Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cancer Sci ; 113(10): 3330-3346, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35792657

ABSTRACT

Colon cancer is the third most common cancer and the second leading cause of cancer-related death worldwide. Dysregulated RNA splicing factors have been reported to be associated with tumorigenesis and development in colon cancer. In this study, we interrogated clinical and RNA expression data of colon cancer patients from The Cancer Genome Atlas (TCGA) dataset and the Gene Expression Omnibus (GEO) database. Genes regulating RNA splicing correlated with survival in colon cancer were identified and a risk score model was constructed using Cox regression analyses. In the risk model, RNA splicing factor peroxisome proliferator-activated receptor-γ coactivator-1α (PPARGC1) is correlated with a good survival outcome, whereas Cdc2-like kinase 1(CLK1), CLK2, and A-kinase anchor protein 8-like (AKAP8L) with a bad survival outcome. The risk model has a good performance for clinical prognostic prediction both in the TCGA cohort and the other two validation cohorts. In the tumor microenvironment (TME) analysis, the immune score was higher in the low-risk group, and TME-related pathway gene expression was also higher in low-risk group. We further verified the mRNA and protein expression levels of these four genes in the adjacent nontumor, tumor, and liver metastasis tissues of colon cancer patients, which were consistent with bioinformatics analysis. In addition, knockdown of AKAP8L can suppress the proliferation and migration of colon cancer cells. Animal studies have also shown that AKAP8L knockdown can inhibit tumor growth in colon cancer in vivo. We established a prognostic risk model for colon cancer based on genes related to RNA splicing regulation and uncovered the role of AKAP8L in promoting colon cancer progression.


Subject(s)
Colonic Neoplasms , Gene Expression Regulation, Neoplastic , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Colonic Neoplasms/genetics , Gene Expression , Humans , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Prognosis , RNA Splicing/genetics , RNA Splicing Factors/genetics , RNA, Messenger/genetics , Tumor Microenvironment
2.
Dig Dis Sci ; 66(2): 474-482, 2021 02.
Article in English | MEDLINE | ID: mdl-32193860

ABSTRACT

BACKGROUND: Liver metastasis is an indicator of unfavorable responses to immunotherapy in colorectal cancer patients. However, the difference of immune microenvironment between primary tumors and liver metastases has not been well understood. PATIENTS AND METHODS: Fifty-four colon cancer with liver metastasis patients who received resection of both primary and metastasis lesions have been analyzed. The immune score is based on the density of infiltrating immune cells (CD3+ cell, CD8+ cell, CD11b+ cell, CD11c+ cell, and CD33+ cell) in the center and margin of the tumor. The expression of immune markers between the primary tumor and hepatic metastases was analyzed using Wilcoxon's signed rank test. RESULTS: All the five markers had higher expression in tumor margins than center tumor in both primary tumor and hepatic metastases lesions. The expression of CD11c and CD11b had no difference between metastatic lesions and primary tumor. In tumor margins, except CD11b, all the other 4 markers expressed significantly higher in hepatic metastases than in primary tumor. Intra-tumor, CD3 had higher expression in primary tumor than in hepatic metastases, while CD33 had higher expression in hepatic metastases than in primary tumor. CD8+ CD3+ cells of the total CD8+ cell population in primary tumor was significantly higher than in hepatic metastases (36.42% vs. 24.88%, p = 0.0069). CONCLUSIONS: The immune microenvironment between primary tumor and hepatic metastasis is different. More immunosuppressing cells in liver may partially explain why immunotherapy in colon cancer is less effective with liver metastatic disease.


Subject(s)
Biomarkers , Colonic Neoplasms/immunology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Tumor Microenvironment/physiology , Colonic Neoplasms/pathology , Female , Humans , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Margins of Excision , Middle Aged , Retrospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/pathology
3.
Clin Res Hepatol Gastroenterol ; 45(5): 101574, 2021 09.
Article in English | MEDLINE | ID: mdl-33272890

ABSTRACT

BACKGROUND: The mechanism of PD-L1 expression in gastric cancer patients remains unclear. microRNAs (miRs) have been reported to be crucial components of the crosstalk between tumor-immune cells and emerging evidence suggests that microRNA-375 (miR-375) is significantly downregulated in digestive system tumors, but its association with PD-L1 expression in gastric cancer remains to be determined. METHODS: The expression level of miR-375 was first investigated in human gastric cancer tissues and cell lines. Its effect on gastric cancer cell proliferation, migration, invasion, and apoptosis were evaluated in vitro via CCK8, colony formation assays, wound healing assays, transwell assays, and flow cytometry. In vivo experiments using immunodeficient BALB/c nude female mice were also performed. Luciferase reporter assays were employed to identify interactions between miR-375 and its target genes. RESULTS: Quantitative real-time PCR showed that the expression of miR-375 was negatively correlated with PD-L1 in gastric cancer tissues. The overexpression of miR-375 inhibited gastric cancer cell proliferation, migration, invasion, and the knockdown of miR-375 demonstrated opposite effects, both in vitro and in vivo. Luciferase reporter assays showed that miR-375 could bind to the 3'-UTR regions of JAK2 and an inverse association between miR-375 and JAK2/STAT3/PD-L1 expression in gastric cancer cell lines was also observed. In vivo experiments showed that miR-375-overexpression decreased the growth of xenograft tumors in immunodeficient BALB/c mice. CONCLUSIONS: miR-375 negatively regulates PD-L1 expression in gastric cancer via the JAK2/STAT3 signaling pathway and could be a promising novel therapeutic target in gastric cancer.


Subject(s)
B7-H1 Antigen , Gene Expression Regulation, Neoplastic , MicroRNAs , Stomach Neoplasms , Animals , B7-H1 Antigen/metabolism , Female , Janus Kinase 2/metabolism , Luciferases , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
4.
Biochem Pharmacol ; 183: 114318, 2021 01.
Article in English | MEDLINE | ID: mdl-33159967

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most common types of cancer in China, with poor prognosis and lack of effective targeted therapy. It has been reported that ibrutinib possesses anticancer activity in ESCC with MYC and/or ERBB2 amplification. Here we explored the synergistic antitumor effect of a novel multi-kinase inhibitor APG-2449 with ibrutinib in ESCC and clarified the mechanism of the combination effect through in vitro and in vivo experiment. We found that APG-2449 exerted antitumor effect in ESCC. APG-2449 combined with ibrutinib showed synergistic inhibition of cell viability in ESCC cell lines. APG-2449 combined with ibrutinib dramatically inhibited the proliferation and migration of ESCC cells. Furthermore, we observed that ibrutinib combined with APG-2449 could induce more cancer cells arrested in the G1/S phase and apoptosis. In terms of mechanism, ibrutinib alone could decrease the phosphorylation level of EGFR and its downstream pathway of MEK/ERK. The combination therapy of APG-2449 and ibrutinib could significantly down-regulate the phosphorylation level of MEK/ERK and AKT. In ESCC xenotransplantation models, single therapy with either ibrutinib or APG-2449 was equivalent in delaying tumor growth, while the combination therapy suppressed tumor growth more significantly. Our data strongly suggest that the combination therapy of APG-2449 and ibrutinib can provide an effective therapeutic strategy for ESCC patients, which deserved further clinical investigation.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/administration & dosage , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Focal Adhesion Kinase 1/metabolism , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Adenine/administration & dosage , Adenine/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Piperidines/chemistry , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Xenograft Model Antitumor Assays/methods
5.
Cell Death Dis ; 12(8): 772, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354046

ABSTRACT

Advanced nasopharyngeal carcinoma (NPC) has a poor prognosis, with an unfavorable response to palliative chemotherapy. Unfortunately, there are few effective therapeutic regimens. Therefore, we require novel treatment strategies with enhanced efficacy. The present study aimed to investigate the antitumor efficacy of APG-1252-M1, a dual inhibitor of BCL-2/BCL-XL, as a single agent and combined with gemcitabine. We applied various apoptotic assays and used subcutaneous transplanted NPC model to assess the in vitro and in vivo antitumor activity. Moreover, phospho-tyrosine kinase array was used to investigate the combined therapy's potential synergistic mechanism. In addition, further validation was performed using immunohistochemistry and western blotting. In vitro, we observed that APG-1252-M1 had moderate antitumor activity toward NPC cells; however, it markedly improved gemcitabine's ability to promote NPC cell apoptosis and suppress invasion, migration, and proliferation. Specifically, APG-1252 plus gemcitabine exhibited even remarkable antitumor activity in vivo. Mechanistically, the drug combination synergistically suppressed NPC by activating caspase-dependent pathways, blocking the phospho (p)-JAK-2/STAT3/MCL-1 signaling pathway, and inhibiting epithelial-mesenchymal transition. In conclusion, the results indicated that the combination of APG-1252 and gemcitabine has synergistic anticancer activities against NPC, providing a promising treatment modality for patients with NPC.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Deoxycytidine/analogs & derivatives , Janus Kinase 2/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Piperidines/pharmacology , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/pharmacology , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Humans , Models, Biological , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Piperidines/therapeutic use , Signal Transduction/drug effects , bcl-X Protein/metabolism , Gemcitabine
6.
Front Oncol ; 10: 513, 2020.
Article in English | MEDLINE | ID: mdl-32391262

ABSTRACT

Background: Increasing evidences from phase II or III trials have proved that salvage systematic therapy, including chemotherapy, target therapy, or checkpoint inhibitor therapy can prolong survival in patients who do not succeed with second line therapy, yet there are no guidelines for the optimum third-line treatments. To compare the effectiveness and safety of current third-line therapies for metastatic Gastric Cancer (mGC), we conducted this network analysis. Methods: Literature up to Sep 30, 2019 were systematically searched and analyzed by a Bayesian fixed-effect model. Results: This study included seven randomized clinical trails which involved 2,655 patients. It turns out that for overall survival, nivolumab has the highest probability to be the optimal choice for overall survival (OS). For patients with no peritoneal metastases, the network meta-analysis showed that Nivolumab (HR:0.64; 95% CI: 0.48-0.85) and Trifluridine/tipiacil (HR:0.66; 95% CI: 0.51-0.86) were associated with significantly higher improvement in OS than placebo. However, patients with peritoneal metastases could not benefit from nivolumab, ramucirumab, or Trifluridine/tipiacil, when compared with a placebo. For progression-free survival, apatinib (850 mg) was the most likely candidate, followed by ramucirumab. Statistically, Apatinib (850 mg), Trifluridine/tipiacil, and SLC had higher incidences of high-grade adverse events (AEs) than placebo. Conclusion: Our findings demonstrate that nivolumab has the best balance between acceptability and effectiveness in the third line therapy for mGC.

7.
Cancer Med ; 7(4): 1141-1150, 2018 04.
Article in English | MEDLINE | ID: mdl-29533001

ABSTRACT

Mountain of studies has showed that right-sided colon cancer (RSCC) and left-sided colon cancer (LSCC) have different clinical presentation and biologic features and should be considered as two distinct disease entities. The survival difference between RSCC and LSCC remains controversial. Using Surveillance, Epidemiology, and End Results (SEER) database, we identified colon adenocarcinoma patients from 2004 to 2013. The 5-year cause-specific survival (CSS) was our primary endpoint. All statistical analyses were performed using the Intercooled Stata 13.0. All statistical tests were two-sided. The study included 95,847 (58.72%) RSCC and 67,385 (41.28%) LSCC patients. RSCC patients were older, more often females, more Caucasian, more unmarried, more advanced T and N stage, larger tumor sizes, and more poorly differentiated tumor, while LSCC patients had more stage IV diseases. Location was an independent prognostic factor in the multivariable analysis. Compared with RSCC patients, the hazard ratio for LSCC was 0.87, 95% CI: 0.85-0.89 P < 0.001. There was no survival difference between RSCC and LSCC in the following situations: older than 68 years old, T3-4, N0, poorly differentiated, and undifferentiated diseases. We firstly reported that RSCC patients had a better prognosis than LSCC in mucinous adenocarcinoma/signet ring cell carcinoma patients. RSCC patients also had a better prognosis than LSCC in stage II disease. There is a need for further subdivisions when analyzing the survival difference between RSCC and LSCC patients. RSCC had lower mortality rate than LSCC in stage II disease and mucinous adenocarcinoma/signet ring cell carcinoma patients.


Subject(s)
Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Aged , Aged, 80 and over , Colonic Neoplasms/epidemiology , Colonic Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Proportional Hazards Models , Public Health Surveillance , SEER Program
8.
J Exp Clin Cancer Res ; 37(1): 53, 2018 Mar 12.
Article in English | MEDLINE | ID: mdl-29530056

ABSTRACT

BACKGROUND: Ovarian cancer is a deadly disease. Inhibitors of apoptosis proteins (IAPs) are key regulators of apoptosis and are frequently dysregulated in ovarian cancer. Overexpression of IAPs proteins has been correlated with tumorigenesis, treatment resistance and poor prognosis. Reinstalling functional cell death machinery by pharmacological inhibition of IAPs proteins may represent an attractive therapeutic strategy for treatment of ovarian cancer. METHODS: CCK-8 and colony formation assay was performed to examine cytotoxic activity. Apoptosis was analyzed by fluorescence microscopy, flow cytometry and TUNEL assay. Elisa assay was used to determine TNFα protein. Caspase activity assay was used for caspase activation evaluation. Immunoprecipitation and siRNA interference were carried out for functional analysis. Western blotting analysis were carried out to test protein expression. Ovarian cancer cell xenograft nude mice model was used for in vivo efficacy evaluation. RESULTS: APG-1387 demonstrated potent inhibitory effect on ovarian cancer cell growth and clonogenic cell survival. APG-1387 induced RIP1- and TNFα-dependent apoptotic cell death in ovarian cancer through downregulation of IAPs proteins and induction of caspase-8/FADD/RIP1 complex, which drives caspase-8 activation. NF-κB signaling pathway was activated upon APG-1387 treatment and RIP1 contributed to NF-κB activation. APG-1387 induced cytoprotective autophagy while triggering apoptosis in ovarian cancer cells and inhibition of autophagy enhanced APG-1387-induced apoptotic cell death. APG-1387 exhibited potent antitumor activity against established human ovarian cancer xenografts. CONCLUSIONS: Our results demonstrate that APG-1387 targets IAPs proteins to potently elicit apoptotic cell death in vitro and in vivo, and provide mechanistic and applicable rationale for future clinical evaluation of APG-1387 in ovarian cancer.

10.
Oncotarget ; 7(23): 35181-7, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27174916

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is the most common type of kidney tumor with increasing incidence. Tyrosine Kinase Inhibitors (TKIs) are considered important treatment in the management of metastatic RCC. Some previous studies demonstrated that sorafenib treatment is associated with a significantly increased risk of potentially life-threatening adverse events, like bleeding. But bleeding at the fundus site is the rarest type of hemorrhage. As for TKIs' risk of bleeding, how we distinguish the degree of bleeding and what optimal strategies should we take to manage bleeding, needs to be studied systematically. RESULTS: With a long-term exposure (17 months) to sorafenib, he experienced blurred vision in his right eye and was hospitalized. The patient's diagnosis was central retinal vein occlusion (CRVO) of the right eye. Unfortunately sorafenib was terminated. MATERIALS AND METHODS: The authors describe the first case of unilateral fundus hemorrhage induced by sorafenib. A 42-year-old man was diagnosed metastatic left RCC, with clinical stage and prognostic risk being assessed as T4N1M1 and intermediate. He received a radical left nephrectomy and retroperitoneal lymph node dissection, with taking the oral multi-targeted TKI, sorafenib (800 mg daily) from 7 months to 7 days before the surgery and 7 days after the surgery restarting again until the occurrence of fundus hemorrhage. CONCLUSIONS: In this patient, long-term exposure to sorafenib possibly has increased the risk of fundus hemorrhage. This article provides us a previously undescribed morbidity associated with sorafenib, which reminds us of understanding the risk of bleeding and how this complication might be managed systematically.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/drug therapy , Eye Hemorrhage/chemically induced , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/adverse effects , Adult , Carcinoma, Renal Cell/pathology , Fundus Oculi , Humans , Liver Neoplasms/pathology , Male , Niacinamide/adverse effects , Sorafenib
SELECTION OF CITATIONS
SEARCH DETAIL