Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Oncol ; 11: 685972, 2021.
Article in English | MEDLINE | ID: mdl-34249734

ABSTRACT

OBJECTIVE: To explore the suitable cases for vaginal cuff brachytherapy (VCB) combined with external beam radiation therapy (EBRT) in the postoperative treatment of cervical cancer. METHODS: We retrospectively analyzed the clinical data of 214 postoperative cervical cancer patients who received radiotherapy from January 2008 to December 2015. Among them, 146 patients received postoperative EBRT, 68 received EBRT plus VCB. There was no statistical difference in clinical and pathological characteristics between these two groups. Those who with negative vaginal cuff underwent supplemented 12-18 Gy/2-3 Fx VCB. Survival analyses were performed using Kaplan-Meier method, and Cox model was used to analyze prognostic factors. RESULTS: The median follow-up was 52 months (9-136 months), and 4-year RFS (recurrence-free survival) was 77%. Among them, 58 patients (27.10%) had local or distant recurrences, 29 (13.55%) in pelvis, six (2.80%) with metastases to para-aortic, 19 (8.88%) with distant metastases (including inguinal lymph nodes) and four (1.87%) with both local and distant recurrences. The postoperative brachytherapy boost did not improve RFS or OS (overall survival) among the investigated subjects, P = 0.77, P = 0.99, respectively. Neither it decreased the local relapse in the pelvis or vaginal cuff, P = 0.56, P = 0.59. Subgroup analyses showed that brachytherapy boost improved RFS in patients who had bulky mass (>4 cm) as well as 1) with deep stromal invasion (>50% stromal invasion), P = 0.012 or 2) received low EBRT dose (≤45 Gy), P = 0.033, and in patients with deep stromal invasion as well as received low EBRT dose (P = 0.018). CONCLUSIONS: We first proposed the case selection model for postoperative EBRT plus VCB. Brachytherapy boost were considered in the setting of postoperative radiotherapy if the patients had at least two out of these following factors: bulky mass, deep stromal invasion and low EBRT dose.

2.
Oncol Lett ; 21(1): 67, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33365078

ABSTRACT

The presence of hypoxia in solid tumors is considered one of the major factors that contribute to radiation resistance. The aim of the present study was to establish a therapeutic system, which can be controlled by radiation itself, to enhance radiosensitivity. For this purpose, a lentiviral gene therapy vector containing the human inhibitor of growth 4 (ING4) and its upstream promoter, human early growth response factor-1 (EGR1), which possesses the radiation-inducible characteristics to activate the transcription of its downstream genes, was constructed. Downstream fluorescence proteins were investigated to ensure that the EGR1 promoter was induced by irradiation. Furthermore, ING4 open reading frame (ORF) expression was detected by western blotting. The cell cycle was analyzed by fluorescence-activated cell sorting analysis 48 h after the cells were exposed to X-rays ranging between 0 and 8 Gy. In cells stably and transiently transfected with reporter plasmids, the EGR1-driver gene was sensitive to ionizing irradiation. Furthermore, irradiation-induced ING4 gene expression was observed. The enhanced ING4 expression increased the number of cells in the G2/M phase and decreased the proportion of cells in the G1/S phase. Therefore, ING4 expression inhibited cell proliferation and was associated with less colonies being formed. Furthermore, ING4 suppressed hypoxia-inducible factor 1α expression under hypoxic conditions and promoted cell apoptosis. Overall, these results revealed that combining the EGR1 promoter and ING4 ORF using a lentivirus system may be a promising therapeutic strategy with which to enhance radiosensitivity controlled by radiation. However, further studies using in vivo models are required to confirm these findings.

3.
Zhonghua Zhong Liu Za Zhi ; 36(5): 329-34, 2014 May.
Article in Chinese | MEDLINE | ID: mdl-25030586

ABSTRACT

OBJECTIVE: To explore the combined anti-tumor effect of radiation therapy and gene-targeted suppression of tumor neovasculature in lung adenocarcinoma in vivo, and to explore the feasibility of micro-PET/CT in dynamic evaluation of treatment effectiveness. METHODS: Thirty 5-6-week old male BALB/c nude mice were used in this study. The mouse models of xenotransplanted human lung adenocarcinoma were divided into 5 groups at random, six mice in each group: the control group, radiation treatment alone group and three groups of recombinant baculovirus plus radiation treatment (intratumoral injection, tail vein injection, and intramuscular injection). The tumor volume was measured every 2 days. Growth delay time (GD) and growth inhibition rate was calculated. FDG metabolism was evaluated by micro-PET-CT before and after treatment. The expressions of VEGF, CD31 and Ki-67 were detected by immunohistochemistry (IHC). RESULTS: The tumor growth delay was >12 days, and the tumor inhibition rate was >45% in the recombinant baculovirus combined with radiotherapy groups, significantly higher than that of the radiotherapy alone group (P < 0.05). Immunohistochemical analysis showed that the expressions of VEGF, CD31 and Ki-67 were significantly lower than that in other groups (P < 0.05). The micro-PET-CT assessment showed that the FDG-metabolism in the recombinant baculovirus combined with radiotherapy groups was significantly reduced (P < 0.05), and the SUVmax (FDG metabolism) of transplanted tumors after treatment was also markedly decreased in comparison with that of the control group. The tumor volume after treatment was significantly correlated with SUVmax in the recombinant baculovirus intratumoral injection + radiotherapy group(r = 0.976), recombinant baculovirus intravenous injection + radiotherapy group (r = 0.954), recombinant baculovirus intramuscular injection + radiotherapy group (r = 0.929), and radiotherapy alone group (r = 0.871, P < 0.05). CONCLUSIONS: The recombinant baculovirus containing Egr1 promoter and K5 gene combined with radiotherapy enhances the suppressing effect on the growth of lung adenocarcinoma in the tumor-bearing nude mice. The inducibility of Egr1 promoter by radiation allows the targeting and controllability of treatment. Micro-PET-CT results have a good correlation with the treatment effectiveness. Therefore, it can be used in real-time evaluation of tumor metabolic function in vivo.


Subject(s)
Adenocarcinoma/radiotherapy , Early Growth Response Protein 1/genetics , Genetic Therapy , Lung Neoplasms/radiotherapy , Molecular Targeted Therapy , Peptide Fragments/genetics , Plasminogen/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Baculoviridae/genetics , Cell Line, Tumor , Combined Modality Therapy , Early Growth Response Protein 1/physiology , Fluorodeoxyglucose F18 , Genetic Vectors , Humans , Ki-67 Antigen/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Peptide Fragments/physiology , Plasminogen/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Positron-Emission Tomography , Promoter Regions, Genetic , Random Allocation , Recombinant Proteins/genetics , Tomography, X-Ray Computed , Tumor Burden , Vascular Endothelial Growth Factor A/metabolism
4.
Tumour Biol ; 35(2): 1287-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24068565

ABSTRACT

The purposes of this study were to determine the expression profiles of microRNA-34a (miR-34a) in human gastric cancer cell line (SGC-7901) and cisplatin-resistant cell lines (SGC-7901/DDP), and to establish the correlation between miR-34a expression profile and the sensitivity of human gastric cancer cell to cisplatin-based pattern, thereby providing new methods and strategies for treating gastric cancer. Gastric cancer cell line (SGC-7901) and cisplatin-resistant cell line (SGC-7901/DDP) were cultivated in vitro, respectively. Quantitative real-time PCR (qRT-PCR) and Western blot were utilized to determine the expression profiles of miR-34a and survivin in both gastric cancer cell lines. With miR-34a mimic and miR-34a inhibitor transfected into SGC-7901 and SGC-7901/DDP for 48 h, post-transfection changes of miR-34a expression was determined; the effects of miR-34a ectopic expression on the viability of cisplatin-induce gastric cancer cell were assayed by the MTT method. The effects of miR-34a ectopic expression on apoptosis of cisplatin-induce gastric cancer cell were determined by Annexin V/propidium iodide (PI) double staining method and flow cytometry. The effects of miR-34a ectopic expression on the AKT and p-AKT expression of cisplatin-induce gastric cancer cells were determined by Western blot and flow cytometry with the PI3K pathway inhibitor Wortmannin. As shown by qRT-PCR and Western blot analyses, the expression of miR-34a in cisplatin-resistant cell lines decreased significantly in comparison to that of SGC-7901 cell line (p < 0.05), while significant up-regulation of survivin expression was also observed (p < 0.05). Compared with the control group, the expression of miR-34a increased significantly in SGC-7901 cells transfected with miR-34a mimic for 48 h (p < 0.01). After miR-34a inhibitor transfection, the expression of miR-34a decreased significantly (p < 0.05). The viability of cisplatin-induce gastric cancer cells increased significantly (p < 0.05) with significant decrease of apoptosis after miR-34a expression inhibition, as demonstrated by MTT and flow cytometry with miR-34a over-expression, the viability of cisplatin-induce gastric cancer cells decreased significantly (p < 0.05), with significant apoptosis increase (p < 0.05). As shown by Western blot and flow cytometry, in comparison to the control group, Wortmannin could inhibit miR-34a inhibitor and DDP induced up-regulation of p-AKT significantly (p < 0.05) and stimulated apoptosis. In conclusion, miR-34a expression was down-regulated in cisplatin-resistant cell lines. miR-34a over-expression could improve the sensitivity of gastric cancer cells against cisplatin-based chemotherapies, with PI3K/AKT/survivin signaling pathway possibly involved in the mechanism.


Subject(s)
Cisplatin/pharmacology , Elafin/genetics , MicroRNAs/genetics , Stomach Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Elafin/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
5.
World J Gastroenterol ; 19(37): 6245-57, 2013 Oct 07.
Article in English | MEDLINE | ID: mdl-24115823

ABSTRACT

AIM: To reveal the clinicopathological features and risk factors for lymph node metastases in gastric cardiac adenocarcinoma of male patients. METHODS: We retrospective reviewed a total of 146 male and female patients with gastric cardiac adenocarcinoma who had undergone curative gastrectomy with lymphadenectomy in the Department of Surgery, Xin Hua Hospital and Rui Jin Hospital of Shanghai Jiaotong University Medical School between November 2001 and May 2012. Both the surgical procedure and extent of lymph node dissection were based on the recommendations of Japanese gastric cancer treatment guidelines. Univariate and multivariate analyses of lymph node metastases and the clinicopathological features were undertaken. RESULTS: The rate of lymph node metastases in male patients with gastric cardiac adenocarcinoma was 72.1%. Univariate analysis showed an obvious correlation between lymph node metastases and tumor size, gross appearance, differentiation, pathological tumor depth, and lymphatic invasion in male patients. Multivariate logistic regression analysis revealed that tumor differentiation and pathological tumor depth were the independent risk factors for lymph node metastases in male patients. There was an obvious relationship between lymph node metastases and tumor size, gross appearance, differentiation, pathological tumor depth, lymphatic invasion at pN1 and pN2, and nerve invasion at pN3 in male patients. There were no significant differences in clinicopathological features or lymph node metastases between female and male patients. CONCLUSION: Tumor differentiation and tumor depth were risk factors for lymph node metastases in male patients with gastric cardiac adenocarcinoma and should be considered when choosing surgery.


Subject(s)
Adenocarcinoma/secondary , Cardia/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Cardia/surgery , Cell Differentiation , Chi-Square Distribution , Female , Gastrectomy , Humans , Logistic Models , Lymph Node Excision , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Retrospective Studies , Risk Factors , Sex Factors , Stomach Neoplasms/surgery , Young Adult
6.
Lancet Oncol ; 14(10): 953-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23948351

ABSTRACT

BACKGROUND: Icotinib, an oral EGFR tyrosine kinase inhibitor, had shown antitumour activity and favourable toxicity in early-phase clinical trials. We aimed to investigate whether icotinib is non-inferior to gefitinib in patients with non-small-cell lung cancer. METHODS: In this randomised, double-blind, phase 3 non-inferiority trial we enrolled patients with advanced non-small-cell lung cancer from 27 sites in China. Eligible patients were those aged 18-75 years who had not responded to one or more platinum-based chemotherapy regimen. Patients were randomly assigned (1:1), using minimisation methods, to receive icotinib (125 mg, three times per day) or gefitinib (250 mg, once per day) until disease progression or unacceptable toxicity. The primary endpoint was progression-free survival, analysed in the full analysis set. We analysed EGFR status if tissue samples were available. All investigators, clinicians, and participants were masked to patient distribution. The non-inferiority margin was 1·14; non-inferiority would be established if the upper limit of the 95% CI for the hazard ratio (HR) of gefitinib versus icotinib was less than this margin. This study is registered with ClinicalTrials.gov, number NCT01040780, and the Chinese Clinical Trial Registry, number ChiCTR-TRC-09000506. FINDINGS: 400 eligible patients were enrolled between Feb 26, 2009, and Nov 13, 2009; one patient was enrolled by mistake and removed from the study, 200 were assigned to icotinib and 199 to gefitinib. 395 patients were included in the full analysis set (icotinib, n=199; gefitinib, n=196). Icotinib was non-inferior to gefitinib in terms of progression-free survival (HR 0·84, 95% CI 0·67-1·05; median progression-free survival 4·6 months [95% CI 3·5-6·3] vs 3·4 months [2·3-3·8]; p=0·13). The most common adverse events were rash (81 [41%] of 200 patients in the icotinib group vs 98 [49%] of 199 patients in the gefitinib group) and diarrhoea (43 [22%] vs 58 [29%]). Patients given icotinib had less drug-related adverse events than did those given gefitinib (121 [61%] vs 140 [70%]; p=0·046), especially drug-related diarrhoea (37 [19%] vs 55 [28%]; p=0·033). INTERPRETATION: Icotinib could be a new treatment option for pretreated patients with advanced non-small-cell lung cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Crown Ethers/therapeutic use , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Crown Ethers/adverse effects , Disease-Free Survival , Double-Blind Method , ErbB Receptors/genetics , Female , Gefitinib , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Middle Aged , Mutation , Quinazolines/adverse effects
7.
Zhonghua Zhong Liu Za Zhi ; 35(4): 295-300, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23985260

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of trastuzumab in combination with chemotherapy versus chemotherapy alone in the first-line treatment of HER-2-positive advanced gastric or gastro-oesophageal junction cancer. METHODS: Fifteen Chinese research centers are involved in the BO18255 (ToGA) study. Patients with gastric or gastro-oesophageal junction cancer were eligible for inclusion if their tumor showed overexpression of HER-2 protein by immunohistochemistry +++ or FISH-positive. Patients were randomly assigned in a 1:1 ratio to receive a chemotherapy regimen consisting of capecitabine or 5-FU plus cisplatin or chemotherapy in combination with intravenous trastuzumab. The primary endpoint was overall survival. RESULTS: Eighty-five Chinese patients were enrolled in this study, of whom 84 were included in the primary analysis: trastuzumab plus chemotherapy (FP/H) (n = 36) and chemotherapy alone (FP)(n = 48). The median follow-up was 15.2 months in the FP/H group and 14.2 months in the FP group. The median survival time was 12.6 months in the FP/H group compared with 9.7 months in the FP group [hazard ratio 0.72, 95%CI (0.40; 1.29)]. Grade 3/4 adverse events were higher in the FP/H(63.9%)than FP (47.9%) groups, including neutropenia, vomiting and nausea. Two mild cardiac adverse events occurred in the FP/H group. Severe adverse events occurred in 3 cases of both two groups, respectively. CONCLUSIONS: Addition of trastuzumab to chemotherapy is well tolerated and shows improved survival in Chinese patients with advanced gastric or gastro-oesophageal junction cancer. These results are consistent with the results of ToGA whole population trial. Trastuzumab in combination with chemotherapy can be considered as a new option for patients with HER-2-positive advanced gastric or gastro-oesophageal junction cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Esophagogastric Junction , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine , China , Cisplatin/administration & dosage , Cisplatin/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Esophageal Neoplasms/pathology , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/analogs & derivatives , Follow-Up Studies , Humans , Male , Middle Aged , Nausea/chemically induced , Neoplasm Staging , Neutropenia/chemically induced , Remission Induction , Retrospective Studies , Stomach Neoplasms/pathology , Survival Rate , Trastuzumab , Vomiting/chemically induced
8.
Exp Ther Med ; 5(5): 1418-1426, 2013 May.
Article in English | MEDLINE | ID: mdl-23737892

ABSTRACT

The current study aimed to evaluate the efficacy and safety of palonosetron hydrochloride injection for preventing chemotherapy-induced moderate and severe nausea and vomiting. A multi-centered, randomly stratified, double-blind, double-dummy, parallel-group and positive-controlled trial was performed. A total of 240 patients who underwent chemotherapy treatment which induced moderate or severe vomiting were divided into the experimental and control groups. Half an hour before chemotherapy, the experimental group received a 0.25-mg palonosetron hydrochloride injection, whereas the control group received a 3-mg granisetron injection. The acute vomiting complete remission rate (CRR) of the experimental group was not significantly different compared with that of the control group (P=0.35). The delayed vomiting CRR of the experimental group was significantly higher compared with that of the control group (P=0.002). No difference in full course vomiting CRR, vomiting control time, treatment failure time or acute nausea CRR was identified between the two groups. No significant differences in adverse events were observed between the experimental group and the control group. No significant differences in adverse reactions occurred between the experimental group and the control group (12.50%). Palonosetron hydrochloride injection had a better effect on delayed vomiting CRR than granisetron hydrochloride injection. The two injections exhibited similar effects on acute vomiting CRR, full course vomiting CRR, vomiting control time, treatment failure time (days), acute nausea CRR and adverse events.

9.
World J Gastroenterol ; 19(20): 3096-107, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23716990

ABSTRACT

AIM: To explore risk factors for lymph node metastases in early gastric cancer (EGC) and to confirm the appropriate range of lymph node dissection. METHODS: A total of 202 patients with EGC who underwent curative gastrectomy with lymphadenectomy in the Department of Surgery, Xinhua Hospital and Ruijin Hospital of Shanghai Jiaotong University Medical School between November 2003 and July 2009, were retrospectively reviewed. Both the surgical procedure and the extent of lymph node dissection were based on the recommendations of the Japanese gastric cancer treatment guidelines. The macroscopic type was classified as elevated (type I or IIa), flat (IIb), or depressed (IIc or III). Histopathologically, papillary and tubular adenocarcinomas were grouped together as differentiated adenocarcinomas, and poorly differentiated and signet-ring cell adenocarcinomas were regarded as undifferentiated adenocarcinomas. Univariate and multivariate analyses of lymph node metastases and patient and tumor characteristics were undertaken. RESULTS: The lymph node metastases rate in patients with EGC was 14.4%. Among these, the rate for mucosal cancer was 5.4%, and 8.9% for submucosal cancer. Univariate analysis showed an obvious correlation between lymph node metastases and tumor location, depth of invasion, morphological classification and venous invasion (χ(2) = 122.901, P = 0.001; χ(2) = 7.14, P = 0.008; χ(2) = 79.523, P = 0.001; χ(2) = 8.687, P = 0.003, respectively). In patients with submucosal cancers, the lymph node metastases rate in patients with venous invasion (60%, 3/5) was higher than in those without invasion (20%, 15/75) (χ(2) = 4.301, P = 0.038). Multivariate logistic regression analysis revealed that the depth of invasion was the only independent risk factor for lymph node metastases in EGC [P = 0.018, Exp (B) = 2.744]. Among the patients with lymph node metastases, 29 cases (14.4%) were at N1, seven cases were at N2 (3.5%), and two cases were at N3 (1.0%). Univariate analysis of variance revealed a close relationship between the depth of invasion and lymph node metastases at pN1 (P = 0.008). CONCLUSION: The depth of invasion was the only independent risk factor for lymph node metastases. Risk factors for metastases should be considered when choosing surgery for EGC.


Subject(s)
Adenocarcinoma/secondary , Lymph Nodes/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/surgery , Adenocarcinoma, Papillary/secondary , Adenocarcinoma, Papillary/surgery , Adult , Aged , Aged, 80 and over , Carcinoma, Signet Ring Cell/secondary , Carcinoma, Signet Ring Cell/surgery , Cell Differentiation , Chi-Square Distribution , Early Detection of Cancer , Female , Gastrectomy , Humans , Logistic Models , Lymph Node Excision , Lymph Nodes/surgery , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Risk Factors , Stomach Neoplasms/surgery
10.
J Laparoendosc Adv Surg Tech A ; 23(3): 267-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23272726

ABSTRACT

OBJECTIVE: To report the feasibility of laparoscopic resection with intraoperative radiotherapy for local advanced rectal cancer in an Asian man. PATIENT AND METHODS: A 55-year-old man with adenocarcinoma of the rectum presented at Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China. The tumor, with a size of 5 × 5 cm, was located 3 cm from the anus and covered a circular area around approximately two-thirds of the bowel. The carcinoembryonic antigen level was 29.86 ng/mL; a nodule was detected, but no distant metastasis was detected. Preoperative staging was T4N1M0. After the patient signed the consent form, laparoscopic resection with intraoperative radiotherapy was performed. RESULTS: The operation time was about 180 minutes, intraoperative blood loss was 100 mL, and postoperative hospital stay was 8 days. The patient had no postoperative complications. conclusions: Performance of laparoscopic resection with intraoperative radiotherapy for local advanced rectal cancer is feasible in selected patients.


Subject(s)
Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Laparoscopy , Rectal Neoplasms/radiotherapy , Rectal Neoplasms/surgery , Adenocarcinoma/pathology , Combined Modality Therapy , Humans , Intraoperative Period , Male , Middle Aged , Neoplasm Staging , Rectal Neoplasms/pathology
11.
Tumour Biol ; 34(2): 963-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23264087

ABSTRACT

We aimed to investigate the expression of microRNA-34a (miR-34a) in human gastric cancer cells and to evaluate the effects of miR-34a, acting via its gene survivin, on gastric cancer cell HGC-27 to provide potential new strategies for treating gastric cancer. In vitro cultures of the human gastric cancer cell lines MGC80-3, HGC-27, NCI-N87, and SGC-7901 and the normal human gastric epithelial cell line GES-1 were established. The expression of miR-34a in each gastric cancer cell line and GES-1 normal human gastric epithelial cell line was detected using quantitative real-time polymerase chain reaction (qRT-PCR). After the HGC-27 cells were transfected with a miR-34a mimic for 48 h, the changes in the expression levels of miR-34a were detected using qRT-PCR. The effect of miR-34a on HGC-27 cell viability was measured using a tetrazolium-based colorimetric [-(4,5)-dimethylthiahiazo-(-z-y1)-3,5-di-phenytetrazoliumromide (MTT)] assay. Flow cytometry was used to analyze the effects of miR-34a on HGC-27 cell proliferation. Annexin V/propidium iodide double staining and flow cytometry were used to analyze the effects of miR-34a on HGC-27 cell apoptosis. A Transwell invasion chamber was used to detect the effects of miR-34a on HGC-27 cell invasion. Finally, western blotting was used to analyze the effects of miR-34a on survivin protein expression. The qRT-PCR test determined that miR-34a expression in gastric cancer cells was significantly reduced compared to the normal gastric epithelial cell line GES-1 (p < 0.01). Compared to the control group, cellular miR-34a expression levels were significantly increased in HGC-27 human gastric carcinoma cells after transfection with a miR-34a mimic for 48 h (p < 0.01). The MTT assay demonstrated that after overexpressing miR-34a in HGC-27 cells, cellular viability was significantly reduced (p < 0.05). Flow cytometry analysis determined that upon miR-34a overexpression, the proliferation index decreased significantly (p < 0.05), and cellular apoptosis was significantly increased (p < 0.01). The Transwell invasion chamber assay illustrated that after increasing the expression of miR-34a, the number of cells passing through the Transwell chamber was significantly reduced (p < 0.01). Based on western blotting, compared with the control group, survivin protein expression levels were significantly decreased in the HGC-27 cells transfected with the miR-34a mimic for 48 h (p < 0.01). In conclusion, the expression level of miR-34a was downregulated in human gastric cancer cell lines. miR-34a can negatively regulate survivin protein expression and inhibit gastric cancer cell proliferation and invasion. Therapeutically enhancing miR-34a expression or silencing the survivin gene may benefit patients with gastric cancer.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Inhibitor of Apoptosis Proteins/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Flow Cytometry , Humans , Inhibitor of Apoptosis Proteins/genetics , MicroRNAs/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survivin , Tumor Cells, Cultured
12.
Zhonghua Zhong Liu Za Zhi ; 34(8): 577-81, 2012 Aug.
Article in Chinese | MEDLINE | ID: mdl-23158989

ABSTRACT

OBJECTIVE: To investigate the essential role and mechanism of TRPC6 gene in the development of gastric cancer. METHODS: The expression of TRPC6 protein was assessed in gastric cancer tissues and normal tissues adjacent to the cancer from 30 patients with gastric cancer. The inhibiting effect of TRPC6 activity on cell growth, cell cycle of a human gastric cancer cell line AGS cells, tumor progression and development of xenografted human gastric cancer in a mouse model was tested using dominant-negative mutant TRPC6 (DNC6). The survival of mice bearing xenografted tumors in the GFP and DNC6 was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS: The TRPC6 protein in the tumor tissues and para-tumor tissues was (21.60 ± 8.32)% versus (7.14 ± 2.24)%. After transfection of DNC6 virus for 24 hours, 48 hours, 72 hours and 96 hours, the growth inhibition rates of gastric cancer cells were (36.90 ± 1.13)%, (44.06 ± 2.17)%, (52.12 ± 2.76)% and (50.89 ± 1.97)%, respectively. The clone formation rates of control group and DNC6 group were (14.70 ± 3.00)% versus (43.80 ± 7.00)%. After transfection with DNC6 virus for 0, 24, 36 and 48 hours, the G(2)/M phase arrest was (20.34 ± 1.98)%, (24.31 ± 2.37)%, (27.70 ± 2.36)%, (35.10 ± 3.0)% in the DNC6 group and (18.40 ± 2.01)%, (18.0% ± 1.72)%, (17.50 ± 1.74)%, (16.80 ± 1.71)% in the control group, respectively. Inhibition of TRPC6 activity also reduced the subcutaneous tumor volume in the mouse models with xenografted human tumors (P < 0.05). CONCLUSION: In the preclinical models tested, TRPC6 channels are essential for gastric cancer development via regulation of G(2)/M phase transition.


Subject(s)
Cell Cycle , Cell Proliferation , Stomach Neoplasms/pathology , TRPC Cation Channels/metabolism , Adenoviridae/genetics , Animals , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin B/metabolism , Cyclin-Dependent Kinases , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Recombinant Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , TRPC6 Cation Channel , Transfection , Tumor Burden
13.
J Surg Oncol ; 103(2): 110-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21259243

ABSTRACT

BACKGROUND AND OBJECTIVES: The present study was designed to investigate the clinicopathological role of survivin-expressing circulating tumor cells (CTCs) and to determine whether the presence of survivin-expressing CTCs is an independent predictor of tumor recurrence following curative resection of gastric cancer. METHODS: This study included 98 patients who underwent potentially curative resection. Reverse transcription polymerase chain reaction enzyme linked immunosorbent assay (RT-PCR ELISA) was used to measure survivin mRNA in peripheral blood. RESULTS: Of the 98 patients studied, 45 (45.9%) were positive for survivin mRNA. Survivin mRNA expression correlated with Lauren classification (P < 0.001), pathological tumor (pT) stage (P < 0.001), pathological tumor node metastasis (pTNM) stage (P = 0.009), and degree of differentiation (P = 0.001). The pTNM stage and the status of survivin mRNA were independent prognostic factors of disease-free survival (P = 0.007 and <0.001, respectively). CONCLUSIONS: The detection of CTCs expressing survivin mRNA could be a good clinical biomarker used to predict the prognosis of patients with curatively resected gastric cancer.


Subject(s)
Biomarkers, Tumor/analysis , Microtubule-Associated Proteins/analysis , Neoplasm Recurrence, Local/chemistry , Neoplasm Recurrence, Local/diagnosis , Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/pathology , Stomach Neoplasms/surgery , Adult , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Inhibitor of Apoptosis Proteins , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Predictive Value of Tests , Prognosis , ROC Curve , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Survivin
14.
J Natl Cancer Inst ; 102(14): 1052-68, 2010 Jul 21.
Article in English | MEDLINE | ID: mdl-20554944

ABSTRACT

BACKGROUND: Patients with glioblastoma multiforme, the most aggressive form of glioma, have a median survival of approximately 12 months. Calcium (Ca(2+)) signaling plays an important role in cell proliferation, and some members of the Ca(2+)-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of many types of cancer cells. In this study, we investigated the role of TRPC6 in cell cycle progression and in the development of human glioma. METHODS: TRPC6 protein and mRNA expression were assessed in glioma (n = 33) and normal (n = 17) brain tissues from patients and in human glioma cell lines U251, U87, and T98G. Activation of TRPC6 channels was tested by platelet-derived growth factor-induced Ca(2+) imaging. The effect of inhibiting TRPC6 activity or expression using the dominant-negative mutant TRPC6 (DNC6) or RNA interference, respectively, was tested on cell growth, cell cycle progression, radiosensitization of glioma cells, and development of xenografted human gliomas in a mouse model. The green fluorescent protein (GFP) and wild-type TRPC6 (WTC6) were used as controls. Survival of mice bearing xenografted tumors in the GFP, DNC6, and WTC6 groups (n = 13, 15, and 13, respectively) was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS: Functional TRPC6 was overexpressed in human glioma cells. Inhibition of TRPC6 activity or expression attenuated the increase in intracellular Ca(2+) by platelet-derived growth factor, suppressed cell growth and clonogenic ability, induced cell cycle arrest at the G2/M phase, and enhanced the antiproliferative effect of ionizing radiation. Cyclin-dependent kinase 1 activation and cell division cycle 25 homolog C expression regulated the cell cycle arrest. Inhibition of TRPC6 activity also reduced tumor volume in a subcutaneous mouse model of xenografted human tumors (P = .014 vs GFP; P < .001 vs WTC6) and increased mean survival in mice in an intracranial model (P < .001 vs GFP or WTC6). CONCLUSIONS: In this preclinical model, TRPC6 channels were essential for glioma development via regulation of G2/M phase transition. This study suggests that TRPC6 might be a new target for therapeutic intervention of human glioma.


Subject(s)
Brain Neoplasms/metabolism , Cell Division , Cell Transformation, Neoplastic/metabolism , G2 Phase , Glioma/metabolism , TRPC Cation Channels/metabolism , Adenoviridae , Animals , Base Sequence , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Genetic Vectors , Glioma/genetics , Glioma/pathology , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Lentivirus , Mice , Molecular Sequence Data , Phenotype , RNA, Messenger/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , TRPC Cation Channels/immunology , TRPC6 Cation Channel , Transplantation, Heterologous , Up-Regulation
15.
Int J Cancer ; 125(10): 2281-7, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19610066

ABSTRACT

Channels formed by the canonical transient receptor potential (TRPC) subfamily of proteins are Ca(2+)-permeable, nonselective cation channels with various functions. Through a phospholipase C (PLC)-dependent mechanism TRPC6, a member of TRPC subfamily, can be activated by receptor tyrosine kinases (RTK) or G protein-coupled receptors (GPCR), which are implicated in cell proliferation and human malignancies. Here, we report that TRPC6 has a critical role in human gastric cancer development. Expression of TRPC6 was greatly upregulated in human gastric cancer epithelial cells compared with that in normal gastric epithelial cells. Treatment of AGS or MKN45 cells, human gastric cancer cell lines, with SKF96365, an agent known to inhibit TRPC channels, arrested cell cycle in G2/M phase and suppressed cell growth. Importantly, expressing a dominant negative mutant of TRPC6 (DNC6) in these cells also arrested cell cycle in G2/M phase and inhibited cell growth. The Ca(2+) elevation in the MKN45 cells evoked by histamine was inhibited by SKF96365 and DNC6. Moreover, inhibition of TRPC6 suppressed the formation of gastric tumors in nude mice. These results suggest that Ca(2+) elevation regulated by TRPC6 channels is essential for G2/M phase transition and for the development of gastric cancers.


Subject(s)
Cell Division , G2 Phase , Stomach Neoplasms/prevention & control , TRPC Cation Channels/antagonists & inhibitors , Adenoviridae/genetics , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Proliferation , Electrophysiology , Flow Cytometry , Humans , Imidazoles/pharmacology , Immunoenzyme Techniques , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , TRPC Cation Channels/genetics , TRPC6 Cation Channel
16.
Jpn J Clin Oncol ; 39(9): 582-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19509000

ABSTRACT

OBJECTIVE: The aim of this study was to assess the efficacy and toxicity of the combination of paclitaxel and nedaplatin as a first-line chemotherapy for patients with advanced esophageal cancer. METHODS: Patients with advanced esophageal cancer received 175 mg/m(2) of paclitaxel over a 3 h infusion, followed by nedaplatin 80 mg/m(2) in a 1 h infusion on day 1 every 3 weeks until the documented disease progression, unacceptable toxicity or patient's refusal. RESULTS: Between March 2005 and December 2007, 48 patients entered in the study. Forty-six (95.8%) of the 48 patients were assessable for response. The overall response rate was 41.7% (95% CI, 27.8-55.7%) with 2 complete responses and 18 partial responses. The median follow-up period was 20.5 months (range, 12.5-27.2 months). The median overall time to progression and overall survival (OS) were 6.1 months (95% CI, 4.8-7.4 months) and 11.5 months (95% CI, 9.1-13.9 months), respectively. The estimate of OS at 12 and 24 months was 43.8% (95% CI, 29.7-77.8%) and 10.4% (95% CI, 1.8-19.1%), respectively. Most patients experienced anemia, during their course of therapy with 6 (13.0%) patients for grade 3/4 anemia, and grade 1 or 2 anemia was detected in 23 (50%) patients. Grade 3 leucopenia, neutropenia and thrombocytopenia were documented in 8 (17.4%), 9 (17.4%) and 2 (4.3%) patients, respectively. Grade 3 nausea and vomiting were detected in 3 (6.5%) and 2 (4.3%) patients, respectively. Two patients (4.3%) were hospitalized because of treatment-related complications. The treatment was well tolerated and no toxic death occurred. CONCLUSIONS: Combination of paclitaxel and nedaplatin is a tolerated treatment modality with promising activity in previously untreated advanced esophageal cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Carcinoma, Squamous Cell/pathology , Dose-Response Relationship, Drug , Esophageal Neoplasms/pathology , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Organoplatinum Compounds/administration & dosage , Paclitaxel/administration & dosage , Prognosis , Survival Rate , Treatment Outcome
17.
Anticancer Drugs ; 20(4): 287-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19177020

ABSTRACT

The efficacy of chemotherapy for advanced gastric cancer with palliative intent compared with supportive care alone is now widely accepted. However, the best choice of chemotherapy regimen for patients with advanced gastric cancer is still a matter of controversy and requires further investigation. This study is performed to evaluate the efficacy and safety of the FOLFOXIRI regimen (oxaliplatin 85 mg/m as a 2-h intravenous infusion, irinotecan 165 mg/m as a 90-min infusion, leucovorin 200 mg/m as a 2-h infusion, 5-fluorouracil 3200 mg/m as a 48-h continuous infusion on day 1, every 2 weeks) as first-line treatment for advanced gastric cancer. Forty-seven (95.9%) of the 49 patients were assessable for response. Two cases of complete response and 29 cases of partial response were confirmed, giving an overall response rate of 63.3% [95% confidence interval (CI): 49.8-76.8%]. The median time to progression and overall survival for all patients were 7.3 months (95% CI: 6.0-8.6 months) and 11.9 months (95% CI: 9.4-14.4 months), respectively. The estimate of overall survival at 12 months was 42.9% (95% CI: 29.0-56.7%). Most patients experienced neutropenia during their course of therapy with 49% of patients (n=23) for grade 3/4 neutropenia. Grade 3 nausea/vomiting, stomatitis, and diarrhea were observed in 20 (42.6%), two (4.3%), and five (10.6%) patients, respectively. Yet, no grade 4 nonhematologic toxicity was observed. The FOLFOXIRI combination is a tolerated treatment modality with promising activity in previously untreated advanced gastric cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Stomach Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Disease Progression , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Gastrointestinal Diseases/chemically induced , Humans , Infusions, Intravenous , Leucovorin/administration & dosage , Male , Middle Aged , Neutropenia/chemically induced , Organoplatinum Compounds/administration & dosage , Survival Rate , Treatment Outcome , Young Adult
18.
Ai Zheng ; 26(12): 1330-5, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18076795

ABSTRACT

BACKGROUND & OBJECTIVE: Epidermal growth factor receptor (EGFR) is expressed in most human epithelial cancers and is involved in the development of cancer cell resistance to irradiation. We used gefitinib, a selective EGFR tyrosine kinase inhibitor (EGFR-TKI), to investigate its effects and mechanisms in enhancing the radiosensitivity of human gastric cancer cell lines in vitro. METHODS: The expression of EGFR protein in 7 human gastric cell lines (MKN45, SGC7901, SNU-1, N87, AGS, SNU-16, and KATO-III) was determined by Western blot, in which 2 cell lines with high expression of EGFR were selected for additional test. The inhibitory effect of gefitinib on cell proliferation was measured by MTT assay. Cell survival was determined by clonogenic assay, and then the radiosensitivity parameters were calculated. The effects of gefitinib in combination with radiation on cell apoptosis and cell cycle distribution were analyzed by flow cytometry. RESULTS: Of the 7 gastric cancer cell lines, the expression of EGFR in MKN45 and SGC7901 cells were the highest. The 50% inhibition concentrations (IC(50)) of gefitinib were 0.4 mmol/L for MKN45 cells and 0.8 mmol/L for SGC7901 cells. Cell survival was significantly decreased with the elevation of gefitinib concentration or radiation dose (P<0.05). When treated with 0.1x and 0.2 x IC(50) of gefitinib, the radiosensitization enhancement ratios (SER) of MKN45 cells were 1.102 and 1.154, and those of SGC7901 cells were 1.092 and 1.176, respectively. Either gefitinib or radiation induced cell apoptosis, reduced the percentage of cells at S phase and increased the percentage of cells at G(2)/M phase (P<0.01). CONCLUSIONS: Gefitinib followed by radiation could increase the radiosensitivity of MKN45 and SGC7901 cells with high expression of EGFR and inhibit cell proliferation, induce apoptosis, and alter cell phase distribution. Gefitinib could be a radiation sensitizer for gastric tumors with high expression of EGFR.


Subject(s)
ErbB Receptors/metabolism , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Stomach Neoplasms/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Particle Accelerators , Protein Kinase Inhibitors/pharmacology
19.
World J Gastroenterol ; 13(39): 5267-72, 2007 Oct 21.
Article in English | MEDLINE | ID: mdl-17876899

ABSTRACT

AIM: To evaluate the relationship between changes in serum transforming growth factor beta1 (TGFbeta1) level and curative effect of radiotherapy (RT) in patients with esophageal carcinoma. METHODS: Ninety patients with histologically confirmed esophageal carcinoma were enrolled. Serum samples for TGFbeta1 analysis were obtained before and at the end of RT. An enzyme-linked immunosorbent assay was used to measure serum TGFbeta1 level. Multivariate analysis was performed to investigate the relationship between disease status and changes in serum TGFbeta1 level. RESULTS: Serum TGFbeta1 level in patients with esophageal carcinoma before RT was significantly higher than that in healthy controls (P < 0.001). At the end of RT, serum TGFbeta1 level was decreased in 67.82% (59/87) of the patients. The overall survival rate at 1, 3 and 5 years was 48.28% (42/87), 19.54% (17/87) and 12.64% (11/87), respectively. Main causes of death were local failure and regional lymph node metastasis. In patients whose serum TGFbeta1 level decreased after RT, the survival rate at 1, 3 and 5 years was 61.02% (36/59), 28.81% (17/59) and 18.64% (11/59), respectively. The survival rate at 1 year was 17.86% (5/28) in patients whose serum TGFbeta1 level increased after RT, and all died within 18 mo (P < 0.01). CONCLUSION: Serum TGFbeta1 level may be a useful marker for monitoring disease status after RT in patients with esophageal carcinoma.


Subject(s)
Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/radiotherapy , Carcinoma/blood , Carcinoma/radiotherapy , Esophageal Neoplasms/blood , Esophageal Neoplasms/radiotherapy , Transforming Growth Factor beta1/blood , Adult , Aged , Biomarkers, Tumor/blood , Carcinoma/diagnosis , Carcinoma, Squamous Cell/diagnosis , Case-Control Studies , Disease Progression , Dose-Response Relationship, Radiation , Esophageal Neoplasms/diagnosis , Female , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Severity of Illness Index , Survival Rate
20.
Zhonghua Zhong Liu Za Zhi ; 29(3): 225-7, 2007 Mar.
Article in Chinese | MEDLINE | ID: mdl-17649643

ABSTRACT

OBJECTIVE: To investigate the effect of preoperative radiotherapy or chemoradiotherapy on the sphincter preservation and local tumor control as well as survival for the patient with locally advanced middle-low rectal cancer. METHODS: 121 locally advanced middle-low rectal cancer patients were treated with preoperative radiotherapy or chemoradiotherapy followed by surgery after rest of 4 to 6 weeks. 103 of these patients who underwent radical surgery were finally included in this study. The irradiation regimen was: 40 Gy/4 - 5 weeks, whereas 57 of these 103 patients received concurrent chemotherapy of 5-Fu or Xeloda. Sphincter-preserving surgery was performed in 59 patients and abdominoperineal resection in 44 patients. The survival was estimated by Kaplan-Meier model, and the differences between groups were compared using Log rank test. Multivariate analysis was performed by Cox's model. RESULTS: Ten patients (9.7%) achieved a complete pathological response (pCR) to preoperative radiotherapy or chemoradiotherapy. The sphincter preservation rate was 57.3%. The 3-year overall survival (OS) and disease free survival (DFS) was 66.3% and 59.5%, respectively. Univariate analysis showed that pCR and postoperative pTNM stage were prognostic factors affecting survival, whereas, only pTNM stage was an independent prognostic factor (P = 0.003) by multivariate analysis. CONCLUSION: Neoadjuvant preoperative radiotherapy and chemoradiotherapy is effective in local tumor control and improving survival for locally advanced middle-low rectal cancer, which can raise the rate of sphincter-preserving surgery, and achieve comparable result to abdominoperineal resection.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Radiotherapy, High-Energy/methods , Rectal Neoplasms/therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Capecitabine , Combined Modality Therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Male , Middle Aged , Multivariate Analysis , Neoadjuvant Therapy , Particle Accelerators , Preoperative Care , Rectal Neoplasms/pathology , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...