首页 > 最新文献

Cancer Chemotherapy and Pharmacology最新文献

英文 中文
Avermectin B1 mediates antitumor activity and induces autophagy in osteosarcoma through the AMPK/ULK1 signaling pathway. 阿维菌素 B1 通过 AMPK/ULK1 信号通路介导骨肉瘤的抗肿瘤活性并诱导自噬。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-09-05 DOI: 10.1007/s00280-024-04695-z
Xiang Fei, Zhaohui Li, Zhen Pan, Yonghui Liang, Chen Tan, Dongdong Cheng, Qingcheng Yang

Background: Osteosarcoma is the most common malignant bone tumor in children and adolescents. Conventional chemotherapy remains unsatisfactory due to drug toxicity and resistance issues. Therefore, there is an urgent need to develop more effective treatments for advanced osteosarcoma. In the current study, we focused on evaluating the anticancer efficacy of avermectin B1, a novel avermectin analog, against osteosarcoma cells.

Methods: The half-inhibitory concentration of avermectin B1 was calculated in three osteosarcoma cell lines. Then, functional experiments were conducted to evaluate the effects of avermectin B1 on cell proliferation, the cell cycle, apoptosis and autophagy. Moreover, the AMPK/ULK1 signaling pathway was detected by Western blot assay. Finally, the in vivo effect of avermectin B1 on tumor growth and metastasis was investigated using the xenograft mouse model. To examine the role of the AMPK/ULK1 pathway, an AMPK-specific inhibitor (dorsomorphin) was used in combination with avermectin B1.

Results: Avermectin B1 inhibited the proliferation of osteosarcoma cells in a dose-dependent manner based on CCK8 and colony formation assays. Then, it was found to inhibit migration and invasion by wound healing assay and cell migration and invasion assay. In addition, avermectin B1 induced osteosarcoma cell apoptosis and autophagy. In vivo, avermectin B1 effectively inhibited osteosarcoma cell growth and pulmonary metastasis. Mechanistically, avermectin B1 activated the AMPK/ULK1 pathway to exert antitumor activity in vitro and in vivo. Dorsomorphin significantly attenuated the Avermectin B1-induced antitumor activities.

Conclusion: Our study suggests that avermectin B1 is a potential agent to treat osteosarcoma cells through the AMPK/ULK1 signaling pathway.

背景:骨肉瘤是儿童和青少年最常见的恶性骨肿瘤:骨肉瘤是儿童和青少年中最常见的恶性骨肿瘤。由于药物毒性和耐药性问题,传统化疗效果仍不理想。因此,开发更有效的晚期骨肉瘤治疗方法迫在眉睫。在本研究中,我们重点评估了新型阿维菌素类似物阿维菌素 B1 对骨肉瘤细胞的抗癌疗效:方法:计算了阿维菌素 B1 在三种骨肉瘤细胞系中的半抑制浓度。然后,通过功能实验评估阿维菌素 B1 对细胞增殖、细胞周期、细胞凋亡和自噬的影响。此外,还通过 Western 印迹检测了 AMPK/ULK1 信号通路。最后,利用异种移植小鼠模型研究了阿维菌素 B1 对肿瘤生长和转移的体内影响。为了研究AMPK/ULK1通路的作用,研究人员将AMPK特异性抑制剂(多索吗啉)与阿维菌素B1联合使用:结果:根据CCK8和集落形成试验,阿维菌素B1以剂量依赖的方式抑制骨肉瘤细胞的增殖。然后,通过伤口愈合试验和细胞迁移与侵袭试验发现,阿维菌素 B1 可抑制迁移和侵袭。此外,阿维菌素 B1 还能诱导骨肉瘤细胞凋亡和自噬。在体内,阿维菌素 B1 能有效抑制骨肉瘤细胞的生长和肺转移。从机理上讲,阿维菌素 B1 可激活 AMPK/ULK1 通路,从而在体外和体内发挥抗肿瘤活性。多索吗啡可明显减弱阿维菌素B1诱导的抗肿瘤活性:我们的研究表明,阿维菌素 B1 是一种通过 AMPK/ULK1 信号通路治疗骨肉瘤细胞的潜在药物。
{"title":"Avermectin B1 mediates antitumor activity and induces autophagy in osteosarcoma through the AMPK/ULK1 signaling pathway.","authors":"Xiang Fei, Zhaohui Li, Zhen Pan, Yonghui Liang, Chen Tan, Dongdong Cheng, Qingcheng Yang","doi":"10.1007/s00280-024-04695-z","DOIUrl":"10.1007/s00280-024-04695-z","url":null,"abstract":"<p><strong>Background: </strong>Osteosarcoma is the most common malignant bone tumor in children and adolescents. Conventional chemotherapy remains unsatisfactory due to drug toxicity and resistance issues. Therefore, there is an urgent need to develop more effective treatments for advanced osteosarcoma. In the current study, we focused on evaluating the anticancer efficacy of avermectin B1, a novel avermectin analog, against osteosarcoma cells.</p><p><strong>Methods: </strong>The half-inhibitory concentration of avermectin B1 was calculated in three osteosarcoma cell lines. Then, functional experiments were conducted to evaluate the effects of avermectin B1 on cell proliferation, the cell cycle, apoptosis and autophagy. Moreover, the AMPK/ULK1 signaling pathway was detected by Western blot assay. Finally, the in vivo effect of avermectin B1 on tumor growth and metastasis was investigated using the xenograft mouse model. To examine the role of the AMPK/ULK1 pathway, an AMPK-specific inhibitor (dorsomorphin) was used in combination with avermectin B1.</p><p><strong>Results: </strong>Avermectin B1 inhibited the proliferation of osteosarcoma cells in a dose-dependent manner based on CCK8 and colony formation assays. Then, it was found to inhibit migration and invasion by wound healing assay and cell migration and invasion assay. In addition, avermectin B1 induced osteosarcoma cell apoptosis and autophagy. In vivo, avermectin B1 effectively inhibited osteosarcoma cell growth and pulmonary metastasis. Mechanistically, avermectin B1 activated the AMPK/ULK1 pathway to exert antitumor activity in vitro and in vivo. Dorsomorphin significantly attenuated the Avermectin B1-induced antitumor activities.</p><p><strong>Conclusion: </strong>Our study suggests that avermectin B1 is a potential agent to treat osteosarcoma cells through the AMPK/ULK1 signaling pathway.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438708/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131945","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
Pharmacokinetic and bioequivalence study of two capecitabine tablets in Chinese patients with solid tumor cancer. 两种卡培他滨片剂在中国实体瘤癌症患者中的药代动力学和生物等效性研究
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-03-07 DOI: 10.1007/s00280-024-04652-w
Guangnan Zong, Jinlin Li, Xiushan Gong, Yuanyuan Liu, Xiuli Guo

Capecitabine (CAP) is one of the fluoropyrimidine deoxynucleoside carbamates, which can be converted to 5-fluorouracil (5-FU) by thymine deoxynucleoside phosphorylase (dThdPase) to exert antitumor effects. The purpose of this study is to compare the pharmacokinetics (PK), bioequivalence (BE), and safety of two CAP tablets in Chinese patients with solid tumor cancer. The results showed that the geometric mean ratios (GMRs) of Cmax, AUC0-t and AUC0-∞ of CAP T/R reagent were 90.26%, 95.27%, and 95.07, respectively. The values and 90% confidence intervals (CI) of AUC0-t, AUC0-∞, and Cmax all fall within the range of 80.00-125.00%. In addition, a total of 22 subjects in this study had 30 adverse events, with an incidence of 45.83%, and there were no serious adverse events and adverse events that led to withdrawal from the trial.

卡培他滨(CAP)是氟嘧啶脱氧核苷氨基甲酸酯类药物之一,可在胸腺嘧啶脱氧核苷磷酸酶(dThdPase)的作用下转化为5-氟尿嘧啶(5-FU),从而发挥抗肿瘤作用。本研究旨在比较两种CAP片剂在中国实体瘤癌症患者中的药代动力学(PK)、生物等效性(BE)和安全性。结果显示,CAP T/R试剂的Cmax、AUC0-t和AUC0-∞的几何平均比(GMRs)分别为90.26%、95.27%和95.07。AUC0-t、AUC0-∞ 和 Cmax 的值和 90% 置信区间(CI)均在 80.00-125.00% 的范围内。此外,本研究中共有 22 名受试者发生了 30 起不良事件,发生率为 45.83%,没有出现严重不良事件和导致退出试验的不良事件。
{"title":"Pharmacokinetic and bioequivalence study of two capecitabine tablets in Chinese patients with solid tumor cancer.","authors":"Guangnan Zong, Jinlin Li, Xiushan Gong, Yuanyuan Liu, Xiuli Guo","doi":"10.1007/s00280-024-04652-w","DOIUrl":"10.1007/s00280-024-04652-w","url":null,"abstract":"<p><p>Capecitabine (CAP) is one of the fluoropyrimidine deoxynucleoside carbamates, which can be converted to 5-fluorouracil (5-FU) by thymine deoxynucleoside phosphorylase (dThdPase) to exert antitumor effects. The purpose of this study is to compare the pharmacokinetics (PK), bioequivalence (BE), and safety of two CAP tablets in Chinese patients with solid tumor cancer. The results showed that the geometric mean ratios (GMRs) of C<sub>max</sub>, AUC<sub>0-t</sub> and AUC<sub>0-∞</sub> of CAP T/R reagent were 90.26%, 95.27%, and 95.07, respectively. The values and 90% confidence intervals (CI) of AUC<sub>0-t</sub>, AUC<sub>0-∞</sub>, and C<sub>max</sub> all fall within the range of 80.00-125.00%. In addition, a total of 22 subjects in this study had 30 adverse events, with an incidence of 45.83%, and there were no serious adverse events and adverse events that led to withdrawal from the trial.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140058689","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
Ferroptosis - a potential feature underlying neratinib-induced colonic epithelial injury. 铁蛋白沉积--奈拉替尼诱导的结肠上皮损伤的潜在特征。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-13 DOI: 10.1007/s00280-024-04699-9
Triet P M Nguyen, Susan L Woods, Kate R Secombe, Simon Tang, Aurelia S Elz, Scott Ayton, John Finnie, Aadya Nagpal, Normand Pouliot, Joanne M Bowen

Purpose: Neratinib, a small-molecule tyrosine kinase inhibitor (TKI) that irreversibly binds to human epidermal growth factor receptors 1, 2 and 4 (HER1/2/4), is an approved extended adjuvant therapy for patients with HER2-amplified or -overexpressed (HER2-positive) breast cancers. Patients receiving neratinib may experience mild-to-severe symptoms of gut toxicity including abdominal pain and diarrhoea. Despite being a highly prevalent complication in gut health, the biological processes underlying neratinib-induced gut injury, especially in the colon, remains unclear.

Methods: Real-time quantitative polymerase chain reaction (RT-qPCR) and histology were integrated to study the effect of, and type of cell death induced by neratinib on colonic tissues collected from female Albino Wistar rats dosed with neratinib (50 mg/kg) daily for 28 days. Additionally, previously published bulk RNA-sequencing and CRISPR-screening datasets on human glioblastoma SF268 cell line and glioblastoma T895 xenograft, and mouse TBCP1 breast cancer cell line were leveraged to elucidate potential mechanisms of neratinib-induced cell death.

Results: The severity of colonic epithelial injury, especially degeneration of surface lining colonocytes and infiltration of immune cells, was more pronounced in the distal colon than the proximal colon. Sequencing showed that apoptotic gene signature was enriched in neratinib-treated SF268 cells while ferroptotic gene signature was enriched in neratinib-treated TBCP1 cells and T895 xenograft. However, we found that ferroptosis, but less likely apoptosis, was a potential histopathological feature underlying colonic injury in rats treated with neratinib.

Conclusion: Ferroptosis is a potential feature of neratinib-induced colonic injury and that targeting molecular machinery governing neratinib-induced ferroptosis may represent an attractive therapeutic approach to ameliorate symptoms of gut toxicity.

目的:奈拉替尼是一种与人类表皮生长因子受体1、2和4(HER1/2/4)不可逆结合的小分子酪氨酸激酶抑制剂(TKI),已被批准用于HER2扩增或表达(HER2阳性)乳腺癌患者的延长辅助治疗。接受奈拉替尼治疗的患者可能会出现轻度至严重的肠道毒性症状,包括腹痛和腹泻。尽管奈拉替尼是肠道健康的一种高发并发症,但奈拉替尼诱发肠道损伤(尤其是结肠损伤)的生物学过程仍不清楚:方法:将实时定量聚合酶链反应(RT-qPCR)和组织学相结合,研究奈拉替尼对雌性白化Wistar大鼠结肠组织诱导细胞死亡的影响和类型。此外,还利用以前发表的关于人胶质母细胞瘤SF268细胞系和胶质母细胞瘤T895异种移植以及小鼠TBCP1乳腺癌细胞系的大量RNA测序和CRISPR筛选数据集来阐明奈拉替尼诱导细胞死亡的潜在机制:结果:结肠上皮损伤的严重程度,尤其是结肠表面内膜细胞的变性和免疫细胞的浸润,远端结肠比近端结肠更明显。测序结果显示,奈拉替尼处理的SF268细胞富集了凋亡基因特征,而奈拉替尼处理的TBCP1细胞和T895异种移植物富集了铁蛋白基因特征。然而,我们发现,铁卟啉沉积(但不一定是细胞凋亡)是使用奈拉替尼治疗的大鼠结肠损伤的潜在组织病理学特征:结论:铁细胞凋亡是奈拉替尼诱导的结肠损伤的潜在特征,针对奈拉替尼诱导的铁细胞凋亡的分子机制可能是改善肠道毒性症状的一种有吸引力的治疗方法。
{"title":"Ferroptosis - a potential feature underlying neratinib-induced colonic epithelial injury.","authors":"Triet P M Nguyen, Susan L Woods, Kate R Secombe, Simon Tang, Aurelia S Elz, Scott Ayton, John Finnie, Aadya Nagpal, Normand Pouliot, Joanne M Bowen","doi":"10.1007/s00280-024-04699-9","DOIUrl":"10.1007/s00280-024-04699-9","url":null,"abstract":"<p><strong>Purpose: </strong>Neratinib, a small-molecule tyrosine kinase inhibitor (TKI) that irreversibly binds to human epidermal growth factor receptors 1, 2 and 4 (HER1/2/4), is an approved extended adjuvant therapy for patients with HER2-amplified or -overexpressed (HER2-positive) breast cancers. Patients receiving neratinib may experience mild-to-severe symptoms of gut toxicity including abdominal pain and diarrhoea. Despite being a highly prevalent complication in gut health, the biological processes underlying neratinib-induced gut injury, especially in the colon, remains unclear.</p><p><strong>Methods: </strong>Real-time quantitative polymerase chain reaction (RT-qPCR) and histology were integrated to study the effect of, and type of cell death induced by neratinib on colonic tissues collected from female Albino Wistar rats dosed with neratinib (50 mg/kg) daily for 28 days. Additionally, previously published bulk RNA-sequencing and CRISPR-screening datasets on human glioblastoma SF268 cell line and glioblastoma T895 xenograft, and mouse TBCP1 breast cancer cell line were leveraged to elucidate potential mechanisms of neratinib-induced cell death.</p><p><strong>Results: </strong>The severity of colonic epithelial injury, especially degeneration of surface lining colonocytes and infiltration of immune cells, was more pronounced in the distal colon than the proximal colon. Sequencing showed that apoptotic gene signature was enriched in neratinib-treated SF268 cells while ferroptotic gene signature was enriched in neratinib-treated TBCP1 cells and T895 xenograft. However, we found that ferroptosis, but less likely apoptosis, was a potential histopathological feature underlying colonic injury in rats treated with neratinib.</p><p><strong>Conclusion: </strong>Ferroptosis is a potential feature of neratinib-induced colonic injury and that targeting molecular machinery governing neratinib-induced ferroptosis may represent an attractive therapeutic approach to ameliorate symptoms of gut toxicity.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438713/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141603364","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
Darifenacin: a promising chitinase 3-like 1 inhibitor to tackle drug resistance in pancreatic ductal adenocarcinoma. 达非那新:一种有望解决胰腺导管腺癌耐药性的几丁质酶 3-like 1 抑制剂。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-09-03 DOI: 10.1007/s00280-024-04712-1
Sofia M Sousa, Helena Branco, Amir Avan, Andreia Palmeira, Luca Morelli, Lúcio L Santos, Elisa Giovannetti, M Helena Vasconcelos, Cristina P R Xavier

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is among the most aggressive malignancies. Our previous work revealed Chitinase 3-like 1 (CHI3L1) involvement in PDAC resistance to gemcitabine, identifying it as a promising therapeutic target. Here, we aimed to identify putative CHI3L1 inhibitors and to investigate their chemosensitizing potential in PDAC.

Methods: Docking analysis for CHI3L1 identified promising CHI3L1 inhibitors, including darifenacin (muscarinic receptor antagonist). PDAC cell lines (BxPC-3, PANC-1) and primary PDAC cells were used to evaluate darifenacin's effects on cell growth (Sulforhodamine B, SRB), alone or in combination with gemcitabine or gemcitabine plus paclitaxel. Cytotoxicity against normal immortalized pancreatic ductal cells (HPNE) was assessed. Recombinant protein was used to confirm the impact of darifenacin on CHI3L1-induced PDAC cellular resistance to therapy (SRB assay). Darifenacin's effect on Akt activation was analysed by ELISA. The association between cholinergic receptor muscarinic 3 (CHRM3) expression and therapeutic response was evaluated by immunohistochemistry of paraffin-embedded tissues from surgical resections of a 68 patients' cohort.

Results: In silico screening revealed the ability of darifenacin to target CHI3L1 with high efficiency. Darifenacin inhibited PDAC cell growth, with a GI50 of 26 and 13.6 µM in BxPC-3 and PANC-1 cells, respectively. These results were confirmed in primary PDAC-3 cells, while darifenacin showed no cytotoxicity against HPNE cells. Importantly, darifenacin sensitized PDAC cells to standard chemotherapies, reverted CHI3L1-induced PDAC cellular resistance to therapy, and decreased Akt phosphorylation. Additionally, high CHMR3 expression was associated with low therapeutic response to gemcitabine.

Conclusion: This work highlights the potential of darifenacin as a chemosensitizer for PDAC treatment.

目的:胰腺导管腺癌(PDAC)是侵袭性最强的恶性肿瘤之一。我们之前的研究发现几丁质酶 3-like 1 (CHI3L1) 参与了 PDAC 对吉西他滨的耐药性,并将其确定为一个有前景的治疗靶点。在此,我们旨在确定推定的 CHI3L1 抑制剂,并研究它们在 PDAC 中的化疗增敏潜力:方法:CHI3L1的对接分析确定了有希望的CHI3L1抑制剂,包括达非那新(毒蕈碱受体拮抗剂)。用 PDAC 细胞系(BxPC-3、PANC-1)和原代 PDAC 细胞来评估 darifenacin 单独或与吉西他滨或吉西他滨加紫杉醇联用对细胞生长的影响(磺基罗丹明 B,SRB)。对正常永生胰腺导管细胞(HPNE)的细胞毒性进行了评估。重组蛋白用于证实达非那新对CHI3L1诱导的PDAC细胞耐药性的影响(SRB测定)。通过 ELISA 分析了达非那新对 Akt 活化的影响。通过对68例患者的手术切除石蜡包埋组织进行免疫组化,评估了胆碱能受体毒蕈碱3(CHRM3)的表达与治疗反应之间的关系:硅学筛选显示,达非那新能够高效靶向CHI3L1。达非那新抑制了 PDAC 细胞的生长,在 BxPC-3 和 PANC-1 细胞中的 GI50 分别为 26 µM 和 13.6 µM。这些结果在原代 PDAC-3 细胞中得到了证实,而 darifenacin 对 HPNE 细胞没有细胞毒性。重要的是,达非那新能使 PDAC 细胞对标准化疗敏感,逆转 CHI3L1 诱导的 PDAC 细胞耐药性,并降低 Akt 磷酸化。此外,CHMR3的高表达与吉西他滨的低治疗反应相关:这项研究强调了达非那新作为化疗增敏剂治疗 PDAC 的潜力。
{"title":"Darifenacin: a promising chitinase 3-like 1 inhibitor to tackle drug resistance in pancreatic ductal adenocarcinoma.","authors":"Sofia M Sousa, Helena Branco, Amir Avan, Andreia Palmeira, Luca Morelli, Lúcio L Santos, Elisa Giovannetti, M Helena Vasconcelos, Cristina P R Xavier","doi":"10.1007/s00280-024-04712-1","DOIUrl":"10.1007/s00280-024-04712-1","url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic ductal adenocarcinoma (PDAC) is among the most aggressive malignancies. Our previous work revealed Chitinase 3-like 1 (CHI3L1) involvement in PDAC resistance to gemcitabine, identifying it as a promising therapeutic target. Here, we aimed to identify putative CHI3L1 inhibitors and to investigate their chemosensitizing potential in PDAC.</p><p><strong>Methods: </strong>Docking analysis for CHI3L1 identified promising CHI3L1 inhibitors, including darifenacin (muscarinic receptor antagonist). PDAC cell lines (BxPC-3, PANC-1) and primary PDAC cells were used to evaluate darifenacin's effects on cell growth (Sulforhodamine B, SRB), alone or in combination with gemcitabine or gemcitabine plus paclitaxel. Cytotoxicity against normal immortalized pancreatic ductal cells (HPNE) was assessed. Recombinant protein was used to confirm the impact of darifenacin on CHI3L1-induced PDAC cellular resistance to therapy (SRB assay). Darifenacin's effect on Akt activation was analysed by ELISA. The association between cholinergic receptor muscarinic 3 (CHRM3) expression and therapeutic response was evaluated by immunohistochemistry of paraffin-embedded tissues from surgical resections of a 68 patients' cohort.</p><p><strong>Results: </strong>In silico screening revealed the ability of darifenacin to target CHI3L1 with high efficiency. Darifenacin inhibited PDAC cell growth, with a GI<sub>50</sub> of 26 and 13.6 µM in BxPC-3 and PANC-1 cells, respectively. These results were confirmed in primary PDAC-3 cells, while darifenacin showed no cytotoxicity against HPNE cells. Importantly, darifenacin sensitized PDAC cells to standard chemotherapies, reverted CHI3L1-induced PDAC cellular resistance to therapy, and decreased Akt phosphorylation. Additionally, high CHMR3 expression was associated with low therapeutic response to gemcitabine.</p><p><strong>Conclusion: </strong>This work highlights the potential of darifenacin as a chemosensitizer for PDAC treatment.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11438711/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142119076","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
Impact of ALDH1A1 and NQO1 gene polymorphisms on the response and toxicity of chemotherapy in Bangladeshi breast cancer patients. ALDH1A1 和 NQO1 基因多态性对孟加拉乳腺癌患者化疗反应和毒性的影响。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-16 DOI: 10.1007/s00280-024-04700-5
Md Siddiqul Islam, Ferdowsi Akter, Md Mosiqur Rahman, Md Rajdoula Rafe, Md Abdul Aziz, Salma Parvin, Abu Syed Md Mosaddek, Mohammad Safiqul Islam, Md Wahid Akter

Purpose: Cyclophosphamide, Epirubicin/Doxorubicin, 5-fluorouracil (CEF or CAF) chemotherapy has long been a standard first-line treatment for breast cancer. The genetic variations of enzymes that are responsible for the metabolism of these drugs have been linked to altered treatment response and toxicity. Two drug-metabolizing enzymes ALDH1A1 and NQO1 are critically involved in the pathways of CEF/CAF metabolism. This study aimed to evaluate the effect of ALDH1A1 (rs13959) and NQO1 (rs1800566) polymorphisms on treatment response and toxicities caused by adjuvant (ACT) and neoadjuvant chemotherapy (NACT) where CEF/CAF combination was used to treat Bangladeshi breast cancer patients.

Methods: A total of 330 patients were recruited from various hospitals, with 150 receiving neoadjuvant chemotherapy and 180 receiving adjuvant chemotherapy. To extract genomic DNA, a non-enzymatic simple salting out approach was adopted. The polymerase chain reaction-restriction fragment length polymorphism method was used to detect genetic polymorphisms. Unconditional logistic regression was used to derive odds ratios (ORs) with 95% confidence intervals (CIs) to study the association between genetic polymorphisms and clinical outcome and toxicity.

Results: A statistically significant association was observed between ALDH1A1 (rs13959) polymorphism and treatment response (TT vs. CC: aOR = 6.40, p = 0.007; recessive model: aOR = 6.38, p = 0.002; allele model: p = 0.032). Patients with the genotypes TT and CT + TT of the NQO1 (rs1800566) polymorphism had a significantly higher risk of toxicities such as anemia (aOR = 0.34, p = 0.006 and aOR = 0.58, p = 0.021), neutropenia (aOR = 0.42, p = 0.044 and aOR = 0.57, p = 0.027), leukopenia (aOR = 0.33, p = 0.010 and aOR = 0.46, p = 0.005), and gastrointestinal toxicity (aOR = 0.30, p = 0.02 and aOR = 0.38, p = 0.006) when compared to the wild CC genotype, while patients with the genotype CT had a significant association with gastrointestinal toxicity (aOR = 0.42, p = 0.02) and leukopenia (aOR = 0.52, p = 0.010). The TT and CT + TT genotypes of rs13959 had a significantly higher risk of anemia (aOR = 2.00, p = 0.037 and aOR = 1.68, p = 0.029). There was no significant association between rs1800566 polymorphism and treatment response.

Conclusion: Polymorphisms in ALDH1A1 (rs13959) and NQO1 (rs1800566) may be useful in predicting the probability of treatment response and adverse effects from CEF or CAF-based chemotherapy in breast cancer patients.

目的:环磷酰胺、表柔比星/多柔比星、5-氟尿嘧啶(CEF 或 CAF)化疗长期以来一直是乳腺癌的标准一线治疗方法。负责这些药物代谢的酶的基因变异与治疗反应和毒性的改变有关。ALDH1A1 和 NQO1 这两种药物代谢酶在 CEF/CAF 代谢途径中起着关键作用。本研究旨在评估 ALDH1A1(rs13959)和 NQO1(rs1800566)多态性对孟加拉乳腺癌患者辅助化疗(ACT)和新辅助化疗(NACT)的治疗反应和毒性的影响:从多家医院共招募了 330 名患者,其中 150 人接受新辅助化疗,180 人接受辅助化疗。提取基因组 DNA 时,采用了非酶简单盐析法。聚合酶链反应-限制性片段长度多态性法用于检测基因多态性。采用无条件逻辑回归法得出几率比(ORs)和 95% 置信区间(CIs),以研究基因多态性与临床结果和毒性之间的关系:ALDH1A1(rs13959)多态性与治疗反应之间存在统计学意义上的显著关联(TT vs. CC:aOR = 6.40,p = 0.007;隐性模型:aOR = 6.38,p = 0.002;等位基因模型:p = 0.032)。NQO1(rs1800566)多态性基因型为 TT 和 CT + TT 的患者出现贫血(aOR = 0.34,p = 0.006 和 aOR = 0.58,p = 0.021)、中性粒细胞减少(aOR = 0.42,p = 0.044 和 aOR = 0.57,p = 0.027)、白细胞减少(aOR = 0.33,p = 0.与野生 CC 基因型相比,基因型为 CT 的患者与胃肠毒性(aOR = 0.42,p = 0.02)和白细胞减少症(aOR = 0.52,p = 0.010)显著相关。rs13959的TT和CT + TT基因型发生贫血的风险明显更高(aOR = 2.00,p = 0.037和aOR = 1.68,p = 0.029)。rs1800566多态性与治疗反应无明显关联:结论:ALDH1A1(rs13959)和NQO1(rs1800566)的多态性可能有助于预测乳腺癌患者的治疗反应概率以及基于CEF或CAF化疗的不良反应。
{"title":"Impact of ALDH1A1 and NQO1 gene polymorphisms on the response and toxicity of chemotherapy in Bangladeshi breast cancer patients.","authors":"Md Siddiqul Islam, Ferdowsi Akter, Md Mosiqur Rahman, Md Rajdoula Rafe, Md Abdul Aziz, Salma Parvin, Abu Syed Md Mosaddek, Mohammad Safiqul Islam, Md Wahid Akter","doi":"10.1007/s00280-024-04700-5","DOIUrl":"10.1007/s00280-024-04700-5","url":null,"abstract":"<p><strong>Purpose: </strong>Cyclophosphamide, Epirubicin/Doxorubicin, 5-fluorouracil (CEF or CAF) chemotherapy has long been a standard first-line treatment for breast cancer. The genetic variations of enzymes that are responsible for the metabolism of these drugs have been linked to altered treatment response and toxicity. Two drug-metabolizing enzymes ALDH1A1 and NQO1 are critically involved in the pathways of CEF/CAF metabolism. This study aimed to evaluate the effect of ALDH1A1 (rs13959) and NQO1 (rs1800566) polymorphisms on treatment response and toxicities caused by adjuvant (ACT) and neoadjuvant chemotherapy (NACT) where CEF/CAF combination was used to treat Bangladeshi breast cancer patients.</p><p><strong>Methods: </strong>A total of 330 patients were recruited from various hospitals, with 150 receiving neoadjuvant chemotherapy and 180 receiving adjuvant chemotherapy. To extract genomic DNA, a non-enzymatic simple salting out approach was adopted. The polymerase chain reaction-restriction fragment length polymorphism method was used to detect genetic polymorphisms. Unconditional logistic regression was used to derive odds ratios (ORs) with 95% confidence intervals (CIs) to study the association between genetic polymorphisms and clinical outcome and toxicity.</p><p><strong>Results: </strong>A statistically significant association was observed between ALDH1A1 (rs13959) polymorphism and treatment response (TT vs. CC: aOR = 6.40, p = 0.007; recessive model: aOR = 6.38, p = 0.002; allele model: p = 0.032). Patients with the genotypes TT and CT + TT of the NQO1 (rs1800566) polymorphism had a significantly higher risk of toxicities such as anemia (aOR = 0.34, p = 0.006 and aOR = 0.58, p = 0.021), neutropenia (aOR = 0.42, p = 0.044 and aOR = 0.57, p = 0.027), leukopenia (aOR = 0.33, p = 0.010 and aOR = 0.46, p = 0.005), and gastrointestinal toxicity (aOR = 0.30, p = 0.02 and aOR = 0.38, p = 0.006) when compared to the wild CC genotype, while patients with the genotype CT had a significant association with gastrointestinal toxicity (aOR = 0.42, p = 0.02) and leukopenia (aOR = 0.52, p = 0.010). The TT and CT + TT genotypes of rs13959 had a significantly higher risk of anemia (aOR = 2.00, p = 0.037 and aOR = 1.68, p = 0.029). There was no significant association between rs1800566 polymorphism and treatment response.</p><p><strong>Conclusion: </strong>Polymorphisms in ALDH1A1 (rs13959) and NQO1 (rs1800566) may be useful in predicting the probability of treatment response and adverse effects from CEF or CAF-based chemotherapy in breast cancer patients.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619309","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
Effect of midostaurin on the pharmacokinetics of P-gp, BCRP, and CYP2D6 substrates: assessing potential drug-drug interactions in healthy participants : Brief title: Drug-drug interaction of midostaurin. 米哚妥林对P-gp、BCRP和CYP2D6底物药代动力学的影响:评估健康参与者体内潜在的药物相互作用 :简短标题:米哚妥林的药物相互作用。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-07 DOI: 10.1007/s00280-024-04683-3
Romain Sechaud, Helen Gu, Gholamreza Rahmanzadeh, Ovidiu Chiparus, Astrid Breitschaft, Hans D Menssen

Purpose: Midostaurin, approved for FLT3-mutated acute myeloid leukemia and advanced systemic mastocytosis, is mainly metabolized by cytochrome P450 (CYP) 3A4. Midostaurin exhibited potential inhibitory effects on P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion-transporting polyprotein 1B1, and CYP2D6 in in vitro studies. This study investigated the pharmacokinetic (PK) effects of midostaurin on P-gp (digoxin), BCRP (rosuvastatin) and CYP2D6 (dextromethorphan) substrates in healthy adults.

Methods: This was an open-label, single-sequence, phase I clinical study evaluating the effect of single-dose midostaurin (100 mg) on the PK of digoxin and rosuvastatin (Arm 1), and dextromethorphan (Arm 2). Participants were followed up for safety 30 days after last dose. In addition, the effect of midostaurin on the PK of dextromethorphan metabolite (dextrorphan) was assessed in participants with functional CYP2D6 genes in Arm 2.

Results: The effect of midostaurin on digoxin was minor and resulted in total exposure (AUC) and peak plasma concentration (Cmax) that were only 20% higher. The effect on rosuvastatin was mild and led to an increase in AUCs of approximately 37-48% and of 100% in Cmax. There was no increase in the primary PK parameters (AUCs and Cmax) of dextromethorphan in the presence of midostaurin. The study treatments were very well tolerated with no occurance of severe adverse events (AEs), AEs of grade ≥ 2, or deaths.

Conclusion: Midostaurin showed only a minor inhibitory effect on P-gp, a mild inhibitory effect on BCRP, and no inhibitory effect on CYP2D6. Study treatments were well tolerated in healthy adults.

目的:米多司林被批准用于治疗FLT3突变的急性髓性白血病和晚期系统性肥大细胞增多症,它主要通过细胞色素P450(CYP)3A4代谢。在体外研究中,米哚妥林对 P-糖蛋白(P-gp)、乳腺癌抗性蛋白(BCRP)、有机阴离子转运多聚蛋白 1B1 和 CYP2D6 具有潜在的抑制作用。本研究调查了米哚妥林在健康成人中对 P-gp(地高辛)、BCRP(洛伐他汀)和 CYP2D6(右美沙芬)底物的药代动力学(PK)影响:这是一项开放标签、单序列、I期临床研究,评估单剂量米哚妥林(100毫克)对地高辛、洛伐他汀(试验组1)和右美沙芬(试验组2)PK的影响。在最后一次给药后 30 天对参与者进行安全性随访。此外,还评估了米哚妥林对右美沙芬代谢物(右美沙芬)PK的影响:结果:米多司他林对地高辛的影响较小,导致总暴露量(AUC)和血浆峰浓度(Cmax)仅高出20%。对罗伐他汀的影响轻微,导致AUCs增加约37-48%,Cmax增加100%。米哚妥林存在时,右美沙芬的主要 PK 参数(AUCs 和 Cmax)没有增加。研究治疗的耐受性非常好,没有发生严重不良事件(AE)、AE≥2级或死亡:结论:米多司林对P-gp只有轻微的抑制作用,对BCRP有轻微的抑制作用,对CYP2D6没有抑制作用。健康成人对研究治疗的耐受性良好。
{"title":"Effect of midostaurin on the pharmacokinetics of P-gp, BCRP, and CYP2D6 substrates: assessing potential drug-drug interactions in healthy participants : Brief title: Drug-drug interaction of midostaurin.","authors":"Romain Sechaud, Helen Gu, Gholamreza Rahmanzadeh, Ovidiu Chiparus, Astrid Breitschaft, Hans D Menssen","doi":"10.1007/s00280-024-04683-3","DOIUrl":"10.1007/s00280-024-04683-3","url":null,"abstract":"<p><strong>Purpose: </strong>Midostaurin, approved for FLT3-mutated acute myeloid leukemia and advanced systemic mastocytosis, is mainly metabolized by cytochrome P450 (CYP) 3A4. Midostaurin exhibited potential inhibitory effects on P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion-transporting polyprotein 1B1, and CYP2D6 in in vitro studies. This study investigated the pharmacokinetic (PK) effects of midostaurin on P-gp (digoxin), BCRP (rosuvastatin) and CYP2D6 (dextromethorphan) substrates in healthy adults.</p><p><strong>Methods: </strong>This was an open-label, single-sequence, phase I clinical study evaluating the effect of single-dose midostaurin (100 mg) on the PK of digoxin and rosuvastatin (Arm 1), and dextromethorphan (Arm 2). Participants were followed up for safety 30 days after last dose. In addition, the effect of midostaurin on the PK of dextromethorphan metabolite (dextrorphan) was assessed in participants with functional CYP2D6 genes in Arm 2.</p><p><strong>Results: </strong>The effect of midostaurin on digoxin was minor and resulted in total exposure (AUC) and peak plasma concentration (C<sub>max</sub>) that were only 20% higher. The effect on rosuvastatin was mild and led to an increase in AUCs of approximately 37-48% and of 100% in C<sub>max</sub>. There was no increase in the primary PK parameters (AUCs and C<sub>max</sub>) of dextromethorphan in the presence of midostaurin. The study treatments were very well tolerated with no occurance of severe adverse events (AEs), AEs of grade ≥ 2, or deaths.</p><p><strong>Conclusion: </strong>Midostaurin showed only a minor inhibitory effect on P-gp, a mild inhibitory effect on BCRP, and no inhibitory effect on CYP2D6. Study treatments were well tolerated in healthy adults.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896856","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
Phase I-II study of OBI-888, a humanized monoclonal IgG1 antibody against the tumor-associated carbohydrate antigen Globo H, in patients with advanced solid tumors. 针对肿瘤相关碳水化合物抗原 Globo H 的人源化单克隆 IgG1 抗体 OBI-888 在晚期实体瘤患者中的 I-II 期研究。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-23 DOI: 10.1007/s00280-024-04714-z
Apostolia Maria Tsimberidou, Axel Grothey, Darren Sigal, Heinz-Josef Lenz, Howard S Hochster, Yee Chao, Li-Yuan Bai, Chia-Jui L Yen, Dong Xu, M Wayne Saville

Purpose: OBI-888 is a humanized, monoclonal IgG1 antibody specific to the tumor-associated carbohydrate antigen Globo H. We conducted a phase I-II study of OBI-888 in patients with advanced cancer.

Methods: Patients were treated with OBI-888 5, 10, or 20 mg/kg IV weekly in Part A ("3 + 3" design) and 20 mg/kg IV weekly in Part B (Simon's 2-stage design) (1 cycle = 28 days).

Results: Overall, 54 patients were treated (Part A, n = 14; Part B, n = 40). OBI-888 was safe and well tolerated across the doses studied, with a low incidence of OBI-888-related treatment emergent adverse events. The maximum tolerated dose of OBI-888 was not reached. No dose-limiting toxicities were noted up to the 20 mg/kg dose level (recommended phase 2 dose). Stable disease (SD) was noted in 28.6% and 20% of Parts A and B, respectively, including three patients with SD for 6+, 7+, and 9 months. Antibody-dependent cellular cytotoxicity (ADCC) was induced after each OBI-888 treatment (average increase, 3.8-fold and 4.7-fold in Parts A and B, respectively), suggesting that ADCC induction is a potential mechanism of action of OBI-888.

Conclusions: OBI-888 was well tolerated. Prolonged SD was noted in three patients. ADCC was induced after each OBI-888 treatment.

目的:OBI-888是一种特异于肿瘤相关碳水化合物抗原Globo H的人源化单克隆IgG1抗体:方法:在A部分("3 + 3 "设计)中,患者每周接受5、10或20毫克/千克的OBI-888静脉注射治疗;在B部分(西蒙两阶段设计)中,患者每周接受20毫克/千克的OBI-888静脉注射治疗(1个周期=28天):共有54名患者接受了治疗(A部分,14人;B部分,40人)。在所有研究剂量中,OBI-888均安全且耐受性良好,与OBI-888相关的治疗突发不良事件发生率较低。未达到OBI-888的最大耐受剂量。在20毫克/千克剂量水平(第二阶段推荐剂量)以下,未发现剂量限制性毒性反应。A部分和B部分分别有28.6%和20%的患者病情稳定(SD),其中有3名患者病情稳定时间分别为6个月、7个月和9个月。每次OBI-888治疗后都会诱导抗体依赖性细胞毒性(ADCC)(A部分和B部分的平均增幅分别为3.8倍和4.7倍),这表明ADCC诱导是OBI-888的一种潜在作用机制:结论:OBI-888的耐受性良好。结论:OBI-888的耐受性良好,有3名患者的SD延长。每次OBI-888治疗后都能诱导ADCC。
{"title":"Phase I-II study of OBI-888, a humanized monoclonal IgG1 antibody against the tumor-associated carbohydrate antigen Globo H, in patients with advanced solid tumors.","authors":"Apostolia Maria Tsimberidou, Axel Grothey, Darren Sigal, Heinz-Josef Lenz, Howard S Hochster, Yee Chao, Li-Yuan Bai, Chia-Jui L Yen, Dong Xu, M Wayne Saville","doi":"10.1007/s00280-024-04714-z","DOIUrl":"https://doi.org/10.1007/s00280-024-04714-z","url":null,"abstract":"<p><strong>Purpose: </strong>OBI-888 is a humanized, monoclonal IgG1 antibody specific to the tumor-associated carbohydrate antigen Globo H. We conducted a phase I-II study of OBI-888 in patients with advanced cancer.</p><p><strong>Methods: </strong>Patients were treated with OBI-888 5, 10, or 20 mg/kg IV weekly in Part A (\"3 + 3\" design) and 20 mg/kg IV weekly in Part B (Simon's 2-stage design) (1 cycle = 28 days).</p><p><strong>Results: </strong>Overall, 54 patients were treated (Part A, n = 14; Part B, n = 40). OBI-888 was safe and well tolerated across the doses studied, with a low incidence of OBI-888-related treatment emergent adverse events. The maximum tolerated dose of OBI-888 was not reached. No dose-limiting toxicities were noted up to the 20 mg/kg dose level (recommended phase 2 dose). Stable disease (SD) was noted in 28.6% and 20% of Parts A and B, respectively, including three patients with SD for 6+, 7+, and 9 months. Antibody-dependent cellular cytotoxicity (ADCC) was induced after each OBI-888 treatment (average increase, 3.8-fold and 4.7-fold in Parts A and B, respectively), suggesting that ADCC induction is a potential mechanism of action of OBI-888.</p><p><strong>Conclusions: </strong>OBI-888 was well tolerated. Prolonged SD was noted in three patients. ADCC was induced after each OBI-888 treatment.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280630","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
Hypoalbuminemia in children with acute lymphoblastic leukemia: relation to asparaginase therapy and impact on high dose methotrexate elimination. 急性淋巴细胞白血病患儿的低白蛋白血症:与天冬酰胺酶疗法的关系以及对大剂量甲氨蝶呤消除的影响。
IF 2.7 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-21 DOI: 10.1007/s00280-024-04713-0
Sophie Rex Christensen, Christina Friis Jensen, Jesper Heldrup, Zachary Taylor, Laura B Ramsey, Steen Rosthøj

Purpose: High-dose methotrexate (HDMTX) therapy is an important component in treatment regimens for acute lymphoblastic leukemia (ALL). Courses are associated with a risk of renal injury, delayed elimination, and increased systemic toxicity. Recently hypoalbuminemia has been recognized as yet another risk factor.

Methods: To examine the impact of serum albumin we reviewed 325 HDMTX 5 g/m2 courses in a cohort of 51 children treated on the NOPHO ALL 2008 protocol, dividing the courses into four groups with different levels of baseline albumin (A < 25 g/L, B 25-29 g/L, C 30-34 g/L and D ≥ 35 g/L).

Results: Hypoalbuminemia was present in 51% of the courses, mostly in the early phases of chemotherapy while asparaginase therapy is ongoing, and especially if given less than 2 weeks after a dose (78%). Hypoalbuminemia had a significant impact on the end-of-infusion serum MTX, depending on the degree of hypoalbuminemia: MTX > 150 µM was seen in 37%, 32%, 20% and 8% in groups A to D. Serum albumin < 30 g/L was significantly associated with low MTX clearance < 10 L/h/1.73m2 (78% vs. 36%) and high AUC ≥ 1000 µM*h (44% vs. 31%). The frequency of rising creatinine or prolonged elimination was not increased, but the risk of stomatitis was significantly higher (42% vs. 19%).

Conclusion: Low serum albumin is caused by concurrent asparaginase therapy and has a clinically significant impact on MTX disposition. Guidelines for administering HDMTX may need adjustment if serum albumin < 30 g/L, and, if possible, HDMTX courses should not be scheduled soon after asparaginase doses.

目的:大剂量甲氨蝶呤(HDMTX)疗法是急性淋巴细胞白血病(ALL)治疗方案的重要组成部分。疗程与肾损伤风险、消除延迟和全身毒性增加有关。最近,低白蛋白血症被认为是另一个风险因素:为了研究血清白蛋白的影响,我们回顾了根据 NOPHO ALL 2008 方案接受治疗的 51 名儿童中的 325 个 HDMTX 5 g/m2 疗程,并将这些疗程分为四组,每组的基线白蛋白水平不同(A 结果:51%的疗程存在低白蛋白血症,主要发生在天冬酰胺酶治疗进行中的化疗早期,尤其是在用药后不到两周的情况下(78%)。低白蛋白血症对输注末血清 MTX 有显著影响,具体取决于低白蛋白血症的程度:在 A 组至 D 组中,MTX > 150 µM 的比例分别为 37%、32%、20% 和 8%:低血清白蛋白是由同时接受天冬酰胺酶治疗引起的,对 MTX 的处置有显著的临床影响。如果血清白蛋白偏低,可能需要调整 HDMTX 的用药指南。
{"title":"Hypoalbuminemia in children with acute lymphoblastic leukemia: relation to asparaginase therapy and impact on high dose methotrexate elimination.","authors":"Sophie Rex Christensen, Christina Friis Jensen, Jesper Heldrup, Zachary Taylor, Laura B Ramsey, Steen Rosthøj","doi":"10.1007/s00280-024-04713-0","DOIUrl":"https://doi.org/10.1007/s00280-024-04713-0","url":null,"abstract":"<p><strong>Purpose: </strong>High-dose methotrexate (HDMTX) therapy is an important component in treatment regimens for acute lymphoblastic leukemia (ALL). Courses are associated with a risk of renal injury, delayed elimination, and increased systemic toxicity. Recently hypoalbuminemia has been recognized as yet another risk factor.</p><p><strong>Methods: </strong>To examine the impact of serum albumin we reviewed 325 HDMTX 5 g/m2 courses in a cohort of 51 children treated on the NOPHO ALL 2008 protocol, dividing the courses into four groups with different levels of baseline albumin (A < 25 g/L, B 25-29 g/L, C 30-34 g/L and D ≥ 35 g/L).</p><p><strong>Results: </strong>Hypoalbuminemia was present in 51% of the courses, mostly in the early phases of chemotherapy while asparaginase therapy is ongoing, and especially if given less than 2 weeks after a dose (78%). Hypoalbuminemia had a significant impact on the end-of-infusion serum MTX, depending on the degree of hypoalbuminemia: MTX > 150 µM was seen in 37%, 32%, 20% and 8% in groups A to D. Serum albumin < 30 g/L was significantly associated with low MTX clearance < 10 L/h/1.73m2 (78% vs. 36%) and high AUC ≥ 1000 µM*h (44% vs. 31%). The frequency of rising creatinine or prolonged elimination was not increased, but the risk of stomatitis was significantly higher (42% vs. 19%).</p><p><strong>Conclusion: </strong>Low serum albumin is caused by concurrent asparaginase therapy and has a clinically significant impact on MTX disposition. Guidelines for administering HDMTX may need adjustment if serum albumin < 30 g/L, and, if possible, HDMTX courses should not be scheduled soon after asparaginase doses.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280629","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
Combination of S-1 and the oral ATR inhibitor ceralasertib is effective against pancreatic cancer cells S-1 和口服 ATR 抑制剂 ceralasertib 的组合能有效对抗胰腺癌细胞
IF 3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-14 DOI: 10.1007/s00280-024-04716-x
Yoshihito Morimoto, Kimihiko Takada, Ami Nakano, Osamu Takeuchi, Kazuhiro Watanabe, Masayoshi Hirohara, Yutaka Masuda

Purpose

In our previous study, we found that the Chk1 inhibitor prexasertib enhances the antitumour effect of the oral anticancer drug S-1 against pancreatic cancer cells. In this study, we investigated the effect of combining S-1 and ceralasertib, an oral inhibitor of ATR, which is located upstream of Chk1. Ceralasertib is currently being investigated in multiple clinical trials for various cancers.

Methods

The cell-proliferation inhibitory effect was measured by MTT assay, using the pancreatic cancer cell lines BxPC-3, SUIT-2, PANC-1, and MIA PaCa-2, while apoptosis was measured by flow cytometry using PI/Annexin staining. The mechanism underlying the combined effect was analysed using western blotting, and the antitumor effect was analysed using a mouse xenograft model.

Results

MTT assay revealed that the combination of S-1 and ceralasertib had a synergistic effect, leading to the suppression of cell proliferation. Measurement with PI/Annexin staining revealed that the combination of S-1 and ceralasertib induced apoptosis more efficiently than either drug alone. Western blotting results showed that ceralasertib inhibited S-1-induced activation of ATR and Chk1. The average estimated tumour volume after 3 weeks of administration was 601 mm3 in the S-1 group, 580 mm3 in the ceralasertib group, and 298 mm3 in the combination group.

Conclusion

The combination of S-1 and ceralasertib demonstrated a high antiproliferative effect in inhibiting tumour growth in vitro.

目的 在之前的研究中,我们发现 Chk1 抑制剂 prexasertib 能增强口服抗癌药物 S-1 对胰腺癌细胞的抗肿瘤作用。在本研究中,我们探讨了 S-1 与位于 Chk1 上游的 ATR 口服抑制剂 ceralasertib 联用的效果。Ceralasertib目前正在多项针对各种癌症的临床试验中进行研究。方法采用MTT法测定S-1和Ceralasertib对胰腺癌细胞株BxPC-3、SUIT-2、PANC-1和MIA PaCa-2的细胞增殖抑制作用,同时采用PI/Annexin染色法用流式细胞仪测定细胞凋亡。结果MTT试验显示,S-1和ceralasertib的组合具有协同作用,可抑制细胞增殖。用 PI/Annexin 染色法测量发现,S-1 和 ceralasertib 联用比单独使用其中一种药物更有效地诱导细胞凋亡。Western 印迹结果显示,ceralasertib 可抑制 S-1 诱导的 ATR 和 Chk1 激活。给药 3 周后,S-1 组的平均估计肿瘤体积为 601 立方毫米,ceralasertib 组为 580 立方毫米,联合用药组为 298 立方毫米。
{"title":"Combination of S-1 and the oral ATR inhibitor ceralasertib is effective against pancreatic cancer cells","authors":"Yoshihito Morimoto, Kimihiko Takada, Ami Nakano, Osamu Takeuchi, Kazuhiro Watanabe, Masayoshi Hirohara, Yutaka Masuda","doi":"10.1007/s00280-024-04716-x","DOIUrl":"https://doi.org/10.1007/s00280-024-04716-x","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Purpose</h3><p>In our previous study, we found that the Chk1 inhibitor prexasertib enhances the antitumour effect of the oral anticancer drug S-1 against pancreatic cancer cells. In this study, we investigated the effect of combining S-1 and ceralasertib, an oral inhibitor of ATR, which is located upstream of Chk1. Ceralasertib is currently being investigated in multiple clinical trials for various cancers.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>The cell-proliferation inhibitory effect was measured by MTT assay, using the pancreatic cancer cell lines BxPC-3, SUIT-2, PANC-1, and MIA PaCa-2, while apoptosis was measured by flow cytometry using PI/Annexin staining. The mechanism underlying the combined effect was analysed using western blotting, and the antitumor effect was analysed using a mouse xenograft model.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>MTT assay revealed that the combination of S-1 and ceralasertib had a synergistic effect, leading to the suppression of cell proliferation. Measurement with PI/Annexin staining revealed that the combination of S-1 and ceralasertib induced apoptosis more efficiently than either drug alone. Western blotting results showed that ceralasertib inhibited S-1-induced activation of ATR and Chk1. The average estimated tumour volume after 3 weeks of administration was 601 mm<sup>3</sup> in the S-1 group, 580 mm<sup>3</sup> in the ceralasertib group, and 298 mm<sup>3</sup> in the combination group.</p><h3 data-test=\"abstract-sub-heading\">Conclusion</h3><p>The combination of S-1 and ceralasertib demonstrated a high antiproliferative effect in inhibiting tumour growth in vitro.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268980","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
Assessing pharmacokinetics and drug-drug interactions of the combination therapy of myelofibrosis with ruxolitinib and lenalidomide by a new eco-friendly HPLC method for their simultaneous determination in plasma 用一种新型环保高效液相色谱法同时测定血浆中的芦可利替尼和来那度胺,评估芦可利替尼和来那度胺联合治疗骨髓纤维化的药代动力学和药物相互作用
IF 3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-11 DOI: 10.1007/s00280-024-04715-y
Rashed N. Herqash, Fai A. Alkathiri, Ibrahim A. Darwish

Ruxolitinib (RUX), a Janus kinase 2 (JAK2) inhibitor, and lenalidomide (LEN), an immunomodulatory agent, have recently been proposed as a combined treatment for myelofibrosis (MF). This combination has demonstrated improved efficacy, safety, and tolerability compared to monotherapy. To further refine these findings, an efficient analytical tool is needed to simultaneously determine RUX and LEN concentrations in blood plasma. This tool would enable the study of their pharmacokinetics, drug-drug interactions, and therapeutic monitoring during MF therapy. Unfortunately, such a method has not been existed in the literature. This study presents the first HPLC method with UV detection for the simultaneous quantitation of RUX and LEN in plasma. The method was validated according to the ICH guidelines for bioanalytical method validation. It exhibited linearity in the concentration ranges of 10 to 3150 ng mL− 1 for RUX and 80 to 5200 ng mL− 1 for LEN. The limits of quantitation were determined to be 25 and 90 ng mL− 1 for RUX and LEN, respectively. All other validation parameters were satisfactory. The HPLC-UV method was successfully employed to study the pharmacokinetics and drug-drug interactions of RUX and LEN in rats following oral administration of single doses. The results demonstrated that the pharmacokinetics of both drugs were changed substantially by their coadministration. LEN exhibited synergistic effects on the maximum plasma concentration (Cmax) and total bioavailability of RUX, meanwhile it exhibited diminishing effect on the values of volume of distribution (Vd) and clearance (CL). Additionally, RUX decreased the Cmax and total bioavailability of LEN, meanwhile it increased its Vd and CL. These data suggest that the use of RUX, as a combination with LEN, is a better therapeutic approach for MF, compared with RUX as a monotherapy. The effects of LEN on the pharmacokinetics of RUX should be considered and can be useful in determining the appropriate RUX dosage and dosing regimen to achieve the desired therapeutic effect when used as a combination therapy with LEN. The method’s environmental friendliness was confirmed through three comprehensive tools. This method represents a valuable tool for determining the appropriate dosage and dosing regimen of RUX in combination therapy with LEN to achieve the desired therapeutic effect. Furthermore, it can aid in predicting drug distribution in different patients and assessing the drug accumulation or insufficient drug levels in specific body compartments.

最近,有人提出将Janus激酶2(JAK2)抑制剂鲁索利替尼(Ruxolitinib,RUX)和免疫调节剂来那度胺(Lenalidomide,LEN)作为骨髓纤维化(MF)的联合疗法。与单一疗法相比,这种联合疗法在疗效、安全性和耐受性方面均有改善。为了进一步完善这些研究结果,需要一种高效的分析工具来同时测定血浆中 RUX 和 LEN 的浓度。这种工具将有助于研究它们的药代动力学、药物间相互作用以及中频治疗期间的治疗监测。遗憾的是,文献中还没有这种方法。本研究首次采用高效液相色谱法和紫外检测法同时定量检测血浆中的 RUX 和 LEN。该方法根据 ICH 生物分析方法验证指南进行了验证。该方法在 RUX 10 至 3150 ng mL- 1 和 LEN 80 至 5200 ng mL- 1 的浓度范围内呈线性关系。RUX 和 LEN 的定量限分别为 25 和 90 ng mL-1。所有其他验证参数均令人满意。采用HPLC-UV方法成功地研究了大鼠口服单剂量RUX和LEN的药代动力学和药物间相互作用。结果表明,两种药物的药代动力学因同时给药而发生了很大变化。LEN 对 RUX 的最大血浆浓度(Cmax)和总生物利用度有协同作用,同时对分布容积(Vd)和清除率(CL)有降低作用。此外,RUX 降低了 LEN 的 Cmax 和总生物利用度,同时增加了其 Vd 和 CL。这些数据表明,与 RUX 作为单一疗法相比,RUX 与 LEN 联用是治疗 MF 的更好方法。在与 LEN 联合使用时,应考虑 LEN 对 RUX 药代动力学的影响,这有助于确定适当的 RUX 剂量和给药方案,以达到预期的治疗效果。该方法的环保性通过三种综合工具得到了证实。该方法是一种有价值的工具,可用于确定 RUX 与 LEN 联合治疗时的适当剂量和给药方案,以达到预期的治疗效果。此外,它还有助于预测药物在不同患者体内的分布情况,并评估药物在特定体腔中的蓄积或不足。
{"title":"Assessing pharmacokinetics and drug-drug interactions of the combination therapy of myelofibrosis with ruxolitinib and lenalidomide by a new eco-friendly HPLC method for their simultaneous determination in plasma","authors":"Rashed N. Herqash, Fai A. Alkathiri, Ibrahim A. Darwish","doi":"10.1007/s00280-024-04715-y","DOIUrl":"https://doi.org/10.1007/s00280-024-04715-y","url":null,"abstract":"<p>Ruxolitinib (RUX), a Janus kinase 2 (JAK2) inhibitor, and lenalidomide (LEN), an immunomodulatory agent, have recently been proposed as a combined treatment for myelofibrosis (MF). This combination has demonstrated improved efficacy, safety, and tolerability compared to monotherapy. To further refine these findings, an efficient analytical tool is needed to simultaneously determine RUX and LEN concentrations in blood plasma. This tool would enable the study of their pharmacokinetics, drug-drug interactions, and therapeutic monitoring during MF therapy. Unfortunately, such a method has not been existed in the literature. This study presents the first HPLC method with UV detection for the simultaneous quantitation of RUX and LEN in plasma. The method was validated according to the ICH guidelines for bioanalytical method validation. It exhibited linearity in the concentration ranges of 10 to 3150 ng mL<sup>− 1</sup> for RUX and 80 to 5200 ng mL<sup>− 1</sup> for LEN. The limits of quantitation were determined to be 25 and 90 ng mL<sup>− 1</sup> for RUX and LEN, respectively. All other validation parameters were satisfactory. The HPLC-UV method was successfully employed to study the pharmacokinetics and drug-drug interactions of RUX and LEN in rats following oral administration of single doses. The results demonstrated that the pharmacokinetics of both drugs were changed substantially by their coadministration. LEN exhibited synergistic effects on the maximum plasma concentration (C<sub>max</sub>) and total bioavailability of RUX, meanwhile it exhibited diminishing effect on the values of volume of distribution (Vd) and clearance (CL). Additionally, RUX decreased the C<sub>max</sub> and total bioavailability of LEN, meanwhile it increased its Vd and CL. These data suggest that the use of RUX, as a combination with LEN, is a better therapeutic approach for MF, compared with RUX as a monotherapy. The effects of LEN on the pharmacokinetics of RUX should be considered and can be useful in determining the appropriate RUX dosage and dosing regimen to achieve the desired therapeutic effect when used as a combination therapy with LEN. The method’s environmental friendliness was confirmed through three comprehensive tools. This method represents a valuable tool for determining the appropriate dosage and dosing regimen of RUX in combination therapy with LEN to achieve the desired therapeutic effect. Furthermore, it can aid in predicting drug distribution in different patients and assessing the drug accumulation or insufficient drug levels in specific body compartments.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142190754","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
期刊
Cancer Chemotherapy and Pharmacology
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