Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Oncol (Dordr) ; 42(5): 705-715, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31250364

ABSTRACT

PURPOSE: The cyclin-dependent kinases (CDKs) CDK4 and CDK6 are important regulators of the cell cycle and represent promising targets in cancer treatment. We aimed to investigate the relevance of CDK4/6 in the development of hepatocellular carcinoma (HCC) and the potential of ribociclib, a novel orally available CDK4/6 inhibitor, as a treatment for HCC. METHODS: The effect of ribociclib was assessed in native and sorafenib-resistant HCC cell lines using viability assays, colony formation assays and FACS-based analyses. The expression of potential biomarkers of ribociclib response was assessed in cell lines and primary human hepatocytes using Western blotting. In addition, the prognostic relevance of the cyclin D-CDK4/6-retinoblastoma protein (Rb) pathway was assessed by analysing mRNA expression data from The Cancer Genome Atlas (TCGA). RESULTS: We found that ribociclib downregulated Rb and caused a profound loss of cell viability by inducing G1 cell cycle arrest in HCC cell lines exhibiting Rb-high/p16-low protein expression profiles, but not in Rb-low/p16-high cells, regardless their sensitivity to sorafenib. siRNA-based Rb silencing decreased cell proliferation, but did not diminish the sensitivity of HCC cells to ribociclib. Furthermore, we found that ribociclib synergized with sorafenib to cause cell death. mRNA analysis of primary human HCC specimens showed that CDK4 expression was correlated with patient survival and that the expression of Rb and the p16-encoding CDKN2A gene were inversely correlated. CONCLUSIONS: From our data we conclude that impairment of the cyclin D-CDK4/6-Rb pathway is a frequent feature of HCC and that it is associated with a unfavourable prognosis. We also found that ribociclib exhibits a preferential antineoplastic activity in Rb-high HCC cells. Our results warrant further investigation of Rb and p16 expression as markers of HCC sensitivity to ribociclib.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Liver Neoplasms/metabolism , Purines/pharmacology , Retinoblastoma Protein/metabolism , Apoptosis/drug effects , Biomarkers, Tumor , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Drug Resistance, Neoplasm , G1 Phase Cell Cycle Checkpoints/drug effects , Hepatocytes/drug effects , Humans , Liver Neoplasms/mortality , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Sorafenib/pharmacology
2.
Rev Endocr Metab Disord ; 19(2): 145-158, 2018 06.
Article in English | MEDLINE | ID: mdl-29464446

ABSTRACT

Neuroendocrine tumors (NETs) are a group of rare and heterogeneous malignancies that can develop in various organs. A significant number of gastroenteropancreatic neuroendocrine tumours (GEP-NETs) is functionally active and presents with symptoms related to the secretion of biologically active substances, leading to the development of distinct clinical syndromes. There are various therapeutic approaches for GEP-NETs, including curative surgery, palliative surgery, local-ablative and loco-regional therapies as well as systemic therapeutic options including peptide receptor radionuclide therapy, cytotoxic therapy, and molecularly targeted therapies. Specific supportive therapy of patients with NETs includes management or prevention of hormone-related clinical syndromes and paraneoplastic states. Supportive therapy plays a key role in NET treatment. Supportive therapy includes debulking surgery and interventional radiologic techniques to reduce tumour bulk or load, as well as systemic medical treatment options to manage or prevent hypersecretion syndromes and treatment-related side effects. Supportive therapies are a type of of comprehensive treatment addressing the patient as a whole person throughout the process of NET treatment. Therefore, supportive therapy also encompasses psychosocial support, expert nursing, nutritional support and management of cancer related pain.


Subject(s)
Gastrinoma/therapy , Glucagonoma/therapy , Insulinoma/therapy , Intestinal Neoplasms/therapy , Malignant Carcinoid Syndrome/therapy , Neuroendocrine Tumors/therapy , Palliative Care/methods , Pancreatic Neoplasms/therapy , Paraneoplastic Syndromes/therapy , Stomach Neoplasms/therapy , Vipoma/therapy , Humans
3.
Neuroendocrinology ; 103(3-4): 383-401, 2016.
Article in English | MEDLINE | ID: mdl-26338447

ABSTRACT

BACKGROUND/AIMS: The hepatocyte growth factor/transmembrane tyrosine kinase receptor c-Met has been defined as a potential target in antitumoral treatment of various carcinomas. We aimed to investigate the direct effect of c-Met inhibition on neuroendocrine tumor cells in vitro. METHODS: The effects of the multi-tyrosine kinase inhibitors cabozantinib and tivantinib and of the highly specific c-Met inhibitor INC280 were investigated in human pancreatic neuroendocrine BON1, bronchopulmonary NCI-H727 and midgut GOT1 cells in vitro. RESULTS: INC280, cabozantinib and tivantinib inhibited c-Met phosphorylation, respectively. However, while equimolar concentrations (10 µM) of cabozantinib and tivantinib inhibited cell viability and cell migration, INC280 had no inhibitory effect. Knockdown experiments with c-Met siRNA also did not demonstrate effects on cell viability. Cabozantinib and tivantinib caused a G2 arrest in neuroendocrine tumor cells. CONCLUSIONS: Our in vitro data suggest that c-Met inhibition alone is not sufficient to exert direct antitumoral or antimigratory effects in neuroendocrine tumor cells. The multi-tyrosine kinase inhibitors cabozantinib and tivantinib show promising antitumoral and antimigratory effects in neuroendocrine tumor cells, which are most probably 'off-target' effects, not mediated by c-Met.


Subject(s)
Anilides/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pyridines/pharmacology , Pyrrolidinones/pharmacology , Quinolines/pharmacology , Benzamides , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Flow Cytometry , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuroendocrine Tumors/pathology , Oncogene Protein v-akt/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Triazines/pharmacology
4.
Hormones (Athens) ; 13(4): 498-508, 2014.
Article in English | MEDLINE | ID: mdl-25402373

ABSTRACT

Metformin is a widely used oral antidiabetic drug with good tolerability. Recent studies suggest that it also possesses adjuvant potent anticancer properties in a variety of tumors. Neuroendocrine tumors (NETs) of the gastro-entero-pancreatic system (GEP) comprise a heterogeneous group of tumors with increasing incidence and limited effective therapeutic options. Here we report the antiproliferative effects of metformin in neuroendocrine tumor cells in vitro. Treatment of human pancreatic BON1, bronchopulmonary NCI-H727, and midgut GOT1 neuroendocrine tumor cells with increasing concentrations of metformin (0.1-10 mM) dose-dependently suppressed cell viability and cell counts. Metformin induced AMPK phosphorylation in pancreatic BON1 and midgut GOT1 but suppressed AMPK activity in bronchopulmonary NCI-H727. Thus, AMPK-dependent and AMPK-independent properties may be operative in NETs of different origin. Metformin suppressed mTORC1 signaling in all three tumor cell types, evidenced by suppression of 4EBP1, pP70S6K, and S6 phosphorylation, and was associated with compensatory AKT activity. We observed induction of ERK phosphorylation in BON1 and NCI-H727 and inhibition of ERK in midgut GOT1 cells, while all three tumor cell types responded with induction of GSK3 phosphorylation. This suggests a central role for GSK3 in metformin-mediated signal transduction. Inhibition of cell proliferation by metformin was associated with apoptosis induction only in midgut GOT1, evidenced by increased subG0/1 fraction and PARP cleavage. These results suggest a potential role of metformin as a (adjuvant) therapeutic for patients with NETs.


Subject(s)
Antineoplastic Agents/pharmacology , Metformin/pharmacology , Neuroendocrine Cells/drug effects , Neuroendocrine Tumors/pathology , AMP-Activated Protein Kinases/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Neuroendocrine Cells/pathology , Neuroendocrine Cells/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
5.
World J Gastroenterol ; 20(29): 10038-49, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25110431

ABSTRACT

AIM: To investigate the effect of aspirin on neuroendocrine tumor (NET) cell growth and signaling in vitro. METHODS: Human pancreatic BON1, bronchopulmonary NCI-H727 and midgut GOT1 neuroendocrine tumor cells were treated with different concentrations of aspirin (from 0.001 to 5 mmol/L), and the resulting effects on metabolic activity/cell proliferation were measured using cell proliferation assays and SYBR-DNA-labeling after 72, 144 and 216 h of incubation. The effects of aspirin on the expression and phosphorylation of several critical proteins that are involved in the most common intracellular growth factor signaling pathways (especially Akt protein kinase B) and mammalian target of rapamycin (mTOR) were determined by Western blot analyses. Propidium iodide staining and flow cytometry were used to evaluate changes in cell cycle distribution and apoptosis. Statistical analysis was performed using a 2-tailed Student's t-test to evaluate the proliferation assays and cell cycle analyses. The results are expressed as the mean ± SD of 3 or 4 independently performed experiments. Statistical significance was set at P < 0.05. RESULTS: Treatment with aspirin suppressed the viability/proliferation of BON1, NCI-H727 and GOT1 cells in a time- and dose-dependent manner. Significant effects were observed at starting doses of 0.5-1 mmol/L and peaked at 5 mmol/L. For instance, after treatment with 1 mmol/L aspirin for 144 h, the viability of pancreatic BON1 cells decreased to 66% ± 13% (P < 0.05), the viability of bronchopulmonary NCI-H727 cells decreased to 53% ± 8% (P < 0.01) and the viability of midgut GOT1 cells decreased to 89% ± 6% (P < 0.01). These effects were associated with a decreased entry into the S phase, the induction of the cyclin-dependent kinase inhibitor p21 and reduced expression of cyclin-dependent kinase 4 and cyclin D3. Aspirin suppressed mTOR downstream signaling, evidenced by the reduced phosphorylation of the mTOR substrates 4E binding protein 1, serine/threonine kinase P70S6K and S6 ribosomal protein and inhibited glycogen synthase kinase 3 activity. We observed the (compensatory) activation of tuberous sclerosis 2, the serine/threonine specific protein kinase AKT and extracellular signal-regulated kinases. CONCLUSION: Aspirin demonstrates promising anticancer properties for NETs in vitro. Further preclinical and clinical studies are needed.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Lung Neoplasms/enzymology , Neuroendocrine Tumors/enzymology , Pancreatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/pathology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors
6.
J Cancer Res Ther ; 10(2): 443-5, 2014.
Article in English | MEDLINE | ID: mdl-25022420

ABSTRACT

Both multiple endocrine neoplasia type 1 (MEN1)-related gastrinomas and gastrointestinal stromal tumors (GISTs) are rare neoplasms, and their association has been rarely reported. We describe an unusual association between a GIST and a MEN1-related gastrinoma. A 44-year-old man had undergone surgical removal of a pancreatic gastrinoma in 2004 and was then administered long-term somatostatin analogs, and diagnosed as having MEN1 syndrome. Following an uneventful follow-up, in April 2009, an upper gastrointestinal tract endoscopy showed esophageal narrowing, with evidence of a 2-cm solid mass on endoscopic ultrasonography. Histology revealed a tumor composed of elongated cells with plump cytoplasm arranged in a storiform pattern. The immunophenotype of the lesion was CD117 and Platelet Derived Growth Factor (PDGF) positive, whereas alpha-1 muscle actin and S-100 protein were negative. Due to morphological and immunohistochemical results, a final diagnosis of esophageal GIST was made. The association between GISTs and MEN1 could be casual, although a single case of the coexistence of a GIST and a MEN1-related gastrinoma has already been reported. A role of the MEN1 gene in the pathogenesis of GISTs could be hypothesized.


Subject(s)
Esophageal Neoplasms/diagnostic imaging , Gastrinoma/diagnostic imaging , Gastrointestinal Stromal Tumors/diagnostic imaging , Liver Neoplasms/diagnostic imaging , Multiple Endocrine Neoplasia Type 1/diagnostic imaging , Neoplasms, Second Primary/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Adult , Esophageal Neoplasms/pathology , Gastrinoma/secondary , Gastrointestinal Stromal Tumors/pathology , Humans , Liver Neoplasms/secondary , Male , Multiple Endocrine Neoplasia Type 1/pathology , Neoplasms, Second Primary/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Ultrasonography
7.
Eur J Endocrinol ; 168(5): 755-61, 2013 May.
Article in English | MEDLINE | ID: mdl-23447517

ABSTRACT

DESIGN: The coexistence of chronic autoimmune atrophic gastritis (CAAG) and primary hyperparathyroidism (PHPT) has been described previously, even if its extent and underlying mechanisms remain poorly understood. We therefore prospectively evaluated this association in two series of patients, one with CAAG and the other with sporadic PHPT. METHODS: From January 2005 to March 2012, 107 histologically confirmed CAAG patients and 149 PHPT patients were consecutively enrolled. Routine laboratory assays included serum calcium, parathyroid hormone (PTH), plasma gastrin and chromogranin A (CgA). In CAAG patients with high PTH levels, ionized calcium and 25(OH)-vitamin D were evaluated. All CAAG and hypergastrinemic PHPT patients received an upper gastrointestinal endoscopy. Exclusion criteria were familial PHPT, MEN1 syndrome, treatment with proton pump inhibitor drugs, Helicobacter pylori infection and renal failure. RESULTS: Of the 107 CAAG patients, nine (8.4%) had PHPT and 13 (12.1%) had secondary hyperparathyroidism stemming from vitamin D deficiency. Among the 149 PHPT patients, 11 (7.4%) had CAAG. Gastrin and CgA levels were similar in the CAAG patients with vs those without hyperparathyroidism (either primary or secondary), and calcium and PTH levels were similar in the PHPT patients with vs those without CAAG. CONCLUSIONS: This study confirms a non-casual association between PHPT and CAAG. The prevalence of PHPT in CAAG patients is threefold that of the general population (8.4 vs 1-3%), and the prevalence of CAAG in PHPT patients is fourfold that of the general population (7.4 vs 2%). The mechanisms underlying this association remain unknown, but a potential role for autoimmunity is suggested.


Subject(s)
Autoimmune Diseases/epidemiology , Gastritis, Atrophic/epidemiology , Hyperparathyroidism, Primary/epidemiology , Adult , Aged , Autoimmune Diseases/blood , Calcium/blood , Comorbidity , Female , Gastritis, Atrophic/blood , Humans , Hyperparathyroidism, Primary/blood , Male , Middle Aged , Prevalence , Prospective Studies , Vitamin D/blood
8.
J Gastrointest Surg ; 16(10): 1981-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22829240

ABSTRACT

BACKGROUND: Liver metastases are a strong prognostic indicator in patients with gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs). Therapeutic options for metastatic NETs are expanding and not mutually exclusive. AIMS: This paper reviews the literature relating to multidisciplinary approach towards GEP-NET metastases, to highlight advances in knowledge regarding these tumors, and to understand the interdisciplinary management of individual patients. METHODS: A PubMed search was performed for English-language publications from 1995 through 2012. Reference lists from studies selected were manually searched to identify further relevant reports. Manuscripts comparing different therapeutic options and advances for GEP-NET-related liver metastases were selected. RESULTS: There is considerable controversy regarding the optimal management of GEP-NET metastases. Although radical surgery still remains the gold standard, a variety of other therapeutic options are available for metastatic GEP-NETs, including loco-regional chemotherapy/radiotherapy, radioembolization, systemic peptide receptor radionuclide therapy, biotherapy, and chemotherapy. In selected patients, liver transplantation should also be considered. Systemic somatostatin analogues and/or interferon show anti-proliferative effects, representing an appropriate first-line treatment for most patients. In advanced metastatic NETs, recent options include targeted therapies (i.e., everolimus and sunitinib). CONCLUSIONS: It is evident that multidisciplinary care and multimodality treatments remain the cornerstone of management of NET patients. Since NETs often show a more indolent behavior compared to other malignancies, physicians should aim to preserve a satisfactory quality of life for the patient by personalizing the therapeutic approach according to the tumor's features and prognostic factors.


Subject(s)
Digestive System Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Neuroendocrine Tumors/secondary , Neuroendocrine Tumors/therapy , Ablation Techniques , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Embolization, Therapeutic/methods , Hepatectomy , Humans , Liver Transplantation , Radiotherapy/methods , Treatment Outcome
9.
Scand J Gastroenterol ; 47(6): 640-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22512436

ABSTRACT

BACKGROUND: Liver involvement in celiac disease (CD) is clinically relevant and could require specific treatment in addition to gluten-free diet (GFD). Transient elastography (TE), a noninvasive tool for assessing liver stiffness (LS), has widely been reported as an accurate surrogate marker of liver fibrosis. AIMS: To prospectively identify celiac patients with liver involvement by TE and to assess the effect of GFD. MATERIAL AND METHODS: Ninety-five histologically confirmed CD patients (24 newly diagnosed) were consecutively evaluated by TE and compared with 146 patients with chronic hepatitis C (HCV) and 54 healthy subjects. RESULTS: LS ranged between 2.8 and 6.7 kPa (median 4.9) in healthy subjects, defining 6.9 kPa as the upper reference limit (2 SD above the mean levels). TE was above 6.9 kPa in 10 (10.5%) CD patients. Median TE values resulted significantly higher in CD patients with hypertransaminasemia than those without [6.1 vs. 4.2 kPa (p < 0.01)]. Among the 24 newly diagnosed patients with CD, median TE values declined from 4.4 to 4 kPa, after 6 months of GFD, resulting below 6.9 kPa in 100% of the patients. CONCLUSIONS: A subset of CD patients with hypertransaminasemia showed liver involvement by TE. Accordingly, based on its accuracy in predicting liver fibrosis, TE could be used to identify those CD patients suitable for liver biopsy.


Subject(s)
Celiac Disease/complications , Elasticity Imaging Techniques , Liver Cirrhosis/diagnostic imaging , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Celiac Disease/diet therapy , Diet, Gluten-Free , Female , Hepatitis C, Chronic/diagnostic imaging , Hepatitis C, Chronic/pathology , Humans , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Middle Aged , Prospective Studies , Treatment Outcome , Young Adult
10.
Eur J Gastroenterol Hepatol ; 24(5): 589-93, 2012 May.
Article in English | MEDLINE | ID: mdl-22465973

ABSTRACT

Gastric carcinoids are rare tumors of the stomach. Gastric carcinoid type 1 is associated with chronic atrophic gastritis, and because of a low metastatic potential, is the most benign type. Death from metastatic disease has been reported in only three patients in a review including 724 cases. The present report refers to a 60-year-old man who was affected by type 2 diabetes mellitus and pernicious anemia and died from metastatic gastric carcinoid type 1. In 1998, a well-differentiated 1.2 cm gastric neuroendocrine tumor, immunoreactive for chromogranin A, with a Ki-67 index less than 2% and with infiltration to the submucosal layer was diagnosed and enucleated. In 2002, a new well-differentiated gastric endocrine tumor 6 mm in size with a Ki-67 of approximately 2% was detected, and endoscopic ultrasound confirmed it to be limited to the submucosal layer. The patient refused antrectomy and started long-acting somatostatin analog (lanreotide) in 2005 when the Ki-67 index was 7%, but he stopped the treatment after 4 months. In 2007, despite previous endoscopic stability, endoscopic ultrasound showed an infiltrating gastric lesion of 7 cm. At surgery, the disease appeared to be extended to the liver and to the peritoneum (well-differentiated endocrine carcinoma, Ki-67 40%) with both hepatic and massive peritoneal metastases. A regimen of somatostatin analog was soon restarted; however, the disease continued to spread, and the patient died 6 months later. Overall, despite their generally benign course, type 1 gastric carcinoids may have malignant potential, a finding that should be considered when planning the medical workup of these patients.


Subject(s)
Carcinoid Tumor/secondary , Stomach Neoplasms/diagnosis , Carcinoid Tumor/diagnosis , Carcinoid Tumor/pathology , Disease Progression , Endosonography , Fatal Outcome , Humans , Liver Neoplasms/secondary , Male , Middle Aged , Peritoneal Neoplasms/secondary , Prognosis , Stomach Neoplasms/pathology
11.
Am J Gastroenterol ; 105(9): 2072-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20372113

ABSTRACT

OBJECTIVES: Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) expressing somatostatin receptors may be treated with somatostatin analogs (SSAs). Selection criteria are a positive Octreoscan or a >50% hormone level decrease after octreotide subcutaneous (s.c.) injection (octreotide test) (OT). Plasma chromogranin A (CgA) is the best general GEP-NET marker, but data on CgA response to OT are scanty. Thus, we evaluated whether plasma CgA response to OT could predict the clinical response to SSAs. METHODS: At diagnosis, 38 GEP-NET patients received octreotide 200 microg s.c., with plasma CgA determination at 0, 3, and 6 h. Long-term SSA treatment was then given by monitoring symptomatic, biochemical, and objective responses, and survival. RESULTS: Basal plasma CgA levels were significantly higher in patients with functioning than non-functioning tumors (median (range): 220 (18-2,230) vs. 46 (25-8,610) U/l, P=0.03) and in those with than without metastases (171 (18-8,610) vs. 43 (28-220) U/l, P=0.04). CgA levels significantly correlated with WHO classification, clinical TNM staging, and Ki-67 proliferative index. After OT, CgA levels decreased from 146 (18-8,610) to 61 (10-8,535) U/l (basal and nadir values), P<0.001. In patients responsive to OT, a successful objective response occurred in 21/31 patients (68%). Successful symptomatic response occurred in 13/18 patients (72%), biochemical response in 25/31 (81%), and objective response in 21/31 (68%). In the remaining seven unresponsive cases, with CgA decrement <30%, disease progressed to death in six (86%). Median survival from enrollment was 48 months (6-138) in responsive and 6 (6-30) in unresponsive patients (P=0.0005). CONCLUSIONS: In GEP-NETs, plasma CgA is a reliable marker, and a >30% decrease after OT has a relevant prognostic meaning allowing the identification of the subgroup of patients most likely to be responsive to chronic SSAs.


Subject(s)
Chromogranin A/blood , Gastrointestinal Neoplasms/diagnosis , Neuroendocrine Tumors/diagnosis , Octreotide , Pancreatic Neoplasms/diagnosis , Adult , Aged , Female , Gastrointestinal Neoplasms/blood , Humans , Male , Middle Aged , Neuroendocrine Tumors/blood , Pancreatic Neoplasms/blood , Patient Selection , Predictive Value of Tests , Prognosis
12.
Dig Liver Dis ; 42(9): 635-41, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20172770

ABSTRACT

OBJECTIVES: At presentation, gastroenteropancreatic neuroendocrine tumours (GEP NETs) frequently show prognostically negative hepatic involvement. The aim of this study was to characterise hepatic metastases of GEP NETs as revealed by contrast-enhanced ultrasonography (CEUS), which allows the fine definition of the microvascular system, and to correlate these findings to the biological behaviour of the tumour. METHODS: Eighteen out of 62 GEP NET patients examined between January 2007 and September 2008 had histologically proven hepatic metastases from primary ileal (#6), gastric (#1) or rectal (#1) carcinoids, pancreatic tumours (#7), or primary duodenal (#2) or occult gastrinomas (#1), and all underwent low mechanical index real-time CEUS with SonoVue injection. RESULTS: Strong early enhancement in the arterial phase was observed in 15 cases (83%), and a rapid wash-out in the portal venous phase in 14 (78%). In the late venous phase, the lesions were hypoechoic in 12 cases (67%), isoechoic in five (28%), and hyperechoic in one (0.05%). The time of arterial enhancement correlated with the Ki-67 proliferative index (r(s)=0.516; p=0.028). CONCLUSIONS: Most of the neuroendocrine liver metastases showed increased arterial enhancement at CEUS, a behaviour that is similar to that of hepatocellular carcinomas and the opposite of that of other metastases. CEUS can be a useful diagnostic means of characterising such metastases.


Subject(s)
Carcinoma, Neuroendocrine/diagnostic imaging , Contrast Media , Gastrointestinal Neoplasms/diagnostic imaging , Liver Neoplasms/blood supply , Liver Neoplasms/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Adult , Aged , Aged, 80 and over , Carcinoma, Neuroendocrine/pathology , Gastrointestinal Neoplasms/pathology , Humans , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/pathology , Ultrasonography , Young Adult
13.
World J Gastroenterol ; 15(18): 2177-83, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19437556

ABSTRACT

Gastric carcinoids (GCs), which originate from gastric enterochromaffin-like (ECL) mucosal cells and account for 2.4% of all carcinoids, are found increasingly in the course of upper gastrointestinal tract endoscopy. Current nosography includes those occurring in chronic conditions with hypergastrinemia, as the type 1 associated with chronic atrophic gastritis, and the type 2 associated with Zollinger-Ellison syndrome in multiple endocrine neoplasia type 1, and type 3, which is unrelated to hypergastrinemia and is frequently malignant, with distant metastases. The optimal clinical approach to GCs remains to be elucidated, depending upon type, size and number of carcinoids. While there is agreement concerning the treatment of type 3 carcinoids, for types 1 and 2, current possibilities include simple surveillance, endoscopic polypectomy, surgical excision, associated or not with surgical antrectomy, or total gastrectomy. Moreover, the recent introduction of somatostatin analogues represents a therapeutic option of possibly outstanding relevance.


Subject(s)
Carcinoid Tumor/pathology , Carcinoid Tumor/therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Carcinoid Tumor/diagnosis , Carcinoid Tumor/epidemiology , Female , Humans , Male , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology
14.
World J Gastroenterol ; 14(35): 5377-84, 2008 Sep 21.
Article in English | MEDLINE | ID: mdl-18803349

ABSTRACT

Gastro-entero-pancreatic (GEP) neuroendocrine tumors (NETs) are rare neoplasms, although their prevalence has increased substantially over the past three decades. Moreover, there has been an increased clinical recognition and characterization of these neoplasms. They show extremely variable biological behavior and clinical course. Most NETs have endocrine function and secrete peptides and neuroamines that cause distinct clinical syndromes, including carcinoid syndrome; however, many are clinically silent until late presentation with mass effects. Investigation and management should be individualized for each patient, taking into account the likely natural history of the tumor and general health of the patient. Management strategies include surgery for cure or palliation, and a variety of other cytoreductive techniques, and medical treatment including chemotherapy, and biotherapy to control symptoms due to hormone release and tumor growth, with somatostatin analogues (SSAs) and alpha-interferon. New biological agents and somatostatin-tagged radionuclides are under investigation. Advances in the therapy and development of centers of excellence which coordinate multicenter studies, are needed to improve diagnosis, treatment and therefore survival of patients with GEP NETs.


Subject(s)
Digestive System Neoplasms/diagnosis , Neuroendocrine Tumors/diagnosis , Digestive System Neoplasms/classification , Digestive System Neoplasms/therapy , Enteric Nervous System , Humans , Neuroendocrine Tumors/classification , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Stomach Neoplasms/diagnosis , Stomach Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...