1.Tyro3 and CDK9 as biomarkers for drug resistance to breast cancer anti-PD-1 therapies.
Chinese Journal of Oncology 2023;45(8):651-656
		                        		
		                        			
		                        			Objective: PD-1/PD-L1 immune checkpoint treatment is effective for some triple-negative breast cancer populations with PD-L1 expression, but the response rate is still not satisfactory. This study aims to explore the mechanism of drug resistance to breast cancer anti-PD-1 therapies and the strategies for overcoming the resistance to PD-1therapies. Methods: By constructing a human triple-negative breast cancer drug-resistant cell line called BT-549R5 and a mouse breast cancer drug-resistant cell line called 4T1R3, and applying the whole-gene shRNA library screening, candidate drug resistance-associated molecules were obtained and verified by cytological experiments. The expression of Tyro3, Axl and MerTK of the TAM family in the 4T1R3 group was tested using the Western blot method. The down-regulation of CDK9 on the effect of T cells killing the BT-549R5 cells was observed through T cell killing tests, while the down-regulation of Tyro3 and CDK9 on the effect of anti-PD-1 therapies for transplanted breast tumors was observed in mouse tumor formation experiments. Results: The cell lines and animal models of breast cancer resistant to PD-1 treatment were successfully constructed. Tyro3, Axl and MerTK were highly expressed in 4T1R3 cells. Whole genome sequencing showed that Tyro3 and CDK9 were highly expressed in BT-549R5 cells. T cell killing experiment showed that the survival rate of BT-549R5 cells in the CDK9 down-regulated group and the control group decreased gradually with the increase of T cells, but the survival rate of BT-549R5 cells in the CDK9 down-regulated group decreased rapidly. Tumor formation experiment in mice showed that under anti-PD-1 treatment, the transplanted tumor in the 4T1R3 cell group grew rapidly compared with the 4T1 cell group (P<0.05), and the tumor volume of the 4T1R3 group was larger than that of the 4T1 group on Day 20. Nevertheless, the tumor growth rates in the CDK9-knockdown 4T1R3 cell group and the Tyro3-knockdown 4T1R3 cell group were similar to that of the 4T1 cell group, and the tumor volumes at day 20 were signiference lower than that of 4T1R3 cell group(P<0.05). Conclusions: Tyro3 and CDK9 are associated with the drug resistance to anti-PD-1 therapies for breast cancer. Inhibiting the expression of Tyro3 and CDK9 can reverse the drug resistance to breast cancer treatment.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			c-Mer Tyrosine Kinase/metabolism*
		                        			;
		                        		
		                        			Receptor Protein-Tyrosine Kinases/genetics*
		                        			;
		                        		
		                        			Axl Receptor Tyrosine Kinase
		                        			;
		                        		
		                        			Proto-Oncogene Proteins/metabolism*
		                        			;
		                        		
		                        			B7-H1 Antigen/genetics*
		                        			;
		                        		
		                        			Triple Negative Breast Neoplasms/genetics*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Biomarkers
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase 9
		                        			
		                        		
		                        	
2.Tyro3 and CDK9 as biomarkers for drug resistance to breast cancer anti-PD-1 therapies.
Chinese Journal of Oncology 2023;45(8):651-656
		                        		
		                        			
		                        			Objective: PD-1/PD-L1 immune checkpoint treatment is effective for some triple-negative breast cancer populations with PD-L1 expression, but the response rate is still not satisfactory. This study aims to explore the mechanism of drug resistance to breast cancer anti-PD-1 therapies and the strategies for overcoming the resistance to PD-1therapies. Methods: By constructing a human triple-negative breast cancer drug-resistant cell line called BT-549R5 and a mouse breast cancer drug-resistant cell line called 4T1R3, and applying the whole-gene shRNA library screening, candidate drug resistance-associated molecules were obtained and verified by cytological experiments. The expression of Tyro3, Axl and MerTK of the TAM family in the 4T1R3 group was tested using the Western blot method. The down-regulation of CDK9 on the effect of T cells killing the BT-549R5 cells was observed through T cell killing tests, while the down-regulation of Tyro3 and CDK9 on the effect of anti-PD-1 therapies for transplanted breast tumors was observed in mouse tumor formation experiments. Results: The cell lines and animal models of breast cancer resistant to PD-1 treatment were successfully constructed. Tyro3, Axl and MerTK were highly expressed in 4T1R3 cells. Whole genome sequencing showed that Tyro3 and CDK9 were highly expressed in BT-549R5 cells. T cell killing experiment showed that the survival rate of BT-549R5 cells in the CDK9 down-regulated group and the control group decreased gradually with the increase of T cells, but the survival rate of BT-549R5 cells in the CDK9 down-regulated group decreased rapidly. Tumor formation experiment in mice showed that under anti-PD-1 treatment, the transplanted tumor in the 4T1R3 cell group grew rapidly compared with the 4T1 cell group (P<0.05), and the tumor volume of the 4T1R3 group was larger than that of the 4T1 group on Day 20. Nevertheless, the tumor growth rates in the CDK9-knockdown 4T1R3 cell group and the Tyro3-knockdown 4T1R3 cell group were similar to that of the 4T1 cell group, and the tumor volumes at day 20 were signiference lower than that of 4T1R3 cell group(P<0.05). Conclusions: Tyro3 and CDK9 are associated with the drug resistance to anti-PD-1 therapies for breast cancer. Inhibiting the expression of Tyro3 and CDK9 can reverse the drug resistance to breast cancer treatment.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			c-Mer Tyrosine Kinase/metabolism*
		                        			;
		                        		
		                        			Receptor Protein-Tyrosine Kinases/genetics*
		                        			;
		                        		
		                        			Axl Receptor Tyrosine Kinase
		                        			;
		                        		
		                        			Proto-Oncogene Proteins/metabolism*
		                        			;
		                        		
		                        			B7-H1 Antigen/genetics*
		                        			;
		                        		
		                        			Triple Negative Breast Neoplasms/genetics*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Biomarkers
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase 9
		                        			
		                        		
		                        	
3.FNDC1 is highly expressed in lung adenocarcinoma and closely related with poor prognosis.
Hai Ning HONG ; Hao Nan ZHU ; Chao LI ; Chao ZANG ; Hai Wei SANG ; Li Wei CHEN ; An Sheng WANG
Journal of Southern Medical University 2022;42(8):1182-1190
		                        		
		                        			OBJECTIVE:
		                        			To explore the expression of fibronectin type Ⅲ domain containing 1(FNDC1) protein in lung adenocarcinoma and its prognostic significance.
		                        		
		                        			METHODS:
		                        			The expression of FNDC1 in lung adenocarcinoma was predicted by analysis of data from GEO database and GEPIA, and the results were verified by immunohistochemical staining in 92 pairs of clinical specimens of lung adenocarcinoma and adjacent tissues.We further analyzed the correlation of FNDC1 expression with the clinicopathological features of the patients, and evaluated its prognostic value using Cox survival analysis.
		                        		
		                        			RESULTS:
		                        			Analysis of the data form GEO database and GEPIA showed a significantly higher expression level of FNDC1 in lung adenocarcinoma than in matched normal tissues (P < 0.05).Kaplan-Meier survival analysis suggested that a high expression of FNDC1 protein was associated with a significantly shorter overall survival time of the patients (P < 0.05).Immunohistochemistry of the clinical specimens also showed a significantly higher protein expression of FNDC1 in lung adenocarcinoma tissues than in paired adjacent tissues (P < 0.001).A high expression of FNDC1 protein was significantly correlated with advanced clinical stage, T stage and N stage (P < 0.05).Cox univariate and multivariate regression survival analysis indicated that an increased expression of FNDC1 was an independent risk factor for poor prognosis of the patients with lung adenocarcinoma (P < 0.05).
		                        		
		                        			CONCLUSION
		                        			FNDC1 protein is highly expressed in patients with lung adenocarcinoma and in closely related with the occurrence, progression and prognosis of the tumor, suggesting the value of FNDC1 protein as a potential biomarker for assessment of the survival and prognosis of patients with lung adenocarcinoma.
		                        		
		                        		
		                        		
		                        			Adenocarcinoma of Lung/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Kaplan-Meier Estimate
		                        			;
		                        		
		                        			Lung Neoplasms/metabolism*
		                        			;
		                        		
		                        			Neoplasm Proteins/genetics*
		                        			;
		                        		
		                        			Prognosis
		                        			;
		                        		
		                        			Proteins
		                        			
		                        		
		                        	
4.Uptake and transport of Laportea bulbifera extract in Caco-2 cell model.
Jing HUANG ; Hong-Qin XIAO ; Ying LI ; Yi CHEN ; Si-Ying CHEN ; Yue-Ting LI ; Yong HUANG ; Lin ZHENG ; Yong-Lin WANG ; Zi-Peng GONG
China Journal of Chinese Materia Medica 2022;47(20):5617-5626
		                        		
		                        			
		                        			Laportea bulbifera extract is effective in resisting inflammation and shows a good therapeutic effect on rheumatoid arthritis in rats. However, the absorption characteristics of active components in L. bulbifera extract in Caco-2 cells are still unclear, which limits the in-depth development of L. bulbifera resources. The purpose of this study was to investigate the absorption and transport mechanism of the active components of L. bulbifera extract in the Caco-2 cell model and explore the effects of different factors(concentration, time, pH value, temperature, and efflux transporter inhibitor) on its uptake and transport. The results showed that L. bulbifera extract at the concentration of 2.0-8.0 mg·mL~(-1) showed no toxicity to Caco-2 cells. The uptake and transport of L. bulbifera extract in the Caco-2 cell model were concentration-dependent and time-dependent. The main absorption mechanism was passive diffusion, and acidic condition(pH 5.0-6.0) and 37 ℃ were more favorable for drug absorption. P_(app)>1.0×10~(-6 )cm·s~(-1) of each component indicated that L. bulbifera was a moderately absorbed drug. P-gp, MRP2, and BCRP were not involved in its uptake and transport.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Caco-2 Cells
		                        			;
		                        		
		                        			ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics*
		                        			;
		                        		
		                        			Intestinal Absorption
		                        			;
		                        		
		                        			Neoplasm Proteins/metabolism*
		                        			;
		                        		
		                        			Urticaceae
		                        			;
		                        		
		                        			Biological Transport
		                        			;
		                        		
		                        			Plant Extracts/pharmacology*
		                        			
		                        		
		                        	
5.Effect of RBP2 gene silencing on proliferation, migration and invasion of ovarian epithelial cancer SKOV3/DDP cells and its mechanism.
Tong Fu FENG ; Dong Mei YAO ; Rong ZHENG ; Duo Sheng JIANG ; Ling Li ZHANG ; Qi XING ; Li LI
Chinese Journal of Oncology 2022;44(2):139-146
		                        		
		                        			
		                        			Objective: To explore the effect of down-regulation of retinol binding protein 2 (RBP2) expression on the biological characteristics of ovarian cancer cells and its mechanism. Methods: Knockdown of RBP2 and cisplatin (DDP)-resistant ovarian cancer cell line SKOV3/DDP-RBP2i was established, the negative control group and blank control group were also set. Cell counting kit 8 (CCK-8) was used to detect the cell proliferation ability, flow cytometry was used to detect cell apoptosis, scratch test and Transwell invasion test were used to detect cell migration and invasion ability, real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) and western blot were used to detect the expressions of molecular markers related to epithelial-mesenchymal transition (EMT). The effect of RBP2 on the growth of ovarian cancer was verified through experiment of transplanted tumors in nude mice, and the relationships between RBP2 expression and tumor metastasis and patient prognosis were analyzed using the clinical data of ovarian cancer in TCGA database. Results: After down-regulating the expression of RBP2, the proliferation ability of SKOV3/DDP cell was significantly reduced. On the fifth day, the proliferation activities of SKOV3/DDP-RBP2i group, negative control group and blank control group were (56.67±4.16)%, (84.67±3.51) and (87.00±4.00)% respectively, with statistically significant difference (P<0.001). The apoptosis rate of SKOV3/DDP-RBP2i group was (14.19±1.50)%, higher than (8.77±0.75)% of the negative control group and (7.48±0.52)% of the blank control group (P<0.001). The number of invasive cells of SKOV3/DDP-RBP2i group was (55.20±2.39), lower than (82.60±5.18) and (80.80±7.26) of the negative control group and the blank control group, respectively (P<0.001). The scratch healing rate of SKOV3/DDP-RBP2i group was (28.47±2.72)%, lower than (50.58±4.06)% and (48.92±4.63)% of the negative control group and the blank control group, respectively (P<0.001). The mRNA and protein expressions of E-cadherin in the SKOV3/DDP-RBP2i group were higher than those in the negative control group (P=0.015, P<0.001) and the blank control group (P=0.006, P<0.001). The mRNA and protein expression of N-cadherin in SKOV3/DDP-RBP2i group were lower than those in the negative control group (P=0.012, P<0.001) and the blank control group (P=0.005, P<0.001). The mRNA and protein expressions of vimentin in SKOV3/DDP-RBP2i group were also lower than those in the negative control group (P=0.016, P=0.001) and the blank control group (P=0.011, P=0.001). Five weeks after the cells inoculated into the nude mice, the tumor volume of SKOV3/DDP-RBP2i group, negative control group and blank control group were statistically significant different. The tumor volume of SKOV3/DDP-RBP2i group was smaller than those of negative control group and blank control group (P=0.001). Bioinformatics analysis showed that the expression of RBP2 in patients with metastatic ovarian cancer was higher than that without metastasis (P=0.043), and the median overall survival of ovarian cancer patients with high RBP2 expression was 41 months, shorter than 69 months of low RBP2 expression patients (P<0.001). Conclusion: Downregulation of the expression of RBP2 in SKOV3/DDP cells can inhibit cell migration and invasion, and the mechanism may be related to the inhibition of EMT.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Carcinoma, Ovarian Epithelial/genetics*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Cisplatin/pharmacology*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm/genetics*
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Gene Silencing
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			Ovarian Neoplasms/pathology*
		                        			;
		                        		
		                        			Retinol-Binding Proteins, Cellular/metabolism*
		                        			
		                        		
		                        	
6.The effect of HOXC10 gene on biological behaviors of glioma cells and mechanism in tumor microenvironment.
Wen Yi JIANG ; Qing Yang LEI ; Sha Sha LIU ; Li YANG ; Bo YANG ; Yi ZHANG
Chinese Journal of Oncology 2022;44(3):228-237
		                        		
		                        			
		                        			Objective: To study the effects of Homeobox C10 (HOXC10) on biological characteristics such as migration, invasion and proliferation of glioma cancer cells and to explore the role of HOXC10 gene in glioma microenvironment. Methods: The expression level of HOXC10 in high grade glioma (glioblastoma) and low grade glioma and its effect on patient survival were analyzed by using The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) database. Hoxc10-siRNA-1, HOXC10-siRNA-2 and siRNA negative control (NC) were transfected into U251 cells according to the operation instructions of HOXC10-siRNA transfection. 100 ng/ mL recombinant protein chemokine ligand 2 (reCCL2) was added into the transfection group, and was labeled as HOXC10-siRNA-1+ reCCL2 and HOXC10-siRNA-2+ reCCL2 groups. The expressions of HOXC10 mRNA and target protein in each group was detected by real-time fluorescence quantitative polymerase chain reaction (qRT-PCR) and western blot. The proliferation ability of cells in each group was detected by cell counting kit 8 (CCK8) method. The migration ability of cells was detected by Transwell assay and Nick assay, and cell apoptosis was detected by flow cytometry. The expression of chemokines in each group was detected by multiple factors. Co-incubation assays were performed to determine the role of HOXC10 and chemokine ligand 2 (CCL2) in recruiting and polarizing tumor-associated macrophages (M2-type macrophages). Results: The median expression level of HOXC10 in high grade gliomas was 8.51, higher than 1.00 in low grade gliomas (P<0.001) in TCGA database. The median expression level of HOXC10 in high grade gliomas was 0.83, higher than 0.00 in low grade gliomas (P=0.002) in CGGA database. The 5-year survival rate of patients with high HOXC10 expression in TCGA database was 28.2%, lower than 78.7% of those with low HOXC10 expression (P<0.001), and the 5-year survival rate of patients with high HOXC10 expression in CGGA database was 20.3%, lower than 58.0% of those with low HOXC10 expression (P<0.001). The numbers of cell migration in HOXC10-siRNA-1 group and HOXC10-siRNA-2 group were (45±3) and (69±4) respectively, lower than (159±3) in NC group (P<0.05). The cell mobility of HOXC10-siRNA-1 group and HOXC10-siRNA-2 group at 48 hours were (15±2)% and (28±4)% respectively, lower than (80±5)% of NC group (P<0.05). The expressions of vimentin in HOXC10-siRNA-1 group and HOXC10-siRNA-2 group were (141 740.00±34 024.56) and (94 655.00±5 687.97), N-cadherin were (76 810.00±14.14) and (94 254.00±701.45), β-catenin were (75 786.50±789.84) and (107 296.50±9 614.53), lower than (233 768.50±34 114.37), (237 154.50±24 715.50) and (192 449.50±24 178.10) of NC group (P<0.05). The A value of HOXC10-siRNA-1 group and HOXC10-siRNA-2 group were (0.44±0.05) and (0.32±0.02) at 96 hours, lower than 0.92±0.12 of NC group (P<0.05). The apoptosis rates of HOXC10-siRNA-1 group and HOXC10 siRNA-2 group were (10.23±1.24)% and (13.81±2.16)%, higher than (4.60±0.07)% of NC group (P<0.05). The expression levels of CCL2 in U251 cells in HOXC10-siRNA-1 and HOXC10-siRNA-2 groups were (271.63±44.27) and (371.66±50.21), lower than (933.93±29.84) in NC group (P<0.05). The expression levels of CCL5 (234.81±5.95 and 232.62±5.72), CXCL10 (544.13±48.14 and 500.87±15.65) and CXCL11 (215.75±15.30 and 176.18±16.49) in HOXC10-siRNA-1 and HOXC10-siRNA-2 groups were higher than those in NC group (9.98±0.71, 470.54±18.84 and 13.55±0.73, respectively, P<0.05). The recruited numbers of CD14(+) THP1 in HOXC10-siRNA-1 and HOXC10-siRNA-2 groups were (159.33±1.15) and (170.67±1.15), respectively, lower than (360.00±7.81) in NC group (P<0.05), while addition of reCCL2 promoted the recruitment of CD14(+) THP1 cells (287.00±3.61 and 280.67±2.31 in HOXC10-siRNA-1+ reCCL2 group and HOXC10-siRNA-2+ reCCL2 group, respectively, P<0.05). The expressions level of M2-type macrophage-related gene TGF-β in HOXC10-siRNA-1 group and HOXC10-siRNA-2 group were (0.30±0.02) and (0.28±0.02), respectively, lower than (1.06±0.10) in NC group (P<0.05). The expressions level of M1-related gene NOS2 in HOXC10-siRNA-1 and HOXC10-siRNA-2 were (11 413.95±1 911.85) and (5 894.00±945.21), respectively, higher than (13.39±4.32) in NC group (P<0.05). Conclusions: The expression of HOXC10 in glioma is high and positively correlated with the poor prognosis of glioma patients. Knockdown of HOXC10 can inhibit the proliferation, migration and metastasis of human glioma U251 cells. HOXC10 may play an immunosuppressive role in glioma microenvironment by promoting the expression of CCL2 and recruiting and polarizing tumor-associated macrophages (M2 macrophages).
		                        		
		                        		
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation/genetics*
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Genes, Homeobox
		                        			;
		                        		
		                        			Glioma/pathology*
		                        			;
		                        		
		                        			Homeodomain Proteins/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Neoplasm Invasiveness/genetics*
		                        			;
		                        		
		                        			Tumor Microenvironment
		                        			
		                        		
		                        	
8.Influence of 6-shogaol potentiated on 5-fluorouracil treatment of liver cancer by promoting apoptosis and cell cycle arrest by regulating AKT/mTOR/MRP1 signalling.
Yi ZHANG ; Yong QU ; Yun-Zhong CHEN
Chinese Journal of Natural Medicines (English Ed.) 2022;20(5):352-363
		                        		
		                        			
		                        			Currently, chemoresistance seriously attenuates the curative outcome of liver cancer. The purpose of our work was to investigate the influence of 6-shogaol on the inhibition of 5-fluorouracil (5-FU) in liver cancer. The cell viability of cancer cells was determined by MTT assay. Liver cancer cell apoptosis and the cell cycle were examined utilizing flow cytometry. Moreover, qRT-PCR and western blotting was used to analyse the mRNA and protein expression levels, respectively. Immunohistochemistry assays were used to examine multidrug resistance protein 1 (MRP1) expression in tumour tissues. In liver cancer cells, we found that 6-shogaol-5-FU combination treatment inhibited cell viability, facilitated G0/G1 cell cycle arrest, and accelerated apoptosis compared with 6-shogaol or 5-FU treatment alone. In cancer cells cotreated with 6-shogaol and 5-FU, AKT/mTOR pathway- and cell cycle-related protein expression levels were inhibited, and MRP1 expression was downregulated. AKT activation or MRP1 increase reversed the influence of combination treatment on liver cancer cell viability, apoptosis and cell cycle arrest. The inhibition of AKT activation to the anticancer effect of 6-shogaol-5-FU could be reversed by MRP1 silencing. Moreover, our results showed that 6-shogaol-5-FU combination treatment notably inhibited tumour growth in vivo. In summary, our data demonstrated that 6-shogaol contributed to the curative outcome of 5-FU in liver cancer by inhibiting the AKT/mTOR/MRP1 signalling pathway.
		                        		
		                        		
		                        		
		                        			ATP Binding Cassette Transporter, Subfamily B, Member 1
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Catechols
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			Cell Cycle Checkpoints
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Fluorouracil/pharmacology*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Liver Neoplasms/genetics*
		                        			;
		                        		
		                        			Multidrug Resistance-Associated Proteins
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/metabolism*
		                        			;
		                        		
		                        			TOR Serine-Threonine Kinases/metabolism*
		                        			
		                        		
		                        	
9.A review: drug-drug interactions of epithelial growth factor receptor-tyrosine kinase inhibitors.
Chinese Journal of Oncology 2022;44(7):717-724
		                        		
		                        			
		                        			Mutations in the epithelial growth factor receptor (EGFR) is a driving factor that causes non-small cell lung carcinoma (NSCLC). The epithelial growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) is a crucial discovery in the treatment of lung cancer, particularly the efficacy of EGFR-TKIs is superior to that of the standard chemotherapy for patients with EGFR mutation-positive advanced NSCLC. Patients with NSCLC use EGFR-TKIs and other medications simultaneously is commonly seen, especially among those with comorbidities, which increases the risk of drug-drug interactions (DDIs) of EGFR-TKIs. The most common mechanisms underlying the DDIs of EGFR-TKIs are modulations of cytochrome P450 (CYP) and drug transporters [including P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP)], as well as gastrointestinal acid-inhibitory drugs [proton pump inhibitors (PPIs) and H(2) receptor antagonists (H(2)RA)]. Inhibitors or inducers of CYP enzymes and drug transporters can inhibit or accelerate the metabolism of EGFR-TKIs, which increase or reduce the exposure of EGFR-TKIs, thereby affect the efficacy and safety of EGFR-TKIs. In addition, PPIs or H(2)RA can decrease the solubility, bioavailability and efficacy of EGFR-TKIs. This review summarizes the mechanisms of DDIs of gefitinib, erlotinib, icotinib, afatinib, dacomitinib and osimertinib; the management recommendations for DDIs of those EGFR-TKIs from the Chinese and global guideline, as well as from the recent pre-clinical and clinical studies, which provide the reference and evidence for managing the combination therapies of EGFR-TKIs and other medications in clinics.
		                        		
		                        		
		                        		
		                        			ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics*
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/pathology*
		                        			;
		                        		
		                        			Drug Interactions
		                        			;
		                        		
		                        			ErbB Receptors/genetics*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Neoplasm Proteins/metabolism*
		                        			;
		                        		
		                        			Protein Kinase Inhibitors/adverse effects*
		                        			
		                        		
		                        	
10.KIF2C: a novel link between Wnt/β-catenin and mTORC1 signaling in the pathogenesis of hepatocellular carcinoma.
Shi WEI ; Miaomiao DAI ; Chi ZHANG ; Kai TENG ; Fengwei WANG ; Hongbo LI ; Weipeng SUN ; Zihao FENG ; Tiebang KANG ; Xinyuan GUAN ; Ruihua XU ; Muyan CAI ; Dan XIE
Protein & Cell 2021;12(10):788-809
		                        		
		                        			
		                        			Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is the fourth-leading cause of cancer-related deaths worldwide. HCC is refractory to many standard cancer treatments and the prognosis is often poor, highlighting a pressing need to identify biomarkers of aggressiveness and potential targets for future treatments. Kinesin family member 2C (KIF2C) is reported to be highly expressed in several human tumors. Nevertheless, the molecular mechanisms underlying the role of KIF2C in tumor development and progression have not been investigated. In this study, we found that KIF2C expression was significantly upregulated in HCC, and that KIF2C up-regulation was associated with a poor prognosis. Utilizing both gain and loss of function assays, we showed that KIF2C promoted HCC cell proliferation, migration, invasion, and metastasis both in vitro and in vivo. Mechanistically, we identified TBC1D7 as a binding partner of KIF2C, and this interaction disrupts the formation of the TSC complex, resulting in the enhancement of mammalian target of rapamycin complex1 (mTORC1) signal transduction. Additionally, we found that KIF2C is a direct target of the Wnt/β-catenin pathway, and acts as a key factor in mediating the crosstalk between Wnt/β-catenin and mTORC1 signaling. Thus, the results of our study establish a link between Wnt/β-catenin and mTORC1 signaling, which highlights the potential of KIF2C as a therapeutic target for the treatment of HCC.
		                        		
		                        		
		                        		
		                        			Adult
		                        			;
		                        		
		                        			Aged
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular/pathology*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/genetics*
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Intracellular Signaling Peptides and Proteins/metabolism*
		                        			;
		                        		
		                        			Kinesins/metabolism*
		                        			;
		                        		
		                        			Liver Neoplasms/pathology*
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred BALB C
		                        			;
		                        		
		                        			Middle Aged
		                        			;
		                        		
		                        			Neoplasm Staging
		                        			;
		                        		
		                        			Prognosis
		                        			;
		                        		
		                        			Protein Binding
		                        			;
		                        		
		                        			RNA, Small Interfering/metabolism*
		                        			;
		                        		
		                        			Survival Analysis
		                        			;
		                        		
		                        			Tumor Burden
		                        			;
		                        		
		                        			Wnt Signaling Pathway
		                        			;
		                        		
		                        			Xenograft Model Antitumor Assays
		                        			;
		                        		
		                        			beta Catenin/metabolism*
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail