights into potential mechanisms of chemotherapy resistance in NSCLC when treated with the combination of bevacizumab and pemetrexed. These findings provide a theoretical foundation for advancing clinical research on diagnosis and treatment.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The ER Stress-related Gene Prognostic Signature for Predicting Chemosensitivity and Prognosis in AML.
Pub Date : 2025-02-07 DOI: 10.2174/0115748928349165250202172440
Jie Guo, Hongwei Peng, Luyao Long, Li Sun, Lin Yang, Simei Ren

Introduction: Acute myeloid leukemia is characterized by high heterogeneity, and the current European Leukemia Net (ELN) risk stratification system is not universally applicable to all AML patients, requiring approximately three weeks for testing.

Aim: This study aimed to develop an applicable prognostic tool capable of addressing the limitations of current methods. We selected AML patients from the clinic and TCGA database to explore the role of ER stress in response to chemotherapy.

Methods: Patients from the TCGA database were employed as the training cohort, and two GEO datasets were used as external validation cohorts. Univariate/multivariate COX and LASSO regression were exemplified to establish the prognostic model. Kaplan-Meier and timedependent ROC were used to assess and compare the efficiency of the model with ELN stratification and other models. In the training cohort, we selected 5 ER stress-related genes to predict chemosensitivity and establish the ERS-5 prognostic model.

Results: The model successfully predicted the overall survival of patients (p < 0.0001, HR = 4.86 (2.79-8.44); AUC = 0.83). It was verified in validation cohorts and could further stratify the risk of various AML subgroups. It also enhanced the ability of ELN to predict the response of patients with AML to main chemotherapeutic drugs. Finally, an "ERS-5" risk score was constructed by the nomogram based on the ERS-5 model and age.

Conclusion: Consequently, in this study, the ERS-5 model was constructed, which allowed more rapid (about 3 hours) and accurate risk stratification and complemented the ability of ELN to assess chemosensitivity.

简介急性髓性白血病具有高度异质性,目前的欧洲白血病网(ELN)风险分层系统并不适用于所有急性髓性白血病患者,大约需要三周时间进行检测。我们从临床和TCGA数据库中选取了急性髓细胞白血病患者,探讨ER应激在化疗反应中的作用:方法:将TCGA数据库中的患者作为训练队列,将两个GEO数据集作为外部验证队列。通过单变量/多变量 COX 回归和 LASSO 回归建立预后模型。Kaplan-Meier 和时间依赖性 ROC 用于评估和比较该模型与 ELN 分层和其他模型的效率。在训练队列中,我们选择了5个ER应激相关基因来预测化疗敏感性,并建立了ERS-5预后模型:结果:该模型成功预测了患者的总生存期(p < 0.0001,HR = 4.86 (2.79-8.44);AUC = 0.83)。该模型在验证队列中得到了验证,并能进一步对各种急性髓细胞性白血病亚组进行风险分层。它还提高了 ELN 预测急性髓细胞性白血病患者对主要化疗药物反应的能力。最后,基于ERS-5模型和年龄的提名图构建了 "ERS-5 "风险评分:因此,本研究建立了ERS-5模型,该模型可以更快速(约3小时)、准确地进行风险分层,并补充了ELN评估化疗敏感性的能力。
{"title":"The ER Stress-related Gene Prognostic Signature for Predicting Chemosensitivity and Prognosis in AML.","authors":"Jie Guo, Hongwei Peng, Luyao Long, Li Sun, Lin Yang, Simei Ren","doi":"10.2174/0115748928349165250202172440","DOIUrl":"https://doi.org/10.2174/0115748928349165250202172440","url":null,"abstract":"<p><strong>Introduction: </strong>Acute myeloid leukemia is characterized by high heterogeneity, and the current European Leukemia Net (ELN) risk stratification system is not universally applicable to all AML patients, requiring approximately three weeks for testing.</p><p><strong>Aim: </strong>This study aimed to develop an applicable prognostic tool capable of addressing the limitations of current methods. We selected AML patients from the clinic and TCGA database to explore the role of ER stress in response to chemotherapy.</p><p><strong>Methods: </strong>Patients from the TCGA database were employed as the training cohort, and two GEO datasets were used as external validation cohorts. Univariate/multivariate COX and LASSO regression were exemplified to establish the prognostic model. Kaplan-Meier and timedependent ROC were used to assess and compare the efficiency of the model with ELN stratification and other models. In the training cohort, we selected 5 ER stress-related genes to predict chemosensitivity and establish the ERS-5 prognostic model.</p><p><strong>Results: </strong>The model successfully predicted the overall survival of patients (p < 0.0001, HR = 4.86 (2.79-8.44); AUC = 0.83). It was verified in validation cohorts and could further stratify the risk of various AML subgroups. It also enhanced the ability of ELN to predict the response of patients with AML to main chemotherapeutic drugs. Finally, an \"ERS-5\" risk score was constructed by the nomogram based on the ERS-5 model and age.</p><p><strong>Conclusion: </strong>Consequently, in this study, the ERS-5 model was constructed, which allowed more rapid (about 3 hours) and accurate risk stratification and complemented the ability of ELN to assess chemosensitivity.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143416404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Prognostic Role of Interferon Gamma-inducible Protein 30 in Clear Cell Renal Cell Carcinoma with Immune Infiltrates.
Pub Date : 2025-02-06 DOI: 10.2174/0115748928367598250124123411
Wu Xu, Taihong Wu, Yufeng Liu, Yang Luo, Dawei Liu, Lingfei Yan, Qing Li, Tao Wang

Background: Recent research has demonstrated the significance of Interferon Gamma- Inducible Protein 30 (IFI30), an interferon gamma-induced protein, in the immune response to cancerous growths. However, the relationship between IFI30 expression levels, patient prognosis, and tumor-infiltrating lymphocytes in clear cell renal cell carcinoma (ccRCC) remains inadequately defined.

Methods: To ascertain the potential link between IFI30 expression, clinical data, and overall survival (OS) in ccRCC patients, we employed diverse databases, which include TCGA, Gene Expression Profiling Interaction Analysis (GEPIA), and UALCAN. Furthermore, an in-depth analysis of the link between tumor-infiltrating immune cells (TIIC) and IFI30 was carried out using the TIMER, GEPIA, and TISIDB databases. Immunohistochemistry (IHC) was utilized to identify the IFI30 and PD-1 expression levels in a tissue microarray. Patents about molecular classification and drugs in ccRCC were reviewed through Worldwide Espacenet®.

Results: The expression of IFI30 demonstrated a strong association with sample type, lymph node stage, tumor grade, and cancer stage. Elevated IFI30 expression was linked to unfavorable Disease- Specific Survival (DSS) and Overall Survival (OS) outcomes (p <0.01). Furthermore, overexpression of IFI30 was strongly linked to immunomodulatory molecules, chemokines, and increased infiltration of regulatory T cells (Tregs), natural killer (NK) CD56 cells, T helper 1 (Th1) cells, cytotoxic T cells, and T helper cells. IHC analysis confirmed a robust correlation between IFI30 and PD-1 expression.

Conclusion: IFI30 is a prognostic biomarker for ccRCC patients. Targeting IFI30 may provide new strategies for cancer therapy and improve the prognosis of ccRCC patients.

{"title":"The Prognostic Role of Interferon Gamma-inducible Protein 30 in Clear Cell Renal Cell Carcinoma with Immune Infiltrates.","authors":"Wu Xu, Taihong Wu, Yufeng Liu, Yang Luo, Dawei Liu, Lingfei Yan, Qing Li, Tao Wang","doi":"10.2174/0115748928367598250124123411","DOIUrl":"https://doi.org/10.2174/0115748928367598250124123411","url":null,"abstract":"<p><strong>Background: </strong>Recent research has demonstrated the significance of Interferon Gamma- Inducible Protein 30 (IFI30), an interferon gamma-induced protein, in the immune response to cancerous growths. However, the relationship between IFI30 expression levels, patient prognosis, and tumor-infiltrating lymphocytes in clear cell renal cell carcinoma (ccRCC) remains inadequately defined.</p><p><strong>Methods: </strong>To ascertain the potential link between IFI30 expression, clinical data, and overall survival (OS) in ccRCC patients, we employed diverse databases, which include TCGA, Gene Expression Profiling Interaction Analysis (GEPIA), and UALCAN. Furthermore, an in-depth analysis of the link between tumor-infiltrating immune cells (TIIC) and IFI30 was carried out using the TIMER, GEPIA, and TISIDB databases. Immunohistochemistry (IHC) was utilized to identify the IFI30 and PD-1 expression levels in a tissue microarray. Patents about molecular classification and drugs in ccRCC were reviewed through Worldwide Espacenet®.</p><p><strong>Results: </strong>The expression of IFI30 demonstrated a strong association with sample type, lymph node stage, tumor grade, and cancer stage. Elevated IFI30 expression was linked to unfavorable Disease- Specific Survival (DSS) and Overall Survival (OS) outcomes (p <0.01). Furthermore, overexpression of IFI30 was strongly linked to immunomodulatory molecules, chemokines, and increased infiltration of regulatory T cells (Tregs), natural killer (NK) CD56 cells, T helper 1 (Th1) cells, cytotoxic T cells, and T helper cells. IHC analysis confirmed a robust correlation between IFI30 and PD-1 expression.</p><p><strong>Conclusion: </strong>IFI30 is a prognostic biomarker for ccRCC patients. Targeting IFI30 may provide new strategies for cancer therapy and improve the prognosis of ccRCC patients.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143401122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Present State and Potential Applications of Artificial Intelligence in Cancer Diagnosis and Treatment.
Pub Date : 2025-02-03 DOI: 10.2174/0115748928361472250123105507
Anuja Mishra, Srishti Sharma, Swaroop Kumar Pandey

An aberrant increase in cancer incidences has demanded extreme attention globally despite advancements in diagnostic and management strategies. The high mortality rate is concerning, and tumour heterogeneity at the genetic, phenotypic, and pathological levels exacerbates the problem. In this context, lack of early diagnostic techniques and therapeutic resistance to drugs, sole awareness among the public, coupled with the unavailability of these modern technologies in developing and low-income countries, negatively impact cancer management. One of the prime necessities of the world today is the enhancement of early detection of cancers. Several independent studies have shown that screening individuals for cancer can improve patient survival but are bogged down by risk classification and major problems in patient selection. Artificial intelligence (AI) has significantly advanced the field of oncology, addressing various medical challenges, particularly in cancer management. Leveraging extensive medical datasets and innovative computational technologies, AI, especially through deep learning (DL), has found applications across multiple facets of oncology research. These applications range from early cancer detection, diagnosis, classification, and grading, molecular characterization of tumours, prediction of patient outcomes and treatment responses, personalized treatment, and novel anti-cancer drug discovery. Over the past decade, AI/ML has emerged as a valuable tool in cancer prognosis, risk assessment, and treatment selection for cancer patients. Several patents have been and are being filed and granted. Some of those inventions were explored and are being explored in clinical settings as well. In this review, we will discuss the current status, recent advancements, clinical trials, challenges, and opportunities associated with AI/ML applications in cancer detection and management. We are optimistic about the potential of AI/ML in improving outcomes for cancer and the need for further research and development in this field.

{"title":"The Present State and Potential Applications of Artificial Intelligence in Cancer Diagnosis and Treatment.","authors":"Anuja Mishra, Srishti Sharma, Swaroop Kumar Pandey","doi":"10.2174/0115748928361472250123105507","DOIUrl":"https://doi.org/10.2174/0115748928361472250123105507","url":null,"abstract":"<p><p>An aberrant increase in cancer incidences has demanded extreme attention globally despite advancements in diagnostic and management strategies. The high mortality rate is concerning, and tumour heterogeneity at the genetic, phenotypic, and pathological levels exacerbates the problem. In this context, lack of early diagnostic techniques and therapeutic resistance to drugs, sole awareness among the public, coupled with the unavailability of these modern technologies in developing and low-income countries, negatively impact cancer management. One of the prime necessities of the world today is the enhancement of early detection of cancers. Several independent studies have shown that screening individuals for cancer can improve patient survival but are bogged down by risk classification and major problems in patient selection. Artificial intelligence (AI) has significantly advanced the field of oncology, addressing various medical challenges, particularly in cancer management. Leveraging extensive medical datasets and innovative computational technologies, AI, especially through deep learning (DL), has found applications across multiple facets of oncology research. These applications range from early cancer detection, diagnosis, classification, and grading, molecular characterization of tumours, prediction of patient outcomes and treatment responses, personalized treatment, and novel anti-cancer drug discovery. Over the past decade, AI/ML has emerged as a valuable tool in cancer prognosis, risk assessment, and treatment selection for cancer patients. Several patents have been and are being filed and granted. Some of those inventions were explored and are being explored in clinical settings as well. In this review, we will discuss the current status, recent advancements, clinical trials, challenges, and opportunities associated with AI/ML applications in cancer detection and management. We are optimistic about the potential of AI/ML in improving outcomes for cancer and the need for further research and development in this field.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143124249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unveiling miR-1468-5p: A New Prognostic Biomarker and Therapeutic Target in Lung Adenocarcinoma.
Pub Date : 2025-02-03 DOI: 10.2174/0115748928344326240927071620
Yi Chen, Dongbing Li, Yueyu Fang

Background: miR-1468-5p, a type of microRNA, is acknowledged for its crucial involvement in a variety of cancerous processes. Nonetheless, the specific impact of this microRNA on lung adenocarcinoma (LUAD) has not yet been clearly defined.

Objective: Our aim was to investigate how miR-1468-5p influences LUAD.

Methods: The Cancer Genome Atlas (TCGA) offered specimens for our research. Employing statistical techniques, we assessed the diagnostic and prognostic significance of miR-1468-5p, as well as its association with clinical characteristics. Our analysis delved into the target genes and the regulatory mechanisms influenced by miR-1468-5p. The expression levels of miR-1468-5p in LUAD cell lines were validated through quantitative reverse transcription polymerase chain reaction (qRT-PCR).

Results: The expression of miR-1468-5p varied significantly across different cancer types. The presence of reduced miR-1468-5p levels was correlated with a lower likelihood of overall survival in LUAD patients, with a statistically significant result (p = 0.005). miR-1468-5p demonstrated independent prognostic significance in LUAD and potentially contributes to disease progression via multiple pathways, including the HIF-1 signaling pathway and more. There was a significant reduction in miR-1468-5p expression in LUAD cell lines when compared to cells of the normal lung epithelium.

Conclusion: miR-1468-5p may serve as a useful patent as a therapeutic intervention target and a prognostic indicator for LUAD patients.

{"title":"Unveiling miR-1468-5p: A New Prognostic Biomarker and Therapeutic Target in Lung Adenocarcinoma.","authors":"Yi Chen, Dongbing Li, Yueyu Fang","doi":"10.2174/0115748928344326240927071620","DOIUrl":"https://doi.org/10.2174/0115748928344326240927071620","url":null,"abstract":"<p><strong>Background: </strong>miR-1468-5p, a type of microRNA, is acknowledged for its crucial involvement in a variety of cancerous processes. Nonetheless, the specific impact of this microRNA on lung adenocarcinoma (LUAD) has not yet been clearly defined.</p><p><strong>Objective: </strong>Our aim was to investigate how miR-1468-5p influences LUAD.</p><p><strong>Methods: </strong>The Cancer Genome Atlas (TCGA) offered specimens for our research. Employing statistical techniques, we assessed the diagnostic and prognostic significance of miR-1468-5p, as well as its association with clinical characteristics. Our analysis delved into the target genes and the regulatory mechanisms influenced by miR-1468-5p. The expression levels of miR-1468-5p in LUAD cell lines were validated through quantitative reverse transcription polymerase chain reaction (qRT-PCR).</p><p><strong>Results: </strong>The expression of miR-1468-5p varied significantly across different cancer types. The presence of reduced miR-1468-5p levels was correlated with a lower likelihood of overall survival in LUAD patients, with a statistically significant result (p = 0.005). miR-1468-5p demonstrated independent prognostic significance in LUAD and potentially contributes to disease progression via multiple pathways, including the HIF-1 signaling pathway and more. There was a significant reduction in miR-1468-5p expression in LUAD cell lines when compared to cells of the normal lung epithelium.</p><p><strong>Conclusion: </strong>miR-1468-5p may serve as a useful patent as a therapeutic intervention target and a prognostic indicator for LUAD patients.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143124252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Organosulfur Compounds in Garlic for Gastric Cancer Treatment: Anticancer Effects, Overcoming Drug Resistance, and Mechanisms.
Pub Date : 2025-01-29 DOI: 10.2174/0115748928367900250116074925
Xudong Yi, Jinrui Zhang, Yiman Yao, Junyu Liu, Mingyue Li, Min Zhang, Anat Elmann, Haixia Chen

Garlic (Allium sativum L.) has been consumed globally as a functional food and traditional medicine for various ailments. Its active organosulfur compounds (OSCs) have demonstrated significant anticancer properties, particularly against gastric cancer. However, a comprehensive review of these effects and the underlying molecular mechanisms, including their role in overcoming drug resistance, is currently lacking. This review systematically examines both preclinical and clinical studies on the anticancer effects of garlic and its organosulfur compounds against gastric cancer, with a focus on patents. Emphasis is placed on explaining the mechanisms of action, exploring how these compounds can overcome drug resistance, and highlighting relevant patents that have been granted in this field. The literature search included databases, like PubMed, Web of Science, Google Scholar, ScienceDirect, and patent databases, including articles and patents published up to October 2024. Preclinical studies demonstrate that garlic-derived organosulfur compounds possess anticancer activities against gastric cancer. They work through multiple mechanisms, including inducing apoptosis, causing cell cycle arrest, inhibiting cancer stem cell properties, suppressing epithelial-mesenchymal transition, and modulating key signaling pathways, like PI3K/Akt and NF-κB. These compounds also show potential in overcoming drug resistance by downregulating multidrug resistance proteins and enhancing the effectiveness of standard chemotherapy drugs. Clinical studies suggest that regular garlic consumption may reduce the risk of gastric cancer and improve outcomes in patients undergoing chemotherapy. This review highlights the significant potential of garlic's organosulfur compounds as complementary agents in gastric cancer prevention and treatment and emphasizes the relevance of existing patents and the need for further clinical trials to confirm these effects and develop effective therapeutic strategies.

{"title":"Organosulfur Compounds in Garlic for Gastric Cancer Treatment: Anticancer Effects, Overcoming Drug Resistance, and Mechanisms.","authors":"Xudong Yi, Jinrui Zhang, Yiman Yao, Junyu Liu, Mingyue Li, Min Zhang, Anat Elmann, Haixia Chen","doi":"10.2174/0115748928367900250116074925","DOIUrl":"https://doi.org/10.2174/0115748928367900250116074925","url":null,"abstract":"<p><p>Garlic <i>(Allium sativum L.)</i> has been consumed globally as a functional food and traditional medicine for various ailments. Its active organosulfur compounds (OSCs) have demonstrated significant anticancer properties, particularly against gastric cancer. However, a comprehensive review of these effects and the underlying molecular mechanisms, including their role in overcoming drug resistance, is currently lacking. This review systematically examines both preclinical and clinical studies on the anticancer effects of garlic and its organosulfur compounds against gastric cancer, with a focus on patents. Emphasis is placed on explaining the mechanisms of action, exploring how these compounds can overcome drug resistance, and highlighting relevant patents that have been granted in this field. The literature search included databases, like PubMed, Web of Science, Google Scholar, ScienceDirect, and patent databases, including articles and patents published up to October 2024. Preclinical studies demonstrate that garlic-derived organosulfur compounds possess anticancer activities against gastric cancer. They work through multiple mechanisms, including inducing apoptosis, causing cell cycle arrest, inhibiting cancer stem cell properties, suppressing epithelial-mesenchymal transition, and modulating key signaling pathways, like PI3K/Akt and NF-κB. These compounds also show potential in overcoming drug resistance by downregulating multidrug resistance proteins and enhancing the effectiveness of standard chemotherapy drugs. Clinical studies suggest that regular garlic consumption may reduce the risk of gastric cancer and improve outcomes in patients undergoing chemotherapy. This review highlights the significant potential of garlic's organosulfur compounds as complementary agents in gastric cancer prevention and treatment and emphasizes the relevance of existing patents and the need for further clinical trials to confirm these effects and develop effective therapeutic strategies.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143070488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Three-agent Regimen for Triple Negative Breast Cancer Treatment. 三阴性乳腺癌的三药治疗方案。
Pub Date : 2025-01-17 DOI: 10.2174/0115748928350267250105153944
Shaojun Wang, Congxiu Huang, Ying Zhu, Min Wang, Wei Wang, Caixia Liu, Wuyun Su

Background: Triple-negative breast cancer (TNBC) has a poor prognosis with current treatment options. Novel therapeutic strategies are urgently needed to enhance treatment outcomes for TNBC.

Objective: This study evaluated the efficacy of a three-agent regimen compared to existing treatment regimens in a TNBC mouse model, and elucidated its potential mechanisms of action.

Methods: The TNBC xenograft tumor mouse model was established using a 4T1 cell line in female BALB/c mice. Mice were treated with the three-agent regimen and other comparative treatments. Tumor volume was monitored to assess the anti-tumor effects. Biochemical and pathological evaluations were conducted to examine the impact of the regimen on anti-tumor immunity, anti- tumor angiogenesis, and tumor cell apoptosis.

Results: The three-agent regimen consisting of SIN+BEV+PAB demonstrated significant anti-tumor efficacy compared to controls, PAB alone, SIN+PAB, and BEV+PAB groups from day 9 of drug administration. The superior anti-tumor effect of SIN+BEV+PAB was primarily attributed to enhanced anti-tumor immunity, evidenced by increased percentages of CD4+ and CD8+ T cells, elevated IFN-γ levels, and decreased percentages of Tregs, reduced levels of TGF-β, IL-6, and IL-10. Additionally, the regimen showed potent anti-angiogenic effects by reducing VEGF expression and micro vessel density (MVD). Furthermore, it promoted tumor cell apoptosis through upregulation of BAX and cleaved caspase3, while downregulating Bcl2.

Conclusion: These findings suggest that the novel three-agent combination of SIN+BEV+PAB may prove beneficial in improving treatment outcomes for patients with TNBC. The development of this regimen, which may be eligible for patent protection, could facilitate its introduction as a new therapeutic option for advanced TNBC in clinical practice.

背景:三阴性乳腺癌(TNBC)在目前的治疗方案下预后较差。迫切需要新的治疗策略来提高TNBC的治疗效果。目的:本研究在TNBC小鼠模型中评估三药方案与现有治疗方案的疗效,并阐明其潜在的作用机制。方法:采用BALB/c雌性小鼠4T1细胞系建立TNBC异种移植瘤小鼠模型。小鼠采用三药方案和其他比较治疗。监测肿瘤体积以评估抗肿瘤效果。通过生化和病理评价来检验该方案对抗肿瘤免疫、抗肿瘤血管生成和肿瘤细胞凋亡的影响。结果:从给药第9天起,与对照组、单独PAB组、SIN+PAB组和BEV+PAB组相比,SIN+BEV+PAB三药方案的抗肿瘤效果显著。SIN+BEV+PAB的优越抗肿瘤效果主要归因于增强的抗肿瘤免疫,其表现为CD4+和CD8+ T细胞百分比增加,IFN-γ水平升高,Tregs百分比降低,TGF-β, IL-6和IL-10水平降低。此外,该方案通过降低VEGF表达和微血管密度(MVD)显示出有效的抗血管生成作用。通过上调BAX和裂解caspase3,下调Bcl2,促进肿瘤细胞凋亡。结论:这些研究结果表明,SIN+BEV+PAB的新型三药联合可能有助于改善TNBC患者的治疗结果。该方案的开发可能有资格获得专利保护,可以促进其作为晚期TNBC临床实践的新治疗选择。
{"title":"A Three-agent Regimen for Triple Negative Breast Cancer Treatment.","authors":"Shaojun Wang, Congxiu Huang, Ying Zhu, Min Wang, Wei Wang, Caixia Liu, Wuyun Su","doi":"10.2174/0115748928350267250105153944","DOIUrl":"https://doi.org/10.2174/0115748928350267250105153944","url":null,"abstract":"<p><strong>Background: </strong>Triple-negative breast cancer (TNBC) has a poor prognosis with current treatment options. Novel therapeutic strategies are urgently needed to enhance treatment outcomes for TNBC.</p><p><strong>Objective: </strong>This study evaluated the efficacy of a three-agent regimen compared to existing treatment regimens in a TNBC mouse model, and elucidated its potential mechanisms of action.</p><p><strong>Methods: </strong>The TNBC xenograft tumor mouse model was established using a 4T1 cell line in female BALB/c mice. Mice were treated with the three-agent regimen and other comparative treatments. Tumor volume was monitored to assess the anti-tumor effects. Biochemical and pathological evaluations were conducted to examine the impact of the regimen on anti-tumor immunity, anti- tumor angiogenesis, and tumor cell apoptosis.</p><p><strong>Results: </strong>The three-agent regimen consisting of SIN+BEV+PAB demonstrated significant anti-tumor efficacy compared to controls, PAB alone, SIN+PAB, and BEV+PAB groups from day 9 of drug administration. The superior anti-tumor effect of SIN+BEV+PAB was primarily attributed to enhanced anti-tumor immunity, evidenced by increased percentages of CD4+ and CD8+ T cells, elevated IFN-γ levels, and decreased percentages of Tregs, reduced levels of TGF-β, IL-6, and IL-10. Additionally, the regimen showed potent anti-angiogenic effects by reducing VEGF expression and micro vessel density (MVD). Furthermore, it promoted tumor cell apoptosis through upregulation of BAX and cleaved caspase3, while downregulating Bcl2.</p><p><strong>Conclusion: </strong>These findings suggest that the novel three-agent combination of SIN+BEV+PAB may prove beneficial in improving treatment outcomes for patients with TNBC. The development of this regimen, which may be eligible for patent protection, could facilitate its introduction as a new therapeutic option for advanced TNBC in clinical practice.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143019146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Recent patents on anti-cancer drug discovery
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