Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Endocr Pract ; 21(5): 546-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25962093

ABSTRACT

Incidental detection of pancreatic neuroendocrine tumors (PNETs) has substantially increased over the last decade due to widespread use of advanced imaging studies. Reliable initial imaging-based characterization is crucial for the differential diagnosis from other exocrine neoplasms and to determine the appropriate management plan. Measurements of chromogranin A, pancreatic polypeptide, and calcitonin are recommended for the biochemical evaluation. A thorough medical history needs to be performed to rule out multiple endocrine neoplasia (MEN) type 1. The European Neuroendocrine Tumor Society (ENETS)/Tumor Node Metastasis (TNM) staging system together with a grading based on the Ki-67 proliferation index and mitotic counts has proven to give more appropriate prognostic information than the World Health Organization (WHO)/American Joint Committee on Cancer (AJCC) TNM staging but may still fail to safely differentiate benign from malignant lesions. Poorly differentiated PNETs generally present with metastases and are rarely amenable for resection. Well- or intermediately differentiated tumors ≥2 cm with imaging evidence of malignancy or with a Ki-67 >2% should be resected. It has been suggested that non-MEN related, nonfunctioning, and asymptomatic PNETs <2 cm with a Ki-67 index ≤2% carry a low risk of metastasis and may be observed in the absence of clinical or radiologic criteria of malignancy or progression, especially in older patients. However, because metastases may occur with long delay with smaller PNETS, physicians should consider patient age, lesion location, and the risks of operation, and patients not undergoing surgery need to be closely followed closely.


Subject(s)
Incidental Findings , Neuroendocrine Tumors/diagnosis , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/blood , Biopsy, Fine-Needle , Calcitonin/blood , Chromogranin A/blood , Diagnostic Imaging , Humans , Ki-67 Antigen/analysis , Neoplasm Metastasis/pathology , Neoplasm Staging , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Pancreatic Polypeptide/blood
2.
Proc Natl Acad Sci U S A ; 112(13): 4062-7, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25787250

ABSTRACT

Insulinomas are pancreatic islet tumors that inappropriately secrete insulin, producing hypoglycemia. Exome and targeted sequencing revealed that 14 of 43 insulinomas harbored the identical somatic mutation in the DNA-binding zinc finger of the transcription factor Yin Yang 1 (YY1). Chromatin immunoprecipitation sequencing (ChIP-Seq) showed that this T372R substitution changes the DNA motif bound by YY1. Global analysis of gene expression demonstrated distinct clustering of tumors with and without YY1(T372R) mutations. Genes showing large increases in expression in YY1(T372R) tumors included ADCY1 (an adenylyl cyclase) and CACNA2D2 (a Ca(2+) channel); both are expressed at very low levels in normal ß-cells and show mutation-specific YY1 binding sites. Both gene products are involved in key pathways regulating insulin secretion. Expression of these genes in rat INS-1 cells demonstrated markedly increased insulin secretion. These findings indicate that YY1(T372R) mutations are neomorphic, resulting in constitutive activation of cAMP and Ca(2+) signaling pathways involved in insulin secretion.


Subject(s)
Gene Expression Regulation , Insulinoma/genetics , Mutation, Missense , Pancreatic Neoplasms/genetics , YY1 Transcription Factor/genetics , Adenylyl Cyclases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Binding Sites , Blood Glucose/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cohort Studies , Cyclic AMP/metabolism , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulinoma/metabolism , Male , Middle Aged , Molecular Sequence Data , Pancreatic Neoplasms/metabolism , Protein Binding , YY1 Transcription Factor/metabolism
3.
Surgery ; 156(6): 1512-21; discussion 1521-2, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456945

ABSTRACT

BACKGROUND: One-fifth of all patients with small-intestinal neuroendocrine tumors (SI-NETs) present with or develop peritoneal carcinomatosis (PC). Our aim was to determine the prognosis and genetic profiles of tumors in patients with PC compared with tumors in patients without PC. METHODS: We included SI-NET patients (cases with PC, n = 73, and controls without PC, n = 468) who underwent operation between 1985 and 2012. The Lyon prognostic index was used to correlate the amount of PC to survival. DNA samples from patients with (n = 8) and without (n = 7) PC were analyzed with a single-nucleotide polymorphism array (HumanOmni2.5 BeadChip, Illumina) to investigate genetic disparities between groups. RESULTS: Patients with PC had poorer survival (median 5.1 years) than controls (11.1 years). An advanced postoperative Lyon prognostic index was a negative prognostic marker for survival by multivariable analysis (P = .042). Patients with and without PC clustered differently based on loss of heterozygosity and copy number variation data from single-nucleotide polymorphism array of the primary tumors (P = .042). CONCLUSION: SI-NET patients with PC have poor survival, which diminishes with increasing PC load after surgery. Clustering based on copy number variation and loss of heterozygosity data suggests different genotypes in primary tumors comparing patients with and without PC.


Subject(s)
Carcinoma/secondary , Genetic Variation , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Peritoneal Neoplasms/secondary , Adult , Aged , Carcinoma/genetics , Carcinoma/mortality , Cohort Studies , DNA Copy Number Variations , Disease-Free Survival , Female , Humans , Intestinal Neoplasms/mortality , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Middle Aged , Multivariate Analysis , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/mortality , Prognosis , Reference Values , Retrospective Studies , Risk Assessment , Survival Analysis
4.
J Clin Endocrinol Metab ; 99(10): 3595-606, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25162669

ABSTRACT

OBJECTIVE: The surgical management of primary hyperparathyroidism (PHPT) has undergone considerable advances over the past two decades. The purpose of this report is to review these advances. PARTICIPANTS: This subgroup was constituted by the Steering Committee of the Fourth International Workshop on the Management of Asymptomatic Primary Hyperparathyroidism to address key questions related to the surgical management of PHPT. EVIDENCE: Data since the last International Workshop were presented and discussed in detail. The topics included improvements in preoperative imaging, intraoperative adjuncts, refinements in local and regional anesthesia, and rapid intraoperative PTH assays. CONSENSUS PROCESS: Questions were developed by the International Task Force on PHPT. A comprehensive literature search for relevant studies was undertaken. After extensive review and discussion, the subgroup agreed on what recommendations should be made to the Expert Panel regarding surgical approaches to parathyroidectomy. CONCLUSIONS: 1) All patients with PHPT who meet surgical criteria should be referred to an experienced endocrine surgeon to discuss the risks, benefits, and potential complications of surgery. 2) Patients who do not meet surgical criteria and in whom there are no medical contraindications to surgery may request a visit with an experienced endocrine surgeon. Alternatively, a multidisciplinary endocrine conference with surgeon involvement could be employed to address all relevant issues. 3) Imaging is not a diagnostic procedure; it is a localization procedure to help the surgeon optimize the operative plan. 4) The frequency of hereditary forms of PHPT may be underappreciated and needs to be assessed with increased vigilance. And 5) surgery is likely to benefit patients due to high cure rates, low complication rates, and the likelihood of reversing skeletal manifestations.


Subject(s)
Asymptomatic Diseases , Endocrinology/standards , Evidence-Based Medicine/standards , Hyperparathyroidism, Primary/surgery , Practice Guidelines as Topic , Education , Humans
5.
Nat Genet ; 46(6): 613-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24747643

ABSTRACT

Adrenal tumors autonomously producing cortisol cause Cushing's syndrome. We performed exome sequencing of 25 tumor-normal pairs and identified 2 subgroups. Eight tumors (including three carcinomas) had many somatic copy number variants (CNVs) with frequent deletion of CDC42 and CDKN2A, amplification of 5q31.2 and protein-altering mutations in TP53 and RB1. Seventeen tumors (all adenomas) had no somatic CNVs or TP53 or RB1 mutations. Six of these had known gain-of-function mutations in CTNNB1 (ß-catenin) or GNAS (Gαs). Six others had somatic mutations in PRKACA (protein kinase A (PKA) catalytic subunit) resulting in a p.Leu206Arg substitution. Further sequencing identified this mutation in 13 of 63 tumors (35% of adenomas with overt Cushing's syndrome). PRKACA, GNAS and CTNNB1 mutations were mutually exclusive. Leu206 directly interacts with the regulatory subunit of PKA, PRKAR1A. Leu206Arg PRKACA loses PRKAR1A binding, increasing the phosphorylation of downstream targets. PKA activity induces cortisol production and cell proliferation, providing a mechanism for tumor development. These findings define distinct mechanisms underlying adrenal cortisol-producing tumors.


Subject(s)
Adrenal Gland Neoplasms/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , DNA Copy Number Variations , Hydrocortisone/metabolism , Mutation , Adolescent , Adult , Aged , Amino Acid Sequence , Base Sequence , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Exome , Female , Gene Deletion , Gene Dosage , HEK293 Cells , Humans , Male , Middle Aged , Molecular Sequence Data , Phosphorylation , Sequence Analysis, DNA , Sequence Homology, Amino Acid , cdc42 GTP-Binding Protein/genetics
6.
World J Surg ; 38(3): 742-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24233660

ABSTRACT

BACKGROUND: A majority of patients with small intestinal neuroendocrine tumors (SI-NETs) present with or develop liver metastases (LM). A number of treatments for LM are used clinically, including liver transplantation (LTx). Indications for LTx are under debate; young age(<65 years), absence of extrahepatic disease, resected primary tumor and limited extent of LM have been suggested as inclusion criteria for LTx with the aim to optimize outcome. MATERIALS AND METHODS: From our series of 672 patients with SI-NET treated at the University Hospital in Uppsala between 1985 and 2012, we identified 78 patients according to the following criteria: <65 years of age, locoregional surgery (LRS) of the primary tumor and mesenteric metastases successfully performed, LM present but no extrahepatic disease. Baseline was chosen as the first date the following points were met: First visit to our center,LRS performed, LM present. The patients underwent treatment according to the standard clinical protocols at our center, and during this time period we did not perform or refer any SI-NET patients for LTx. Kaplan-Meier survival analyses were performed in three different groups based on hypothetical criteria for LTx. RESULTS: Five-year overall survival rates for patients <65 years (n = 78) and <55 years (n = 36) of age were 84 ± 8 and 92 ± 9 %, respectively. For patients fulfilling the Milan criteria (n = 33) the 5-year survival was 97 ± 6 %. CONCLUSIONS: Most young patients (<65 years) with SINET and LM have a favorable survival with standardized multimodality treatment. Indeed, most survival figures reported after LTx of NET do not surpass these figures.


Subject(s)
Intestinal Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Liver Transplantation , Neuroendocrine Tumors/secondary , Neuroendocrine Tumors/surgery , Adult , Female , Follow-Up Studies , Humans , Intestinal Neoplasms/mortality , Intestinal Neoplasms/surgery , Intestine, Small/pathology , Intestine, Small/surgery , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Male , Middle Aged , Neuroendocrine Tumors/mortality , Survival Rate , Treatment Outcome
8.
Endocr Relat Cancer ; 21(2): 275-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24351681

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs), formerly known as midgut carcinoids, are rare and slow-growing neoplasms. Frequent loss of one copy of chromosome 18 in primary tumors and metastases has been observed. The aim of the study was to investigate a possible role of TCEB3C (Elongin A3), currently the only imprinted gene on chromosome 18, as a tumor suppressor gene in SI-NETs, and whether its expression is epigenetically regulated. Primary tumors, metastases, the human SI-NET cell line CNDT2.5, and two other cell lines were included. Immunohistochemistry, gene copy number determination by PCR, colony formation assay, western blotting, real-time quantitative RT-PCR, RNA interference, and quantitative CpG methylation analysis by pyrosequencing were performed. A large majority of tumors (33/43) showed very low to undetectable Elongin A3 expression and as expected 89% (40/45) displayed one gene copy of TCEB3C. The DNA hypomethylating agent 5-aza-2'-deoxycytidine induced TCEB3C expression in CNDT2.5 cells, in primary SI-NET cells prepared directly after surgery, but not in two other cell lines. Also siRNA to DNMT1 and treatment with the general histone methyltransferase inhibitor 3-deazaneplanocin A induced TCEB3C expression in a cell type-specific way. CpG methylation at the TCEB3C promoter was observed in all analyzed tissues and thus not related to expression. Overexpression of TCEB3C resulted in a 50% decrease in clonogenic survival of CNDT2.5 cells, but not of control cells. The results support a putative role of TCEB3C as a tumor suppressor gene in SI-NETs. Epigenetic repression of TCEB3C seems to be tumor cell type-specific and involves both DNA and histone methylation.


Subject(s)
Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Transcription Factors/genetics , Cell Line, Tumor , CpG Islands , Elongin , Genes, Tumor Suppressor , Histones/metabolism , Humans , Intestinal Neoplasms/metabolism , Intestine, Small , Methylation , Neuroendocrine Tumors/metabolism , Promoter Regions, Genetic
9.
Endocr Relat Cancer ; 21(2): 231-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24292603

ABSTRACT

Primary hyperparathyroidism (pHPT) resulting from parathyroid tumors is a common endocrine disorder with incompletely understood etiology. In renal failure, secondary hyperparathyroidism (sHPT) occurs with multiple tumor development as a result of calcium and vitamin D regulatory disturbance. The aim of this study was to investigate a potential role of the histone 3 lysine 27 methyltransferase EZH2 in parathyroid tumorigenesis. Parathyroid tumors from patients with pHPT included adenomas and carcinomas. Hyperplastic parathyroid glands from patients with HPT secondary to uremia and normal parathyroid tissue specimens were included in this study. Quantitative RT-PCR, western blotting, bisulfite pyrosequencing, colony formation assay, and RNA interference were used. EZH2 was overexpressed in a subset of the benign and in all malignant parathyroid tumors as determined by quantitative RT-PCR and western blotting analyses. Overexpression was explained by EZH2 gene amplification in a large fraction of tumors. EZH2 depletion by RNA interference inhibited sHPT-1 parathyroid cell line proliferation as determined by tritium-thymidine incorporation and colony formation assays. EZH2 depletion also interfered with the Wnt/ß-catenin signaling pathway by increased expression of growth-suppressive AXIN2, a negative regulator of ß-catenin stability. Indeed, EZH2 contributed to the total level of aberrantly accumulated transcriptionally active (nonphosphoylated) ß-catenin in the parathyroid tumor cells. To our knowledge EZH2 gene amplification presents the first genetic aberration common to parathyroid adenomas, secondary hyperplastic parathyroid glands, and parathyroid carcinomas. This supports the possibility of a common pathway in parathyroid tumor development.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Hyperparathyroidism, Secondary/genetics , Parathyroid Glands/metabolism , Parathyroid Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , Adenoma/metabolism , Carcinoma/metabolism , Enhancer of Zeste Homolog 2 Protein , Humans , Oncogenes , Parathyroid Neoplasms/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Messenger/metabolism , Wnt Signaling Pathway
10.
Nat Genet ; 45(9): 1050-4, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913001

ABSTRACT

Adrenal aldosterone-producing adenomas (APAs) constitutively produce the salt-retaining hormone aldosterone and are a common cause of severe hypertension. Recurrent mutations in the potassium channel gene KCNJ5 that result in cell depolarization and Ca(2+) influx cause ∼40% of these tumors. We identified 5 somatic mutations (4 altering Gly403 and 1 altering Ile770) in CACNA1D, encoding a voltage-gated calcium channel, among 43 APAs without mutated KCNJ5. The altered residues lie in the S6 segments that line the channel pore. Both alterations result in channel activation at less depolarized potentials; Gly403 alterations also impair channel inactivation. These effects are inferred to cause increased Ca(2+) influx, which is a sufficient stimulus for aldosterone production and cell proliferation in adrenal glomerulosa. We also identified de novo germline mutations at identical positions in two children with a previously undescribed syndrome featuring primary aldosteronism and neuromuscular abnormalities. These findings implicate gain-of-function Ca(2+) channel mutations in APAs and primary aldosteronism.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Calcium Channels, L-Type/genetics , Germ-Line Mutation , Hyperaldosteronism/genetics , Mutation , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/biosynthesis , Amino Acid Sequence , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Cell Line , Child , Child, Preschool , Female , Humans , Hyperaldosteronism/metabolism , Male , Molecular Sequence Data , Pedigree , Protein Conformation , Sequence Alignment
11.
PLoS One ; 7(7): e41926, 2012.
Article in English | MEDLINE | ID: mdl-22848660

ABSTRACT

BACKGROUND: Aldosterone producing lesions are a common cause of hypertension, but genetic alterations for tumorigenesis have been unclear. Recently, either of two recurrent somatic missense mutations (G151R or L168R) was found in the potassium channel KCNJ5 gene in aldosterone producing adenomas. These mutations alter the channel selectivity filter and result in Na(+) conductance and cell depolarization, stimulating aldosterone production and cell proliferation. Because a similar mutation occurs in a mendelian form of primary aldosteronism, these mutations appear to be sufficient for cell proliferation and aldosterone production. The prevalence and spectrum of KCNJ5 mutations in different entities of adrenocortical lesions remain to be defined. MATERIALS AND METHODS: The coding region and flanking intronic segments of KCNJ5 were subjected to Sanger DNA sequencing in 351 aldosterone producing lesions, from patients with primary aldosteronism and 130 other adrenocortical lesions. The specimens had been collected from 10 different worldwide referral centers. RESULTS: G151R or L168R somatic mutations were identified in 47% of aldosterone producing adenomas, each with similar frequency. A previously unreported somatic mutation near the selectivity filter, E145Q, was observed twice. Somatic G151R or L168R mutations were also found in 40% of aldosterone producing adenomas associated with marked hyperplasia, but not in specimens with merely unilateral hyperplasia. Mutations were absent in 130 non-aldosterone secreting lesions. KCNJ5 mutations were overrepresented in aldosterone producing adenomas from female compared to male patients (63 vs. 24%). Males with KCNJ5 mutations were significantly younger than those without (45 vs. 54, respectively; p<0.005) and their APAs with KCNJ5 mutations were larger than those without (27.1 mm vs. 17.1 mm; p<0.005). DISCUSSION: Either of two somatic KCNJ5 mutations are highly prevalent and specific for aldosterone producing lesions. These findings provide new insight into the pathogenesis of primary aldosteronism.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Aldosterone/biosynthesis , Mutation , Sequence Analysis , Adolescent , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Adult , Aged , Base Sequence , Cohort Studies , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Mutation Rate , Sex Characteristics , Young Adult
12.
Genes Chromosomes Cancer ; 51(10): 949-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22733721

ABSTRACT

The molecular pathogenesis of benign and malignant adrenocortical tumors (ACT) is incompletely clarified. The role of DNA methylation in adrenocortical tumorigenesis has not been analyzed in an unbiased, systematic fashion. Using the Infinium HumanMethylation27 BeadChip, the DNA methylation levels of 27,578 CpG sites were investigated in bisulfite-modified DNA from 6 normal adrenocortical tissue samples, 27 adrenocortical adenomas (ACA), and 15 adrenocortical carcinomas (ACC). Genes involved in cell cycle regulation, apoptosis, and transcriptional regulation of known or putative importance in the development of adrenal tumors showed significant and frequent hypermethylation. Such genes included CDKN2A, GATA4, BCL2, DLEC1, HDAC10, PYCARD, and SCGB3A1/HIN1. Comparing benign versus malignant ACT, a total of 212 CpG islands were identified as significantly hypermethylated in ACC. Gene expression studies of selected hypermethylated genes (CDKN2A, GATA4, DLEC1, HDAC10, PYCARD, SCGB3A1/HIN1) in 6 normal and 16 neoplastic adrenocortical tissues (10 ACA and 6 ACC), displayed reduced gene expression in benign and malignant ACT versus normal adrenocortical tissue. Treatment with 5-aza-2'-deoxycytidine of adrenocortical cancer H-295R cells increased expression of the hypermethylated genes CDKN2A, GATA4, DLEC1, HDAC10, PYCARD, and SCGB3A1/HIN1. In conclusion, the current study represents the first unbiased, quantitative, genome-wide study of adrenocortical tumor DNA methylation. Genes with altered DNA methylation patterns were identified of putative importance to benign and malignant adrenocortical tumor development.


Subject(s)
Adenoma/genetics , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex/metabolism , Adrenocortical Carcinoma/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Adenoma/metabolism , Adenoma/pathology , Adrenal Cortex/pathology , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/pathology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , CpG Islands , DNA Methylation/drug effects , Decitabine , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Genome-Wide Association Study , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic
13.
Endocr Relat Cancer ; 19(5): R163-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22645227

ABSTRACT

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) represent a heterogenous group of tumors arising from a variety of neuroendocrine cell types. The incidence and prevalence of GEP-NENs have markedly increased over the last three decades. Symptoms are often absent in early disease, or vague and nonspecific even in advanced disease. Delayed diagnosis is thus common. Chromogranin A is the most commonly used biomarker but has limitations as does the proliferative marker Ki-67%, which is often used for tumor grading and determination of therapy. The development of a multidimensional prognostic nomogram may be valuable in predicting tumor behavior and guiding therapy but requires validation. Identification of NENs that express somatostatin receptors (SSTR) allows for SSTR scintigraphy and positron emission tomography imaging using novel radiolabeled compounds. Complete surgical resection of limited disease or endoscopic ablation of small lesions localized in stomach or rectum can provide cure; however, the majority of GEP-NENs are metastatic (most frequently the liver and/or mesenteric lymph nodes) at diagnosis. Selected patients with metastatic disease may benefit from advanced surgical techniques including hepatic resection or liver transplantation. Somatostatin analogs are effective for symptomatic treatment and exhibit some degree of antiproliferative activity in small intestinal NENs. There is a place for streptozotocin, temozolomide, and capecitabine in the management of pancreatic NENs, while new agents targeting either mTOR (everolimus) or angiogenic (sunitinib) pathways have shown efficacy in these lesions.


Subject(s)
Gastrointestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/therapy , Humans , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy
14.
Endocrine ; 42(3): 612-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22576020

ABSTRACT

Aberrant accumulation of ß-catenin plays an important role in a variety of human neoplasms. This can be caused by stabilizing mutation of ß-catenin (CTNNB1, exon 3) or by mutation or deregulated expression of other components of the WNT/ß-catenin signaling pathway. Accumulation of non-phosphorylated active ß-catenin has been reported to commonly occur in parathyroid adenomas from patients with primary hyperparathyroidism (pHPT), either due to the aberrantly spliced internally truncated WNT receptor LRP5 (LRP5Δ) or to a stabilizing mutation of ß-catenin. The S37A mutation was reported to occur in 7.3 % in a single study of parathyroid adenomas, while in other studies no stabilizing mutations of ß-catenin exon 3 were identified. The aim of this study was to determine the mutational frequency of the CTNNB1 gene, specifically exon 3 in a large series of parathyroid adenomas. One hundred and eighty sporadic parathyroid adenomas were examined for mutations in exon 3 of CTNNB1 by direct DNA sequencing, utilizing previously published primer sequences. The mutation S33C (TCT>TGT) was detected by direct-DNA sequencing of PCR fragments in 1 out of 180 sporadic parathyroid adenomas (0.68 %). Like serine 37, mutations of serine 33 have been reported in many neoplasms with resulting ß-catenin stabilization, enhanced transcription, and oncogenic activities. Immunohistochemical analysis revealed an overexpression of the ß-catenin protein in the lone mutant tumor. Taking also previous studies into account we conclude that activating mutations of the regulatory GSK-3ß phosphorylation sites serine 33 and 37, encoded by CTNNB1 exon 3, rarely occur in parathyroid adenomas from patients with pHPT.


Subject(s)
Adenoma/genetics , Mutation/genetics , Mutation/physiology , Parathyroid Neoplasms/genetics , beta Catenin/genetics , DNA/genetics , Exons/genetics , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction
15.
Clinics (Sao Paulo) ; 67 Suppl 1: 173-8, 2012.
Article in English | MEDLINE | ID: mdl-22584725

ABSTRACT

Pancreatico-duodenal tumors are the second most common endocrinopathy in multiple endocrine neoplasia syndrome type 1, and have a pronounced effect on life expectancy as the principal cause of disease-related death. Previous discussions about surgical management have focused mainly on syndromes of hormone excess and, in particular, the management of multiple endocrine neoplasia syndrome type 1-related Zollinger-Ellison syndrome. Since hormonal syndromes tend to occur late and indicate the presence of metastases, screening with biochemical markers and endoscopic ultrasound is recommended for early detection of pancreatico-duodenal tumors, and with early surgery before metastases have developed. Surgery is recommended in patients with or without hormonal syndromes in the absence of disseminated liver metastases. The suggested operation includes distal 80% subtotal pancreatic resection together with enucleation of tumors in the head of the pancreas, and in cases with Zollinger-Ellison syndrome, excision of duodenal gastrinomas together with clearance of regional lymph node metastases. This strategy, with early and aggressive surgery before metastases have developed, is believed to reduce the risks for tumor recurrence and malignant progression.


Subject(s)
Duodenal Neoplasms/surgery , Multiple Endocrine Neoplasia Type 1/surgery , Pancreatic Neoplasms/surgery , Duodenal Neoplasms/diagnosis , Humans , Liver Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Multiple Endocrine Neoplasia Type 1/genetics , Pancreatic Neoplasms/diagnosis
16.
J Clin Endocrinol Metab ; 97(7): E1307-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22544915

ABSTRACT

CONTEXT: Primary hyperparathyroidism (pHPT) resulting from parathyroid tumors is a common endocrine disorder with incompletely understood etiology. In renal failure, secondary hyperparathyroidism (sHPT) occurs with multiple tumor development as a result of calcium and vitamin D regulatory disturbance. OBJECTIVE: The aim of the study was to investigate whether HIC1 may act as a tumor suppressor in the parathyroid glands and whether deregulated expression involves epigenetic mechanisms. PATIENTS AND METHODS: Parathyroid tumors from patients with pHPT included single adenomas, multiple tumors from the same patient, and cancer. Hyperplastic parathyroid glands from patients with sHPT and hypercalcemia and normal parathyroid tissue specimens were included in the study. Quantitative RT-PCR, bisulfite pyrosequencing, colony formation assay, chromatin immunoprecipitation, and RNA interference was used. RESULTS: HIC1 was generally underexpressed regardless of the hyperparathyroid disease state including multiple parathyroid tumors from the same patient, and overexpression of HIC1 led to a decrease in clonogenic survival of parathyroid tumor cells. Only the carcinomas showed a high methylation level and reduced HIC1 expression. Cell culture experiments, including use of primary parathyroid tumor cells prepared directly after operation, the general histone methyltransferase inhibitor 3-deazaneplanocin A, chromatin immunoprecipitation, and RNA interference of DNA methyltransferases and EZH2 (enhancer of zeste homolog 2), supported a role of repressive histone H3 modifications (H3K27me2/3) rather than DNA methylation in repression of HIC1. CONCLUSIONS: The results strongly support a growth-regulatory role of HIC1 in the parathyroid glands and suggest that perturbed expression of HIC1 may represent an early event during tumor development. Repressive histone modification H3K27me2/3 is involved in repression of HIC1 expression in hyperparathyroid tumors.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Histones/metabolism , Hyperparathyroidism, Primary/genetics , Kruppel-Like Transcription Factors/genetics , Parathyroid Neoplasms/genetics , Adenoma/complications , Adenoma/metabolism , Carcinoma/complications , Carcinoma/metabolism , Cell Proliferation , Epigenesis, Genetic/physiology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Histones/physiology , Humans , Hyperparathyroidism, Primary/complications , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/physiology , Lysine/metabolism , Parathyroid Neoplasms/complications , Parathyroid Neoplasms/metabolism , Transfection , Tumor Cells, Cultured
17.
World J Surg ; 36(6): 1419-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21984144

ABSTRACT

BACKGROUND: Small intestinal neuroendocrine tumors (SI-NETs) are uncommon, with an annual incidence of about 1 per 100,000 individuals. The primary tumor (PT) is generally small, but nevertheless the majority of patients have mesenteric lymph node metastases and liver metastases at diagnosis. Our aim was to identify prognostic factors for survival and to evaluate outcome after surgery in SI-NET patients. MATERIAL AND METHODS: We included 603 consecutive patients (325 men; age at diagnosis 63 ± 11 years [mean ± SD]) with histopathologically verified SI-NET, who were diagnosed between 1985 and 2010. Hospital charts were reviewed and were scrutinized for carcinoid heart disease (CHD), flush and/or diarrhea, proliferation by Ki-67 index, mesenteric lymph node metastases (m.lgllm), distant abdominal lymph node metastases (da.lgllm), liver tumor load (LTL), extra-abdominal metastases (EAM), locoregional resective surgery, as well as debulking of LTL, and adverse events after surgery. RESULTS: Median overall survival (OS) was 8.4 years; 5-year OS was 67%, and 5-year relative survival was 74%. Independent prognostic factors by univariate and multivariate analysis were age at diagnosis, CHD, m.lgllm, da.lgllm, LTL, EAM, peritoneal carcinomatosis (PC), and proliferation. Locoregional resective surgery was associated with increased survival on crude and multivariate analysis. The 30-day mortality in our institution after initial locoregional resective surgery was 0.5% (1/205). CONCLUSIONS: For the first time, m.lgllm and da.lgllm, LTL, PC, and EAM are demonstrated to be independent prognostic factors by multivariate analysis. Locoregional removal of the PT/m.lgllm. was a positive prognostic factor by crude and adjusted analysis and may influence survival.


Subject(s)
Intestinal Neoplasms/surgery , Intestine, Small/surgery , Neuroendocrine Tumors/surgery , Aged , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Intestinal Neoplasms/mortality , Intestinal Neoplasms/pathology , Intestinal Neoplasms/therapy , Intestine, Small/pathology , Laparotomy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Liver Neoplasms/therapy , Lymph Node Excision , Lymphatic Metastasis , Male , Middle Aged , Multivariate Analysis , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/therapy , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/surgery , Peritoneal Neoplasms/therapy , Prognosis , Retrospective Studies , Survival Analysis
18.
Cancers (Basel) ; 4(1): 100-12, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-24213229

ABSTRACT

Positron emission tomography (PET) with 11C-labeled 5-hydroxytryptophane (5-HTP) is a sensitive technique to visualize neuroendocrine tumours (NETs), due to high intracellular uptake of amine-precursors like L-dihydroxyphenylalanine (L-DOPA) and 5-HTP. NETs are often small and difficult to localize in spite of overt clinical symptoms due to hormonal excess. In our study, 38 consecutive NET patients underwent 11C-5-HTP-PET and morphological imaging by CT within 12 weeks prior to surgery. Surgical, histopathological and 5-HTP PET findings were correlated. 11C-5-HTP-PET corresponded to the surgical findings in 31 cases, was false negative in six, and true negative in one case resulting in 83.8% sensitivity and 100% specificity. Positive predicted value was 100%. In 11 patients 11C-5-HTP-PET was the only imaging method applied to localize the tumour. Thus, we could demonstrate that functional imaging by 11C-5-HTP-PET in many cases adds vital preoperative diagnostic information and in more than every fourth patient was the only imaging method that will guide the surgeon in finding the NET-lesion. Although the present results demonstrates that 11C-5-HTP may be used as an universal NET tracer, the sensitivity to visualize benign insulinomas and non functioning pancreatic NETs was lower.

19.
Clinics ; 67(supl.1): 173-178, 2012. graf, tab
Article in English | LILACS | ID: lil-623149

ABSTRACT

Pancreatico-duodenal tumors are the second most common endocrinopathy in multiple endocrine neoplasia syndrome type 1, and have a pronounced effect on life expectancy as the principal cause of disease-related death. Previous discussions about surgical management have focused mainly on syndromes of hormone excess and, in particular, the management of multiple endocrine neoplasia syndrome type 1-related Zollinger-Ellison syndrome. Since hormonal syndromes tend to occur late and indicate the presence of metastases, screening with biochemical markers and endoscopic ultrasound is recommended for early detection of pancreatico-duodenal tumors, and with early surgery before metastases have developed. Surgery is recommended in patients with or without hormonal syndromes in the absence of disseminated liver metastases. The suggested operation includes distal 80% subtotal pancreatic resection together with enucleation of tumors in the head of the pancreas, and in cases with Zollinger-Ellison syndrome, excision of duodenal gastrinomas together with clearance of regional lymph node metastases. This strategy, with early and aggressive surgery before metastases have developed, is believed to reduce the risks for tumor recurrence and malignant progression.


Subject(s)
Humans , Duodenal Neoplasms/surgery , Multiple Endocrine Neoplasia Type 1/surgery , Pancreatic Neoplasms/surgery , Duodenal Neoplasms/diagnosis , Lymphatic Metastasis , Liver Neoplasms/secondary , Lymph Nodes/pathology , Multiple Endocrine Neoplasia Type 1/genetics , Pancreatic Neoplasms/diagnosis
20.
Genes Chromosomes Cancer ; 50(9): 735-45, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21638518

ABSTRACT

The role of DNA methylation of CpG islands in parathyroid tumorigenesis has not been analyzed in an unbiased, systematic fashion. DNA was isolated from normal and pathologic parathyroid tissues, bisulphite modified and analyzed using the Infinium HumanMethylation27 BeadChip. Distinct hierarchical clustering of genes with altered DNA methylation profiles in normal and pathologic parathyroid tissue was evident. Comparing normal parathyroid tissue with parathyroid adenomas, 367 genes were significantly altered, while 175 genes significantly differed when comparing parathyroid carcinomas and normal parathyroid tissues. A comparison between parathyroid adenomas and parathyroid carcinomas identified 263 genes with significantly distinct methylation levels. Results were confirmed for certain genes in a validation cohort of 40 parathyroid adenomas by methylation-specific PCR. Genes of known or putative importance in the development of parathyroid tumors showed significant and frequent hypermethylation. DNA hypermethylation of CDKN2B, CDKN2A, WT1, SFRP1, SFRP2, and SFRP4 was associated with reduced gene expression in both benign and malignant parathyroid tumors. Treatment with 5-aza-2'-deoxycytidine of primary cell cultures restores expression of hypermethylated genes in benign and malignant parathyroid tumors. In conclusion, the unbiased, genome-wide study of the parathyroid tumor DNA methylome identified a number of genes with altered DNA methylation patterns of putative importance to benign and malignant parathyroid tumorigenesis.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Parathyroid Neoplasms/genetics , Adenoma/pathology , Azacitidine/pharmacology , Carcinoma/pathology , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic , Genes, Neoplasm , Humans , Parathyroid Neoplasms/pathology , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...