Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancers (Basel) ; 15(13)2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37444451

ABSTRACT

BACKGROUND: FLOT perioperative chemotherapy represents the standard of care in non-metastatic gastric cancer patients. Signet-ring cell positivity is associated with a worse prognosis in patients with gastric cancer treated with chemotherapy. Comparison between FLOT perioperative chemotherapy vs. surgery followed by adjuvant chemotherapy based on signet-ring cell positivity is lacking. The aim of the analysis was to compare perioperative FLOT with adjuvant chemotherapy in gastric cancer patients stratified by signet-ring cell positivity. METHODS: We conducted a retrospective multicenter analysis based on disease-free survival (DFS) and overall survival (OS) in patients with gastric cancer who received perioperative chemotherapy with a FLOT regimen and compared their survival with a historical cohort of patients treated with adjuvant chemotherapy, matched by cT and cN stage and by tumor histological features. RESULTS: Seventy-six patients were enrolled and 24 (32%) were signet-ring cell positive. At a median follow-up time of 39 months, the median DFS was 26.3 months and the median OS was 37.3 months. Signet-ring cell positivity was associated with a shorter OS (median OS: 20.4 vs. 46.9 months, HR: 3.30, 95%CI: 1.56-6.99, p = 0.0018) and DFS (mDFS: 15.2 vs. 38.6 months, HR: 3.18, 95%CI: 1.55-6.54, p = 0.0016). This was confirmed by multivariate analysis for DFS (Exp(B): 2.55) and OS (Exp(B): 2.68). After propensity score matching, statistically significant shorter DFS (HR: 3.30, 95%CI: 1.50-7.35, p = 0.003) and OS (HR: 5.25, 95%CI: 2.18-12-68, p = 0.0002) were observed for patients with signet-ring cell positivity who received perioperative treatment vs. those who received surgery followed by adjuvant chemotherapy. CONCLUSIONS: Signet-ring positivity was associated with shorter DFS and OS in patients who received perioperative treatment with FLOT compared with surgery followed by adjuvant therapy. These data suggest that for patients with signet-ring cell histology, FLOT perioperative treatment might not always be the best choice of treatment, and further research should be focused on this group of patients.

2.
Clin Chem Lab Med ; 45(7): 822-8, 2007.
Article in English | MEDLINE | ID: mdl-17617021

ABSTRACT

BACKGROUND: Gastric cancer is the second highest cause of cancer mortality in the world, despite declining rates of incidence in many industrialized countries. We carried out a case-control study to evaluate whether polymorphisms of DNA repair and glutathione S-transferase (GST) genes modulate the risk of developing diffuse gastric cancer. METHODS: ERCC1 118 T/C, XRCC1 399 G/A, XPD 312 G/A, XPD 751 A/C, XRCC3 241 C/T, MS 919 A/G, GSTP1 105 A/G, GSTM1-null/positive and GSTT1-null/positive genotypes were obtained for a series of 126 Helicobacter pylori-negative diffuse gastric cancer patients and 144 Helicobacter pylori-negative controls sampled from the population of Marche, an area with high gastric cancer risk in central Italy. RESULTS: GSTP1 105 A/G and GSTP1 105 G/G genotypes were identified as protective factors, with odds ratio (OR) of 0.4 (95% CI 0.17-0.81, p=0.01) and OR=0.58 (95% CI 0.33-1, p=0.05), respectively. GSTT1-null genotype was identified as a protective factor, with OR=0.48 (95% CI 0.22-0.99, p=0.04). There was no significant difference between cases and controls for XPD 751 A/C, ERCC1 118 T/C, XRCC3 241 C/T, XRCC1 399 G/A, XPD 312 G/A, GSTM1-null/positive and MS 919 A/G polymorphisms. CONCLUSIONS: This study suggests that GSTP1 105A/G and GSTT1-null/positive genotypes might be associated with a reduced risk for sporadic diffuse gastric cancer. Clin Chem Lab Med 2007;45:822-8.


Subject(s)
DNA Repair/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Xenobiotics/metabolism , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Genotype , Humans , Italy , Male , Middle Aged , Risk Factors
3.
J Clin Oncol ; 25(10): 1247-54, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17401013

ABSTRACT

PURPOSE: The objective is to investigate whether polymorphisms with putative influence on fluorouracil/oxaliplatin activity are associated with clinical outcomes of patients with advanced colorectal cancer treated with first-line oxaliplatin, folinic acid, and fluorouracil palliative chemotherapy. MATERIALS AND METHODS: Consecutive patients were prospectively enrolled onto medical oncology units in Central Italy. Patients were required to have cytologically/histologically confirmed metastatic disease with at least one measurable lesion. Peripheral blood samples were used for genotyping 12 polymorphisms in thymidylate synthase, methylenetetrahydrofolate reductase, xeroderma pigmentosum group D (XPD), excision repair cross complementing group 1 (ERCC1), x-ray cross complementing group 1, x-ray cross complementing protein 3, glutathione S-transferases (GSTs) genes. The primary end point of the study was to investigate the association between genotypes and progression-free survival (PFS). RESULTS: In 166 patients, ERCC1-118 T/T, XPD-751 A/C, and XPD-751 C/C genotypes were independently associated with adverse PFS. The presence of two risk genotypes (ERCC1-118 T/T combined with either XPD-751 A/C or XPD-751 C/C) occurred in 50 patients (31%). This profiling showed an independent role for unfavorable PFS with a hazard ratio of 2.84% and 95% CI of 1.47 to 5.45 (P = .002). Neurotoxicity was significantly associated with GSTP1-105 A/G. Carriers of the GSTP1-105 G/G genotype were more prone to suffer from grade 3 neurotoxicity than carriers of GSTP1-105 A/G and GSTP1-105 A/A genotypes. CONCLUSION: A pharmacogenetic approach may be an innovative strategy for optimizing palliative chemotherapy in patients with advanced colorectal cancer. These findings deserve confirmation in additional prospective studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , 5' Untranslated Regions , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA-Binding Proteins/genetics , Endonucleases/genetics , Female , Fluorouracil/administration & dosage , Genotype , Humans , Leucovorin/administration & dosage , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Pharmacogenetics , Polymorphism, Genetic , Prospective Studies , Thymidylate Synthase/genetics
4.
J Clin Oncol ; 24(12): 1883-91, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16622263

ABSTRACT

PURPOSE: To investigate whether polymorphisms with putative influence on fluorouracil/cisplatin activity are associated with clinical outcomes of patients with advanced gastric cancer (AGC). PATIENTS AND METHODS: Peripheral blood samples from 175 prospectively enrolled AGC patients treated with fluorouracil/cisplatin palliative chemotherapy were used for genotyping 13 polymorphisms in nine genes (TS, MTHFR, XPD, ERCC1, XRCC1, XRCC3, GSTPI, GSTTI, GSTMI). Genotypes were correlated to response and survival. RESULTS: The overall response rate was 41%, the median progression-free survival (PFS) was 24 weeks (range, 4 to 50 weeks), and the median overall survival (OS) was 39 weeks (range, 8 to 72+ weeks). Chemoresistance and poor survival were significantly associated with TS 5'-UTR 3G-genotype (2R/3G, 3C/3G, 3G/3G) and GSTP1 105 A/A homozygous genotype. Sixty-one patients (35%) did not show any of these risk genotypes (group 0), 57 patients (32.5%) showed one of the two risk genotypes (group 1), and 57 patients (32.5%) showed both risk genotypes (group 2). Median PFS and OS in group 0 patients were 32 weeks (range, 8 to 50 weeks) and 49 weeks (range, 18 to 72+ weeks), respectively. Group 1 and group 2 patients showed significantly worse PFS (median, 26 weeks [range, 6 to 44 weeks] and 14 weeks [range, 4 to 38 weeks], respectively) and worse OS (median, 39 weeks [range, 10 to 58 weeks] and 28 weeks [range, 8 to 56 weeks]), respectively, than group 0 patients. This adverse effect was retained in multivariate analysis. CONCLUSION: Specific polymorphisms may influence clinical outcomes of AGC patients. Selecting palliative chemotherapy on the basis of pretreatment genotyping may represent an innovative strategy that warrants prospective studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Palliative Care , Pharmacogenetics , Polymorphism, Genetic , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Case-Control Studies , Cisplatin/administration & dosage , Female , Fluorouracil/administration & dosage , Genotype , Humans , Male , Middle Aged , Patient Selection , Prognosis , Stomach Neoplasms/pathology , Survival Analysis
5.
Int J Cancer ; 118(3): 628-32, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16094648

ABSTRACT

We performed a case-control study to examine the relationship between MTHFR C677T gene polymorphism (MTHFR677C/T) and gastric cancer susceptibility in at-risk populations in central Italy. To explore genomic DNA hypomethylation as a potential etiologic mechanism, this phenomenon was evaluated in carriers of the MTHFR677T/T genotype and carriers of the wild-type MTHFR677C/C genotype. Lymphocyte genomic DNA from 162 gastric cancer patients and 164 controls was used for MTHFR677C/T genotyping. Unconditional regression analysis with ORs and 95% CIs was used to investigate the association of the polymorphism with disease. Genomic DNA methylation status by an established enzymatic assay that measures the DNA accepting capacity of methyl groups (inversely related to endogenous methylation) was assessed in a random sample of 40 carriers of the wild-type MTHFR677C/C genotype and 40 carriers of the MTHFR677T/T genotype. The global allelic distribution was in Hardy-Weinberg equilibrium. The MTHFR677T allele was significantly associated with gastric cancer risk with an OR of 2.49 (95% CI 1.48-4.20) in heterozygous MTHFR677C/T carriers and an OR of 2.85 (95% CI 1.52-5.35) in homozygous MTHFR677T/T carriers. This risk association was retained in subgroup analyses by tumor histotype and location. Genomic DNA hypomethylation status in MTHFR677T/T carriers was significantly higher than in subjects with wild-type MTHF677C/C genotype (p = 0.012). In the studied population, MTHFR677T played the role of a moderate-penetrance gastric cancer susceptibility allele. Possession of the MTHFR677T/T genotype was significantly associated with genomic DNA hypomethylation. These findings deserve further investigation in the context of novel strategies for gastric cancer prevention.


Subject(s)
DNA Methylation , DNA, Neoplasm/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Disease Susceptibility , Female , Humans , Italy/epidemiology , Male , Middle Aged , Risk Factors , Stomach Neoplasms/enzymology , Stomach Neoplasms/epidemiology
6.
Tumour Biol ; 25(3): 106-10, 2004.
Article in English | MEDLINE | ID: mdl-15361706

ABSTRACT

Loss of E-cadherin expression has been related with an adverse outcome in patients with resected gastric cancer. More recently, experimental models with cancer cell lines showed that chemosensitivity may be affected by the E-cadherin expression status. We investigated whether E-cadherin expression is correlated with the response to chemotherapy and the survival of patients with advanced gastric cancer. Consecutive patients with advanced gastric cancer who underwent palliative chemotherapy were considered eligible for study entry. Measurable disease, complete follow-up information and availability of tumor specimens for immunohistochemistry were mandatory inclusion criteria. In 70 assessable patients, 30 patients had locoregional disease and 40 patients had visceral metastases. Chemotherapy consisted of cisplatin/fluorouracil/folinic acid in 33 patients and cisplatin/fluorouracil/epirubicin/folinic acid in 37 patients. There were 13 patients with complete response, 20 with partial response, 20 with stable disease and 17 patients progressed. Thirty-eight patients had > 80% E-cadherin-positive cancer cells (positive E-cadherin expression); 15 cases showed 25-70% (reduced E-cadherin expression), and in the remaining 17 cases E-cadherin expression was < 10% (negative E-cadherin expression). The response to chemotherapy was unrelated to the E-cadherin expression status. Conversely, survival in the 32 patients with reduced/negative E-cadherin expression (25 weeks) was significantly worse than that observed in the 38 patients with preserved E-cadherin expression (36 weeks) (p < 0.01). E-cadherin expression retained its independent prognostic role in the multivariate analysis. E-cadherin expression may give prognostic information in patients with advanced gastric cancer, but it does not seem to possess a predictive role in vivo. Some of the mechanisms inducing E-cadherin downregulation, like hypermethylation, may be potentially reversible, and they deserve further investigation as the target of novel therapeutic strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Cadherins/biosynthesis , Carcinoma/drug therapy , Carcinoma/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Adult , Aged , Cadherins/analysis , Carcinoma/genetics , Cisplatin/administration & dosage , Epirubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Immunohistochemistry , Leucovorin/administration & dosage , Male , Middle Aged , Multivariate Analysis , Palliative Care , Predictive Value of Tests , Prognosis , Retrospective Studies , Stomach Neoplasms/genetics , Survival Analysis
7.
Clin Cancer Res ; 10(8): 2784-9, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15102685

ABSTRACT

PURPOSE: Recent investigations have demonstrated that hypermethylation is a frequent mechanism for silencing tumor suppressor genes. This is a potentially reversible epigenetic change, and it is the target of a novel class of anticancer compounds with demethylating activity. Better understanding of the clinical implications of hypermethylation will allow the optimal planning of future trials with demethylating drugs. In this perspective, we investigated whether hypermethylation in the CDH1 promoter region is correlated with poor prognosis of patients with surgically resected, node-positive, diffuse gastric cancer. EXPERIMENTAL DESIGN: Consecutive cases of diffuse gastric cancer were considered eligible for study entry. Additional inclusion criteria were radical surgery with a minimum of D1 lymphadenectomy, complete follow-up information, and availability of tumor specimens for methylation-specific PCR and immunohistochemistry analyses. RESULTS: CDH1 promoter hypermethylation was found in 40 of 73 cases (54%), and it was significantly associated with worse prognosis. In patients with and without hypermethylation, the 5-year event-free survival rate was 30% and 62%, respectively, and the 5-year overall survival rate was 35% and 67%, respectively. CDH1 promoter hypermethylation retained its prognostic role for disease-free survival (P < 0.001) and overall survival (P < 0.001) in multivariate analysis. Immunohistochemistry showed a significant association between CDH1 methylation and E-cadherin expression (P < 0.001). CONCLUSIONS: This study shows adverse prognostic effect of CDH1 promoter hypermethylation in patients with diffuse gastric cancer. This form of cancer, and other types with frequent hypermethylation and silencing of critical tumor suppressor genes, would make appropriate targets for the testing of novel compounds with demethylating activity.


Subject(s)
Cadherins/biosynthesis , Cadherins/genetics , DNA Methylation , Promoter Regions, Genetic , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Adult , DNA Primers/chemistry , Disease-Free Survival , Female , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Stomach Neoplasms/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...