Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Am J Surg Pathol ; 45(3): 414-420, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33027072

ABSTRACT

Undifferentiated carcinoma of the esophagus and gastroesophageal junction is a recently recognized entity in the fifth edition of the World Health Organization Classification of Digestive Tumors and is diagnostically challenging, particularly on small biopsies. SMARCA4 and SMARCA2 are chromatin remodeling genes with key roles in oncogenesis. We retrieved 14 cases of SMARCA4/SMARCA2-deficient undifferentiated carcinoma of the gastroesophageal junction and esophagus from the authors' institutions. The tumors showed similar histologic findings: the sheet-like proliferation of tumor cells characterized by discohesion, large nuclei, and prominent macronucleoli with many tumor cells exhibiting a rhabdoid appearance. In 8 cases, adjacent specialized intestinal metaplasia was noted and 3 cases exhibited adjacent high-grade dysplasia. Immunohistochemically, tumors variably expressed keratins and disclosed loss of expression of SMARCA4 in 12 and SMARCA2 in 7 cases. In 2 cases SMARCA2 alone was lost without SMARCA4 loss. A mutant p53 immunohistochemical pattern was seen in 4 of 4 cases, 3 of which showed diffuse, strong nuclear expression, and 1 case displayed a complete loss of nuclear expression of p53, including invasive carcinoma and associated dysplasia, when present. Limited clinical follow-up was available, but 3 patients died of disease within 0.6, 2, and 7 months of diagnosis. We present the first series of undifferentiated carcinoma of the esophagus and gastroesophageal junction with this characteristic morphology associated with loss of SMARCA4 and/or SMARCA2 expression. This tumor type likely arises from dedifferentiation of a lower grade carcinoma in some cases, and Barrett esophagus and appears to be associated with an aggressive clinical course.


Subject(s)
Biomarkers, Tumor/deficiency , Carcinoma/enzymology , DNA Helicases/deficiency , Esophageal Neoplasms/enzymology , Esophagogastric Junction/enzymology , Nuclear Proteins/deficiency , Stomach Neoplasms/enzymology , Transcription Factors/deficiency , Aged , Aged, 80 and over , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/surgery , Cell Differentiation , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophagogastric Junction/pathology , Esophagogastric Junction/surgery , Female , Humans , Immunohistochemistry , Male , Middle Aged , New South Wales , Prognosis , Retrospective Studies , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Time Factors , United States
2.
J Clin Oncol ; 38(17): 1919-1927, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32208960

ABSTRACT

PURPOSE: This study evaluated the continuous use of trastuzumab beyond progression (TBP) in human epidermal growth factor receptor 2 (HER2)-positive advanced gastric or gastroesophageal junction (G/GEJ) cancer. PATIENTS AND METHODS: Patients with HER2-positive advanced G/GEJ cancer refractory to first-line chemotherapy with trastuzumab in combination with fluoropyrimidine and platinum were eligible. Patients were randomly assigned to the paclitaxel (80 mg/m2, days 1, 8, and 15, every 4 weeks) or paclitaxel with trastuzumab (PT; initially 8 mg/kg followed by 6 mg/kg, every 3 weeks) arms. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), response rate, and safety. Biomarkers such as HER2 expression status in tumor tissue after first-line treatment, HER2 amplification evaluated in serum cell-free DNA, and soluble HER2 levels were analyzed. RESULTS: Overall, 91 patients were allocated to the paclitaxel (n = 46) and PT (n = 45) arms. The median PFS in the paclitaxel and PT arms was 3.2 and 3.7 months, respectively (hazard ratio [HR], 0.91; 80% CI, 0.67 to 1.22; P = .33), and the median OS in both arms was 10 months (HR, 1.2; 95% CI, 0.75 to 2.0; P = .20). The overall response rates in the paclitaxel and PT arms were 32% and 33%, respectively (P = 1.00), and safety was comparable between the 2 arms. On exploratory analyses, HER2 positivity of tumor tissues was lost after first-line chemotherapy in 11 (69%) of 16 patients whose tumor tissues were available, and circulating HER2 DNA amplification was detected in 41 (60%) of 68 patients. However, no biomarkers associated with efficacy of TBP were found. CONCLUSION: The TBP strategy failed to improve PFS in patients with HER2-positive advanced G/GEJ cancer, and no beneficial biomarkers were found.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Paclitaxel/therapeutic use , Stomach Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Progression-Free Survival , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/blood , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Trastuzumab/administration & dosage , Trastuzumab/adverse effects
3.
Lancet Oncol ; 19(10): 1372-1384, 2018 10.
Article in English | MEDLINE | ID: mdl-30217672

ABSTRACT

BACKGROUND: Adding pertuzumab to trastuzumab and chemotherapy improves survival in HER2-positive early breast cancer and metastatic breast cancer. We assessed the efficacy and safety of pertuzumab versus placebo in combination with trastuzumab and chemotherapy in first-line HER2-positive metastatic gastric or gastro-oesophageal junction cancer. METHODS: JACOB was a double-blind, placebo-controlled, randomised, multicentre, phase 3 trial in patients aged 18 years or older with HER2-positive metastatic gastric or gastro-oesophageal junction cancer. Eligible patients had measurable or evaluable non-measurable disease at baseline, Eastern Cooperative Oncology Group performance status of 0 or 1, and baseline left ventricular ejection fraction of 55% or more. Patients at 197 oncology clinics (in 30 countries) were randomly assigned (1:1) to receive either pertuzumab (840 mg intravenously) or placebo every 3 weeks, with trastuzumab (8 mg/kg loading dose, then 6 mg/kg every 3 weeks intravenously), plus chemotherapy (cisplatin 80 mg/m2 every 3 weeks intravenously, oral capecitabine 1000 mg/m2 twice a day [2000 mg/m2 every 24 h] for 28 doses every 3 weeks, or 5-fluorouracil 800 mg/m2 every 24 h intravenously [120 h continuous infusion] every 3 weeks). Randomisation was by a central permuted block randomisation scheme (block size of 4) with an interactive voice or web response system, stratified by geographical region, previous gastrectomy, and HER2 positivity. The primary endpoint was overall survival in the intention-to-treat population. This trial is registered with Clinicaltrials.gov, number NCT01774786 (ongoing, but closed to enrolment). FINDINGS: Between June 10, 2013, and Jan 12, 2016, of 3287 patients assessed, 780 eligible patients were randomly assigned to receive either pertuzumab plus trastuzumab and chemotherapy (pertuzumab group, n=388) or placebo plus trastuzumab and chemotherapy (control group, n=392). Median duration of follow-up was 24·4 months (95% CI 22·3-26·1) in the pertuzumab group and 25·0 months (22·3-28·9) in the control group. After 242 deaths in the pertuzumab group and 262 deaths in the control group (the study was not stopped at this point), overall survival was not significantly different between treatment groups (median overall survival 17·5 months [95% CI 16·2-19·3] in the pertuzumab group and 14·2 months [12·9-15·5] in the control group; hazard ratio 0·84 [95% CI 0·71-1·00]; p=0·057). Serious adverse events occurred in 175 (45%) of 385 patients in the pertuzumab group and 152 (39%) of 388 patients in the control group. Diarrhoea was the most common serious adverse event in both groups (17 [4%] patients in the pertuzumab group vs 20 [5%] patients in the control group). The most common grade 3-5 adverse events were neutropenia (116 [30%] patients in the pertuzumab group vs 108 [28%] patients in the control group), anaemia (56 [15%] vs 65 [17%]), and diarrhoea (51 [13%] vs 25 [6%]). Treatment-related deaths occurred in seven (2%) patients in the control group; no treatment-related deaths occurred in the pertuzumab group. INTERPRETATION: Adding pertuzumab to trastuzumab and chemotherapy did not significantly improve overall survival in patients with HER2-positive metastatic gastric or gastro-oesophageal junction cancer compared with placebo. Further studies are needed to identify improved first-line treatment options in these types of cancer and to identify patients with HER2-driven tumours who might benefit from dual HER2-targeted therapy. FUNDING: F. Hoffmann-La Roche Ltd.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Esophagogastric Junction/drug effects , Protein Kinase Inhibitors/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Trastuzumab/administration & dosage , Adenocarcinoma/enzymology , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers, Tumor/analysis , Double-Blind Method , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Female , Humans , Male , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Receptor, ErbB-2/analysis , Stomach Neoplasms/enzymology , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Time Factors , Trastuzumab/adverse effects , Trastuzumab/pharmacokinetics
4.
J Clin Pathol ; 71(5): 388-394, 2018 May.
Article in English | MEDLINE | ID: mdl-29439009

ABSTRACT

The use of biologics targeted to the human epidermal growth factor receptor 2 (HER2) protein is the latest addition to the armamentarium used to fight advanced gastric or gastro-oesophageal junction adenocarcinoma. The decision to treat with the biologic trastuzumab is completely dependent on HER2 testing of tumour tissue. In 2017, the College of American Pathologists, American Society for Clinical Pathology and the American Society of Clinical Oncology jointly published guidelines for HER2 testing and clinical decision making in gastro-oesophageal adenocarcinoma. The Association of Clinical Pathologists Molecular Pathology and Diagnostics Committee has issued the following document as a commentary of these guidelines and, in parallel, to provide guidance on HER2 testing in National Health Service pathology departments within the UK. This guidance covers issues related to case selection, preanalytical aspects, analysis and interpretation of such HER2 testing.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/analysis , Esophageal Neoplasms/enzymology , Esophagogastric Junction/enzymology , Immunohistochemistry/standards , Molecular Diagnostic Techniques/standards , Pathology, Clinical/methods , Pathology, Molecular/standards , Receptor, ErbB-2/analysis , Societies, Medical/standards , Stomach Neoplasms/enzymology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Biopsy/standards , Clinical Decision-Making , Consensus , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/drug effects , Esophagogastric Junction/pathology , Humans , Molecular Targeted Therapy , Precision Medicine/standards , Predictive Value of Tests , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Reproducibility of Results , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trastuzumab/therapeutic use
5.
Cancer Invest ; 35(7): 463-472, 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28662341

ABSTRACT

BACKGROUND: This phase I/II study was designed to determine the maximum tolerated dose of tivantinib in combination with standard dose FOLFOX for the treatment of patients with advanced solid tumors and to evaluate the safety and efficacy of this combination for patients with previously untreated metastatic adenocarcinoma of the distal esophagus, gastroesophageal (GE) junction, or stomach. METHODS: Patients with advanced solid tumors for which FOLFOX would be appropriate chemotherapy received escalating doses of tivantinib BID (days 1-14) in a standard 3 + 3 design in phase I. In phase II, patients with advanced GE cancer received standard FOLFOX day 1 and tivantinib (360 mg PO BID) days 1-14 of each 2-week cycle. Restaging occurred every four cycles. The primary phase II endpoint was response rate (RR). RESULTS: Forty-nine patients were enrolled (15 on phase I and 34 on phase II). The expansion dose was established as tivantinib 360 mg BID in combination with FOLFOX. Thirty-two phase II patients were treated for a median of eight cycles (range, 1-38), with an overall RR of 38%. Treatment-related toxicities included neutropenia, fatigue, diarrhea, nausea, and peripheral neuropathy. Median progression-free survival (PFS) was 6.1 hmonths with a median time to progression of 7.0 months. Median overall survival was 9.6 months. Two patients remain on study at the time of this analysis. CONCLUSIONS: The combination treatment of tivantinib plus FOLFOX in patients with advanced GE cancer showed a response and PFS in the range of historical controls for first-line FOLFOX therapy. However, two patients had extended time on study treatment (36 and 45 cycles) at the time of data cutoff.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrrolidinones/administration & dosage , Quinolines/administration & dosage , Stomach Neoplasms/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-met/metabolism , Pyrrolidinones/adverse effects , Quinolines/adverse effects , Signal Transduction/drug effects , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Time Factors , Treatment Outcome , United States
6.
Hematol Oncol Clin North Am ; 31(3): 511-527, 2017 06.
Article in English | MEDLINE | ID: mdl-28501091

ABSTRACT

Gastroesophageal cancer (GEC) remains a major cause of cancer-related mortality worldwide. Although the incidence of distal gastric adenocarcinoma (GC) is declining in the United States, proximal esophagogastric junction adenocarcinoma (EGJ) incidence is rising. GC and EGJ, together, are treated uniformly in the metastatic setting as GEC. Overall survival in the metastatic setting remains poor, with few molecular targeted approaches having been successfully incorporated into routine care to date-only first-line anti-HER2 therapy for ERBB2 amplification and second-line anti-VEGFR2 therapy. This article reviews aberrations in epidermal growth factor receptor, MET, and ERBB2, their therapeutic implications, and future directions in targeting these pathways.


Subject(s)
Adenocarcinoma , Antineoplastic Agents/therapeutic use , Esophageal Neoplasms , Esophagogastric Junction , Stomach Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Gene Amplification , Humans , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Rate , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Surg Oncol Clin N Am ; 26(2): 293-312, 2017 04.
Article in English | MEDLINE | ID: mdl-28279470

ABSTRACT

Gastroesophageal cancer (GEC) remains a major cause of cancer-related mortality worldwide. Although the incidence of distal gastric adenocarcinoma (GC) is declining in the United States, proximal esophagogastric junction adenocarcinoma (EGJ) is increasing in incidence. GEC, including GC and EGJ, is treated uniformly in the metastatic setting. Overall survival in the metastatic setting remains poor. Molecular characterization of GEC has identified mutations and copy number variations, along with other oncogenes, biomarkers, and immuno-oncologic checkpoints that may serve as actionable therapeutic targets. This article reviews these key aberrations, their impact on protein expression, therapeutic implications, and clinical directions within each pathway.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Esophagogastric Junction , Molecular Targeted Therapy/methods , Stomach Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antineoplastic Agents , DNA Copy Number Variations , Disease-Free Survival , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Humans , Neoplasm Staging , Proto-Oncogene Proteins c-met , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Rate
8.
J Clin Oncol ; 35(4): 446-464, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28129524

ABSTRACT

Context ERBB2 (erb-b2 receptor tyrosine kinase 2 or HER2) is currently the only biomarker established for selection of a specific therapy for patients with advanced gastroesophageal adenocarcinoma (GEA). However, there are no comprehensive guidelines for the assessment of HER2 in patients with GEA. Objectives To establish an evidence-based guideline for HER2 testing in patients with GEA, formalize the algorithms for methods to improve the accuracy of HER2 testing while addressing which patients and tumor specimens are appropriate, and to provide guidance on clinical decision making. Design The College of American Pathologists (CAP), American Society for Clinical Pathology (ASCP), and the American Society of Clinical Oncology (ASCO) convened an Expert Panel to conduct a systematic review of the literature to develop an evidence-based guideline with recommendations for optimal HER2 testing in patients with GEA. Results The Panel is proposing 11 recommendations with strong agreement from the open comment participants. Recommendations The Panel recommends that tumor specimen(s) from all patients with advanced GEA, who are candidates for HER2-targeted therapy, should be assessed for HER2 status before the initiation of HER2-targeted therapy. Clinicians should offer combination chemotherapy and an HER2-targeted agent as initial therapy for all patients with HER2-positive advanced GEA. For pathologists, guidance is provided for morphologic selection of neoplastic tissue, testing algorithms, scoring methods, interpretation and reporting of results, and laboratory quality assurance. Conclusion This guideline provides specific recommendations for assessment of HER2 in patients with advanced GEA while addressing pertinent technical issues and clinical implications of the results.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , Esophageal Neoplasms , Esophagogastric Junction , Receptor, ErbB-2 , Stomach Neoplasms , Humans , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Algorithms , Biomarkers, Tumor/analysis , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Receptor, ErbB-2/analysis , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Systematic Reviews as Topic
9.
World J Gastroenterol ; 22(19): 4619-25, 2016 May 21.
Article in English | MEDLINE | ID: mdl-27217694

ABSTRACT

Human epidermal growth factor receptor 2 (HER2) overexpression is increasingly recognized as a frequent molecular abnormality in gastric and gastroesophageal cancer. With the recent introduction of HER2 molecular targeted therapy for patients with advanced gastric cancer, determination of HER2 status is crucial in order to select patients who may benefit from this treatment. This paper provides an update on our knowledge of HER2 in gastric and gastroesophageal cancer, including the prognostic relevance of HER2, the key differences between HER2 protein expression interpretation in breast and gastric cancer, the detection methods and the immunohistochemistry scoring system.


Subject(s)
Biomarkers, Tumor/metabolism , Esophagogastric Junction/enzymology , Immunohistochemistry , Receptor, ErbB-2/metabolism , Stomach Neoplasms/enzymology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Esophagogastric Junction/drug effects , Esophagogastric Junction/pathology , Humans , In Situ Hybridization, Fluorescence , Molecular Targeted Therapy , Predictive Value of Tests , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Reproducibility of Results , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trastuzumab/therapeutic use , Treatment Outcome
10.
Int J Cancer ; 139(7): 1626-31, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27198655

ABSTRACT

Although HER2-positive breast cancers demonstrate a propensity for central nervous system (CNS) metastasis, it is unknown whether other HER2-positive tumors, including adenocarcinomas of the esophagus/gastroesophageal junction (EAC), share this characteristic. Insight into this association may inform the development of HER2-targeted therapies that penetrate the blood-brain barrier. We examined HER2 overexpression and gene amplification in 708 patients with EAC who underwent curative-intent surgery during a time period (1980-1997) when no patient received HER2-targeted therapy. We identified patients whose site of first cancer recurrence was CNS and those who had a CNS relapse at any time. After a median follow-up of 61.2 months, 3.4% (24/708) of patients developed CNS relapse (all involved the brain). Patients with HER2-positive (vs -negative) primary tumors showed a higher 5-year cumulative incidence of CNS relapse as first recurrence (5.8% vs. 1.2%; p = 0.0058) and at any time (8.3% vs. 2.4%; p = 0.0062). In a multivariable model that included covariates previously associated with HER2 or with CNS relapse in breast cancer, HER2 positivity was the only variable that was statistically significantly associated with shorter time to CNS relapse as first recurrence (p = 0.0026) or at any time (hazard ratio 4.3 [95% confidence interval 1.8 to 10.3]; p = 0.001). These are the first data in a non-breast cancer to demonstrate an association between HER2 positivity and higher CNS relapse risk after surgery, and suggest that HER2-positive EACs have a predilection for CNS metastases.


Subject(s)
Adenocarcinoma/enzymology , Central Nervous System Neoplasms/enzymology , Esophageal Neoplasms/enzymology , Esophagogastric Junction/pathology , Receptor, ErbB-2/biosynthesis , Stomach Neoplasms/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Gene Amplification , Humans , Immunohistochemistry , Middle Aged , Proportional Hazards Models , Receptor, ErbB-2/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
11.
Eur Rev Med Pharmacol Sci ; 20(3): 476-85, 2016.
Article in English | MEDLINE | ID: mdl-26914122

ABSTRACT

OBJECTIVE: Reflux is the principal complication for patients after esophagectomy with gastric reconstruction. The aim of this study was to investigate the effect of the modified Nissen fundoplication after resection of adenocarcinoma from the esophagogastric junction (AEG) on controlling the reflux and the role of duodenogastroesophageal reflux (DGER) and cyclooxygenase-2 (COX-2) expression level in the remnant esophagus. PATIENTS AND METHODS: Sixty patients with AEG were randomly divided into two groups: (i) the conventional anastomosis group and (ii) the anti-reflux anastomosis group. Fifty esophagectomized patients were invited to participate in postoperative follow-up after 6 to 12 months. Among those we had 29 cases in the conventional anastomosis group and 21 in the anti-reflux anastomosis group. We used endoscopy, simultaneous 24 hours esophageal pH and bilirubin monitoring in this study. The COX-2 expression level in the remnant esophagus was detected using real-time PCR. RESULTS: The reflux esophagitis prevalence in anti-reflux anastomosis group was comparable to that in the conventional group (p = 0.154). DeMeester score and fraction time of bilirubin abs >0.14 decreased more intensely in the anti-reflux anastomosis group (p < 0.05). The COX-2 expression level in of anti-reflux anastomosis group was evidently lower than that in the conventional anastomosis group (p = 0.022) while it was meaningfully higher compared to the normal control group (p = 0.046). COX-2 up-regulation as well as high prevalence of esophagitis were observed in simultaneous acid reflux and DGER (p < 0.05). CONCLUSIONS: Although modified fundoplication following resection of AEG did not achieve an optimal effect on controlling reflux, it was very effective in decreasing the reflux. COX-2 expression monitoring can be considered as a possible new way to evaluate the impact of anti-reflux surgery. DGER occurring in acidic environment could develop severe reflux esophagitis and up-regulate the COX-2 expression.


Subject(s)
Cyclooxygenase 2/biosynthesis , Duodenogastric Reflux/enzymology , Esophageal Neoplasms/enzymology , Esophagectomy/methods , Gastroesophageal Reflux/enzymology , Stomach Neoplasms/enzymology , Adult , Aged , Anastomosis, Surgical/methods , Duodenogastric Reflux/surgery , Esophageal Neoplasms/surgery , Esophagogastric Junction/enzymology , Esophagogastric Junction/surgery , Female , Fundoplication/methods , Gastroesophageal Reflux/surgery , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Postoperative Complications/etiology , Stomach Neoplasms/surgery
12.
World J Gastroenterol ; 20(16): 4526-35, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24782605

ABSTRACT

Amplification of the human epidermal growth factor receptor 2 (HER2) gene and overexpression of the HER2 protein is found in 15%-20% of patients with gastric and gastroesophageal junction cancer. The degree of HER2 overexpression and amplification varies with the location of the carcinoma, with higher expression in the gastroesophageal and proximal parts compared to the distal parts of the stomach. Further, HER2 overexpression and amplification also seems to be related to the Lauren histological classification, with higher levels found in the intestinal phenotype compared to the diffuse and mixed types. The prognostic properties of HER2 overexpression and amplification are still under debate, but a large number of studies seem to indicate that HER2 is a negative prognostic factor. The usefulness of HER2 targeted therapy in gastric cancer was demonstrated in the ToGA trial, where HER2-positive patients with advanced gastric and gastroesophageal junction adenocarcinoma were randomized to receive 5-FU/capecitabine and cisplatin, either alone or in combination with trastuzumab. A statically significant gain in overall survival was seen in patients who received the combined treatment of trastuzumab and chemotherapy. Patients with a strong overexpression of the HER2 protein (IHC3+) specifically benefited from the treatment, with a median overall survival of 17.9 mo. As a consequence of the positive results of the ToGA trial, patients with advanced gastric or gastroesophageal junction adenocarcinoma are now routinely tested for HER2. The ToGA trial must be characterized as a landmark in the treatment of gastric cancer and it has paved the way for a number of new HER2 targeted compounds such as pertuzumab, ado-trastuzumab emtansine, lapatinib, afatinib, and dacomitinib, which are currently undergoing phase II and III clinical testing. Overall, this review will discuss the current status of HER2 in gastric and gastroesophageal junction cancer and the future direction in relation to HER2 target therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Esophagogastric Junction/drug effects , Stomach Neoplasms/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Treatment Outcome
13.
BMC Cancer ; 12: 524, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23153332

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR) is a potential target of anticancer therapy in gastric cancer. However, its prognostic role in metastatic gastric or gastroesophageal junction (GE) cancer has not been established yet. METHODS: EGFR status was analyzed by immunohistochemistry (IHC) in paraffin-embedded samples from 357 patients who received chemotherapy in 4 first-line trials. Automated RNA extraction from paraffin and RT-quantitative PCR were additionally used to evaluate EGFR mRNA expression in 130 patients. RESULTS: EGFR protein expression (any grade) and overexpression (3+) were observed in 43% and 11% of patients, respectively. EGFR positivity correlated with intestinal type histology (p = 0.05), but not with other clinicopathologic characteristics. Median follow-up was 18.2 months. Median overall survival (OS) was similar in patients with EGFR positive vs. those with EGFR negative tumors, regardless whether positivity was defined as ≥1+ (10.6 vs. 10.9 months, p = 0.463) or as 3+ (8.6 vs. 10.8 months, p = 0.377). The multivariate analysis indicated that EGFR status is not an independent prognostic factor (hazard ratio 0.85, 0.56 to 1.12, p = 0.247). There were also no significant differences in overall survival when patients were categorized according to median (p = 0.116) or quartile (p = 0.767) distribution of EGFR mRNA gene expression. Similar distributions of progression-free survival according to EGFR status were observed. CONCLUSIONS: Unlike different cancer types where EGFR-positive disease is associated with an adverse prognostic value, EGFR positivity is not prognostic of patient outcome in metastatic gastric or GE cancer.


Subject(s)
Biomarkers, Tumor/metabolism , ErbB Receptors/metabolism , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Biomarkers, Tumor/genetics , Disease-Free Survival , ErbB Receptors/genetics , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Female , Follow-Up Studies , Humans , Male , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics
14.
Int J Clin Exp Pathol ; 5(4): 278-89, 2012.
Article in English | MEDLINE | ID: mdl-22670171

ABSTRACT

Human aldo-keto reductase family 1 member C3 (AKR1C3) was initially identified as a critical enzyme in reducing 5α-dihydrotestosterone (5α-DHT) to 5α-androstane-3α,17ß-diol (3α-diol) and oxidizing 3α-diol to androsterone. Based on these enzymatic activities, AKR1C3 was originally named type 2 3α-hydroxysteroid dehydrogenase (HSD)/type 5 17ß-HSD. Additionally, AKR1C3 was demonstrated to be capable of metabolizing other steroids including estrogen and progesterone. Subsequently, AKR1C3 was shown to possess 11-ketoprostaglandin reductase activity in metabolizing prostaglandins and dihydrodiol dehydrogenase x (DDx) activity in metabolizing xenobiotics. Tissue distribution of AKR1C3 has been detected in both sex hormone-dependent organs such as the testis, breast, endometrium, and prostate as well as sex hormone-independent organs including the kidney and urothelium. Although prominent expression of AKR1C isozymes has been reported in human non-small cell lung carcinoma (NSCLC), the expression of AKR1C3 in small cell carcinoma of the lung has not been described. Also, the expression of AKR1C3 in normal lung has not been described. In this study, we demonstrated strong AKR1C3 immunoreactivity in bronchial epithelium but not in bronchial glands or alveolar pneumocytes. Strong AKR1C3 immunoreactivity was also demonstrated in columnar epithelium but only weak immunoreactivity in squamous epithelium of the gastrointestinal junction. Although AKR1C3 immunoreactivity was absent in small cell carcinoma of the lung, positive AKR1C3 immunoreactivity was extensively present in both adenocarcinoma and squamous cell carcinoma arising from the lung and the gastroesophageal junction. AKR1C3 may serve as an adjunct marker for differentiating small cell carcinoma from NSCLC. However, roles of AKR1C3 in adenocarcinoma, squamous cell carcinoma, and small cell carcinoma pathogenesis require further studies.


Subject(s)
3-Hydroxysteroid Dehydrogenases/analysis , Adenocarcinoma/enzymology , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Esophagogastric Junction/enzymology , Hydroxyprostaglandin Dehydrogenases/analysis , Lung Neoplasms/enzymology , Small Cell Lung Carcinoma/enzymology , Stomach Neoplasms/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3 , Carcinoma, Squamous Cell/pathology , Diagnosis, Differential , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Middle Aged , Oklahoma , Predictive Value of Tests , Small Cell Lung Carcinoma/pathology , Stomach Neoplasms/pathology
15.
Klin Khir ; (3): 51-3, 2008 Mar.
Article in Ukrainian | MEDLINE | ID: mdl-18681000

ABSTRACT

Experimental investigation on studying of pro- and antioxidant state in esophageal tissues was performed. It was established, that accumulation of products of the proteins and lipids oxidation during inflammatory process course coincides with significant lowering of the antioxidant enzymes activity. The data obtained trusts the various diagnostic significance of diienic conjugates and malonic dialdehyde (MDA) levels. The increase of MDA concentration as well as of products of the oxidational modifications proteins, owing basic character, was revealed as a typical biochemical marker of acid-peptic injury of esophageal tissues.


Subject(s)
Antioxidants/metabolism , Esophagitis, Peptic/metabolism , Esophagogastric Junction , Free Radicals/metabolism , Animals , Disease Models, Animal , Esophagitis, Peptic/enzymology , Esophagogastric Junction/enzymology , Esophagogastric Junction/metabolism , Lipid Peroxides/metabolism , Rats , Rats, Wistar
16.
Pathol Res Pract ; 203(12): 831-7, 2007.
Article in English | MEDLINE | ID: mdl-17993370

ABSTRACT

OBJECTIVES: Metaplastic glandular mucosa with goblet cells at the squamocolumnar junction is induced either by reflux or by Helicobacter infection. We investigated whether the accompanying inflammation may give information about the etiology of these metaplastic changes and whether there are further criteria which are helpful in differentiating Helicobacter-induced vs. reflux-caused metaplasia. METHODS: One hundred and nine patients with intestinal metaplasia diagnosed in biopsies obtained immediately below the Z-line were evaluated. Further biopsies were taken from the gastric body and antrum. Patients were diagnosed as having a normal Z-line, or as showing short tongues or segments of Barrett's esophagus endoscopically. Inflammation was graded according to the updated Sydney-system. Metaplasia was typed using Gomori's-aldehyde-fuchsin-Alcianblue staining. RESULTS: Compared to patients with Barrett's esophagus, the active (p=0.0002) and chronic inflammation (p=0.0004) at the squamocolumnar junction was higher in patients with a normal Z-line and frequently accompanied by lymphoid aggregates (p<0.0001) and regular cardia- (p=0.0044) and/or corpus-type glands (p=0.0004). Pseudogoblet cells were more frequent in Barrett's esophagus (p=0.0159). CONCLUSIONS: The endoscopic aspect of the Z-line, the inflammatory pattern, and the type of glands in biopsies from the squamocolumnar junction, as well as the presence of pseudogoblet cells are helpful tools in distinguishing Barrett's mucosa from Helicobacter-associated intestinal metaplasia.


Subject(s)
Barrett Esophagus/diagnosis , Esophagitis, Peptic/diagnosis , Esophagogastric Junction/pathology , Gastric Mucosa/pathology , Helicobacter Infections/diagnosis , Adult , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Biomarkers/metabolism , Biopsy , Diagnosis, Differential , Endoscopy, Gastrointestinal , Esophagogastric Junction/enzymology , Female , Goblet Cells/metabolism , Goblet Cells/pathology , Helicobacter pylori/cytology , Helicobacter pylori/enzymology , Helicobacter pylori/isolation & purification , Humans , Male , Metaplasia/etiology , Metaplasia/pathology , Middle Aged , Urease/metabolism
17.
Gastroenterology ; 130(4): 1191-206, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16618413

ABSTRACT

BACKGROUND & AIMS: Although progression to adenocarcinoma at the gastroesophageal junction reflects exposure to acid and bile acids associated with reflux, mechanisms mediating this transformation remain undefined. Guanylyl cyclase C (GC-C), an intestine-specific tumor suppressor, may represent a mechanism-based marker and target of transformation at the gastroesophageal junction. The present studies examine the expression of GC-C in normal tissues and tumors from esophagus and stomach and mechanisms regulating its expression by acid and bile acids. METHODS: Gene expression was examined by reverse-transcription polymerase chain reaction, promoter analysis, immunohistochemistry, immunoblotting, and functional analysis. Promoter transactivation was quantified by using luciferase constructs and mutational analysis. DNA binding of transcription factors was examined by electromobility shift analysis. RESULTS: GC-C mRNA and protein were ectopically expressed in approximately 80% of adenocarcinomas arising in, but not in normal, esophagus and stomach. Similarly, in OE19 human esophageal cancer cells, deoxycholate and acid induced expression of GC-C. This was associated with the induction of expression of Cdx2, a transcription factor required for GC-C expression. In turn, induction of Cdx2 expression by deoxycholate was mediated by binding sites in the proximal promoter for nuclear factor kappaB (NF-kappaB). Furthermore, deoxycholate increased NF-kappaB activity, associated with nuclear translocation and Cdx2 promoter binding of the NF-kappaB subunit p50. Moreover, a dominant negative construct for NF-kappaB prevented deoxycholate-induced p50 nuclear translocation and activation of the Cdx2 promoter. CONCLUSIONS: Transformation associated with reflux at the gastroesophageal junction reflects activation by bile acid and acid of a transcriptional program involving NF-kappaB and Cdx2, which mediate intestinal metaplasia and ectopic expression of GC-C.


Subject(s)
Bile Acids and Salts/pharmacology , Cell Transformation, Neoplastic , Esophagus/metabolism , Guanylate Cyclase/metabolism , Homeodomain Proteins/metabolism , Intestines/enzymology , NF-kappa B/metabolism , Receptors, Peptide/metabolism , Adenocarcinoma/enzymology , CDX2 Transcription Factor , Cell Line, Tumor , Deoxycholic Acid/pharmacology , Esophageal Neoplasms/enzymology , Esophagogastric Junction/enzymology , Esophagogastric Junction/metabolism , Esophagus/enzymology , Esophagus/pathology , Gene Expression , Guanylate Cyclase/genetics , Homeodomain Proteins/genetics , Humans , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Receptors, Peptide/genetics , Tissue Distribution/drug effects , Transcription, Genetic/drug effects
18.
Neoplasia ; 7(9): 854-61, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16229808

ABSTRACT

Chronic gastroesophageal reflux disease is a known risk factor for Barrett's esophagus (BE), which induces oxidative mucosal damage. Glutathione peroxidase-3 (GPx3) is a secretory protein with potent extracellular antioxidant activity. In this study, we have investigated the mRNA and protein expression of GPx3, and explored promoter hypermethylation as an epigenetic mechanism for GPx3 gene inactivation during Barrett's carcinogenesis. Quantitative real-time reverse transcription polymerase chain reaction on 42 Barrett's adenocarcinomas (BAs) revealed consistently reduced levels of GPx3 mRNA in 91% of tumor samples. GPx3 promoter hypermethylation was detected in 62% of Barrett's metaplasia, 82% of dysplasia, and 88% of BA samples. Hypermethylation of both alleles of GPx3 was most frequently seen in BAs (P = .001). Immunohistochemical staining of GPx3 in matching tissue sections (normal, BE, Barrett's dysplasia, and BA) revealed strong immunostaining for GPx3 in normal esophageal and gastric tissues. However, weak to absent GPx3 staining was observed in Barrett's dysplasia and adenocarcinoma samples where the promoter was hypermethylated. The degree of loss of immunohistochemistry correlated with the hypermethylation pattern (monoallelic versus biallelic). The observed high frequency of promoter hypermethylation and progressive loss of GPx3 expression in BA and its associated lesions, together with its known function as a potent antioxidant, suggest that epigenetic inactivation and regulation of glutathione pathway may be critical in the development and progression of BE.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glutathione Peroxidase/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Barrett Esophagus/enzymology , Barrett Esophagus/pathology , DNA Methylation , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagogastric Junction/enzymology , Esophagogastric Junction/pathology , Gastric Mucosa/metabolism , Glutathione Peroxidase/metabolism , Humans , Metaplasia , Promoter Regions, Genetic , RNA, Messenger/metabolism
19.
Am J Physiol Cell Physiol ; 287(2): C384-94, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15128504

ABSTRACT

Contraction of smooth muscle depends on the balance of myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) activities. Because MLCK activation depends on the activation of calmodulin, which requires a high Ca(2+) concentration, phosphatase inhibition has been invoked to explain contraction at low cytosolic Ca(2+) levels. The link between activation of the Ca(2+)-independent protein kinase Cepsilon (PKCepsilon) and MLC phosphorylation observed in the esophagus (ESO) (Sohn UD, Cao W, Tang DC, Stull JT, Haeberle JR, Wang CLA, Harnett KM, Behar J, and Biancani P. Am J Physiol Gastrointest Liver Physiol 281: G467-G478, 2001), however, has not been elucidated. We used phosphatase and kinase inhibitors and antibodies to signaling enzymes in combination with intact and saponin-permeabilized isolated smooth muscle cells from ESO and lower esophageal sphincter (LES) to examine PKCepsilon-dependent, Ca(2+)-independent signaling in ESO. The phosphatase inhibitors okadaic acid and microcystin-LR, as well as an antibody to the catalytic subunit of type 1 protein serine/threonine phosphatase, elicited similar contractions in ESO and LES. MLCK inhibitors (ML-7, ML-9, and SM-1) and antibodies to MLCK inhibited contraction induced by phosphatase inhibition in LES but not in ESO. The PKC inhibitor chelerythrine and antibodies to PKCepsilon, but not antibodies to PKCbetaII, inhibited contraction of ESO but not of LES. In ESO, okadaic acid triggered translocation of PKCepsilon from cytosolic to particulate fraction and increased activity of integrin-linked kinase (ILK). Antibodies to the mitogen-activated protein (MAP) kinases ERK1/ERK2 and to ILK, and the MAP kinase kinase (MEK) inhibitor PD-98059, inhibited okadaic acid-induced ILK activity and contraction of ESO. We conclude that phosphatase inhibition potentiates the effects of MLCK in LES but not in ESO. Contraction of ESO is mediated by activation of PKCepsilon, MEK, ERK1/2, and ILK.


Subject(s)
Esophagogastric Junction/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle Contraction/physiology , Muscle, Smooth/enzymology , Protein Kinase C/metabolism , Animals , Antibodies , Cats , Enzyme Inhibitors/pharmacology , Female , Male , Marine Toxins , Microcystins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/enzymology , Myosin-Light-Chain Kinase/metabolism , Okadaic Acid/pharmacology , Peptides, Cyclic/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/immunology , Phosphoprotein Phosphatases/metabolism , Protein Kinase C-epsilon , Protein Phosphatase 1 , Protein Serine-Threonine Kinases/metabolism
20.
Dis Esophagus ; 16(4): 312-4, 2003.
Article in English | MEDLINE | ID: mdl-14641295

ABSTRACT

The present study explores the effects of nitric oxide synthase inhibitor on esophageal motility in a feline model with esophagitis. Perfusion of the esophagus with acid produced inflammatory changes of esophageal mucosa. The esophageal motility was measured before and after the perfusion. One group of cats was given nitric oxide inhibitor orally at the same time as the perfusion of acid. The control group was given water instead. Esophagitis impairs the motility of the esophagus. However, the esophageal motility of the cats that were given nitric oxide synthase inhibitor decreased less than that of the control group. The results suggested that during esophagitis there is an alteration of the nitric oxide synthase/nitric oxide pathway in the esophagus, which may be one of the important mechanisms of esophageal motility dysfunction.


Subject(s)
Enzyme Inhibitors/pharmacology , Esophagitis/physiopathology , Esophagogastric Junction/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Animals , Cats , Esophagitis/chemically induced , Esophagitis/enzymology , Esophagogastric Junction/enzymology , Esophagogastric Junction/physiopathology , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Hydrochloric Acid , Male , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...