Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Front Oncol ; 12: 905922, 2022.
Article in English | MEDLINE | ID: mdl-35795055

ABSTRACT

Background: Neoadjuvant intraperitoneal and systemic chemotherapy (NIPS) has shown promising results in gastric cancer (GC) with peritoneal metastasis. However, clinical practice experience of NIPS is still lacking in China. In this study, we investigate the efficacy and safety of NIPS in Chinese patients. Methods: Eligible patients received NIPS every 3 weeks. Gastrectomy was performed for patients who met the criteria of conversion surgery. The primary end point was 1-year overall survival (OS) rate. Secondary end points were the response rate, toxic effects, conversion surgery outcomes and median survival time (MST). Results: Sixty-seven patients were enrolled. The primary endpoint was achieved with 1-year OS rate reached 67.2% (95% CI, 56.8%-79.4%). Conversion surgery was performed in 42 patients (62.9%), and R0 resection was achieved in 23 patients (54.8%) with the MST of 31.3 months (95% CI, 24.3-38.3). And the MST was 19.3 months (95% CI, 16.4-22.2) for all patients. Toxicity and surgical complications were well-tolerated. Moreover, sex, R0 resection, pathological nodal stage and tumor regression grade (TRG) were independent prognostic factors for patients who underwent conversion surgery. Conclusion: The NIPS is effective and safe in treating GC patients with peritoneal metastasis. Male patients, patients who underwent R0 resection, patients with ypN0-1 or TRG 1 after conversion surgery are more likely to benefit from the NIPS. Clinical Trial Registration: http://www.chictr.org.cn/, identifier https://clinicaltrials.gov/ ().

2.
Future Oncol ; 18(10): 1175-1183, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35114800

ABSTRACT

Although recent advances in systemic chemotherapy have improved the clinical outcomes of gastric cancer patients with peritoneal metastasis, the peritoneum still represents a common site of treatment failure and disease recurrence. Neoadjuvant intraperitoneal-systemic chemotherapy has been acknowledged as a more aggressive treatment for gastric cancer patients with peritoneal metastasis. In this multicenter phase III randomized controlled trial, 238 patients will be randomly separated into two groups in a 2:1 ratio after laparoscopic exploration. The experimental arm will receive the proposed neoadjuvant intraperitoneal-systemic chemotherapy regimen, whereas the control group will receive a Paclitaxel + S-1 (PS) chemotherapy regimen. The endpoints for the study are overall survival, response rate, gastrectomy radicality rate, progression-free survival and adverse events.


Recent advances in technology have improved the outcomes of stomach cancer patients. However, there are still many patients who die of cancer that has spread from another part of the body. Neoadjuvant intraperitoneal­systemic chemotherapy has been acknowledged as a more aggressive treatment for stomach cancer patients with peritoneal metastasis (cancer that has spread to the very thin layer of tissue on the inside of the abdomen that covers the stomach and other organs). In this study, 238 patients will be randomly separated into two groups in a 2:1 ratio after evaluation. The experimental group will receive the proposed neoadjuvant intraperitoneal­systemic chemotherapy regimen, whereas the control group will receive a Paclitaxel + S-1 (PS) chemotherapy regimen. The endpoints for the study are how long patients live, number of patients who respond to treatment, number of patients who undergo surgery, how long patients live without their disease getting worse and problems caused by treatment. Trial registration number: ChiCTR-IIR-16009802.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoadjuvant Therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Humans , Neoplasm Staging , Paclitaxel/therapeutic use , Prospective Studies
3.
Future Oncol ; 18(39): 4239-4349, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36651765

ABSTRACT

Although gastric cancer with para-aortic lymph node (PAN) metastasis is commonly regarded as unresectable, surgeons have explored the optimal treatment for patients with PAN metastases limited to No.16a2/b1 in the past few decades. Preoperative systemic therapy combined with D2 gastrectomy plus PAN dissection may improve the prognosis of these patients. In this multicenter phase II trial, 29 gastric cancer patients with PAN metastasis limited to No.16a2/b1 will receive preoperative treatment with nab-paclitaxel, oxaliplatin, S-1 (nab-POS: nab-paclitaxel, oxaliplatin, S-1) and sintilimab followed by D2 gastrectomy plus PAN dissection; and postoperative treatment with oral S-1, intravenous sintilimab and intraperitoneal paclitaxel. The end points for the study are 3-year overall survival, 3-year disease-free survival, pathological response rate, incidence of postoperative complications and adverse events.


Stomach cancer with metastases in the para-aortic lymph nodes is usually considered inoperable. Chemotherapy combined with resection of the stomach and more extensive lymph node dissection may prolong the life of these patients. In this multicenter study, 29 stomach cancer patients with para-aortic lymph node metastases will receive preoperative treatment with nab-paclitaxel, oxaliplatin, S-1 and sintilimab, followed by resection of the stomach combined with para-aortic lymph node dissection and use of continued oral, intravenous and intraperitoneal chemotherapy. The study's end points are 3-year overall survival, 3-year disease-free survival, pathological response rate, incidence of postoperative complications and adverse events. Clinical Trial Registration: ChiCTR2200061125 (ChiCTR.org.cn).


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Lymph Node Excision , Lymphatic Metastasis/pathology , Oxaliplatin , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lymph Nodes/pathology , Gastrectomy/adverse effects , Multicenter Studies as Topic , Clinical Trials, Phase II as Topic
4.
Future Oncol ; 17(25): 3301-3307, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34008422

ABSTRACT

Although complete omentectomy is traditionally performed in patients with gastric cancer as part of radical gastrectomy to ensure the elimination of micrometastases, the prognostic value of omentectomy during gastrectomy remains unclear. Retrospective studies have shown that the incidence of metastases in the greater omentum is very low in T1-T3 gastric cancer. Thus radical gastrectomy with D2 lymphadenectomy and preservation of the greater omentum may be a proper curative treatment for gastric cancer patients with T1-T3 tumors. The aim of this article is to describe the design and rationale for this prospective, randomized controlled DRAGON-05 trial, conducted to evaluate the prognostic value of omentum-preserving gastrectomy for patients with T1-T3 gastric cancer. Clinical trial registration: ChiCTR2000040045 (ClinicalTrials.gov).


Subject(s)
Gastrectomy/methods , Neoplasm Recurrence, Local/epidemiology , Omentum/surgery , Organ Sparing Treatments/methods , Stomach Neoplasms/surgery , Adolescent , Adult , Aged , Disease-Free Survival , Female , Gastrectomy/statistics & numerical data , Humans , Lymph Node Excision , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Organ Sparing Treatments/statistics & numerical data , Prognosis , Prospective Studies , Randomized Controlled Trials as Topic , Retrospective Studies , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Rate , Young Adult
5.
Cancer Lett ; 408: 10-21, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28843497

ABSTRACT

Increasing evidence demonstrates that long noncoding RNAs (lncRNAs) regulate gene and protein expression by exerting an influence on transcriptional and post-transcriptional processes. Here, we report that the lncRNA UCA1 increases the metastatic ability of gastric cancer (GC) cells by regulating GRK2 protein stability by promoting Cbl-c-mediated GRK2 ubiquitination and degradation. This process then activates the ERK-MMP9 signalling pathway. Furthermore, we demonstrate that GRK2 is downregulated in GC cells and that silencing of GRK2 might cause similar phenotypic changes and signalling pathway activation as those induced by elevated UCA1 in GC cells. Our results suggest that UCA1 might function as a mediator of protein ubiquitination and may be a promising molecular target for GC therapy.


Subject(s)
Cell Movement , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , RNA, Long Noncoding/genetics , Stomach Neoplasms/pathology , Animals , Apoptosis , Cell Proliferation , G-Protein-Coupled Receptor Kinase 2/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Proteolysis , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 8(6): e2839, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569779

ABSTRACT

Long noncoding RNA UCA1 has emerged as a novel regulator in cancer initiation and progression of various cancers. However, function and underlying mechanism of UCA1 in the progression of gastric cancer (GC) remain unclear. In the present study, we report that UCA1 expressed highly in GC tissues and GC cells, which was partly induced by SP1. UCA1 promoted GC cell proliferation and G1/S transition in vitro and in vivo. Moreover, UCA1 exerted its function through interacting with EZH2, promoting direct interaction with cyclin D1 promoter to activate the translation of cyclin D1. Furthermore, AKT/GSK-3B/cyclin D1 axis was activated to upregulate cyclin D1 due to overexpression of UCA1. In addition, EZH2 and phosphorylated AKT induced by UCA1 could impact each other to form a positive feedback to promote cyclin D1 expression. This study demonstrated that UCA1 as a critical regulator involved in GC proliferation and cell cycle progression by promoting cyclin D1 expression, which indicates that it may be clinically a potential therapeutic target in GC.


Subject(s)
Cyclin D1/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-akt/genetics , RNA, Long Noncoding/genetics , Sp1 Transcription Factor/genetics , Stomach Neoplasms/genetics , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Feedback, Physiological , Female , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Transplantation , Prognosis , Promoter Regions, Genetic , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
7.
Oncotarget ; 7(30): 48346-48359, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27356739

ABSTRACT

Revival of dormant tumor cells may be an important tumor metastasis mechanism. We hypothesized that aurora kinase A (AURKA), a cell cycle control kinase, promotes the transition of laryngeal squamous cell carcinoma (LSCC) cells from G0 phase to active division. We therefore investigated whether AURKA could revive dormant tumor cells to promote metastasis. Western blotting revealed that AURKA expression was persistently low in dormant laryngeal cancer Hep2 (D-Hep2) cells and high in non-dormant (T-Hep2) cells. Decreasing AURKA expression in T-Hep2 cells induced dormancy and reduced FAK/PI3K/Akt pathway activity. Increasing AURKA expression in D-Hep2 cells increased FAK/PI3K/Akt pathway activity and enhanced cellular proliferation, migration, invasion and metastasis. In addition, FAK/PI3K/Akt pathway inhibition caused dormancy-like behavior and reduced cellular mobility, migration and invasion. We conclude that AURKA may revive dormant tumor cells via FAK/PI3K/Akt pathway activation, thereby promoting migration and invasion in laryngeal cancer. AURKA/FAK/PI3K/Akt inhibitors may thus represent potential targets for clinical LSCC treatment.


Subject(s)
Aurora Kinase A/metabolism , Carcinoma, Squamous Cell/enzymology , Focal Adhesion Kinase 1/metabolism , Head and Neck Neoplasms/enzymology , Laryngeal Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Proliferation/physiology , Down-Regulation , Focal Adhesion Kinase 1/antagonists & inhibitors , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Heterografts , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Male , Mice , Mice, Nude , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...