1
|
Li R, Wu X, Zhao P, Xue K, Li J. A pan-cancer analysis identifies HDAC11 as an immunological and prognostic biomarker. FASEB J 2022; 36:e22326. [PMID: 35657209 DOI: 10.1096/fj.202101742rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 11/11/2022]
Abstract
Histone deacetylase 11 (HDAC11) is aberrantly expressed in many types of cancer, and such abnormalities are associated with tumor immunity and heterogeneous clinical outcomes. Here, we explore the prognostic value and immunological function of HDAC11 across 33 cancer types. We observe HDAC11 is aberrantly expressed in 25 cancer types and positively or negatively associated with prognosis in different cancers. HDAC11 played a protective prognostic role in KIRP, KIRC, LGG, PCPG, READ, and UVM, which was contrary to the conventional opinion that HDAC11 was an oncogenic gene. Moreover, HDAC11 is negatively associated with tumor immune components, most immune checkpoint genes, and key cytokine expression. HDAC11 is correlated with tumor mutational burden in 11 cancer types and with microsatellite instability in 9 cancer types, suggesting HDAC11 may affect a patient's response to immune checkpoint inhibitor (ICI) therapy. In addition, HDAC11 is negatively correlated with the drug sensitivity of oxaliplatin, carmustine, ifosfamide, imexon, lomustine, and BN-2629, indicating the potential synergy between HDAC11 inhibitors and these anti-tumor drugs. In vitro assays indicate that HDAC11 inhibitor SIS17 combined with oxaliplatin shows a synergistic cytotoxic role in K562 cells while SIS17 has an antagonistic effect on the cytotoxic role of oxaliplatin in 769P cells. HDAC11 is also associated with hallmark pathways, including epithelial mesenchymal transition, IL-6/JAK/STAT3, and allograft rejection pathways. Overall, we provide clues regarding the key role of HDAC11 in multiple cancers.
Collapse
Affiliation(s)
- Ran Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolu Wu
- Department of Child Health Care, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Ping Zhao
- Department of Biology, University of North Alabama, Florence, Alabama, USA
| | - Kai Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Walker SM, Gautam R, Turkbey B, Malayeri A, Choyke PL. Update on Hereditary Renal Cancer and Imaging Implications. Radiol Clin North Am 2020; 58:951-963. [PMID: 32792126 DOI: 10.1016/j.rcl.2020.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Up to 8% of renal cancers are thought to have a hereditary component. Several hereditary renal cancer syndromes have been identified over the last few decades. It is important for the radiologist to be aware of findings associated with hereditary renal cancer syndromes to detect tumors early, enroll patients in appropriate surveillance programs, and improve outcomes for the patient and affected family members. This review discusses from a radiologist's perspective well-known hereditary renal cancer syndromes and emerging genetic mutations associated with renal cancer that are less well characterized, focusing on imaging features and known associations.
Collapse
Affiliation(s)
- Stephanie M Walker
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Rabindra Gautam
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ashkan Malayeri
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Alterations of chromosome 3p in 24 cases of gastrinomas and their correlations with clinicopathological and prognostic features. JOURNAL OF PANCREATOLOGY 2020. [DOI: 10.1097/jp9.0000000000000034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
4
|
Karakaxas D, Gazouli M, Liakakos T, Vaiopoulou A, Apessou D, Papaparaskeva K, Patapis P, Dervenis C. Pancreatic neuroendocrine tumors: current opinions on a rare, but potentially curable neoplasm. Eur J Gastroenterol Hepatol 2014; 26:826-35. [PMID: 24987821 DOI: 10.1097/meg.0000000000000138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pancreatic neuroendocrine tumors (PNETs) share a unique genetic identity, functional behavior, and clinical course. Compared with tumors of the exocrine pancreas, they are rare and show a different biologic behavior and prognosis. On the basis of data from recent studies, all PNETs, outside of small insulinomas, should be considered potentially malignant and treated accordingly. Untreated tumors have a high possibility to grow locally into adjacent structures or spread to distant organs. Although surgical excision irrespective of tumor functioning or nonfunctioning state remains the cornerstone of therapy, providing the best disease-free and survival rates to date, the understanding of the genetic nature of the disease yields new 'targets' to consider in drug development. The aim of this review is to summarize all recent advances of genetic research and new drug development in terms of PNETs, especially their genetic identity and subsequent alterations leading to the development of near or total malignant activity, and the new medical treatment strategies of this potentially curable disease on the basis of therapeutical agents acting, where possible, at the genetic level.
Collapse
Affiliation(s)
- Dimitrios Karakaxas
- aSurgical Department-HPB Surgical Unit, Konstantopouleion Agia Olga General Hospital bLaboratory of Biology, Department of Basic Medical Science, School of Medicine, University of Athens cThird Department of Surgery, University of Athens School of Medicine, Attikon University Hospital, Athens, Greece
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Meeker A, Heaphy C. Gastroenteropancreatic endocrine tumors. Mol Cell Endocrinol 2014; 386:101-20. [PMID: 23906538 DOI: 10.1016/j.mce.2013.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 02/06/2023]
Abstract
Gastroenteropancreatic endocrine tumors (GEP-NETs) are relatively uncommon; comprising approximately 0.5% of all human cancers. Although they often exhibit relatively indolent clinical courses, GEP-NETs have the potential for lethal progression. Due to their scarcity and various technical challenges, GEP-NETs have been understudied. As a consequence, we have few diagnostic, prognostic and predictive biomarkers for these tumors. Early detection and surgical removal is currently the only reliable curative treatment for GEP-NET patients; many of whom, unfortunately, present with advanced disease. Here, we review the genetics and epigenetics of GEP-NETs. The last few years have witnessed unprecedented technological advances in these fields, and their application to GEP-NETS has already led to important new information on the molecular abnormalities underlying them. As outlined here, we expect that "omics" studies will provide us with new diagnostic and prognostic biomarkers, inform the development of improved pre-clinical models, and identify novel therapeutic targets for GEP-NET patients.
Collapse
Affiliation(s)
- Alan Meeker
- The Johns Hopkins University School of Medicine, Department of Pathology, Bond Street Research Annex Bldg., Room B300, 411 North Caroline Street, Baltimore, MD 21231, United States.
| | - Christopher Heaphy
- The Johns Hopkins University School of Medicine, Department of Pathology, Bond Street Research Annex Bldg., Room B300, 411 North Caroline Street, Baltimore, MD 21231, United States
| |
Collapse
|
6
|
Abstract
The field of epigenetics has evolved rapidly over recent years providing insight into the tumorigenesis of many solid and haematological malignancies. Determination of epigenetic modifications in neuroendocrine tumour (NET) development is imperative if we are to improve our understanding of the biology of this heterogenous group of tumours. Epigenetic marks such as DNA methylation at RASSF1A are frequent findings in NETs of all origins and may be associated with worse prognosis. MicroRNA signatures and histone modifications have been identified which can differentiate subtypes of NET and distinguish NET from adenocarcinoma in cases of diagnostic uncertainty. Historically, candidate gene-driven approaches have yielded limited insight into the epigenetics of NET. Recent progress has been facilitated by development of high-throughput tools including second-generation sequencing and arrays for analysis of the 'epigenome' of tumour and normal tissue, permitting unbiased approaches such as exome sequencing that identified mutations of chromatin-remodelling genes ATRX/DAXX in 44% of pancreatic NETs. Epigenetic changes are reversible and therefore represent an attractive therapeutic target; to date, clinical outcomes of epigenetic therapies in solid tumours have been disappointing; however, in vitro studies on NETs are promising and further clinical trials are required to determine utility of this class of novel agents. In this review, we perform a comprehensive evaluation of epigenetic changes found in NETs to date, including rare NETs such as phaeochromocytoma and adrenocortical tumours. We suggest priorities for future research and discuss potential clinical applications and novel therapies.
Collapse
Affiliation(s)
- A Karpathakis
- University College London Cancer Institute, 72 Huntley Street, London WC1E 6BT, UK
| | | | | |
Collapse
|
7
|
Ro C, Chai W, Yu VE, Yu R. Pancreatic neuroendocrine tumors: biology, diagnosis,and treatment. CHINESE JOURNAL OF CANCER 2012; 32:312-24. [PMID: 23237225 PMCID: PMC3845620 DOI: 10.5732/cjc.012.10295] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic neuroendocrine tumors (PNETs), a group of endocrine tumors arising in the pancreas, are among the most common neuroendocrine tumors. The genetic causes of familial and sporadic PNETs are somewhat understood, but their molecular pathogenesis remains unknown. Most PNETs are indolent but have malignant potential. The biological behavior of an individual PNET is unpredictable; higher tumor grade, lymph node and liver metastasis, and larger tumor size generally indicate a less favorable prognosis. Endocrine testing, imaging, and histological evidence are necessary to accurately diagnose PNETs. A 4-pronged aggressive treatment approach consisting of surgery, locoregional therapy, systemic therapy, and complication control has become popular in academic centers around the world. The optimal application of the multiple systemic therapeutic modalities is under development; efficacy, safety, availability, and cost should be considered when treating a specific patient. The clinical presentation, diagnosis, and treatment of specific types of PNETs and familial PNET syndromes, including the novel Mahvash disease, are summarized.
Collapse
Affiliation(s)
- Cynthia Ro
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | |
Collapse
|
8
|
Sunitinib in pancreatic neuroendocrine tumors. Target Oncol 2012; 7:117-25. [DOI: 10.1007/s11523-012-0220-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 05/04/2012] [Indexed: 02/02/2023]
|
9
|
Philips S, Shah SN, Vikram R, Verma S, Shanbhogue AKP, Prasad SR. Pancreatic endocrine neoplasms: a current update on genetics and imaging. Br J Radiol 2012; 85:682-96. [PMID: 22253347 PMCID: PMC3474103 DOI: 10.1259/bjr/85014761] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pancreatic endocrine neoplasms are rare pancreatic tumours that may occur sporadically or as part of inherited syndromes such as multiple endocrine neoplasia-1 syndrome, von Recklinghausen disease, von Hippel-Lindau syndrome and tuberous sclerosis complex. Recent advances in the genetics and pathology of hereditary syndromes have provided valuable insights into the pathophysiology and biology of sporadic pancreatic endocrine neoplasms. Evolving molecular data on the biology of these neoplasms have the potential for diagnostic, therapeutic and prognostic use.
Collapse
Affiliation(s)
- S Philips
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
10
|
Goldstein R, Meyer T. Role of everolimus in pancreatic neuroendocrine tumors. Expert Rev Anticancer Ther 2011; 11:1653-65. [PMID: 21932937 DOI: 10.1586/era.11.145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Survival from pancreatic neuroendocrine tumors has not improved over the past two decades and, until recently, streptozocin was the last therapeutic agent approved for this malignancy. Everolimus blocks mTOR, which plays an integral role in cell growth, mitosis and angiogenesis. Abnormal PI3K-Akt/PKB-mTOR pathway signaling has been implicated in the pathogenesis of pancreatic neuroendocrine tumors. In a Phase III study, patients with low- and intermediate-grade advanced pancreatic neuroendocrine tumors were randomized to receive everolimus 10 mg/day or placebo. Median progression-free survival was significantly greater in patients treated with everolimus than placebo - 11 versus 4.6 months - and drug-related adverse events were consistent with the known side-effect profile of everolimus. Everolimus represents a significant treatment development for pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Robert Goldstein
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | | |
Collapse
|
11
|
Amato E, Barbi S, Malpeli G, Bersani S, Pelosi G, Capelli P, Scarpa A. Chromosome 3p alterations in pancreatic endocrine neoplasia. Virchows Arch 2010; 458:39-45. [PMID: 20981439 PMCID: PMC3016198 DOI: 10.1007/s00428-010-1001-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 09/28/2010] [Accepted: 10/10/2010] [Indexed: 01/28/2023]
Abstract
Pancreatic endocrine tumors (PET) are rare neoplasms classified as functioning (F-PET) or non-functioning (NF-PET) according to the presence of a clinical syndrome due to hormonal hypersecretion. PETs show variable degrees of clinical aggressiveness and loss of chromosome 3p has been suggested to be associated with an advanced stage of disease. We assessed chromosome 3p copy number in 113 primary PETs and 32 metastases by fluorescence in situ hybridization (FISH) using tissue microarrays. The series included 56 well-differentiated endocrine tumors (WDET), 62 well-differentiated endocrine carcinomas (WDEC), and 6 poorly differentiated endocrine carcinomas (PDEC). Chromosome 3p alterations were found in 23/113 (20%) primary tumors, with losses being predominant over gains (14% vs. 6%). Loss of 3p was found in 5/55 (9%) WDET, 11/52 (21%) WDEC, and never in PDEC. Gains of 3p were detected in 4/55 (7%) WDET, no WDEC, but notably in 3/6 (50%) PDEC (OR 23.6; P = 0.003). Metastases were more frequently monosomic for 3p compared to primary tumors (OR 3.6; P = 0.005). Monosomy was significantly associated with larger tumor size, more advanced tumor stage, and metastasis. No association was found with survival. Chromosome 3p copy number alterations are frequent events in advanced stage PET, with gains prevailing in PDEC while losses are more frequent in WDEC, supporting the view that a specific pattern of alterations are involved in these diverse disease subtypes.
Collapse
Affiliation(s)
- Eliana Amato
- ARC-NET Center for Applied Research on Cancer, Hospital Concern and University School of Medicine, Verona, Italy
| | | | | | | | | | | | | |
Collapse
|
12
|
Capelli P, Martignoni G, Pedica F, Falconi M, Antonello D, Malpeli G, Scarpa A. Endocrine neoplasms of the pancreas: pathologic and genetic features. Arch Pathol Lab Med 2009; 133:350-64. [PMID: 19260741 DOI: 10.5858/133.3.350] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT Pancreatic endocrine neoplasms (PENs) are diagnostically challenging tumors whose natural history is largely unknown. Histopathology allows the distinction of 2 categories: poorly differentiated high-grade carcinomas and well-differentiated neoplasms. The latter include more than 90% of PENs whose clinical behavior varies from indolent to malignant and cannot be predicted by their morphology. OBJECTIVES To review the literature and report on additional primary material about the clinicopathologic features, classification, staging, grading, and genetic features of PENs. DATA SOURCES Literature review of relevant articles indexed in PubMed (US National Library of Medicine) and primary material from the authors' institution. CONCLUSIONS The diagnosis of PEN is generally easy, but unusual features may induce misdiagnosis. Immunohistochemistry solves the issue, provided that the possibility of a PEN has been considered. Morphology allows the distinction of poorly differentiated aggressive carcinomas from well-differentiated neoplasms. The World Health Organization classification criteria allow for the discernment of the latter into neoplasms and carcinomas with either benign or uncertain behavior. The recently proposed staging and grading systems hold great promise for permitting a stratification of carcinomas into clinically significant risk categories. To date, inactivation of the MEN1 gene remains the only ascertained genetic event involved in PEN genesis. It is inactivated in roughly one-third of PENs. The degree of genomic instability correlates with the aggressiveness of the neoplasm. Gene silencing by promoter methylation has been advocated, but a formal demonstration of the involvement of specific genes is still lacking. Expression profiling studies are furnishing valuable lists of mRNAs and noncoding RNAs that may advance further the research to discover novel markers and/or therapeutic targets.
Collapse
Affiliation(s)
- Paola Capelli
- Department of Pathology, Section ofAnatomical Pathology, Policlinico G. B. Rossi, 37134 Verona, Italy.
| | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Jensen RT, Berna MJ, Bingham DB, Norton JA. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies. Cancer 2008; 113:1807-43. [PMID: 18798544 DOI: 10.1002/cncr.23648] [Citation(s) in RCA: 286] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic endocrine tumors (PETs) can occur as part of 4 inherited disorders, including Multiple Endocrine Neoplasia type 1 (MEN1), von Hippel-Lindau disease (VHL), neurofibromatosis 1 (NF-1) (von Recklinghausen disease), and the tuberous sclerosis complex (TSC). The relative frequency with which patients who have these disorders develop PETs is MEN1>VHL>NF-1>TSC. Over the last few years, there have been major advances in the understanding of the genetics and molecular pathogenesis of these disorders as well in the localization and the medical and surgical treatment of PETs in such patients. The study of PETs in these disorders not only has provided insights into the possible pathogenesis of sporadic PETs but also has presented several unique management and treatment issues, some of which are applicable to patients with sporadic PETs. Therefore, the study of PETs in these uncommon disorders has provided valuable insights that, in many cases, are applicable to the general group of patients with sporadic PETs. In this article, these areas are reviewed briefly along with the current state of knowledge of the PETs in these disorders, and the controversies that exist in their management are summarized briefly and discussed.
Collapse
Affiliation(s)
- Robert T Jensen
- Digestive Diseases Branch, National Institutes of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | |
Collapse
|
15
|
Chaturvedi D, Khadgawat R, Kanakamani J, Kulshreshtha B, Gupta DK, Karak AK, Mathur SR, Kulkarni KK, Seith A, Ammini AC. Management challenges in a child with hyperinsulinemic hypoglycemia. Clin Pediatr Endocrinol 2008; 17:61-4. [PMID: 24790364 PMCID: PMC4004855 DOI: 10.1297/cpe.17.61] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 03/31/2008] [Indexed: 11/22/2022] Open
Abstract
Malignant insulinoma is very rare in children. Herein, we present a case of a
child with malignant insulinoma along with islet cell hyperplasia. She initially presented
with features of hyperinsulinemic hypoglycemia at 18 mo of age. Magnetic resonance imaging
(MRI) of the abdomen showed a mass at the junction of the head and body of the pancreas.
The tumor was enucleated. Five months later symptoms of hypoglycemia recurred. A subtotal
pancreatectomy was performed. She continued to have hypoglycemia, although less
frequently. She was put on increasing doses of diazoxide. Seven months later, MRI of the
abdomen and a PET scan revealed metastatic deposits in the liver, which were confirmed by
histopathology and immunostaining. To the best of our knowledge, this is the youngest
child with metastatic insulinoma reported so far.
Collapse
Affiliation(s)
- Deepti Chaturvedi
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Khadgawat
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - Jeyaraman Kanakamani
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - Bindu Kulshreshtha
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | - Devender K Gupta
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Asis Kumar Karak
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalyani K Kulkarni
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashu Seith
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Ariachery C Ammini
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Abstract
Endocrine pancreatic tumors (EPTs) are uncommon, having an incidence of one per 100,000 people. They may appear as sporadic tumors or be associated with hereditary syndromes. EPTs are categorized as functioning or nonfunctioning tumors, based on the presence or absence of clinical syndromes. Among the former, insulinomas and gastrinomas are the most common. For the histopathological investigation of EPTs, chromogranin A and synaptophysin immunostainings are recommended. Measurement of circulating chromogranin A is also the cornerstone for the biochemical diagnosis of these tumors. Furthermore, specific hormones produced and released by the neoplastic cells can be identified by immunostaining and used for biochemical evaluation. To locate EPTs, both noninvasive (ultrasonography, computerized tomography, MRI and radionuclear imaging) and invasive techniques (arterial stimulation with venous sampling) can be used. Debulking procedures (surgery, radiofrequency ablation, embolization/chemoembolization and liver transplantation) and/or medical treatment (chemotherapy, biotherapy and peptide receptor radionuclide therapy) are the options available for the treatment of EPTs. Understanding the molecular events underlying the pathobiology of EPTs will aid the development of more accurate diagnostic/prognostic markers and give guidance for improved therapeutic modalities.
Collapse
Affiliation(s)
- Apostolos V Tsolakis
- a Department of Medical Sciences, Section of Endocrine Oncology, University Hospital, 751 85 Uppsala, Sweden.
| | - Eva T Janson
- b Department of Medical Sciences, Section of Endocrine Oncology, University Hospital, 751 85 Uppsala, Sweden.
| |
Collapse
|
17
|
Kasiyanov A, Fujii N, Tamamura H, Xiong H. Modulation of network-driven, GABA-mediated giant depolarizing potentials by SDF-1alpha in the developing hippocampus. Dev Neurosci 2007; 30:285-92. [PMID: 18073458 DOI: 10.1159/000112520] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Accepted: 09/07/2007] [Indexed: 11/19/2022] Open
Abstract
Chemokine stromal cell-derived factor-1 (SDF-1, or CXCL12) plays an important role in brain development and functioning. Whole-cell patch clamp recordings were conducted on CA3 neurons in hippocampal slices prepared from neonatal rats between postnatal days 2 and 6 to study the modulatory effects of SDF-1alpha on network-driven, gamma-aminobutyric-acid-mediated giant depolarizing potentials (GDPs), a hallmark of the developing hippocampus. We found that SDF-1alpha, the only natural ligand for chemokine CXC motif receptor 4 (CXCR4), decreased GDP firing without significant effects on neuronal passive membrane properties in neonatal hippocampal neurons. The SDF-1alpha-mediated decrease in GDP firing was blocked by T140, a CXCR4 receptor antagonist, suggesting that SDF-1alpha modulates GDP firing via CXCR4. We also showed that endogenous SDF-1 exerts a tonic inhibitory action on GDPs in the developing hippocampus. As SDF-1/CXCR4 are highly expressed in the developing brain and GDPs are involved in activity-dependent synapse formation and functioning, the inhibitory action of SDF-1alpha on GDPs may reflect a potential mechanism for chemokine regulation of neural development in early neonatal life.
Collapse
Affiliation(s)
- Alexander Kasiyanov
- Neurophysiology Laboratory, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
18
|
Perren A, Anlauf M, Komminoth P. Molecular profiles of gastroenteropancreatic endocrine tumors. Virchows Arch 2007; 451 Suppl 1:S39-46. [PMID: 17684763 DOI: 10.1007/s00428-007-0449-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
Neuroendocrine tumors of the gastroenteropancreatic system are defined by their endocrine phenotype and share many histopathological and clinical features. However, the fact that the hormone production of tumors depends on their site of origin, that the tumors differ in their biology, and that the association with familial syndromes is nonrandom suggests heterogeneity. It is therefore conceivable that the gastroenteropancreatic neuroendocrine tumors also differ in their molecular profile. This review summarizes and discusses the available data in this field.
Collapse
Affiliation(s)
- Aurel Perren
- Department of Pathology, University Hospital Zürich, Zurich, Switzerland.
| | | | | |
Collapse
|
19
|
Abstract
Neuroendocrine tumors can develop either sporadically or in association with familial syndromes such as multiple endocrine neoplasia type 1 (MEN1), multiple endocrine neoplasia type 2 (MEN2) or von Hippel-Lindau (VHL). A variety of genetic approaches has been utilized to dissect the underlying molecular pathogenesis of these distinctive tumors, including genome-wide screens such as comparative genomic hybridization, loss of heterozygosity and DNA microarray analysis as well as targeted investigations into specific tumor suppressor gene and oncogene candidates. The identification of the MEN1 tumor suppressor gene that underlies the MEN1 syndrome has provided important new insights into tumor pathogenesis. In addition, a number of independent approaches has converged on a pivotal role for regulators of the cell cycle. However, our understanding of the molecular biology of these tumors remains far from complete. In this review we highlight some of the key approaches, findings and implications of these genetic studies.
Collapse
Affiliation(s)
- Eva-Maria Duerr
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Neuroendocrine tumors of the pancreas are a small subgroup of tumors characterized by a variety of biological behaviors. Recent changes in their classification should help better define the prognosis of this diverse group of tumors. With recent advances in diagnosis and staging, the treatment options for all neuroendocrine tumors have evolved. Presented here is a review of the current-day knowledge for neuroendocrine tumors of the pancreas. RECENT FINDINGS A consensus by leading experts in the neuroendocrine tumors field has proposed an algorithm for the diagnosis, treatment and follow-up of these rare tumors. Surgical resection remains the first-line therapy. Alternative forms of cytoreduction such as radiofrequency ablation and embolization, have increased the ability of the surgeon to debulk these tumors, resulting in improved survival and better palliation. Contrary to adenocarcinoma of the pancreas, hormonal and biotherapy offer unique treatment strategies for these rare tumors. Very recent developments utilizing radionuclide therapy hold promise for not only palliation, but may prove to be a beneficial form of adjuvant therapy. SUMMARY Presented here is a summary of the recent literature on the diagnosis and treatment of neuroendocrine tumors of the pancreas.
Collapse
Affiliation(s)
- Elijah Dixon
- Department of Surgery and Oncology, University of Calgary and Tom Baker Cancer Center, Calgary, Alberta, Canada
| | | |
Collapse
|
21
|
Abstract
Although the majority of pancreatic neoplasms are infiltrating ductal adenocarcinomas or other neoplasms with ductal differentiation, neoplasms with acinar, endocrine, mixed, or uncertain differentiation constitute a diverse and distinctive group. The most common and best-characterized nonductal neoplasms are pancreatic endocrine neoplasm, acinar cell carcinoma, pancreatoblastoma, and solid pseudopapillary neoplasm. This review details the clinical and pathologic features of these nonductal neoplasms, highlighting diagnostic criteria including the use of specific immunohistochemical stains to define the cellular differentiation of the neoplasms.
Collapse
Affiliation(s)
- David S Klimstra
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Lindberg D, Hessman O, Akerström G, Westin G. Cyclin-dependent kinase 4 (CDK4) expression in pancreatic endocrine tumors. Neuroendocrinology 2007; 86:112-8. [PMID: 17664862 DOI: 10.1159/000106762] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 06/26/2007] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIMS Pancreatic endocrine tumors (PETs) occur sporadically, in association with the multiple endocrine neoplasia type 1 (MEN1) and the von Hippel-Lindau syndromes. CDK4 is central to the cell cycle control in pancreatic beta cells, and we have assessed whether CDK4 expression is deregulated in 18 human sporadic or familial PETs. METHODS Real-time quantitative PCR, immunohistochemistry, DNA sequencing, and Western blot analysis were used. RESULTS CDK4 mRNA was expressed in all PETs within the range of the arbitrary control. CDK4 protein was absent in normal pancreatic islets but distinctly expressed in all PETs as determined by immunohistochemistry. CDK4 expression was confirmed by Western blot analysis. No significant differences of CDK4 expression were observed between the groups of benign and malignant PETs or between tumors with or without MEN1 gene mutations. CDK4 expression was not due to gene amplification, and no mutations were identified in coding exons and RNA splice sites. c-Myc is known to be overexpressed in PETs and directly augments CDK4 expression in other cell types. Analysis of consecutive tissue sections for CDK4 and c-Myc showed overlapping homo- or heterogeneous immunostaining in all 18 PETs. CONCLUSION We conclude that CDK4 and c-Myc is generally expressed in benign and malignant PETs, and regardless of MEN1 mutational status. Targeting of CDK4 may present an alternative to traditional chemotherapy of PETs in the future.
Collapse
Affiliation(s)
- Daniel Lindberg
- Department of Surgical Sciences, Endocrine Unit, Uppsala University Hospital, Uppsala, Sweden
| | | | | | | |
Collapse
|
23
|
Abstract
Gastrointestinal and pancreatic neuroendocrine tumors originate from the cells of the diffuse endocrine system. Their molecular genetic mechanism of development and progression is complex and remains largely unknown, and they are different in genetic composition from the gastrointestinal epithelial tumors. The current literature suggests that multiple genes are involved in their tumorigenesis with significant differences for tumors of different embryological derivatives: foregut, midgut and hindgut. The MEN1 gene is involved in initiation of 33% of foregut gastrointestinal neuroendocrine tumors. 18q defects are present almost exclusively in mid/hindgut neuroendocrine tumors. X-chromosome markers are associated with malignant behavior in foregut tumors only. Analysis of poorly differentiated neuroendocrine carcinomas of any site demonstrates high chromosomal instability and frequent p53 alterations similar to other poorly differentiated carcinomas. Several factors played a limiting role in the molecular studies published to date: the tumors are rare and heterogeneous, it is difficult to predict their behavior and prognosis, and several different tumor classifications are used by the investigators in the studies. Future studies need to evaluate molecular genetic composition of large series of gastrointestinal and pancreatic neuroendocrine tumors of each specific tumor type. Understanding of specific genetic alterations characteristic for gastrointestinal and pancreatic neuroendocrine tumors might lead to their improved diagnosis, morphologic and molecular characterization and treatment.
Collapse
Affiliation(s)
- Irina A Lubensky
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis National Cancer Institute, National Institutes of Health, 6130 Executive Blvd, EPN 6032, Rockville, MD 20892, USA.
| | | |
Collapse
|
24
|
Lindberg D, Akerström G, Westin G. Mutational analyses of WNT7A and HDAC11 as candidate tumour suppressor genes in sporadic malignant pancreatic endocrine tumours. Clin Endocrinol (Oxf) 2007; 66:110-4. [PMID: 17201809 DOI: 10.1111/j.1365-2265.2006.02694.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE We and others have reported loss of heterozygosity (LOH) on chromosome 3p25 in sporadic malignant pancreatic endocrine tumours (PETs). A common region of deletion on chromosome 3p25 contains numerous genes, including VHL and PPARgamma, that have been excluded previously as candidate tumour suppressor genes by DNA sequencing analysis. We have analysed whether WNT7A or HDAC11 was biallelically inactivated in a group of well-characterized PETs. PATIENTS AND DESIGN Ten PETs from eight patients were selected from a previous study, where LOH on chromosome 3p25 was found in 11 out of 22 sporadic PETs. These tumours were examined for inactivating mutations of WNT7A and HDAC11 by direct sequencing of all exons and intron-exon boundaries. Inactivation of WNT7A expression by aberrant CpG island methylation and WNT7A protein expression were evaluated by methylation-specific polymerase chain reaction (PCR) and immunohistochemistry, respectively. HDAC11 protein expression was also examined. RESULTS No point mutations, deletion or insertions were detected in either WNT7A or HDAC11 in any of the PETs. Two polymorphisms were identified in the third exon of the WNT7A gene. CpG methylation of the WNT7A gene was not detected and the WNT7A and HDAC11 proteins were normally expressed. CONCLUSION The absence of tumour-specific somatic events in WNT7A and HDAC11 suggests that these genes are unlikely to have a classical tumour suppressor gene role in sporadic malignant PETs. The putative 3p25 tumour suppressor remains to be identified.
Collapse
Affiliation(s)
- Daniel Lindberg
- Department of Surgical Sciences, Endocrine Unit, Uppsala University Hospital, Uppsala, Sweden
| | | | | |
Collapse
|
25
|
Zikusoka MN, Kidd M, Eick G, Latich I, Modlin IM. The molecular genetics of gastroenteropancreatic neuroendocrine tumors. Cancer 2006; 104:2292-309. [PMID: 16258976 DOI: 10.1002/cncr.21451] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pathobiology of neuroendocrine tumors (NETs) is hampered by the lack of scientific tools that define their mechanisms of secretion, proliferation, and metastasis; and, currently, there are no accurate means to assess tumor behavior and disease prognosis. Molecular biologic techniques and genetic analysis may facilitate the delineation of the molecular pathology of NETs and provide novel insights into their cellular mechanisms. The current status and recent advances in assessment of the molecular basis of tumorigenesis of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) were reviewed (1981-2004). The objectives of this retrospective study were to provide a cohesive overview of the current state of knowledge and to develop a molecular understanding of these rare tumor entities to facilitate the establishment of therapeutic targets and rational management strategies. Multiple differences in chromosomal aberration patterns were noted between gastrointestinal (GI) neuroendocrine and pancreatic endocrine tumors (PETs). Divergence in gene expression patterns in the development of GI carcinoids and PETs was identified, whereas examination of the PET and GI carcinoid data demonstrated only few areas of overlap in the accumulation of genetic aberrations. These data suggest that the recent World Health Organization classification of GEP-NETs may require updating. In addition, previous assumptions of tumor similarity (pancreatic vs. GI) may be unfounded when they are examined at a molecular level. On the basis of the evolution of genetic information, enteric neuroendocrine lesions (carcinoids) and PETs may need to be classified as two distinct entities rather than grouped together as the single entity "GEP-NETs."
Collapse
Affiliation(s)
- Michelle N Zikusoka
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut 06520-8062, USA
| | | | | | | | | |
Collapse
|
26
|
Bordi C, D'Adda T, Azzoni C, Pizzi S, Bottarelli L, Mormandi F, Antonetti T, Luong TV, Rindi G. Criteria for malignancy in gastrointestinal endocrine tumors. Endocr Pathol 2006; 17:119-29. [PMID: 17159244 DOI: 10.1385/ep:17:2:119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
In contrast with the large amount of data generated from endocrine tumors of the pancreas, sparse and mostly unconfirmed data are available on the criteria for the assessment of malignancy risk and patient outcome in endocrine tumors of the gastrointestinal tract. In these conditions the 2000 WHO classification with its standardized scheme of pathologic report constitutes a framework facilitating the assessment of tumor malignancy and has been regarded as useful for clinical purposes, providing the basis for proper management of the patients and for the design of treatment protocols. The classification is based on a combination of pathological and clinical features with parameters specific for each organ in which the endocrine tumors originate. Three main categories, one further subdivided into two subgroups, are considered: (1) well-differentiated endocrine tumors, further subdivided into tumors with benign and with uncertain behavior; (2) well-differentiated endocrine carcinomas, low grade; and (3) poorly differentiated endocrine carcinomas, high grade. In this review the differential tumor characteristics between the different categories are summarized. Moreover, the relevance of additional features with respect to tumor prognostication, chiefly the Ki-67 proliferation index and malignancy-associated genetic changes, is discussed with emphasis on the discrepancies emerging between tumors of foregut and of midgut origin.
Collapse
Affiliation(s)
- Cesare Bordi
- Department of Pathology and Laboratory Medicine, Section of Anatomic Pathology, University of Parma, Parma, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Azzoni C, Bottarelli L, Pizzi S, D'Adda T, Rindi G, Bordi C. Xq25 and Xq26 identify the common minimal deletion region in malignant gastroenteropancreatic endocrine carcinomas. Virchows Arch 2005; 448:119-26. [PMID: 16244870 DOI: 10.1007/s00428-005-0058-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Accepted: 08/03/2005] [Indexed: 11/30/2022]
Abstract
Loss of heterozygosity (LOH) for markers on X chromosome are associated with malignancy in endocrine tumors of the stomach and pancreas. The aim of this work is to investigate low-grade, well-differentiated endocrine carcinomas (WDEC) vs high-grade, poorly differentiated endocrine carcinomas (PDEC) of the gastroenteropancreatic (GEP) tract for common deletion regions on X chromosome. We performed a comparative allelotyping analysis with 24 highly polymorphic markers for the X chromosome in 12 WDECs and 5 PDECs. Overall, the LOH frequency in all informative loci investigated was 59% in primary and 61% in metastasis, with a significantly higher rate in PDECs than in WDECs (p<0.015 for primary and p<0.00005 for metastasis). In both WDECs and PDECs, the small Xq25 region as defined by DXS8059, DXS8098, and DXS8009 markers showed higher LOH rate as compared to the rest of the chromosome markers (p<0.04). In addition, LOH was very frequently elevated also in DXS294 and in DXS102 loci mapping the chromosomal region Xq26. In no instances differences were found between primary tumors and metastases. Methylation analysis revealed that Xq25 loss preferentially occurred on the inactive X chromosome, a feature in agreement with findings from other human cancers suggesting escape of tumor suppressor genes to X chromosome inactivation at this region. Overall, our data indicate that the two chromosomal regions, Xq25 and Xq26, may participate to the malignant progression of GEP endocrine carcinomas.
Collapse
Affiliation(s)
- Cinzia Azzoni
- Department of Pathology and Laboratory Medicine, Section of Pathological Anatomy, University of Parma, Via Gramsci 14, 43100 Parma, Italy
| | | | | | | | | | | |
Collapse
|
28
|
van Eeden S, Offerhaus GJA. Historical, current and future perspectives on gastrointestinal and pancreatic endocrine tumors. Virchows Arch 2005; 448:1-6. [PMID: 16220293 DOI: 10.1007/s00428-005-0082-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 08/25/2005] [Indexed: 01/26/2023]
Abstract
Gastrointestinal and pancreatic endocrine tumors are neoplasms of which the pathogenesis is not completely understood and of which the clinical behavior is difficult to predict. Originally, Masson suggested that the cell of origin was an endocrine cell derived from the gastrointestinal epithelium. However, Pearse showed that the endocrine cells throughout the body shared various features, among others the amine precursor uptake and decarboxylation (APUD) capacity, and postulated the neural crest as the common origin for all APUD cells, a hypothesis that received support from the scientific community for many years. Now, biologists start to elucidate the various transcription factors that drive gastrointestinal development, and it has become evident that Masson was presumably right. Transcription factors relevant for development may also operate during tumorigenesis, and their expression may determine tumor biology. With other genetic factors, they may play a role in the pathogenesis of gastrointestinal and pancreatic endocrine tumors, and perhaps, their expression will turn out to be of prognostic or therapeutic value. In this review, current knowledge on the development of endocrine cells, hypotheses on the origin of endocrine tumors, genetic alterations, and prognostic factors are discussed. It is suggested that the increasing understanding of the normal development of gastrointestinal and pancreatic endocrine cells, the accumulating data on genetic alterations in endocrine tumors and the reappraisal of the hypotheses on their pathogenesis formulated in the past may help in elucidating their pathogenesis and in more accurately predicting prognosis.
Collapse
Affiliation(s)
- Susanne van Eeden
- Department of Pathology, Academic Medical Center, Meibergdreef 9, 1105, Amsterdam, The Netherlands.
| | | |
Collapse
|
29
|
Papouchado B, Erickson LA, Rohlinger AL, Hobday TJ, Erlichman C, Ames MM, Lloyd RV. Epidermal growth factor receptor and activated epidermal growth factor receptor expression in gastrointestinal carcinoids and pancreatic endocrine carcinomas. Mod Pathol 2005; 18:1329-35. [PMID: 15920550 DOI: 10.1038/modpathol.3800427] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The epidermal growth factor receptor (EGFR) plays an important role in the pathogenesis of many tumors. To analyze the expression of EGFR and activated EGFR in well-differentiated neuroendocrine carcinomas including primary and metastatic gastrointestinal carcinoid tumors and pancreatic endocrine tumors (PET), we examined 58 gastrointestinal carcinoid tumors and 48 PET using immunohistochemistry, Western blotting, and RT-PCR. EGFR and activated EGFR (P-EGFR) were expressed by both gastrointestinal carcinoids and PET in primary and metastatic tumors, although a higher percentage of gastrointestinal carcinoid tumors expressed EGFR and activated EGFR. Western blotting detected a 170 kDa band for both EGFR and activated EGFR in three primary carcinoid tumors and two metastatic carcinoid tumors to the liver. RT-PCR analysis confirmed the expression of EGFR mRNA in both primary and metastatic carcinoid tumors. Patients with activated EGFR expression in their primary PET had a significantly worse prognosis compared to those who did not express activated-EGFR (P = 0.043). These results indicate that gastrointestinal carcinoid tumors as well as PET express EGFR and activated EGFR, and that expression is more common in gastrointestinal carcinoid tumors compared to pancreatic endocrine tumors. These findings implicate the EGFR and P-EGFR signal transduction pathway in the pathogenesis of these neuroendocrine tumors and suggest that targeted therapy directed against the EGFR tyrosine kinase domain may be a useful therapeutic approach in patients with unresectable metastatic gastrointestinal carcinoid tumors and pancreatic endocrine tumors.
Collapse
Affiliation(s)
- Bettina Papouchado
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Endocrine pancreatic tumours (EPTs) are uncommon tumours occurring in approximately 1 in 100,000 of the population, representing 1-2% of all pancreatic neoplasms. Some of the tumours may be part of multiple endocrine neoplasia type one (MEN-1) syndrome or von Hippel-Lindau (vHL) disease. EPTs are classified as functioning or non-functioning tumours on the basis of their clinical manifestation. The biochemical diagnosis of EPT is based on hormones and amines released. Besides specific markers such as insulin, there are also general tumour markers such as chromogranin A, which is the most valuable marker and has been reported to be increased in plasma in 50-80% of patients with EPTs and correlates with tumour burden. The location of endocrine tumours of the pancreas includes different techniques, from endoscopic investigations to scintigraphy (e.g. somatostatin receptor scintigraphy) and positron emission tomography. The medical treatment of endocrine pancreatic tumours consists of chemotherapy, somatostatin analogues and alpha-interferon. None of these can cure a patient with malignant disease. In future, therapy will be custom-made and based on current knowledge of tumour biology and molecular genetics.
Collapse
Affiliation(s)
- Kjell Oberg
- Department of Medical Sciences, University Hospital, 751 85 Uppsala, Sweden.
| | | |
Collapse
|
31
|
Abstract
The neuroendocrine tumors of the pancreas are rare, but belong to the most common endocrine neoplasms of the abdomen. In most cases they present as solitary tumors, composed of monomorphic cells. Functionally active cells are associated with characteristic hormonal syndromes. Using morphological, immunohistochemical and biological criteria, the classification distinguishes between well differentiated tumors with benign or uncertain behavior, well differentiated (low grade malignant) carcinomas and poorly differentiated (high grade malignant) carcinomas. In recent years the percentage of non-functional tumors has increased.
Collapse
Affiliation(s)
- M Anlauf
- Institut für Allgemeine Pathologie, Universitätsklinikum Schleswig-Holstein--Campus Kiel, Michaelisstrasse 11, 24105 Kiel, Germany.
| | | | | |
Collapse
|
32
|
Pizzi S, Azzoni C, Bottarelli L, Campanini N, D'Adda T, Pasquali C, Rossi G, Rindi G, Bordi C. RASSF1A promoter methylation and 3p21.3 loss of heterozygosity are features of foregut, but not midgut and hindgut, malignant endocrine tumours. J Pathol 2005; 206:409-16. [PMID: 15887288 DOI: 10.1002/path.1784] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Ras-association domain family 1A (RASSF1A) tumour suppressor gene is inactivated in a variety of solid tumours, usually by epigenetic silencing of the promoter and/or allelic loss of its locus at 3p21.3. RASSF1A induces cell cycle arrest through inhibition of cyclin D1 accumulation. In this work, 62 endocrine tumours from different sites in the gut were investigated for methylation of the RASSF1A promoter using the polymerase chain reaction, the presence of 3p21.3 deletions by loss of heterozygosity analysis, and cyclin D1 expression by immunohistochemistry. Methylation was found in 20/62 (32%) cases and was restricted to foregut tumours; deletion at 3p21.3 was found in 15/58 (26%) informative cases and restricted to malignant foregut tumours; cyclin D1 hyper-expression was found in 31/58 (53%) cases and correlated with RASSF1A methylation. Our data suggest that RASSF1A is involved in the development of endocrine tumours derived from the foregut only, and that the presence of both RASSF1A methylation and 3p21.3 deletion is associated with malignancy. These results may provide a rationale for foregut-targeted therapy for aggressive endocrine carcinomas entailing the use of demethylating agents.
Collapse
Affiliation(s)
- S Pizzi
- Department of Pathology and Laboratory Medicine, University of Parma, Parma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rindi G, Bordi C. Endocrine tumours of the gastrointestinal tract: aetiology, molecular pathogenesis and genetics. Best Pract Res Clin Gastroenterol 2005; 19:519-34. [PMID: 16183525 DOI: 10.1016/j.bpg.2005.03.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endocrine tumours of the gut and pancreas originate from cells of the diffuse endocrine system and are characterised by the production of a wide variety of bioactive substances including growth factors. Two major tumour categories are distinguished-well-differentiated and poorly differentiated neoplasms-with distinct phenotypes and significantly diverse clinical behaviour. Here, genetic background data are summarised on an anatomical basis for tumours of foregut, midgut and hindgut derivatives. For well-differentiated tumours, independent techniques identified the abnormality of multiple chromosomal sites and genes, pointing to a complex genetic background. Differences in foregut tumours compared with midgut and hindgut tumours are, however, outlined. The multiple endocrine neoplasia syndrome type 1 (MEN1) gene is reported to be involved in about one-third of sporadic foregut endocrine tumours and exceptionally in midgut and hindgut tumours. Similarly, X chromosome markers are associated with malignant behaviour in foregut tumours only. For poorly differentiated carcinomas, a high degree of chromosomal instability is the common genetic trait independent of tumour site and frequently involving the p53 gene.
Collapse
Affiliation(s)
- Guido Rindi
- Department of Pathology and Laboratory Medicine, Section of Anatomic Pathology, University of Parma, Italy.
| | | |
Collapse
|
34
|
Costa-Guda J, Rosen ED, Jensen RT, Chung DC, Arnold A. Mutational analysis of PPARG as a candidate tumour suppressor gene in enteropancreatic endocrine tumours. Clin Endocrinol (Oxf) 2005; 62:603-6. [PMID: 15853832 DOI: 10.1111/j.1365-2265.2005.02267.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Loss of heterozygosity (LOH) or deletion of chromosome 3p is a frequent finding in enteropancreatic endocrine tumours (EPETs), suggesting the pathogenetic involvement of one or more tumour suppressor genes on 3p. PPARG, the gene encoding the gamma isoform of the peroxisome proliferator-activated receptor (PPARgamma), is highly expressed in normal human pancreatic islet cells, is located at 3p25, and has been reported to sustain loss-of-function mutations in human colorectal carcinomas. Additionally, the development of islet cell hyperplasia in an islet cell-specific pparg knockout mouse has further emphasized the attractiveness of PPARG as a candidate gene important in the pathogenesis of EPETs. Therefore, we sought to examine PPARG for intragenic inactivating mutations, the evidence needed to rigorously establish it as a tumour suppressor in EPETs. PATIENTS AND DESIGN Twenty-three EPETs from 20 patients were examined for coding region mutations in PPARG and for LOH on 3p at microsatellite markers flanking PPARG. RESULTS LOH on 3p was detected in tumours from six patients (30%), but no intragenic mutations were detected in PPARG, whether or not LOH was present. CONCLUSION These findings strongly suggest that PPARG does not commonly function as a classical tumour suppressor gene in the pathogenesis of EPETs.
Collapse
Affiliation(s)
- Jessica Costa-Guda
- Center for Molecular Medicine and Division of Endocrinology and Metabolism, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
Only relatively recently has there been an increased clinical recognition and characterization of the heterogeneous group of rare gastroenteropancreatic neuroendocrine neoplasms. Most have endocrine function and exhibit varying degrees of malignancy. This review summarizes the derivation of these tumors and the advances in their diagnosis and treatment over the past decade and a half. They are varied in their biological behavior and clinical courses and, depending on their cell type, can produce different hormones causing distinct clinical endocrine syndromes (insulinoma [hypoglycemia], gastrinoma [Zollinger-Ellison syndrome (ZES)], vasoactive intestinal peptideoma [VIPoma], watery diarrhea, hypokalemia-achlorhydria [WDHA], glucagonoma [glucagonoma syndrome], and so forth). In addition to surgery for cure or palliation (by excision and a variety of other cytoreductive techniques), they each are treated with anti-hormonal agents or drugs targeted to each tumor's specific product or its effects. The majority have benefited from the gut hormone-inhibiting action of somatostatin analogs. Because of their usual slow rate of growth it is recommended that, even when they are advanced and incurable, unlike in patients with common and more malignant cancers, patients with neuroendocrine tumors often can be palliated and appear to survive longer when managed with an active approach using sequential multimodality treatment. Advances in these various therapies are reviewed and the beneficial emergence of global self-help patient support groups is noted.
Collapse
Affiliation(s)
- Richard R P Warner
- Gastrointestinal Division, Department of Medicine, The Mount Sinai School of Medicine, New York, New York 10128, USA.
| |
Collapse
|
36
|
Kouvaraki MA, Solorzano CC, Shapiro SE, Yao JC, Perrier ND, Lee JE, Evans DB. Surgical treatment of non-functioning pancreatic islet cell tumors. J Surg Oncol 2005; 89:170-85. [PMID: 15719379 DOI: 10.1002/jso.20178] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pancreatic endocrine tumors (PETs) are rare neoplasms originating from the amine precursor uptake and decarboxylation (APUD) stem cells. Although the majority of PETs are sporadic, they frequently occur in familial syndromes. PETs may cause a variety of functional syndromes or symptoms of local progression if they are non-functional. General neuroendocrine tumor markers are highly sensitive in the diagnostic assessment of a PET. Imaging studies for tumor localization and staging include computer tomography (CT) scan, magnetic resonance imaging (MRI), In(111)-octreotide scan, MIBG, and endoscopic ultrasonography (EUS). Treatment of PETs often requires a multi-modality approach; however, surgical resection remains the only curative therapy for localized (non-metastatic) disease. Treatment of metastatic disease includes biologic agents, cytotoxic chemotherapy, and liver-directed therapies.
Collapse
Affiliation(s)
- Maria A Kouvaraki
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Couvelard A, O'Toole D, Turley H, Leek R, Sauvanet A, Degott C, Ruszniewski P, Belghiti J, Harris AL, Gatter K, Pezzella F. Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer 2005; 92:94-101. [PMID: 15558070 PMCID: PMC2361752 DOI: 10.1038/sj.bjc.6602245] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumour-associated angiogenesis is partly regulated by the hypoxia-inducible factor (HIF) pathway. Endocrine tumours are highly vascularised and the molecular mechanisms of their angiogenesis are not fully delineated. The aim of this study is to evaluate angiogenesis and expression of HIF-related molecules in a series of patients with pancreatic endocrine tumours (PETs). The expression of vascular endothelial growth factor (VEGF), HIF-1alpha, HIF-2alpha and carbonic anhydrase 9 (CA9) was examined by immunohistochemistry in 45 patients with PETs and compared to microvascular density (MVD), endothelial proliferation, tumour stage and survival. Microvascular density was very high in PETs and associated with a low endothelial index of proliferation. Microvascular density was significantly higher in benign PETs than in PETs of uncertain prognosis, well-differentiated and poorly differentiated carcinomas (mean values: 535, 436, 252 and 45 vessels mm(-2), respectively, P < 0.0001). Well-differentiated tumours had high cytoplasmic VEGF and HIF-1alpha expression. Poorly differentiated carcinomas were associated with nuclear HIF-1alpha and membranous CA9 expression. Low MVD (P = 0.0001) and membranous CA9 expression (P = 0.0004) were associated with a poorer survival. Contrary to other types of cancer, PETs are highly vascularised, but poorly angiogenic tumours. As they progress, VEGF expression is lost and MVD significantly decreases. The regulation of HIF signalling appears to be specific in pancreatic endocrine tumours.
Collapse
Affiliation(s)
- A Couvelard
- Department of Pathology, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110 Clichy, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Furlan D, Cerutti R, Uccella S, La Rosa S, Rigoli E, Genasetti A, Capella C. Different molecular profiles characterize well-differentiated endocrine tumors and poorly differentiated endocrine carcinomas of the gastroenteropancreatic tract. Clin Cancer Res 2004; 10:947-57. [PMID: 14871972 DOI: 10.1158/1078-0432.ccr-1068-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The molecular pathogenesis of gastroenteropancreatic endocrine tumors (ETs) is still largely unknown. The purpose of this work was a molecular characterization of 38 gastroenteropancreatic ETs with respect to the primary site and to the morphofunctional profile, pointing out useful diagnostic or prognostic molecular markers. EXPERIMENTAL DESIGN Twenty-four well-differentiated ETs or carcinomas (WDET/Cs; 11 pancreatic, 3 gastric, and 10 intestinal) and 14 poorly differentiated endocrine carcinomas (1 pancreatic, 6 gastric, and 7 colorectal) were microallelotyped using 38 polymorphic microsatellite markers covering chromosomes 1, 3, 5q, 6, 11, 17, and 18. RESULTS Regardless of the primary site, a significantly higher percentage of allelic imbalances (AIs) was observed in poorly differentiated endocrine carcinomas than in WDET/Cs (P = 0.012), except for 3 of 8 nonfunctioning pancreatic endocrine tumors and 1 colorectal WDEC, exhibiting multiple AIs on chromosomes 1, 3, 6, and 11. A strong positive correlation between AI percentage and Ki-67 proliferation index was detected considering both the whole series of ETs (P = 0.004) and the group of WDET/Cs alone (P = 0.011). The survival analysis showed a positive correlation between low percentage of AI and longer survival (P = 0.01). No recurrent AIs at specific chromosomal regions were identifiable with respect to the primary site. CONCLUSIONS The malignant progression of endocrine tumors seems to be associated with complex allelotypes and chromosomal instability. Although no specific molecular markers of malignancy can be defined with certainty, the ploidy status and the degree of chromosomal derangements appear to be the most informative genetic factors with prognostic significance.
Collapse
Affiliation(s)
- Daniela Furlan
- Department of Human Morphology, University of Insubria and Ospedale di Circolo, Viale Borri 57, 21100 Varese, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- John C Mansour
- Department of Surgery, University of Wisconsin, Madison, Wisconsin 53792, USA
| | | |
Collapse
|
40
|
Abstract
The morphology of pancreatic endocrine tumors (PETs) is similar to that of endocrine tumors elsewhere in the body. PETs are usually encountered in adults. They may be clinically functional and associated with various syndromes related to hormone excess. However, it must be remembered that absence of obvious clinical symptoms may not necessarily reflect true lack of clinical function, and subtle clinical manifestations may be missed. Current thinking indicates that PETs arise from totipotential stem cells as well as preexisting endocrine cells. PETs may be hereditary or sporadic. The hereditary forms are associated with multiple endocrine neoplasia type 1 (MEN-1), von Hippel-Lindau syndrome, neurofibromatosis, and tuberous sclerosis. In sporadic PETs, the most consistent and recurring chromosomal abnormality is allelic loss of chromosome 11q, which includes the MEN-1 locus. Loss of a sex chromosome has been shown to be associated with metastasis, local invasion, and poor survival.
Collapse
Affiliation(s)
- Runjan Chetty
- Department of Pathology, University Health Network/Toronto Medical Laboratories, University of Toronto, Toronto, Canada
| | | |
Collapse
|
41
|
Perren A, Komminoth P, Heitz PU. Molecular genetics of gastroenteropancreatic endocrine tumors. Ann N Y Acad Sci 2004; 1014:199-208. [PMID: 15153435 DOI: 10.1196/annals.1294.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
To elucidate the molecular background of sporadic gastroenteropancreatic endocrine tumors, several investigations into chromosomal alterations and allelic imbalances have identified several chromosomal regions of interest. These regions might harbor candidate genes important in tumor development and progression. However, only a small number of genes have been thoroughly analyzed, and only very few were shown to be altered in a substantial subset of tumors. Therefore, we are far from understanding the molecular mechanisms of tumor initiation and progression in gastroenteropancreatic endocrine tumors, although some "molecular patterns" are currently emerging. In this review, chromosomal alterations, that is, allelic losses and gene mutations, identified in gastroenteropancreatic endocrine tumors are briefly summarized. Molecular differences among various subtypes of gastroenteropancreatic endocrine tumors are highlighted in view of their role as indicators of separate genetic pathways.
Collapse
Affiliation(s)
- Aurel Perren
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
42
|
Abstract
Human neuroendocrine tumors exhibit unique biological properties, and defining the molecular genetic alterations that underlie these distinctive features remains an important challenge. In addition to the MEN1 tumor suppressor gene, the cyclin D1 oncogene has demonstrated a role in the pathogenesis of parathyroid and gastroenteropancreatic neuroendocrine tumors. Up-regulation of cyclin D1 is observed early in tumor formation, implying a possible role in tumor initiation. Overexpression of cyclin D1 in the parathyroid glands of mice resulted in the tandem regulation of cellular proliferation and hormonal secretion, a feature intrinsic to neuroendocrine tumors.
Collapse
Affiliation(s)
- Daniel C Chung
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| |
Collapse
|
43
|
Rosen ED, Kulkarni RN, Sarraf P, Ozcan U, Okada T, Hsu CH, Eisenman D, Magnuson MA, Gonzalez FJ, Kahn CR, Spiegelman BM. Targeted elimination of peroxisome proliferator-activated receptor gamma in beta cells leads to abnormalities in islet mass without compromising glucose homeostasis. Mol Cell Biol 2003; 23:7222-9. [PMID: 14517292 PMCID: PMC230305 DOI: 10.1128/mcb.23.20.7222-7229.2003] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The nuclear hormone receptor peroxisome proliferator-activated receptor gamma (PPAR gamma) is an important regulator of lipid and glucose homeostasis and cellular differentiation. Studies of many cell types in vitro and in vivo have demonstrated that activation of PPAR gamma can reduce cellular proliferation. We show here that activation of PPAR gamma is sufficient to reduce the proliferation of cultured insulinoma cell lines. We created a model with mice in which the expression of the PPARG gene in beta cells was eliminated (beta gamma KO mice), and these mice were found to have significant islet hyperplasia on a chow diet. Interestingly, the normal expansion of beta-cell mass that occurs in control mice in response to high-fat feeding is markedly blunted in these animals. Despite this alteration in beta-cell mass, no effect on glucose homeostasis in beta gamma KO mice was noted. Additionally, while thiazolidinediones enhanced insulin secretion from cultured wild-type islets, administration of rosiglitazone to insulin-resistant control and beta gamma KO mice revealed that PPAR gamma in beta cells is not required for the antidiabetic actions of these compounds. These data demonstrate a critical physiological role for PPAR gamma function in beta-cell proliferation and also indicate that the mechanisms controlling beta-cell hyperplasia in obesity are different from those that regulate baseline cell mass in the islet.
Collapse
Affiliation(s)
- Evan D Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
House MG, Herman JG, Guo MZ, Hooker CM, Schulick RD, Lillemoe KD, Cameron JL, Hruban RH, Maitra A, Yeo CJ. Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Ann Surg 2003; 238:423-31; discussion 431-2. [PMID: 14501508 PMCID: PMC1422710 DOI: 10.1097/01.sla.0000086659.49569.9e] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Because histologic features can be unreliable in determining the malignant potential of pancreatic endocrine neoplasms (PENs), we characterized the methylation patterns of PENs to develop a molecular marker system useful for clinical prognosis. SUMMARY BACKGROUND DATA Aberrant promoter methylation of tumor suppressor genes is associated with a loss of gene function that can afford selective growth advantages to neoplastic cells. Gene hypermethylation, coupled with sporadic genetic mutations, defines the heterogeneous biology of human neoplasms. METHODS Forty-eight well-differentiated PENs were subjected to methylation-specific polymerase chain reaction to detect aberrant methylation associated with 11 candidate tumor suppressor genes (INK4a/p16, APC, O6-MGMT, hMLH1, p73, E-cadherin, RAR-beta, p14ARF, GST-pi, TIMP3, and RASSF1A). Methylation differences among PENs, subdivided according to tumor size, lymph node status, or liver metastasis, were analyzed, and the association of gene methylation with tumor recurrence and patient survival was evaluated. RESULTS Aberrant hypermethylation of any of the 11 tumor suppressor genes was detected in 87% of the PENs. In decreasing order of frequency, the 5 most commonly methylated genes were: RASSF1A (75%), INK4a/p16 (40%), O6-MGMT (40%), RAR-beta (25%), and hMLH1 (23%). In general, tumors larger than 5 cm and those associated with lymph node or hepatic metastases exhibited a higher frequency of methylation at each promoter site compared with PENs without malignant histologic features. The methylation of specific tumor suppressor genes was an independent predictor of early PEN recurrence and decreased 5-year survival following surgical resection. The accumulation of methylation of multiple tumor suppressor genes was associated with early tumor recurrence and reduced survival among a subpopulation of patients with lymph node-negative PENs. CONCLUSIONS Aberrant methylation of multiple tumor suppressor genes is associated with advanced tumor stage and identifies molecularly distinct PENs with identical histologic characteristics. The methylation status of specific tumor suppressor genes is predictive of PEN behavior.
Collapse
|
45
|
Moore PS, Beghelli S, Zamboni G, Scarpa A. Genetic abnormalities in pancreatic cancer. Mol Cancer 2003; 2:7. [PMID: 12537585 PMCID: PMC149421 DOI: 10.1186/1476-4598-2-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2002] [Accepted: 01/07/2003] [Indexed: 01/30/2023] Open
Abstract
The incidence and mortality of pancreatic adenocarcinoma are nearly coincident having a five-year survival of less than 5%. Enormous advances have been made in our knowledge of the molecular alterations commonly present in ductal cancer and other pancreatic malignancies. One significant outcome of these studies is the recognition that common ductal cancers have a distinct molecular fingerprint compared to other nonductal or endocrine tumors. Ductal carcinomas typically show alteration of K-ras, p53, p16INK4, DPC4 and FHIT, while other pancreatic tumor types show different aberrations. Among those tumors arising from the exocrine pancreas, only ampullary cancers have a molecular fingerprint that may involve some of the same genes most frequently altered in common ductal cancers. Significant molecular heterogeneity also exists among pancreatic endocrine tumors. Nonfunctioning pancreatic endocrine tumors have frequent mutations in MEN-1 and may be further subdivided into two clinically relevant subgroups based on the amount of chromosomal alterations. The present review will provide a brief overview of the genetic alterations that have been identified in the various subgroups of pancreatic tumors. These results have important implications for the development of genetic screening tests, early diagnosis, and prognostic genetic markers.
Collapse
Affiliation(s)
- Patrick S Moore
- Dipartimento di Patologia, Università di Verona, Strada Le Grazie, 37134, Verona, Italy
| | - Stefania Beghelli
- Dipartimento di Patologia, Università di Verona, Strada Le Grazie, 37134, Verona, Italy
| | - Giuseppe Zamboni
- Dipartimento di Patologia, Università di Verona, Strada Le Grazie, 37134, Verona, Italy
| | - Aldo Scarpa
- Dipartimento di Patologia, Università di Verona, Strada Le Grazie, 37134, Verona, Italy
| |
Collapse
|
46
|
The assessment of malignancy in endocrine tumours of the gastrointestinal tract. ACTA ACUST UNITED AC 2002. [DOI: 10.1054/cdip.2002.0145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
47
|
Corleto VD, Delle Fave G, Jensen RT. Molecular insights into gastrointestinal neuroendocrine tumours: importance and recent advances. Dig Liver Dis 2002; 34:668-80. [PMID: 12405256 DOI: 10.1016/s1590-8658(02)80212-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A subset of gastrointestinal neuroendocrine tumours (carcinoids and pancreatic endocrine tumours) show aggressive growth. Early identification of this subset is essential for management; however, clinical, laboratory and histologic features frequently fail to achieve this. Currently, there is an increased understanding of the molecular pathogenesis/changes in neuroendocrine tumours and this may identify important prognostic factors and possibly, new treatments. Recent findings and progress in this area are briefly reviewed in this article.
Collapse
Affiliation(s)
- V D Corleto
- Division of Digestive and Liver Diseases, University La Sapienza, Rome, Italy
| | | | | |
Collapse
|
48
|
Gumbs AA, Moore PS, Falconi M, Bassi C, Beghelli S, Modlin I, Scarpa A. Review of the clinical, histological, and molecular aspects of pancreatic endocrine neoplasms. J Surg Oncol 2002; 81:45-53; discussion 54. [PMID: 12210027 DOI: 10.1002/jso.10142] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pancreatic endocrine neoplasms (PENs) are rare tumors, and little is known about their genetic and chromosomal alterations. Elucidation of the molecular events involved in PEN carcinogenesis has been hindered by the fact that PENs have been considered a single disease entity. The emergence of novel molecular characterization strategies has, however, made it apparent that these lesions exhibit diverse molecular fingerprints, which will facilitate the precise delineation of PEN prognosis, histopathology, and carcinogenesis.
Collapse
Affiliation(s)
- A A Gumbs
- Department of Surgery, University of Verona, Verona, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Guo SS, Arora C, Shimoide AT, Sawicki MP. Frequent deletion of chromosome 3 in malignant sporadic pancreatic endocrine tumors. Mol Cell Endocrinol 2002; 190:109-14. [PMID: 11997184 DOI: 10.1016/s0303-7207(02)00002-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Pancreatic endocrine tumors (PETs) arise from neuroendocrine cells in and around the pancreas. As loss of heterozygosity (LOH) of chromosome 3 has been reported in sporadic PETs, we examined 16 sporadic PETs for LOH of 10 polymorphic DNA markers spanning both arms of chromosome 3. LOH was demonstrated in 4 of 8 (50%) sporadic PETs with hepatic metastasis, but in none of 8 sporadic PETs without hepatic involvement. The smallest common-deleted region (SCDR) mapped to 3q27-qter. Analysis of this data with the status of markers on chromosomes 1, 11, and MEN1 mutations in these 16 sporadic PETs revealed that chromosome 3q loss may be a late event in sporadic PET tumorigenesis. These data, combined with reports from other investigators, indicate that chromosome 3q27-qter may contain a tumor suppressor gene that's important in the tumorigenesis of sporadic PETs.
Collapse
Affiliation(s)
- Sydney S Guo
- Department of Surgery, West Los Angeles VA Medical Center and the UCLA School of Medicine, 90095, USA
| | | | | | | |
Collapse
|
50
|
Calender A, Vercherat C, Gaudray P, Chayvialle JA. Deregulation of genetic pathways in neuroendocrine tumors. Ann Oncol 2002; 12 Suppl 2:S3-11. [PMID: 11762348 DOI: 10.1093/annonc/12.suppl_2.s3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Complexity and redundancy of functional pathways controlled by the human genome explain that a single type of tumor can be induced by independant defective mutations in various genes that encode proteins acting in different parts of the cell physiology. Neuroendocrine tumors represent a powerful model for understanding such a complexity from the fact that at least six unrelated genetic syndromes have been characterized in the last decade which predispose to endocrine cell proliferation with variable penetrance and expressivity. Multiple Endocrine Neoplasia, von Hippel-Lindau. Carney and uncommonly Recklinghausen and Tuberous Sclerosis syndromes represent almost the whole panel of genetic diseases for which genes have been cloned and most of the functional knowledge has been collected. All the endocrine glands are concerned in these diseases, but the cellular pathways that are deregulated downstream from the deleterious mutations occurring in the genes of these autosomal dominant syndromes. might be related to each step of the cell life, from mitosis to DNA transcription, membrane receptor signalling and growth factor production, protein catabolism and extracellular matrix synthesis, and from transcription regulation to apoptosis and response to hypoxia and cellular stress. Here, we present an overview of genes involved in genetic predisposition to neuroendocrine tumors and highlight the complexity of pathways involved and the need of further studies focussing on genes involved in tumoral progression, most neuroendocrine tumors being benign at initial diagnosis but able to produce highly malignant cellular clones related to secondary genetic alterations or deregulation of growth factor production or cell cell adhesion processes.
Collapse
Affiliation(s)
- A Calender
- Department of Genetics, Hĵpital Edouard Herriot, Lyon, France.
| | | | | | | |
Collapse
|