Anti-Cancer Drugs最新文献

筛选
英文 中文
Stereotactic body radiotherapy and poly (ADP-ribose) polymerase inhibitors in ovarian cancer: a knowledge and attitudes survey in collaboration with the Italian Association of Radiation Oncology (AIRO) and Multicenter Italian Trials in Ovarian Cancer (MITO) groups. 卵巢癌的立体定向体放疗和聚(adp -核糖)聚合酶抑制剂:与意大利放射肿瘤学协会(AIRO)和意大利卵巢癌多中心试验(MITO)组合作进行的知识和态度调查。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-08 DOI: 10.1097/CAD.0000000000001684
Gabriella Macchia, Donato Pezzulla, Donatella Russo, Maura Campitelli, Simona Lucci, Mara Fanelli, Francesco Deodato, Anna Fagotti, Maria Antonietta Gambacorta, Antonella Savarese, Sandro Pignata, Cynthia Aristei, Gabriella Ferrandina
{"title":"Stereotactic body radiotherapy and poly (ADP-ribose) polymerase inhibitors in ovarian cancer: a knowledge and attitudes survey in collaboration with the Italian Association of Radiation Oncology (AIRO) and Multicenter Italian Trials in Ovarian Cancer (MITO) groups.","authors":"Gabriella Macchia, Donato Pezzulla, Donatella Russo, Maura Campitelli, Simona Lucci, Mara Fanelli, Francesco Deodato, Anna Fagotti, Maria Antonietta Gambacorta, Antonella Savarese, Sandro Pignata, Cynthia Aristei, Gabriella Ferrandina","doi":"10.1097/CAD.0000000000001684","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001684","url":null,"abstract":"<p><p>The aim of this study was to present a nationwide survey on the specialist's attitudes towards stereotactic body radiotherapy (SBRT) combined with poly (ADP-ribose) polymerase inhibitors (PARPi) with oligometastatic/oligoprogressive/oligorecurrent ovarian cancer (oMPR-OC) patients. The 19-item questionnaire was developed by specialists and distributed online. Replies were stratified by categories and analyzed using descriptive statistics. Respondents (N = 100) were radiation oncologists (57%), medical oncologists (32%), and gynecologic oncologists (11%). Fifty-four percent of respondents considered medical oncologists as the primary oncologists for oMPR-OC, while 23% preferred radiation oncologists and 15% favored gynecologic oncologists. Seventy-three percent discuss these cases in the Multidisciplinary Tumor Board, while 15, 6, and 2% send the patients straight to SBRT, surgery, or chemotherapy, respectively. Seventy-four percent of the experts interviewed were treated with SBRT less than 10 oMPR-OC patients. Concomitant treatment was highly heterogeneous, but it had little to no reported side effects. A significant variation in how PARPi is managed during SBRT was found: 34% do not interrupt the administration, while 52% pause and restart it later. Forty-three percent of respondents believe that the PARPi dosage should not be reduced when administered concurrently with SBRT. Sixty-nine percent of respondents believe that the SBRT dose should not be decreased while receiving PARPi if the constraints are met. The majority of respondents (40%) favored expert consensus for enhancing the clinical management of oMPR-OC, while 34% preferred clinical guidelines. A lack of or low toxicity with the combination of PARPi and SBRT was perceived, and a significant degree of heterogeneity concerning clinical protocols for their combination. Moreover, it emphasizes the low number of patients who have received this treatment approach nationwide.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943248","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transformation of lung adenocarcinoma to small cell lung cancer following osimertinib treatment: a case report and literature review. 奥西替尼治疗后肺腺癌向小细胞肺癌的转化:1例报告及文献复习。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-08 DOI: 10.1097/CAD.0000000000001686
Linwu Kuang, Peng Wang, Lin Zhou, Yangkai Li
{"title":"Transformation of lung adenocarcinoma to small cell lung cancer following osimertinib treatment: a case report and literature review.","authors":"Linwu Kuang, Peng Wang, Lin Zhou, Yangkai Li","doi":"10.1097/CAD.0000000000001686","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001686","url":null,"abstract":"<p><p>Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) effectively treat EGFR-mutant lung adenocarcinoma, demonstrating initial efficacy but eventually leading to acquired resistance. Small cell transformation is a rare resistance mechanism to EGFR-TKIs in lung adenocarcinoma, which can complicate clinical diagnosis and treatment. We present a patient with lung adenocarcinoma who underwent a prior pneumonectomy and adjuvant chemotherapy and was treated with osimertinib after the recurrence of lung cancer. Small cell transformation occurred approximately 20 months after starting osimertinib treatment. After this transformation, the patient underwent lung radiotherapy and cisplatin-etoposide chemotherapy, which stabilized the disease. Following the confirmation of small cell lung cancer (SCLC) via thyroid puncture, treatments with irinotecan, irinotecan plus atezolizumab, thyroid radiotherapy, adrenal radiotherapy, and head radiotherapy were sequentially administered, yet the disease continued to progress. The patient succumbed to the disease in May 2023 because of progression and organ failure, with an overall survival of 52.7 months, including 16 months post small cell transformation. This case highlights the possibility of osimertinib causing lung adenocarcinoma to transform into SCLC and underscores rebiopsies' importance in identifying resistance mechanisms to EGFR-TKIs. Increased levels of neuron-specific enolase and pro-gastrin releasing peptide can signal early transformation into SCLC.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Paeoniflorin sensitizes imatinib mesylate-resistant chronic myeloid leukemia cells via the inhibition of Cyr61 production. 芍药苷通过抑制Cyr61的产生使耐伊马替尼甲磺酸慢性髓系白血病细胞增敏。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-07 DOI: 10.1097/CAD.0000000000001681
Yanfang Song, Li Luo, Zhen Lin, Taigang Zhang, Zhaozhong Li, Yinping Cao, Xianjin Zhu
{"title":"Paeoniflorin sensitizes imatinib mesylate-resistant chronic myeloid leukemia cells via the inhibition of Cyr61 production.","authors":"Yanfang Song, Li Luo, Zhen Lin, Taigang Zhang, Zhaozhong Li, Yinping Cao, Xianjin Zhu","doi":"10.1097/CAD.0000000000001681","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001681","url":null,"abstract":"<p><p>Imatinib mesylate (IM) is a first-line therapy for chronic myeloid leukemia (CML) and exhibits good therapeutic effects, but not in all patients with CML owing to drug resistance. Our previous study showed that Cyr61 plays a key role in IM resistance in CML cells. Paeoniflorin (PF) is a bioactive compound isolated from the traditional Chinese medicine Paeonia lactiflora Pall that displays anticancer activity. Little is, however, known regarding the role of PF in IM-resistant CML cells. This study aimed to evaluate whether PF could decrease Cyr61 production and improve IM-resistant CML cell sensitivity to IM and to investigate the underlying mechanisms. CML cell lines (K562 and KCL22) and IM-resistant cell lines (K562G and KCL22R) were used as CML study models. Cyr61 expression was assessed in both parental and IM-resistant CML cells by western blotting, real-time quantitative PCR , and ELISA. Lentiviral vectors were used to induce the knockdown of Cyr61 expression, followed by a comprehensive evaluation of cell proliferation and apoptosis. The results showed that PF decreased the production of Cyr61 in the presence of IM by inhibiting extracellular regulated protein kinases 1/2 activation. PF significantly decreased the IC50 value of IM and increased IM-induced apoptosis of IM-resistant CML cells. Importantly, PF also improved the sensitivity of CML cells to bosutinib and dasatinib via inhibition of Cyr61 production. In conclusion, we report for the first time that PF may effectively improve the sensitivity of IM-resistant CML cells to IM, bosutinib, and dasatinib, at least in part, by subsequently downregulating Cyr61.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943244","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Albumin-based nanocarriers loaded with novel Zn(II)-thiosemicarbazone compounds chart a new path for precision breast cancer therapy. 新型锌(II)-硫代氨基脲类化合物的白蛋白纳米载体为乳腺癌的精准治疗开辟了新的途径。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-06 DOI: 10.1097/CAD.0000000000001679
Ferdane Danişman-Kalindemirtaş, Dilşad Özerkan, İshak Afşin Kariper, Gökçe Erdemir Cilasun, Bahri Ülküseven, Serap Erdem-Kuruca
{"title":"Albumin-based nanocarriers loaded with novel Zn(II)-thiosemicarbazone compounds chart a new path for precision breast cancer therapy.","authors":"Ferdane Danişman-Kalindemirtaş, Dilşad Özerkan, İshak Afşin Kariper, Gökçe Erdemir Cilasun, Bahri Ülküseven, Serap Erdem-Kuruca","doi":"10.1097/CAD.0000000000001679","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001679","url":null,"abstract":"<p><p>This study explores the therapeutic potential of albumin-bound Zn(II)-thiosemicarbazone compounds (Alb-ZnTcA, Alb-ZnTcB) against breast cancer cells. Previous research indicates that these compounds hinder cancer cell proliferation by blocking DNA synthesis, promoting oxidative stress to induce apoptosis, and disrupting the cell cycle to inhibit cellular division. This study focuses on the loading and characterization of these potentially chemically unstable compounds on bovine serum albumin-based nanocarriers. Accordingly, unlike previous studies using albumin nanoparticles, in this study, ultraviolet light was used to precisely bind the therapeutic agent to albumin during the integration of thiosemicarbazones, achieving controlled nanoparticle size to control nanoparticle size. The mean diameter of Alb-ZnTcA nanoparticles was 32 nm, while Alb-ZnTcB exhibited an average diameter of 43 nm. Notably, Alb-ZnTcA displayed the highest cytotoxicity toward breast cancer cells, suggesting an optimal size for cellular uptake. Additionally, albumin-bound compounds showed enhanced cytotoxicity at lower concentrations, potentially minimizing adverse side effects. Apoptosis analysis indicated that both Alb-ZnTcA and Alb-ZnTcB induce cell death predominantly through apoptosis, effectively preventing the uncontrolled proliferation of cancer cells. These findings demonstrate the potential of Zn(II)-thiosemicarbazone compounds loaded on albumin-based nanocarriers for breast cancer treatment. The increased potency of Alb-ZnTcA and Alb-ZnTcB compared to free compounds, along with their ability to activate apoptotic signaling pathways in MCF-7 breast cancer cells, highlights a promising approach for future cancer therapies. This study suggests that albumin-bound Zn(II)-thiosemicarbazone compounds could offer a targeted and effective strategy in breast cancer treatment, leveraging the advantages of nanocarrier-based delivery systems.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943241","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The impact of bevacizumab intraperitoneal perfusion combined with paclitaxel and platinum-based chemotherapy on serum stromal-derived factor-1α (SDF-1α) and chemokine ligand 5 (CXCL-5) levels in patients with ovarian cancer after tumor cell debulking surgery. 贝伐单抗腹腔灌注联合紫杉醇、铂类化疗对卵巢癌患者肿瘤细胞去膨胀手术后血清基质衍生因子-1α (SDF-1α)和趋化因子配体5 (CXCL-5)水平的影响
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-06 DOI: 10.1097/CAD.0000000000001663
Liangliang Wang, Shanshan Ma, Huiwen Su, Dandan Nie, Lihua Wang
{"title":"The impact of bevacizumab intraperitoneal perfusion combined with paclitaxel and platinum-based chemotherapy on serum stromal-derived factor-1α (SDF-1α) and chemokine ligand 5 (CXCL-5) levels in patients with ovarian cancer after tumor cell debulking surgery.","authors":"Liangliang Wang, Shanshan Ma, Huiwen Su, Dandan Nie, Lihua Wang","doi":"10.1097/CAD.0000000000001663","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001663","url":null,"abstract":"&lt;p&gt;&lt;p&gt;The aim of this study is to investigate the impact of bevacizumab intraperitoneal perfusion combined with paclitaxel and platinum-based chemotherapy on serum stromal-derived factor-1α (SDF-1α) and chemokine ligand 5 (CXCL-5) levels in patients with ovarian cancer after tumor cell debulking surgery. This clinical study was conducted on a cohort of 89 ovarian cancer patients who underwent tumor debulking surgery at our hospital from February 2020 to February 2021. The patients were divided into two groups using a random number table: the control group (n = 44) received postoperative treatment with paclitaxel and platinum-based chemotherapy, while the research group (n = 45) received additional treatment with intraperitoneal perfusion of bevacizumab in addition to the control group's treatment regimen. The analysis included an assessment of the clinical efficacy of both groups, changes in tumor biomarker levels before and after treatment, serum levels of SDF-1α and CXCL-5, T-lymphocyte subset levels, treatment-related adverse reactions, and a 2-year prognosis and survival assessment. The research group showed better performance compared to the control group in terms of disease remission rate (80.00% vs. 59.09%) and treatment effectiveness rate (95.56% vs. 75.00%) (P &lt; 0.05). Before treatment, the levels of tumor biomarkers between the two groups were compared (P &gt; 0.05). After treatment, the levels of serum ferritin, carbohydrate antigen 125, carbohydrate antigen 199, and human epididymis protein 4 in both groups significantly decreased compared to before treatment, with the research group having lower levels (P &lt; 0.05). Before treatment, serum levels of SDF-1α and CXCL-5 between the two groups were compared (P &gt; 0.05). After treatment, however, the levels of SDF-1α and CXCL-5 significantly decreased compared to before treatment, with the research group having lower levels than the control group (P &lt; 0.05). Before treatment, there was no difference in T-lymphocyte levels between the two groups (P &gt; 0.05). In the control group, there was no significant change in T-lymphocyte levels before and after treatment (P &gt; 0.05). In the research group, however, after treatment, each indicator increased compared to before treatment, and posttreatment levels of all indicators were higher than those in the control group (P &lt; 0.05). The adverse reactions were compared between the two groups (P &gt; 0.05). The research group had a longer average survival time than the control group, with 1-year and 2-year survival rates higher than the control group (P &lt; 0.05). There was, however, no significant difference between the two groups in terms of local recurrence and metastasis (P &gt; 0.05). In conclusion, bevacizumab intraperitoneal perfusion combined with paclitaxel and platinum-based chemotherapy shows better clinical efficacy in the treatment of ovarian cancer after tumor cell debulking surgery. It can significantly reduce the levels of serum SDF-1α and CXCL-5 in pa","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142930340","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
WISP1 inhibition of YAP phosphorylation drives breast cancer growth and chemoresistance via TEAD4 activation. WISP1抑制YAP磷酸化通过TEAD4激活驱动乳腺癌生长和化疗耐药。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-06 DOI: 10.1097/CAD.0000000000001687
Tingting Dong, Li Liu, Yikai You, Jin Liu, Fuchao Wang, Shimeng Li, Zhenghong Yu
{"title":"WISP1 inhibition of YAP phosphorylation drives breast cancer growth and chemoresistance via TEAD4 activation.","authors":"Tingting Dong, Li Liu, Yikai You, Jin Liu, Fuchao Wang, Shimeng Li, Zhenghong Yu","doi":"10.1097/CAD.0000000000001687","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001687","url":null,"abstract":"<p><p>Wnt1-inducible signaling pathway protein 1 (WISP1) promotes breast cancer. The Hippo signaling pathway demonstrates a potential connection with WISP1, necessitating an exploration of their interaction. This study hypothesized that WISP1 boosts breast cancer by modulating the Hippo signaling pathway. The Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases were used to analyze WISP1 expression and Hippo signaling in breast cancer patients. WISP1, yes-associated protein (YAP), and domain family member 4 (TEAD4) were overexpressed or silenced in breast cancer cells. Epithelial-mesenchymal transition (EMT), and chemoresistance of breast cancer cells were evaluated. Immunofluorescence, PCR, immunoprecipitation, and western blot were used to detect the expression of WISP1 and key Hippo signaling factors and their interactions. Enrichment analysis indicated activation of WISP1 and Hippo signaling pathway and correlated with a worse prognosis in breast cancer. WISP1 overexpression facilitated EMT and chemotherapy resistance in breast cancer. Importantly, overexpression of WISP1 promoted YAP's nuclear translocation. TEAD4 expression in YAP precipitates from nuclear of WISP1-overexpressing MCF-7 cells increased. The promoting effect of WISP1 on breast cancer was counteracted by silencing YAP or TEAD4. Moreover, in WISP1 small interfering RNA-transfected MCF-7 cells, p-YAP expression increased, while interaction between YAP and TEAD4 decreased. WISP1 silencing led to ubiquitin increase and TEAD reduction in the p-YAP precipitates. In conclusion, WISP1 promotes YAP nuclear translocation and binding with TEAD4 by inhibiting YAP phosphorylation, reducing ubiquitin recruitment, and participating in transcriptional regulation in breast cancer.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KLF4 activates LATS2 to promote cisplatin sensitivity in ovarian cancer through DNA damage. KLF4 通过 DNA 损伤激活 LATS2,促进卵巢癌对顺铂的敏感性。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-01 Epub Date: 2024-09-27 DOI: 10.1097/CAD.0000000000001662
Ling Ma, Xiaoting Zhao, Xiang Lu, Jiahui Shen, Jiankang Huang
{"title":"KLF4 activates LATS2 to promote cisplatin sensitivity in ovarian cancer through DNA damage.","authors":"Ling Ma, Xiaoting Zhao, Xiang Lu, Jiahui Shen, Jiankang Huang","doi":"10.1097/CAD.0000000000001662","DOIUrl":"10.1097/CAD.0000000000001662","url":null,"abstract":"<p><p>We aimed to investigate the role of large tumor suppressor kinase 2 (LATS2) in cisplatin (DDP) sensitivity in ovarian cancer. Bioinformatic analysis explored LATS2 expression, pathways, and regulators. Quantitative reverse transcription-PCR measured LATS2 and KLF4 mRNA levels. Dual-luciferase and chromatin immunoprecipitation assays confirmed their binding relationship. Cell viability, half maximal inhibitory concentration (IC 50 ) values, cell cycle, and DNA damage were assessed using CCK-8, flow cytometry, and comet assays. Western blot analyzed protein expression. The effect of LATS2 on the sensitivity of ovarian cancer to DDP was verified in vivo . LATS2 and KLF4 were downregulated in ovarian cancer, with LATS2 enriched in cell cycle, DNA replication, and mismatch repair pathways. KLF4, an upstream regulator of LATS2, bound to its promoter. Overexpressing LATS2 increased G1-phase cells, reduced cell viability and IC 50 values, and induced DNA damage. Silencing KLF4 alone showed the opposite effect on LATS2 overexpression. Knocking out LATS2 reversed the effects of KLF4 overexpression on cell viability, cell cycle, IC 50 values, and DNA damage in ovarian cancer cells. Inhibiting LATS2 inactivated the Hippo-YAP signaling pathway. In vivo experiments showed that overexpression of LATS2 enhanced the sensitivity of ovarian cancer to DDP. KLF4 activates LATS2 via DNA damage to enhance DDP sensitivity in ovarian cancer, providing a potential target for improving treatment outcomes.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":"49-61"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142374909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SMAC mimetic BV6 acts in synergy with mTOR inhibitor to increase cisplatin sensitivity in ovarian cancer. SMAC 模拟物 BV6 与 mTOR 抑制剂协同作用,可提高卵巢癌患者对顺铂的敏感性。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-01 Epub Date: 2024-10-16 DOI: 10.1097/CAD.0000000000001664
Qi Chen, Hong Zhang
{"title":"SMAC mimetic BV6 acts in synergy with mTOR inhibitor to increase cisplatin sensitivity in ovarian cancer.","authors":"Qi Chen, Hong Zhang","doi":"10.1097/CAD.0000000000001664","DOIUrl":"10.1097/CAD.0000000000001664","url":null,"abstract":"<p><p>The objective of this study is to observe the antitumor efficacy of the second mitochondria-derived activator of caspases (SMAC) mimetic bivalent smac mimetic (BV6) in combination with target of rapamycin (mTOR) inhibitor on DDP (cisplatin) sensitivity. Ovarian cancer cells were exposed to cisplatin, BV6, DDP + BV6, and DDP + BV6 + mTOR inhibitor Rapamycin. Using proteomics and bioinformatics, protein expression profiles in ovarian cancer were determined. Bagg Albino color nude mice were treated with DDP or BV6 alone or in combination, or BV6 + DDP + Rapamycin. The effects of different treatments on ovarian cancer cells and tumor growth were evaluated in vivo and in vitro . Proteomics and bioinformatics analysis revealed significant changes of protein kinase (AKT)/mTOR pathway. Consistently, western blot data indicated that AKT/mTOR axis was gradually activated in BV6-treated ovarian cancer cells and attenuated the cytotoxic effect of BV6. Functional assays showed that DDP or BV6 treatment alone significantly enhanced the sensitivity and inhibited the migration of ovarian cancer cells, but without any synergistic effects. In addition, combination with BV6 and mTOR inhibitor Rapamycin significantly decreased cell viability and inhibited migration of ovarian cancer cells exposed to DDP. Consistently, the xenograft model showed that co-treatment with Rapamycin with BV6 had significantly suppressed tumor growth and metastasis. Our study demonstrated that SMAC analogue BV6 exhibits a strong anticancer effect on ovarian cancer in vitro and in vivo . Combination with Rapamycin overcomes the activation of mTOR pathway by BV6 and increases the chemosensitivity to DDP. These data suggest a potential application of triple combination with DDP + BV6 + Rapamycin in clinical management of ovarian cancer.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":"62-71"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456486","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Screening of a kinase inhibitor library identified novel targetable kinase pathways in triple-negative breast cancer. 激酶抑制剂文库的筛选在三阴性乳腺癌中发现了新的靶向激酶途径。
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-01 Epub Date: 2024-08-21 DOI: 10.1097/CAD.0000000000001658
Caroline H Rinderle, Christopher V Baker, Courtney B Lagarde, Khoa Nguyen, Sara Al-Ghadban, Margarite D Matossian, Van T Hoang, Elizabeth C Martin, Bridgette M Collins-Burow, Simak Ali, David H Drewry, Matthew E Burow, Bruce A Bunnell
{"title":"Screening of a kinase inhibitor library identified novel targetable kinase pathways in triple-negative breast cancer.","authors":"Caroline H Rinderle, Christopher V Baker, Courtney B Lagarde, Khoa Nguyen, Sara Al-Ghadban, Margarite D Matossian, Van T Hoang, Elizabeth C Martin, Bridgette M Collins-Burow, Simak Ali, David H Drewry, Matthew E Burow, Bruce A Bunnell","doi":"10.1097/CAD.0000000000001658","DOIUrl":"10.1097/CAD.0000000000001658","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is a highly invasive breast cancer subtype that is challenging to treat due to inherent heterogeneity and absence of estrogen, progesterone, and human epidermal growth factor 2 receptors. Kinase signaling networks drive cancer growth and development, and kinase inhibitors are promising anti-cancer strategies in diverse cancer subtypes. Kinase inhibitor screens are an efficient, valuable means of identifying compounds that suppress cancer cell growth in vitro , facilitating the identification of kinase vulnerabilities to target therapeutically. The Kinase Chemogenomic Set is a well-annotated library of 187 kinase inhibitor compounds that indexes 215 kinases of the 518 in the known human kinome representing various kinase networks and signaling pathways, several of which are understudied. Our screen revealed 14 kinase inhibitor compounds effectively inhibited TNBC cell growth and proliferation. Upon further testing, three compounds, THZ531, THZ1, and PFE-PKIS 29, had the most significant and consistent effects across a range of TNBC cell lines. These cyclin-dependent kinase (CDK)12/CDK13, CDK7, and phosphoinositide 3-kinase inhibitors, respectively, decreased metabolic activity in TNBC cell lines and promote a gene expression profile consistent with the reversal of the epithelial-to-mesenchymal transition, indicating these kinase networks potentially mediate metastatic behavior. These data identified novel kinase targets and kinase signaling pathways that drive metastasis in TNBC.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":"36 1","pages":"39-48"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11634125/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821722","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combination of anlotinib and sintilimab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma: a single-arm prospective study. anlotinib和sintilmab联合治疗复发或转移性头颈部鳞状细胞癌:单臂前瞻性研究
IF 1.8 4区 医学
Anti-Cancer Drugs Pub Date : 2025-01-01 Epub Date: 2024-12-11 DOI: 10.1097/CAD.0000000000001660
Tianxiao Wang, Jiaxin Wang, Yabing Zhang, Yuntao Song, Guohui Xu, Bin Zhang
{"title":"Combination of anlotinib and sintilimab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma: a single-arm prospective study.","authors":"Tianxiao Wang, Jiaxin Wang, Yabing Zhang, Yuntao Song, Guohui Xu, Bin Zhang","doi":"10.1097/CAD.0000000000001660","DOIUrl":"10.1097/CAD.0000000000001660","url":null,"abstract":"<p><p>To investigate whether blocking both programmed cell death protein and vascular endothelial growth factor receptor could offer superior anticancer activity in these patients without compromising safety. In this study, patients were administered oral anlotinib (12 mg/day) on days 1-14 and intravenous sintilimab (200 mg) on day 1 of a 3-weekly cycle. The primary endpoints included the objective response rate and disease control rate. The secondary endpoints included overall survival (OS) and safety. Ten eligible patients were enrolled between June 2019 and May 2022, and eight patients underwent radiographic assessments. The results showed an objective response rate of 50% (partial and complete response in four and zero patients, respectively) and a disease control rate of 100%; four patients demonstrated stable disease for at least 8 weeks. The median OS was 4.37 (in our study, the score was 7), and the OS rate at 12 months was 37.5%. The Kaplan-Meier survival curve showed that the group with high blood glucose levels had a significantly shorter duration of survival than those with normal blood glucose levels. Adverse events of grade 3 and higher occurred in 50% of patients, and the most common severe adverse events included tumor pain (50%), hypertension (37.5%), tumor hemorrhage (25%), and decreased appetite (25%). The combination of anlotinib and sintilimab showed promising efficacy in controlling tumor size. However, the disappointing OS rate suggests that anti-vascular endothelial growth factor receptor agents should be used cautiously after radical radiation therapy. The combination used in this study demonstrated a toxicity profile comparable to that of other agents used in this setting. These findings warrant further investigation into the potential clinical utility of this combination.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":"36 1","pages":"79-84"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11634082/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821762","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信