1
|
Epidemiology of Large Bowel Carcinoid Tumors in the USA: A Population-Based National Study. Dig Dis Sci 2020; 65:269-275. [PMID: 31300994 DOI: 10.1007/s10620-019-05725-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Prior studies have shown that about 90% of all carcinoid tumors occur in the GI tract. However, epidemiological studies of these tumors have been limited by small sample size. Our aim was to obtain a more robust epidemiologic survey of large bowel carcinoids (LBC), using population-based data in order to more accurately identify risk factors for these tumors. METHODS We used a commercial database (Explorys Inc, Cleveland, OH) which includes electronic health record data from 26 major integrated US healthcare systems. We identified all patients aged 18 and older who were diagnosed with LBC, excluding appendiceal carcinoids, between 1999 and 2018 based on Systematized Nomenclature Of Medicine-Clinical Terms (SNOMED-CT) and evaluated the prevalence of LBC. We also performed univariate analysis to describe age-, race-, and gender-based distributions and to identify potential risk factors. RESULTS Of the 62,817,650 individuals in the database, 4530 were identified to have LBC with an overall prevalence of 7.21/100,000. Individuals with LBC were more likely to be elderly (age > 65) [OR 2.17, CI 2.05-2.31, p < 0.0001], smokers [OR 3.25; 95% CI 3.00-3.53, p < 0.0001], have a history of alcohol use [OR 3.75; 95% CI 3.52-3.99, p < 0.0001], diabetes mellitus (DM) [OR 4.42; 95% CI 4.14-4.72, p < 0.0001], obesity [OR 1.58; 95% CI 1.43-1.74, p < 0.0001], family history of cancer [OR 8.06; 95% CI 7.47-8.71, p < 0.0001], and personal history of ulcerative colitis [OR 6.93; 95% CI 5.55-8.64, p < 0.0001] or Crohn's disease [OR 6.45; 95% CI 5.24-7.95, p < 0.0001]. The prevalence of LBC was less among Caucasians compared to African-Americans [OR 0.57; 95% CI 0.53-0.61, p < 0.0001]. There was no statistically significant gender-based difference; men versus women [OR 1.02; 95% CI 0.96-1.08, p = 0.47]. The most common presenting symptoms included flushing, diarrhea, nausea, weight loss, and abdominal pain, while the most common GI associations included perforation, obstruction, hemorrhage, intussusception, and volvulus. CONCLUSION This is the largest epidemiological study evaluating the prevalence of LBC. We estimated the prevalence rate of LBC to be 7.21/100,000. The presence of significant risk factors with the clinical picture suspicious for a LBC should warrant thorough evaluation as these tumors can lead to life-threatening complications. Further studies are needed to better understand the mechanism of association between these risk factors and LBC.
Collapse
|
2
|
Lew FH, McQuown B, Borrego J, Cunningham S, Burgess KE. Retrospective evaluation of canine heart base tumours treated with toceranib phosphate (Palladia): 2011-2018. Vet Comp Oncol 2019; 17:465-471. [PMID: 31069932 DOI: 10.1111/vco.12491] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 11/30/2022]
Abstract
Heart base tumours (HBT) occur commonly in older, brachycephalic dogs. A presumptive diagnosis is made based on location and appearance of the tumour via echocardiogram. Effective treatment options are limited to surgery (when feasible) or radiation therapy. Benefit of medical management is presently unknown. The goal of this retrospective study was to assess the efficacy and tolerability of toceranib phosphate for dogs with HBT. Twenty-eight dogs with histologically, cytologically confirmed or presumed HBT were evaluated retrospectively. Twenty-seven dogs were treated with single agent toceranib. One dog received combination therapy with concurrent metronomic chemotherapy. This dog was not included in response or survival analysis. Factors assessed included clinical signs, hematologic/biochemical parameters and response to treatment. For the 27 dogs receiving single agent toceranib, an overall response rate of 10% was found. Overall median survival time was 823 days (range, 68-1190 days). The overall response rate for the dogs presenting with metastasis was 28.5%, with a median survival time of 532 days (range, 77-679 days). This was not significantly different than the median survival time of 796 days for dogs who did not present with metastasis. Of the dogs displaying clinical signs at the time of diagnosis, 90% had improvement and 81% had complete resolution of signs after starting toceranib. Toxicity was seen in 54% of dogs with gastrointestinal distress as the most common toxicity but dose reductions were infrequent required. Results demonstrate that toceranib phosphate is a well-tolerated and effective treatment for inoperable canine heart base tumours including dogs with advanced or metastatic disease.
Collapse
Affiliation(s)
- Felicia H Lew
- Department of Clinical Sciences, Cummings Veterinary Medical Center, Tufts University, North Grafton, Massachusetts.,Department of Medical Oncology, MedVet Chicago, Chicago, Illinois
| | - Bobbi McQuown
- Department of Clinical Sciences, Cummings Veterinary Medical Center, Tufts University, North Grafton, Massachusetts.,Department of Medical Oncology, MedVet Dallas, Dallas, Texas
| | - Juan Borrego
- Department of Medical Oncology, Hospital Aúna Especialidades Veterinarias, Valencia, Spain
| | - Suzanne Cunningham
- Department of Clinical Sciences, Cummings Veterinary Medical Center, Tufts University, North Grafton, Massachusetts
| | - Kristine E Burgess
- Department of Clinical Sciences, Cummings Veterinary Medical Center, Tufts University, North Grafton, Massachusetts
| |
Collapse
|
3
|
Cuny T, de Herder W, Barlier A, Hofland LJ. Role of the tumor microenvironment in digestive neuroendocrine tumors. Endocr Relat Cancer 2018; 25:R519-R544. [PMID: 30306777 DOI: 10.1530/erc-18-0025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) represent a group of heterogeneous tumors whose incidence increased over the past few years. Around half of patients already present with metastatic disease at the initial diagnosis. Despite extensive efforts, cytotoxic and targeted therapies have provided only limited efficacy for patients with metastatic GEP-NETs, mainly due to the development of a certain state of resistance. One factor contributing to both the failure of systemic therapies and the emergence of an aggressive tumor phenotype may be the tumor microenvironment (TME), comprising dynamic and adaptative assortment of extracellular matrix components and non-neoplastic cells, which surround the tumor niche. Accumulating evidence shows that the TME can simultaneously support both tumor growth and metastasis and contribute to a certain state of resistance to treatment. In this review, we summarize the current knowledge of the TME of GEP-NETs and discuss the current therapeutic agents that target GEP-NETs and those that could be of interest in the (near) future.
Collapse
Affiliation(s)
- Thomas Cuny
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Marseille, France
- Department of Endocrinology, Assistance Publique - Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Marseille, France
| | - Wouter de Herder
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anne Barlier
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Marseille, France
- Department of Endocrinology, Assistance Publique - Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Marseille, France
| | - Leo J Hofland
- Division Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Grillo F, Florio T, Ferraù F, Kara E, Fanciulli G, Faggiano A, Colao A. Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer 2018; 25:R453-R466. [PMID: 29769293 DOI: 10.1530/erc-17-0531] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 05/16/2018] [Indexed: 12/18/2022]
Abstract
In the last few years, the therapeutic approach for neuroendocrine neoplasms (NENs) has changed dramatically following the approval of several novel targeted treatments. The multitarget tyrosine kinase inhibitor (MTKI), sunitinib malate, has been approved by Regulatory Agencies in pancreatic NENs. The MTKI class, however, includes several other molecules (approved for other conditions), which are currently being studied in NENs. An in-depth review on the studies published on the MTKIs in neuroendocrine tumors such as axitinib, cabozantinib, famitinib, lenvatinib, nintedanib, pazopanib, sorafenib and sulfatinib was performed. Furthermore, we extensively searched on the Clinical Trial Registries databases worldwide, in order to collect information on the ongoing clinical trials related to this topic. Our systematic analysis on emerging MTKIs in the treatment of gastroenteropancreatic and lung NENs identifies in vitro and in vivo studies, which demonstrate anti-tumor activity of diverse MTKIs on neuroendocrine cells and tumors. Moreover, for the first time in the literature, we report an updated view concerning the upcoming clinical trials in this field: presently, phase I, II and III clinical trials are ongoing and will include, overall, a staggering 1667 patients. This fervid activity underlines the increasing interest of the scientific community in the use of emerging MTKIs in NEN treatment.
Collapse
Affiliation(s)
- Federica Grillo
- Pathology UnitDepartment of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
- Ospedale Policlinico San Martino IRCCSGenova, Italy
| | - Tullio Florio
- Pharmacology UnitDepartment of Internal Medicine (DIMI), University of Genova, Genova, Italy
| | - Francesco Ferraù
- Department of Human Pathology of Adulthood and ChildhoodUniversity of Messina, Messina, Italy
| | - Elda Kara
- Unit of EndocrinologyMetabolism, Diabetology and Nutrition, Azienda Sanitaria Universitaria Integrata di Udine, Ospedale Santa Maria della Misericordia, Udine, Italy
| | - Giuseppe Fanciulli
- Neuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, Italy
| | - Antongiulio Faggiano
- Department of Clinical Medicine and SurgeryUniversity 'Federico II', Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and SurgeryUniversity 'Federico II', Naples, Italy
| | | |
Collapse
|
5
|
Blažević A, Hofland J, Hofland LJ, Feelders RA, de Herder WW. Small intestinal neuroendocrine tumours and fibrosis: an entangled conundrum. Endocr Relat Cancer 2018; 25:R115-R130. [PMID: 29233841 DOI: 10.1530/erc-17-0380] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
Small intestinal neuroendocrine tumours (SI-NETs) are neoplasms characterized by their ability to secrete biogenic amines and peptides. These cause distinct clinical pathology including carcinoid syndrome, marked by diarrhoea and flushing, as well as fibrosis, notably mesenteric fibrosis. Mesenteric fibrosis often results in significant morbidity by causing intestinal obstruction, oedema and ischaemia. Although advancements have been made to alleviate symptoms of carcinoid syndrome and prolong the survival of patients with SI-NETs, therapeutic options for patients with mesenteric fibrosis are still limited. As improved insight in the complex pathogenesis of mesenteric fibrosis is key to the development of new therapies, we evaluated the literature for known and putative mediators of fibrosis in SI-NETs. In this review, we discuss the tumour microenvironment, growth factors and signalling pathways involved in the complex process of fibrosis development and tumour progression in SI-NETs, in order to elucidate potential new avenues for scientific research and therapies to improve the management of patients suffering from the complications of mesenteric fibrosis.
Collapse
Affiliation(s)
- Anela Blažević
- Department of Internal MedicineSector Endocrinology, ENETS Centre of Excellence, Erasmus University Medical Center (Erasmus MC) and Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Johannes Hofland
- Department of Internal MedicineSector Endocrinology, ENETS Centre of Excellence, Erasmus University Medical Center (Erasmus MC) and Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Leo J Hofland
- Department of Internal MedicineSector Endocrinology, ENETS Centre of Excellence, Erasmus University Medical Center (Erasmus MC) and Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Richard A Feelders
- Department of Internal MedicineSector Endocrinology, ENETS Centre of Excellence, Erasmus University Medical Center (Erasmus MC) and Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Wouter W de Herder
- Department of Internal MedicineSector Endocrinology, ENETS Centre of Excellence, Erasmus University Medical Center (Erasmus MC) and Erasmus MC Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|
6
|
Use of monoclonal antibodies to detect specific mutations in formalin-fixed, paraffin-embedded tissue sections. Hum Pathol 2016; 53:168-77. [DOI: 10.1016/j.humpath.2016.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/10/2016] [Accepted: 03/12/2016] [Indexed: 02/08/2023]
|
7
|
Barral M, Dohan A, Allez M, Boudiaf M, Camus M, Laurent V, Hoeffel C, Soyer P. Gastrointestinal cancers in inflammatory bowel disease: An update with emphasis on imaging findings. Crit Rev Oncol Hematol 2016; 97:30-46. [DOI: 10.1016/j.critrevonc.2015.08.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 06/15/2015] [Accepted: 08/04/2015] [Indexed: 12/20/2022] Open
|
8
|
Kidd M, Modlin IM, Bodei L, Drozdov I. Decoding the Molecular and Mutational Ambiguities of Gastroenteropancreatic Neuroendocrine Neoplasm Pathobiology. Cell Mol Gastroenterol Hepatol 2015; 1:131-153. [PMID: 28210673 PMCID: PMC5301133 DOI: 10.1016/j.jcmgh.2014.12.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/19/2014] [Indexed: 02/08/2023]
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN), considered a heterogeneous neoplasia, exhibit ill-defined pathobiology and protean symptomatology and are ubiquitous in location. They are difficult to diagnose, challenging to manage, and outcome depends on cell type, secretory product, histopathologic grading, and organ of origin. A morphologic and molecular genomic review of these lesions highlights tumor characteristics that can be used clinically, such as somatostatin-receptor expression, and confirms features that set them outside the standard neoplasia paradigm. Their unique pathobiology is useful for developing diagnostics using somatostatin-receptor targeted imaging or uptake of radiolabeled amino acids specific to secretory products or metabolism. Therapy has evolved via targeting of protein kinase B signaling or somatostatin receptors with drugs or isotopes (peptide-receptor radiotherapy). With DNA sequencing, rarely identified activating mutations confirm that tumor suppressor genes are relevant. Genomic approaches focusing on cancer-associated genes and signaling pathways likely will remain uninformative. Their uniquely dissimilar molecular profiles mean individual tumors are unlikely to be easily or uniformly targeted by therapeutics currently linked to standard cancer genetic paradigms. The prevalence of menin mutations in pancreatic NEN and P27KIP1 mutations in small intestinal NEN represents initial steps to identifying a regulatory commonality in GEP-NEN. Transcriptional profiling and network-based analyses may define the cellular toolkit. Multianalyte diagnostic tools facilitate more accurate molecular pathologic delineations of NEN for assessing prognosis and identifying strategies for individualized patient treatment. GEP-NEN remain unique, poorly understood entities, and insight into their pathobiology and molecular mechanisms of growth and metastasis will help identify the diagnostic and therapeutic weaknesses of this neoplasia.
Collapse
Key Words
- 5-HT, serotonin, 5-hydroxytryptamine
- Akt, protein kinase B
- BRAF, gene encoding serine/threonine-protein kinase B-Raf
- Blood
- CGH, comparative genomic hybridization
- CREB, cAMP response element-binding protein
- Carcinoid
- CgA, chromogranin A
- D cell, somatostatin
- DAG, diacylglycerol
- EC, enterochromaffin
- ECL, enterochromaffin-like
- EGFR, epidermal growth factor receptor
- ERK, extracellular-signal-regulated kinase
- G cell, gastrin
- GABA, γ-aminobutyric acid
- GEP-NEN, gastroenteropancreatic neuroendocrine neoplasms
- GPCR, G-protein coupled receptor
- Gastroenteropancreatic Neuroendocrine Neoplasms
- IGF-I, insulin-like growth factor-I
- ISG, immature secretory vesicles
- Ki-67
- LOH, loss of heterozygosity
- MAPK, mitogen-activated protein kinase
- MEN-1/MEN1, multiple endocrine neoplasia type 1
- MSI, microsatellite instability
- MTA, metastasis associated-1
- NEN, neuroendocrine neoplasms
- NFκB, nuclear factor κB
- PET, positron emission tomography
- PI3, phosphoinositide-3
- PI3K, phosphoinositide-3 kinase
- PKA, protein kinase A
- PKC, protein kinase C
- PTEN, phosphatase and tensin homolog deleted on chromosome 10
- Proliferation
- SD-208, 2-(5-chloro-2-fluorophenyl)-4-[(4-pyridyl)amino]p-teridine
- SNV, single-nucleotide variant
- SSA, somatostatin analog
- SST, somatostatin
- Somatostatin
- TGF, transforming growth factor
- TGN, trans-Golgi network
- TSC2, tuberous sclerosis complex 2 (tuberin)
- Transcriptome
- VMAT, vesicular monoamine transporters
- X/A-like cells, ghrelin
- cAMP, adenosine 3′,5′-cyclic monophosphate
- mTOR, mammalian target of rapamycin
- miR/miRNA, micro-RNA
Collapse
Affiliation(s)
| | - Irvin M. Modlin
- Correspondence Address correspondence to: Irvin M. Modlin, MD, PhD, The Gnostic Consortium, Wren Laboratories, 35 NE Industrial Road, Branford, Connecticut, 06405.
| | | | | |
Collapse
|
9
|
Prommegger R, Obrist P, Ensinger C, Profanter C, Mittermair R, Hager J. Retrospective evaluation of carcinoid tumors of the appendix in children. World J Surg 2014; 26:1489-92. [PMID: 12297915 DOI: 10.1007/s00268-002-6440-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Carcinoids of the appendix are rare in children and are usually diagnosed incidentally on histologic investigation following appendectomy for appendicitis. To investigate the significance of the diagnosis of appendiceal carcinoid in children, we conducted a retrospective study of the treatment and follow-up of 36 children with histologically confirmed carcinoid tumors of the appendix. Between 1970 and 2000 a total of 36 patients (25 girls, 11 boys) were diagnosed with appendiceal carcinoid. The median age of the patients at diagnosis was 12.3 years (range 6-16 years). The indication for appendectomy was acute lower right quadrant pain in 27 cases and chronic right lower quadrant pain in 9 patients. In 27 specimens the tumor was localized at the apex, in 7 at the midportion, and in 2 at the base of the appendix. The median tumor diameter was 6 mm (range 3-17 mm). Concomitant severe appendicitis was diagnosed in 14 patients 2 with a perforated appendicitis. In only one tumor were mucin-producing cells detectable. After a median follow-up of 10 years (range 2 months to 30 years) all patients were tumor-free. None of the patients had a synchronous or metachronous noncarcinoid malignant tumor. Appendiceal carcinoids are usually asymptomatic, and the indication for surgical intervention is acute or chronic abdominal pains in the right lower quadrant. For most patients the prognosis is excellent after appendectomy. As in adults, appendectomy is the appropriate treatment.
Collapse
Affiliation(s)
- Rupert Prommegger
- Department of Surgery, University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
10
|
Kidd M, Schimmack S, Lawrence B, Alaimo D, Modlin IM. EGFR/TGFα and TGFβ/CTGF Signaling in Neuroendocrine Neoplasia: Theoretical Therapeutic Targets. Neuroendocrinology 2013; 97:35-44. [PMID: 22710195 PMCID: PMC3684083 DOI: 10.1159/000334891] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 11/06/2011] [Indexed: 12/17/2022]
Abstract
Neuroendocrine neoplasms (NENs) are a heterogeneous family of malignancies whose proliferation is partially dependent on growth factors secreted by the microenvironment and the tumor itself. Growth factors which were demonstrated to be important in experimental models of NENs include EGF (epidermal growth factor), TGF (transforming growth factor) α, TGFβ and CTGF (connective tissue growth factor). EGF and TGFα bind to the EGF receptor to stimulate an intact RAS/RAF/MAPK pathway, leading to the transcription of genes associated with cell proliferation, invasion and metastasis. Theoretically, TGFα stimulation can be inhibited at several points of the MAPK pathway, but success is limited to NEN models and is not evident in the clinical setting. TGFβ1 stimulates TGFβ receptors (TGFβRI and TGFβRII) resulting in inhibition of neuroendocrine cell growth through SMAD-mediated activation of the growth inhibitor P21(WAF1/CIP1). Although some NENs are inhibited by TGFβ1, paradoxical growth is seen in experimental models of gastric and small intestinal (SI) NENs. Therapeutic targeting of TGFβ1 in NENs is therefore complicated by uncertainty of the effect of TGFβ1 secretion on the direction of proliferative regulation. CTGF expression is associated with more malignant clinical phenotypes in a variety of cancers, including NENs. CTGF promotes growth in gastric and SI-NEN models, and is implicated as a mediator of local and distant fibrosis caused by NENs of enterochromaffin cell origin. CTGF inhibitors are available, but their anti-proliferative effect has not been tested in NENs. In summary, growth factors are essential for NEN proliferation, and although interventions targeting these proteins are effective in experimental models, only limited clinical efficacy has been identified.
Collapse
Affiliation(s)
- M Kidd
- Gastrointestinal Pathobiology Research Group, Department of Gastroenterological Surgery, Yale University School of Medicine, New Haven, CT 06520-8062, USA
| | | | | | | | | |
Collapse
|
11
|
Abstract
Endocrine tumours derived from the small intestine, ileal carcinoids, produce and secrete the hormones tachykinins and serotonin, which induces the specific symptoms related to the tumour. Because of their low proliferation rate, they are often discovered at late stages when metastases have occurred. The biology that characterizes these tumours differs in many ways from what is generally recognized for other malignancies. In this overview, the current knowledge on the development and progression of ileal carcinoids is described.
Collapse
Affiliation(s)
- Janet L Cunningham
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
12
|
Gupta S, Engstrom PF, Cohen SJ. Emerging therapies for advanced gastroenteropancreatic neuroendocrine tumors. Clin Colorectal Cancer 2011; 10:298-309. [PMID: 21813338 DOI: 10.1016/j.clcc.2011.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 12/21/2010] [Accepted: 01/24/2011] [Indexed: 01/17/2023]
Abstract
Neuroendocrine tumors comprise a heterogeneous group of neoplasms derived from peptide- and amine-producing cells of the neuroendocrine system. Gastroenteropancreatic NET are differentiated into tumors and carcinomas based on their malignant potential and subdivided into those arising from the pancreas (islet cell tumors or pancreatic NET) and the more classical gut "carcinoids". Moderate to well differentiated NET have historically been considered rare tumors but recent epidemiological statistics suggest that their frequency has increased substantially over the past three decades. While the incidence of NET is increasing, data from both the US and UK demonstrate no improvement in outcomes over a similar time period. Due to the generally indolent biology of NET, most patients present with advanced disease before symptoms become apparent. In patients with localized NET, the 5-year survival rates after resection range from 60 to 90%, while regional lymph node involvement decreases the 5-year survival rates after surgery to 50-75%. Patients with distant metastases have a 5 year survival rate of approximately 25-40%. Conventional cytotoxic chemotherapy is of unclear benefit in patients with these generally slow growing tumors. Multiple agents have been tested in Phase 2 and Phase 3 trials. In general, the lack of major objective responses with significant toxicities has limited routine use of traditional chemotherapy agents and has emphasized the need to develop new agents in these diseases. This review will focus on emerging molecularly-targeted treatments with an emphasis on their underlying biologic and preclinical rationale.
Collapse
Affiliation(s)
- Sameer Gupta
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
13
|
Grossman EJ, Millis JM. Liver transplantation for non-hepatocellular carcinoma malignancy: Indications, limitations, and analysis of the current literature. Liver Transpl 2010; 16:930-42. [PMID: 20677284 DOI: 10.1002/lt.22106] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Orthotopic liver transplantation (OLT) is currently incorporated into the treatment regimens for specific nonhepatocellular malignancies. For patients suffering from early-stage, unresectable hilar cholangiocarcinoma (CCA), OLT preceded by neoadjuvant radiotherapy has the potential to readily achieve a tumor-free margin, accomplish a radical resection, and treat underlying primary sclerosing cholangitis when present. In highly selected stage I and II patients with CCA, the 5-year survival rate is 80%. As additional data are accrued, OLT with neoadjuvant chemoradiation may become a viable alternative to resection for patients with localized, node-negative hilar CCA. Hepatic involvement from neuroendocrine tumors can be treated with OLT when metastases are unresectable or for palliation of medically uncontrollable symptoms. Five-year survival rates as high as 90% have been reported, and the Ki67 labeling index can be used to predict outcomes after OLT. Hepatic epithelioid hemangioendothelioma is a rare tumor of vascular origin. The data from single-institution series are limited, but compiled reviews have reported 1- and 10-year survival rates of 96% and 72%, respectively. Hepatoblastoma is the most common primary hepatic malignancy in children. There exist subtle differences in the timing of chemotherapy between US and European centers; however, the long-term survival rate after transplantation ranges from 66% to 77%. Fibrolamellar hepatocellular carcinoma is a distinct liver malignancy best treated by surgical resection. However, there is an increasing amount of data supporting OLT when resection is contraindicated. In the treatment of either primary or metastatic hepatic sarcomas, unacceptable survival and recurrence rates currently prohibit the use of OLT.
Collapse
Affiliation(s)
- Eric J Grossman
- Section of Transplantation, Department of Surgery, University of Chicago Medical Center, Chicago, IL 60637, USA
| | | |
Collapse
|
14
|
Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, Bergsland E, Stuart K, Tye L, Huang X, Li JZ, Baum CM, Fuchs CS. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 2008; 26:3403-10. [PMID: 18612155 DOI: 10.1200/jco.2007.15.9020] [Citation(s) in RCA: 431] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Standard cytotoxic chemotherapy has limited efficacy in metastatic neuroendocrine tumor patients. Neuroendocrine tumors express vascular endothelial growth factor (VEGF) and its receptor (VEGFR). Sunitinib malate, an oral tyrosine kinase inhibitor, has activity against VEGFRs as well as platelet-derived growth factor receptors, stem-cell factor receptor, glial cell line-derived neurotrophic factor, and FMS-like tyrosine kinase-3. We evaluated the efficacy of sunitinib in a two-cohort, phase II study of advanced carcinoid and pancreatic neuroendocrine tumor patients. PATIENTS AND METHODS Patients were treated with repeated 6-week cycles of oral sunitinib (50 mg/d for 4 weeks, followed by 2 weeks off treatment). Patients were observed for response, survival, and adverse events. Patient-reported outcomes were assessed. RESULTS Among 109 enrolled patients, 107 received sunitinib (carcinoid, n = 41; pancreatic endocrine tumor, n = 66). Overall objective response rate (ORR) in pancreatic endocrine tumor patients was 16.7% (11 of 66 patients), and 68% (45 of 66 patients) had stable disease (SD). Among carcinoid patients, ORR was 2.4% (one of 41 patients), and 83% (34 of 41 patients) had SD. Median time to tumor progression was 7.7 months in pancreatic neuroendocrine tumor patients and 10.2 months in carcinoid patients. One-year survival rate was 81.1% in pancreatic neuroendocrine tumor patients and 83.4% in carcinoid patients. No significant differences from baseline in patient-reported quality of life or fatigue were observed during treatment. CONCLUSION Sunitinib has antitumor activity in pancreatic neuroendocrine tumors; its activity against carcinoid tumors could not be definitively determined in this nonrandomized study. Randomized trials of sunitinib in patients with neuroendocrine tumors are warranted.
Collapse
Affiliation(s)
- Matthew H Kulke
- Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Zitzmann K, Vlotides G, Göke B, Auernhammer CJ. PI(3)K-Akt-mTOR pathway as a potential therapeutic target in neuroendocrine tumors. Expert Rev Endocrinol Metab 2008; 3:207-222. [PMID: 30764093 DOI: 10.1586/17446651.3.2.207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Constitutive activation of PI(3)K-Akt-mTOR signaling is a frequently occurring event in human cancer and has also been detected in the majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system. Molecular analysis of NETs suggests, that in addition to mutations in certain tumor-suppressor genes (e.g., PTEN), multiple autocrine growth factor loops contribute to hyperactive PI(3)K-Akt-mTOR signaling, thus promoting unrestricted proliferation and resistance to apoptosis. These insights opened new perspectives for targeted therapy in NETs. In particular, several novel small-molecule inhibitors of tyrosine and serine/threonine kinases have demonstrated potent anti-tumor activity. This review will summarize current knowledge on PI(3)K-Akt-mTOR signaling, its role in proliferation and apoptosis, as well as novel therapeutic approaches targeting PI(3)K-Akt-mTOR pathway components in NET disease.
Collapse
Affiliation(s)
- Kathrin Zitzmann
- a Department of Internal Medicine II - Grosshadern, Ludwig-Maximilians- University of Munich, Marchioninistr. 15, 81377 Munich, Germany.
| | - George Vlotides
- b Department of Medicine, Cedars-Sinai Medical Center, University of California School of Medicine, Los Angeles, CA 90048, USA.
| | - Burkhard Göke
- c Department of Internal Medicine II - Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.
| | - Christoph J Auernhammer
- d Department of Internal Medicine II - Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.
| |
Collapse
|
16
|
Relation of neuroendocrine cells to transforming growth factor-alpha and epidermal growth factor receptor expression in gastric adenocarcinomas: prognostic implications. Pathol Oncol Res 2007; 13:215-26. [PMID: 17922051 DOI: 10.1007/bf02893502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 08/21/2007] [Indexed: 01/28/2023]
Abstract
The presence of neuroendocrine (NE) cells in gastric adenocarcinoma (GCa) is well documented, however, their significance is controversial. There is no evidence in the literature concerning the possible effect of these cells on the expression of TGF-alpha and EGFR, which are believed to confer growth advantage to tumor cells. 101 partial or total gastrectomy specimens from patients operated for conventional gastric adenocarcinoma were included in the study. In each case immunohistochemistry was performed on sequential tissue sections for chromogranin A (ChrA), TGF-alpha and EGFR. Samples were graded based on the number of ChrA-positive cells (0-3). TGF-alpha and EGFR expressions were evaluated according to both the intensity (0-2) and quantification of the positively stained areas (0-3). Follow-up data was available in 54 patients. Twenty-seven patients died of disease, while 27 patients were alive with a follow-up of at least 15 months. ChrA expression was detected in 54.4% of the tumor specimens. TGF-alpha was stained positively in 42.6% and EGFR in 49.5% of the cases. NE cells in GCa was related to TGF-alpha (p<0.0001) and EGFR expression (p<0.05), and TGF-alpha/EGFR coexpression (p<0.001). Among histopathologic variables, the presence of NE cells was significantly related to grade, stage and lymph node status. Although the presence of NE cells had no effect on survival, the expression of EGFR (p<0.0001) and TGF-alpha (p=0.002) were related to survival. The results of our study suggest that the presence of NE cells may have an effect on the expression of TGF-alpha and EGFR in GCa, and the autocrine mechanism between TGF-alpha and EGFR plays an important role in the prognosis of gastric carcinoma.
Collapse
|
17
|
West NE, Wise PE, Herline AJ, Muldoon RL, Chopp WV, Schwartz DA. Carcinoid tumors are 15 times more common in patients with Crohn's disease. Inflamm Bowel Dis 2007; 13:1129-34. [PMID: 17538985 DOI: 10.1002/ibd.20172] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND The coexistence of intestinal neoplasms with Crohn's disease (CD) has been reported, but the evidence of an increased risk of carcinoid tumor with Crohn's disease has been mixed. We present 4 patients with CD with associated carcinoid tumor. METHODS The charts of 111 patients with CD who had undergone resection between June 2001 and March 2005 were reviewed. The number of incidental carcinoid tumors in patients who underwent an appendectomy was used as a control. RESULTS Four cases of carcinoid tumor discovered in patients at resection for CD were identified. None had metastatic disease or carcinoid syndrome. These included 1 cecal (1 mm), 2 appendiceal (3 and 7 mm), and 1 transverse colon (7 mm) carcinoid tumors. None of the carcinoid tumors were identified in regions of active Crohn's disease. The incidence of carcinoid tumor in patients with Crohn's disease was 4 of 111 (3.6%). In comparison, 3 of 1199 patients (0.25%) who had appendectomies were identified as having appendiceal carcinoid tumor. Crohn's disease was associated with an increased incidence of carcinoid tumor; OR 14.9 (95% CI 2.5-102.5), P<0.0001. CONCLUSIONS There was a significantly increased incidence of carcinoid tumor in our Crohn's patients compared to the control patients. None of the carcinoid tumors developed in areas of Crohn's disease. This suggests that the development of carcinoid tumors may be secondary to distant proinflammatory mediators, rather than a local inflammatory effect from adjacent Crohn's disease. Patients with CD may be at increased risk of developing a carcinoid tumor.
Collapse
Affiliation(s)
- N E West
- Inflammatory Bowel Disease Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Although endocrine tumors are often slow growing, most can be life threatening and are considered resistant to conventional cytotoxic chemotherapy. The recent emergence of molecularly targeted therapy in oncology has brought renewed interest in the development of novel agents for this rare group of diseases. Preliminary results from phase II studies have shown promising results for VEGF and mTOR inhibitors in carcinoid and islet cell carcinoma and RET inhibitors in medullary thyroid carcinoma. Large confirmatory studies are planned.
Collapse
|
19
|
Chan JA, Kulke MH. Emerging therapies for the treatment of patients with advanced neuroendocrine tumors. Expert Opin Emerg Drugs 2007; 12:253-70. [PMID: 17604500 DOI: 10.1517/14728214.12.2.253] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Patients with neuroendocrine tumors may pursue a number of treatment options, but there is little consensus on a single, standard treatment approach. Somatostatin analogs are generally administered to patients with symptoms of hormonal secretion, and are often highly effective in this regard. However, the administration of somatostatin analogs is only rarely associated with tumor regression, and randomized trials demonstrating a survival benefit associated with their use have not been performed. Selected patients with hepatic metastases may undergo surgical debulking, embolization or other ablative therapies. The clinical benefit associated with administration of systemic agents such as IFN-alpha or cytotoxic chemotherapy has been limited. With the possible exception of streptozocin-based therapy in patients with pancreatic neuroendocrine tumors, the widespread use of standard cytotoxic regimens has been limited by their relatively modest antitumor activity, as well as concerns regarding their potential toxicity. The modest efficacy seen with these agents in patients with advanced neuroendocrine tumors has led to great interest in the development of novel treatment approaches. One such approach is the use of radiolabeled somatostatin analogs. Recently, agents targeting the VEGF pathway and mammalian target of rapamycin have also shown promise in patients with advanced neuroendocrine tumors. Ongoing randomized studies should help better define the role these agents will play in the future treatment of patients with this disease.
Collapse
Affiliation(s)
- Jennifer A Chan
- Dana-Farber Cancer Institute, Department of Medical Oncology, 44 Binney Street, Boston, MA 02115, USA.
| | | |
Collapse
|
20
|
Abstract
Patients with metastatic gastrointestinal neuroendocrine tumors have traditionally been faced with few effective treatment options. Somatostatin analogs often successfully control symptoms of hormonal hypersecretion but seldom result in tumor regression. Some patients with hepatic metastases are also candidates for ablative therapies such as surgical debulking or embolization. The role of systemic agents such as interferon alfa or cytotoxic chemotherapy remains ill defined. The more prevalent use of these modalities has been restricted by low tumor response rates and the potential for toxicity. Novel agents, including radiolabeled somatostatin analogs, inhibitors of the vascular endothelial growth factor pathway, and inhibitors of mammalian target of rapamycin, have shown promising activity in recent clinical studies. Continued investigation of these agents should render a better understanding of their efficacy in patients with advanced neuroendocrine tumors.
Collapse
Affiliation(s)
- Matthew H Kulke
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Zhang J, Jia Z, Li Q, Wang L, Rashid A, Zhu Z, Evans DB, Vauthey JN, Xie K, Yao JC. Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer 2007; 109:1478-86. [PMID: 17340592 DOI: 10.1002/cncr.22554] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a critical proangiogenic factor in solid tumors. However, its expression and role in human neuroendocrine tumor development and progression remains unclear. METHODS Using immunohistochemistry, VEGF and Sp1 expression patterns were investigated in 50 cases of human gastrointestinal neuroendocrine tumor having various clinicopathologic characteristics. RESULTS It was found that strong VEGF expression was detected in tumor cells, whereas no or very weak VEGF expression was detected in stromal cells surrounding or within the tumors. The levels of VEGF expression directly correlated with the expression levels of Sp1 and microvessel density. Strong, weak, and negative VEGF expression was observed in 32%, 54%, and 14% of cases, respectively. Compared with the group with negative VEGF expression, VEGF (weak/strong) expression was associated with metastasis (14% versus 58%; P = .03). The median progression-free survival (PFS) durations of patients with strong and weak VEGF expression were 29 months and 81 months, respectively. With a median follow-up duration of 50 months, the median PFS duration for the group with negative VEGF expression has not been reached. Compared with the log-rank test, VEGF expression was associated with poor PFS (P = .02). Using in vitro and in vivo models, human carcinoid cell lines were treated with bevacizumab, a monoclonal antibody targeting VEGF. Bevacizumab did not inhibit the growth of carcinoid cells in vitro but significantly reduced tumor angiogenesis and impaired tumor growth in animals. CONCLUSIONS The data suggest that overexpression of VEGF promotes the growth of human neuroendocrine tumors in part through up-regulation of angiogenesis.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Bevacizumab
- Biomarkers, Tumor/analysis
- Blotting, Western
- Carcinoma, Neuroendocrine/blood supply
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/mortality
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease-Free Survival
- Female
- Humans
- Immunohistochemistry
- Mice
- Mice, Nude
- Neovascularization, Pathologic/metabolism
- Prognosis
- Vascular Endothelial Growth Factor A/biosynthesis
Collapse
Affiliation(s)
- Jun Zhang
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yao JC. Neuroendocrine tumors. Molecular targeted therapy for carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol Metab 2007; 21:163-72. [PMID: 17382271 DOI: 10.1016/j.beem.2007.01.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Carcinoid and islet-cell carcinoma are often also known as low-grade neuroendocrine carcinomas. They are often slow-growing but can be resistant to standard therapy. While somatostatin analogues are often used to control hormonal syndromes, there is currently no therapy approved in the US for control of carcinoid tumor growth. For islet-cell carcinoma, streptozocin-based chemotherapy may induce tumor shrinkage, but second-line option are limited. This chapter reviews the molecular biology of neuroendocrine tumors, including the roles of MENIN, TSC2, NF-1, vHL, p53, bcl-2, bax, VEGF, IGF, PDGF, EGFR, and mTOR. Recently, there has been interest in developing molecularly targeted therapy for this group of diseases. Phase-II studies with imatinib, bevacizumab, sunitinib, gefitnib, temsirolimus, and everolimus (RAD001) have completed accrual. Encouraging results have been observed in studies with VEGF and mTOR inhibitors. Phase-III study of bevacizumab is planned in the US. Large-scale multinational phase-II and -III studies of everolimus are under way.
Collapse
Affiliation(s)
- James C Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
23
|
|
24
|
|
25
|
Berković M, Cacev T, Zjacić-Rotkvić V, Kapitanović S. TNF-alpha promoter single nucleotide polymorphisms in gastroenteropancreatic neuroendocrine tumors. Neuroendocrinology 2006; 84:346-52. [PMID: 17164537 DOI: 10.1159/000097988] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Accepted: 11/03/2006] [Indexed: 12/17/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) secrete biogenic amines, hormones and growth factors, tumor necrosis factor-alpha (TNF-alpha) being one of them. As the expression of TNF-alpha is mostly regulated at the transcriptional level, its promoter polymorphisms have been intensively studied as a potential determinant of TNF-alpha production and cancer susceptibility. We have analyzed for the first time the potential association between -238, -308, -857 and -1031 TNF-alpha promoter polymorphisms and GEP-NETs. The study included 65 individuals diagnosed with GEP-NET and 154 healthy age- and sex-matched controls. Although most of the patients had solitary GEP-NETs, 6 were diagnosed with GEP-NET as a part of multiple endocrine neoplasia type 1 and 1 as a part of neurofibromatosis type 1. The C allele at the -1031 position was more frequent in GEP-NET patients (p < 0.0005), suggesting its possible role in GEP-NET development. The significant difference between foregut and midgut GEP-NET patients was observed in the -308 high expression genotypes and -308A allele (high expression) which tend to occur more frequently in the foregut GEP-NETs (p = 0.0392 and p = 0.0350, respectively). When functional and nonfunctional pancreatic endocrine tumors were compared, there were no significant differences in the researched TNF-alpha SNPs. The results suggest the putative role of TNF-alpha -1031 polymorphism in the development of GEP-NET.
Collapse
Affiliation(s)
- Maja Berković
- Department of Endocrinology, Diabetes and Metabolism, University Hospital 'Sestre Milosrdnice', Zagreb, Croatia.
| | | | | | | |
Collapse
|
26
|
Granberg D, Wilander E, Oberg K. Expression of Tyrosine Kinase Receptors in Lung Carcinoids. Tumour Biol 2006; 27:153-7. [PMID: 16612146 DOI: 10.1159/000092718] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 09/16/2005] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Typical lung carcinoids are usually relatively benign tumors, but distant metastases are seen in up to 12% of the patients. In contrast, atypical carcinoids are more aggressive tumors, displaying metastases in up to 70%. The current treatment of metastatic lung carcinoids is discouraging. New therapies, such as inhibitors of the tyrosine kinase receptor family c-kit, platelet-derived growth factor receptors (PDGFR) alpha and beta and epidermal growth factor receptor (EGFR) have shown promising results in other malignancies and might be of value in malignant lung carcinoids. PATIENTS AND METHODS Tumor tissue from 51 patients with typical lung carcinoids were immunostained with polyclonal antibodies against c-kit, PDGFRalpha, PDGFRbeta and EGFR. Of the 24 patients who had metastatic disease, 17 had distant metastases. Fifteen of the patients had died from their disease. RESULTS Twelve of the tumors stained positive for c-kit, 44 expressed PDGFRalpha, 30 showed positive immunoreactivity for PDGFRbeta and 26 were EGFR immunoreactive. Among the 17 patients with distant metastases, 5 tumors expressed c-kit, 12 were PDGFRalpha immunoreactive, 9 stained positive for PDGFRbeta, and 7 showed positive immunoreactivity for EGFR. There was no correlation to distant metastases or survival for c-kit, PDGFRbeta or EGFR. CONCLUSIONS Tyrosine kinase receptors such as c-kit, PDGFRalpha, PDGFRbeta and EGFR are expressed in a significant number of patients with metastatic lung carcinoids. Treatment with inhibitors of the tyrosine kinase receptors expressed may be considered.
Collapse
Affiliation(s)
- Dan Granberg
- Department of Endocrine Oncology, University Hospital, Uppsala, Sweden.
| | | | | |
Collapse
|
27
|
Abstract
The fascinating, but often unpredictable, biology of neuroendocrine tumors (NETs) make the management of these malignancies a real challenge. The more recent development of high-throughput genomic and proteomic techniques, have opened a window to an increased knowledge of the biology of NETs. This review will discuss genes thought to play a role in the context of NE tumor biology, with particularly attention to those that may be potential new diagnostic and prognostic markers, as well as therapeutic targets. NETs constitute a heterogeneous group of neoplasm that may arise in virtually every topographic localization in the body, as a consequence of malignant transformation of various types of NE cells. Since NETs arising in the gastroenteropancreatic (GEP) or bronchopulmonary system are by far the most common, this review focuses on these entities, but lines are drawn to other NETs as well. Although large-scale gene expression analysis undoubtly have raised interesting new hypothesis concerning genes thought to play a role in tumor biology, discrepancies observed between studies and various platforms used, emphasizes the need to not only standardize the way microarray data are reported, but also to introduce standards in sample taking, processing and study design. In addition, the recognition of the complexity of the human proteome, with regard to generation of multiple isoforms from one gene, has created additional challenges. However,some goals have been reached already, as new knowledge has been translated into development of novel promising therapeutics.
Collapse
Affiliation(s)
- Eva Hofsli
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim N-7489, Norway.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Scott N, Millward E, Cartwright EJ, Preston SR, Coletta PL. Gastrin releasing peptide and gastrin releasing peptide receptor expression in gastrointestinal carcinoid tumours. J Clin Pathol 2004; 57:189-92. [PMID: 14747448 PMCID: PMC1770197 DOI: 10.1136/jcp.2003.10660] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS To establish whether gastrin releasing peptide (GRP) and the GRP receptor (GRPR) are expressed together in gastrointestinal carcinoid tumours. METHODS Twenty six carcinoid tumours from the stomach, small intestine, appendix, and colorectum were investigated by immunohistochemistry for GRP and GRPR. RESULTS GRP was detected in nine of 19 tumours and GRPR in 22 of 26. Coexpression of both the ligand and receptor was seen in six of 19 cases. GRPR but not GRP was more strongly expressed in appendix and colonic tumours. CONCLUSIONS GRP and GRPR are produced by a large number of gastrointestinal carcinoid tumours. An autocrine/paracrine pathway may exist for GRP stimulated cell proliferation in some of these neoplasms, analogous to that seen in small cell anaplastic carcinoma of the lung.
Collapse
Affiliation(s)
- N Scott
- Department of Pathology, St James's University Hospital, United Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Carcinoid tumors originate from the neuroendocrine cells throughout the body and are capable of producing various peptides. Their clinical course is often indolent but can also be aggressive and resistant to therapy. We examined all aspects of carcinoid tumors including the molecular biology oncogenesis, role of angiogenesis, recent advances in imaging, and therapy. The Medline and Cancerlit databases were searched using carcinoid as the keyword. English language manuscripts were reviewed and relevant references from a total of 7741 were found. All titles were screened and all the relevant manuscripts were analyzed; we found 307 references pertinent to the history, epidemiology, clinical behavior, pathology, pathophysiology, molecular biology, radiologic imaging, supportive care of carcinoid syndrome, and results of therapeutic clinical trials. Management of patients with carcinoid tumors requires an understanding of the disease process and a multimodality approach. Introduction of long-acting somatostatin analogues has resulted in significant advances in the palliative care of patients with carcinoid syndrome. However, advanced carcinoid tumor remains incurable. Existing therapies for advanced disease have low biologic activity, high toxicity, or both. Clearly, more research is necessary in the areas of molecular biology, targeted therapy, and development of new drugs Future advances in this field need to focus on clinical and biological predictors of outcome. Early works in the area of tumor biology such as the role of p53, bcl-2, bax, MEN1, FGF TGF PDGF and VEGF expression are of interest and need to be explored further.
Collapse
Affiliation(s)
- Isac I Schnirer
- Department of Gastrointestinal Oncology and Digestive Diseases, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77005-4341, USA
| | | | | |
Collapse
|
32
|
Abstract
AIM Transforming growth factor alpha (TGF-alpha) has been localized in neuroendocrine L-cells of the colon and rectum in previous studies. We examined whether neuroendocrine tumours with L-cell differentiation express TGF-alpha. EXPERIMENTAL DESIGN Immunohistochemistry was performed for proglucagon- and pro-pancreatic polypeptide derivatives, as well as for TGF-alpha, and epidermal growth factor receptor (EGFR) using paraffin sections from 16 neuroendocrine tumours of the colon and rectum. Also, in situ hybridization for TGF-alpha and proglucagon was carried out. MAIN RESULTS A strong expression of TGF-alpha at the protein level can be shown for neuroendocrine tumours of the hindgut. In one third of our cases we found a strong hybridization signal and in two thirds a moderate signal for TGF-alpha. The immunohistological phenotype concerning gut hormones is highly heterogeneous. Glucagon-like peptide 2 (GLP2) in our series was the most sensitive immunohistological hormone marker. MAJOR CONCLUSIONS The immunophenotype of colorectal neuroendocrine tumours regarding hormone markers is heterogeneous. The expression of TGF-alpha corresponds to the immunohistological profile of normal L-cells. TGF-alpha, especially in the neuroendocrine L-cells, most probably acts as a multifunctional trophic factor responsible for cellular integrity and survival, and not as an oncogenic growth factor.
Collapse
Affiliation(s)
- Walter Back
- Pathologisches Institut, Universitätsklinikum Mannheim, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Mannheim, Germany.
| | | | | |
Collapse
|
33
|
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
|
34
|
La Rosa S, Uccella S, Erba S, Capella C, Sessa F. Immunohistochemical detection of fibroblast growth factor receptors in normal endocrine cells and related tumors of the digestive system. Appl Immunohistochem Mol Morphol 2001; 9:319-28. [PMID: 11759058 DOI: 10.1097/00129039-200112000-00006] [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: 11/26/2022]
Abstract
Endocrine tumors (ETs) of the digestive system produce several growth factors including acidic and basic fibroblast growth factors (aFGF and bFGF, respectively), which are thought to be involved in the growth of tumor cells and in the proliferation of tumor stromal cells. Their actions depend on binding to four specific receptors--FGFR1, FGFR2, FGFR3, and FGFR4--whose distribution in normal endocrine cells and related tumors of the gastroenteropancreatic (GEP) system has previously been examined. Formalin-fixed, paraffin-embedded normal tissues and 60 well-characterized GEP endocrine tumors were immunostained using specific antibodies directed against various GEP hormones, aFGF, FGFR1, FGFR2, FGFR3, and FGFR4. Acidic FGF immunoreactivity (IR) was found in gut EC cells; FGFR1 immunoreactivity in rare duodenal endocrine cells and in pancreatic A cells; FGFR2 immunoreactivity in gastric and duodenal G cells, pancreatic B cells, and rectal EC cells; FGFR3 immunoreactivity in duodenal G cells; and FGFR4 immunoreactivity in rectal L cells and in pancreatic B, PP, and A cells. Immunoreactivity for at least one of the four FGFRs was found in all tumors, independently of FGFR expression in the putative cell of origin. EC cell tumors, which were all positive for aFGF, were found to express at least three different FGFRs. FGFRs also were localized in the stromal cells of all the tumors examined. The tumor stroma was more abundant in EC cell tumors than in other types of neoplasms. The results suggest that aFGF-FGFR interaction may be involved in the modulation of normal endocrine cell functions and in the regulation of tumor growth and stromal proliferation of EC cell carcinoids.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Diagnostic Techniques, Endocrine
- Digestive System/chemistry
- Digestive System/cytology
- Digestive System/pathology
- Digestive System Neoplasms/chemistry
- Digestive System Neoplasms/pathology
- Endocrine Gland Neoplasms/chemistry
- Endocrine Gland Neoplasms/pathology
- Enteroendocrine Cells/chemistry
- Enteroendocrine Cells/cytology
- Enteroendocrine Cells/pathology
- Female
- Humans
- Immunohistochemistry
- Intestinal Neoplasms/chemistry
- Intestinal Neoplasms/pathology
- Male
- Middle Aged
- Pancreatic Neoplasms/chemistry
- Pancreatic Neoplasms/pathology
- Protein-Tyrosine Kinases
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptor, Fibroblast Growth Factor, Type 2
- Receptor, Fibroblast Growth Factor, Type 3
- Receptor, Fibroblast Growth Factor, Type 4
- Receptors, Fibroblast Growth Factor/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- S La Rosa
- Department of Pathology, Ospedale di Circolo, Varese, Italy
| | | | | | | | | |
Collapse
|
35
|
Srivastava A, Alexander J, Lomakin I, Dayal Y. Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in pancreatic endocrine tumors. Hum Pathol 2001; 32:1184-9. [PMID: 11727256 DOI: 10.1053/hupa.2001.28959] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Coexpression of transforming growth factor alpha (TGF-alpha) and its receptor epidermal growth factor receptor (EGFR) is known to be associated with aggressive biologic behavior and adverse clinical outcome in a variety of tumors, including pancreatic adenocarcinomas. However, very little information is currently available as to whether this is true of pancreatic endocrine tumors (PETs) as well. Thirty-five PETs were retrospectively studied for immunohistochemical expression of TGF-alpha, the intracellular and extracellular domains of EGFR, and various hormonal secretory products. Proliferative activity was additionally studied (in 20 cases only) using the MIB-1 antibody. Thirty-one (89%) of 35 tumors were reactive for 1 or more of the peptide hormones tested; 22 (63%) tumors were positive for TGF-alpha; and 23 (65%) were positive for the intracellular and/or extracellular domain of EGFR. Based on their TGF-alpha and EGFR expression, these tumors could be classified into 4 groups. Of the 10 tumors in group I (positive for TGF-alpha and the complete EGFR molecule), 3 were malignant, 6 were >2 cm in diameter, 5 were functional, and 1 had a proliferative index of >40%. The 12 tumors in group II (positive for TGF-alpha but negative for the intracellular and/or extracellular domain of EGFR) included 4 malignant tumors, 4 PETs >2 cm in diameter, 8 functional, and 1 with a proliferative index of >40%. The 7 PETs in group III (positive for the intracellular/extracellular domain of EGFR alone) included 3 malignant tumors, 3 PETs >2 cm in diameter, and 3 functional tumors. The 6 tumors in group IV (completely negative for both TGF-alpha and EGFR) included 4 malignant tumors, 3 PETs >2 cm in diameter, and 4 functional lesions. Therefore, immunohistochemical expression of TGF-alpha and EGFR, either alone or in concert, shows no correlation with size, functional status, secretory profile, or biologic behavior and hence cannot be used as a marker of malignancy in this group of tumors.
Collapse
Affiliation(s)
- A Srivastava
- Department of Pathology, New England Medical Center, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
36
|
Abrahams NA, Vesoulis Z, Petras RE. Angiogenic polypoid proliferation adjacent to ileal carcinoid tumors: a nonspecific finding related to mucosal prolapse. Mod Pathol 2001; 14:821-7. [PMID: 11557776 DOI: 10.1038/modpathol.3880397] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Case reports have highlighted angiogenic polypoid proliferation in the mucosa adjacent to ileal carcinoid tumors, describing them as granulation tissue polyposis and florid angiogenesis. Some authors have proposed that the ileal carcinoid tumors themselves produce growth factors that cause the change. The purpose of this study was to determine the frequency of angiogenic polypoid proliferation in a large cohort of resected ileal carcinoid tumors compared with control groups. Search of the Cleveland Clinic and Summa Health System pathology files (1985 to 1999) yielded 65 resected ileal carcinoid tumors. Mucosal abnormalities adjacent to the ileal carcinoid tumors were graded 0 to 4+. Twenty ileal resection margins from colonic carcinoma cases served as normal controls. Ileal mucosa adjacent to 22 noncarcinoid neoplasms were also examined. The mucosa adjacent to 54/65 ileal carcinoid tumors (83%) showed mucosal abnormalities (vs. 3/20 normal controls), including mucosal edema, capillary ectasia, muscularis mucosae hypertrophy, fibrosis/smooth muscle proliferation within the lamina propria, club-shaped villi, and intramucosal capillary proliferation. Forty ileal carcinoid tumor cases (61%) showed some degree of angiogenic polypoid proliferation characterized by club-shaped villi and prominent intramucosal capillaries, with 17 (26%) graded as 3+ or 4+. Angiogenic polypoid proliferation was associated with hypertrophy of the muscularis mucosae, lamina proprial fibrosis/smooth muscle proliferation, and capillary ectasia similar to that described with gastrointestinal mucosal trauma/prolapse. This trauma/prolapse change was identified in 45 cases (69%) and was graded 3+ or 4+ in 23 (35%). Seventeen (77%) of the noncarcinoid neoplasms showed trauma/prolapse changes, with 7 (32%) graded as 3+ or 4+. Angiogenic polypoid proliferation also correlated with trauma/prolapse change in the noncarcinoid neoplasm controls. Neither APP (P =.24) nor the prolapse changes (P =.33) were found to be statistically different between the two tumor groups. Angiogenic polypoid proliferation of the adjacent ileal mucosa is common in patients with ileal carcinoid tumors and with noncarcinoid neoplasms. Angiogenic polypoid proliferation almost invariably coexists with fibromuscular change and capillary ectasia within the lamina propria, suggesting that mucosal trauma/prolapse plays a role in the histogenesis. The association of angiogenic polypoid proliferation with a variety of different neoplasms makes it unlikely that the tumors themselves secrete growth factors.
Collapse
Affiliation(s)
- N A Abrahams
- Department of Anatomic Pathology, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | |
Collapse
|
37
|
Gibbons MD, Manne U, Carroll WR, Peters GE, Weiss HL, Grizzle WE. Molecular differences in mucoepidermoid carcinoma and adenoid cystic carcinoma of the major salivary glands. Laryngoscope 2001; 111:1373-8. [PMID: 11568571 DOI: 10.1097/00005537-200108000-00011] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE/HYPOTHESIS Mucoepidermoid carcinoma (MEC) and adenoid cystic carcinoma (ACC), the most common malignancies of the major salivary glands, are clinically and pathologically different. To determine whether MEC and ACC have different molecular characteristics, we examined the expression of erbB-2, erbB-3, epidermal growth factor receptor (EGFR), and transforming growth factor-alpha (TGF-alpha), important molecular features in other malignancies. STUDY DESIGN/METHODS Archival tissue sections of 22 MEC and 6 ACC tumors of the major salivary glands were evaluated immunohistochemically for expression of erbB-2, erbB-3, EGRF, and TGF-alpha. A differential immunostaining score, reflecting the difference in immunostaining between carcinoma and uninvolved salivary gland tissue, was calculated for cytoplasmic and membranous staining. RESULTS Positive immunostaining for all biomarkers was observed in the cytoplasm and membrane of both tumors. However, expression was higher in MEC than in ACC tumors and was statistically significant for cytoplasmic EGFR (P =.009), TGF-alpha (P =.041), and membranous EGFR (P =.004). A significantly higher percentage of MEC cells also demonstrated positive immunostaining for cytoplasmic erbB-3 (P =.022), EGFR (P =.005), membranous erbB-3 (P =.022), and EGFR (P =.013). The differential immunostaining score was significantly higher for MEC compared with uninvolved alveolar tissue and the membranes of uninvolved ductal tissue. There were no statistically positive differential immunostaining scores for ACC. CONCLUSIONS There is a clear difference in the molecular phenotypes of MEC and ACC. The lack of statistically significant expression in ACC, when compared with similar uninvolved salivary gland tissue, suggests minimal involvement for these molecular structures in the pathogenesis of ACC. Conversely, erbB-2, erbB-3, EGFR, and TGF-alpha may have a role in the development and progression of MEC. These results have therapeutic implications for MEC of the major salivary glands.
Collapse
Affiliation(s)
- M D Gibbons
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Alabama at Birmingham, 1501 Fifth Avenue South, Birmingham, AL 35249-6889, U.S.A
| | | | | | | | | | | |
Collapse
|
38
|
Sakorafas GH, Tsiotou AG, Tsiotos GG. Molecular biology of pancreatic cancer; oncogenes, tumour suppressor genes, growth factors, and their receptors from a clinical perspective. Cancer Treat Rev 2000; 26:29-52. [PMID: 10660490 DOI: 10.1053/ctrv.1999.0144] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pancreatic cancer represents the fourth leading cause of cancer death in men and the fifth in women. Prognosis remains dismal, mainly because the diagnosis is made late in the clinical course of the disease. The need to improve the diagnosis, detection, and treatment of pancreatic cancer is great. It is in this type of cancer, in which the mortality is so great and the clinical detection so difficult that the recent advances of molecular biology may have a significant impact. Genetic alterations can be detected at different levels. These alterations include oncogene mutations (most commonly, K-ras mutations, which occur in 75% to more than 95% of pancreatic cancer tissues), tumour suppressor genes alterations (mainly, p53, p16, DCC, etc.), overexpression of growth factors (such as EGF, TGF alpha, TGF beta 1-3, aFGF, bTGF, etc.) and their receptors (i.e., EGF receptor, TGF beta receptor I-III, etc.). Insights into the molecular genetics of pancreatic carcinogenesis are beginning to form a genetic model for pancreatic cancer and its precursors. These improvements in our understanding of the molecular biology of pancreatic cancer are not simply of research interest, but may have clinical implications, such as risk assessment, early diagnosis, treatment, and prognosis evaluation.
Collapse
Affiliation(s)
- G H Sakorafas
- Department of Surgery, 251 Hellenic Air Force (HAF) Hospital, Messogion and Katehaki, Athens, 115 25 (Papagos), Greece
| | | | | |
Collapse
|
39
|
McCluggage WG, McConnell L, Sloan JM, Ellis PK, Irwin ST. Small intestinal ulceration secondary to carcinoid tumour arising in a Meckel's diverticulum. J Clin Pathol 1999; 52:72-4. [PMID: 10343617 PMCID: PMC501012 DOI: 10.1136/jcp.52.1.72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
A solitary small intestinal ulcer associated with a carcinoid tumour in a nearby Meckel's diverticulum was found in a 77 year old man presenting with massive rectal bleeding. Angiography and a radioisotope study localised the bleeding to the ileum. At operation, the Meckel's diverticulum was identified, with bleeding from an ulcer just distal to it. Pathological examination revealed a small carcinoid tumour confined to the Meckel's diverticulum. Close to the opening of the diverticulum, within the ileum, a well demarcated ulcer was present. Histology showed a non-specific ulcer which eroded a large blood vessel. This is the first documented occurrence of solitary small intestinal ulceration in association with a carcinoid tumour. Carcinoid tumour should be added to the list of possible causes of small intestinal ulceration. The ulceration may be secondary to release of cytokines by the tumour.
Collapse
Affiliation(s)
- W G McCluggage
- Department of Pathology, Royal Group of Hospitals Trust, Belfast, UK
| | | | | | | | | |
Collapse
|
40
|
Wulbrand U, Wied M, Zöfel P, Göke B, Arnold R, Fehmann H. Growth factor receptor expression in human gastroenteropancreatic neuroendocrine tumours. Eur J Clin Invest 1998; 28:1038-49. [PMID: 9893017 DOI: 10.1046/j.1365-2362.1998.00397.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Human gastroenteropancreatic neuroendocrine tumours are functionally and biologically heterogeneous, but their exact growth factor receptor expression pattern, important for onco- and carcinogenesis, remains unknown. METHODS This study searched for the mRNA expression pattern of six tyrosine- and serine/threonine kinase receptors [hepatocyte growth factor (HGFR), fibroblast growth factor (FGFR), epidermal growth factor (EGFR), insulin-like growth factor (IGF)-1R, transforming growth factor (TGF)-betaR1, TGF-betaR2] together with the five somatostatin receptors in human gastroenteropancreatic neuroendocrine tumours (gastrinomas, insulinomas, tumours with carcinoid syndrome, functionally inactive neuroendocrine tumours) using reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS EGF receptor was expressed almost exclusively in gastrinomas. Among the four tumour subtypes, expression frequencies of the somatostatin receptors 1 and 5, HGF-, IGF-1-, TGF-betaR1, TGF-betaR2 and the EGF-receptor varied significantly. CONCLUSIONS In spite of the common cellular origin of these tumours, differences in growth factor receptor expression suggest the existence of different pathways during tumour subtype development.
Collapse
|
41
|
|
42
|
Localization of Acidic Fibroblast Growth Factor, Fibroblast Growth Factor Receptor-4, Transforming Growth Factor-??, and Epidermal Growth Factor Receptor in Human Endocrine Cells of the Gut and Related Tumors: An Immunohistochemical Study. ACTA ACUST UNITED AC 1998. [DOI: 10.1097/00022744-199812000-00005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|