101
|
Current management of gastrointestinal stromal tumors: Surgery, current biomarkers, mutations, and therapy. Surgery 2015; 158:1149-64. [DOI: 10.1016/j.surg.2015.06.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 12/11/2022]
|
102
|
Hygum K, Wulff CN, Harsløf T, Boysen AK, Rossen PB, Langdahl BL, Safwat AA. Hypercalcemia in metastatic GIST caused by systemic elevated calcitriol: a case report and review of the literature. BMC Cancer 2015; 15:788. [PMID: 26499069 PMCID: PMC4619287 DOI: 10.1186/s12885-015-1823-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/16/2015] [Indexed: 01/17/2023] Open
Abstract
Background Hypercalcemia is the most common oncologic metabolic emergency but very rarely observed in patients with gastrointestinal stromal tumour, which is a rare mesenchymal malignancy of the gastrointestinal tract. We describe a case of hypercalcemia caused by elevated levels of activated vitamin D in a patient with gastrointestinal tumour. Prior to this case report, only one paper has reported an association between hypercalcemia, gastrointestinal stromal tumours and elevated levels of vitamin D. Case presentation An otherwise healthy 70-year-old Caucasian woman, previously treated for duodenal gastrointestinal stromal tumour, was diagnosed with liver metastasis, and relapse of gastrointestinal stromal tumour was confirmed by biopsy. At presentation, the patient suffered from severe symptoms of hypercalcemia. The most common causes of hypercalcemia, hyperparathyrodism, parathyroid hormone-related peptide secretion from tumour cells, and metastatic bone disease, were all dismissed as the etiology. Analysis of vitamin D subtypes revealed normal levels of both 25-OH Vitamin D2 and 25-OH Vitamin D3, whereas the level of activated vitamin D, 1,25 OH Vitamin D3, also referred to as calcitriol, was elevated. Conclusion The fact that plasma calcitriol decreased after initiation of oncological treatment and the finding that hypercalcemia did not recur during treatment support the conclusion that elevated calcitriol was a consequence of the gastrointestinal stromal tumour. We suggest that gastrointestinal stromal tumours should be added to the list of causes of humoral hypercalcemia in malignancy, and propose that gastrointestinal stromal tumour tissue may have high activity of the specific enzyme 1α-hydroxylase, which can lead to increased levels of calcitriol and secondarily hypercalcemia.
Collapse
Affiliation(s)
- Katrine Hygum
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Tage-Hansens Gade 2, DK-8000, Aarhus, C, Denmark.
| | - Christian Nielsen Wulff
- Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus, C, Denmark.
| | - Torben Harsløf
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Tage-Hansens Gade 2, DK-8000, Aarhus, C, Denmark.
| | - Anders Kindberg Boysen
- Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus, C, Denmark.
| | - Philip Blach Rossen
- Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus, C, Denmark.
| | - Bente Lomholt Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Tage-Hansens Gade 2, DK-8000, Aarhus, C, Denmark.
| | - Akmal Ahmed Safwat
- Department of Oncology, Aarhus University Hospital, Nørrebrogade 44, DK-8000, Aarhus, C, Denmark.
| |
Collapse
|
103
|
Brahmi M, Alberti L, Dufresne A, Ray-Coquard I, Cassier P, Meeus P, Decouvelaere AV, Ranchère-Vince D, Blay JY. KIT exon 10 variant (c.1621 A > C) single nucleotide polymorphism as predictor of GIST patient outcome. BMC Cancer 2015; 15:780. [PMID: 26498480 PMCID: PMC4619434 DOI: 10.1186/s12885-015-1817-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 10/16/2015] [Indexed: 11/25/2022] Open
Abstract
Background Tumor genotype plays a crucial role in clinical management of GIST. Whether genetic polymorphism of KIT may influence GIST patient outcome is unclear. Methods We investigated the biological and clinical significance of the presence of KIT exon 10 variant (c.1621 A > C), KITL541, in a transfected cell line (3 T3 L541) and in two retrospectively collected series of 109 GIST patients in total. The control group consisted of 60 healthy donors collected at the French department of blood transfusion. Results In the 3 T3 L541 cell line, KITL541 protein exhibited a spontaneous phosphorylation status comparable to that of wild-type KIT but displayed a phosphorylation pattern of AKT and ERK1/2 that was found similar to that of the classical mutated forms of the KIT receptor. Of 109 patients enrolled in this retrospective translational research study, 24 (22 %) harboured KITL541, similarly to the control group of healthy donors (n = 10 of 60, 17 %). A higher prevalence of the variant KITL541 was observed in patients with metastatic status at diagnosis (KITL541 correlated nine of 22 versus 15 of 87, p = 0.02). In addition, patients with KITL541 and localized GIST had a higher rate of relapse at 5 years and lower relapse free survival at 5 years in univariate, as well as in multivariate analysis. Response rate and duration of response to imatinib was similar in KITL541 and KITM541 patients. Conclusion KITL541 genotype is associated with a higher risk of metastasis at diagnosis and a higher risk of relapse in GIST patients. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1817-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mehdi Brahmi
- Department of medical oncology, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | - Laurent Alberti
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | - Armelle Dufresne
- Cancer Research Center of Lyon, INSERM UMR 1052, CNRS UMR 5286, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | - Isabelle Ray-Coquard
- Department of medical oncology, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | - Philippe Cassier
- Department of medical oncology, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | - Pierre Meeus
- Department of medical oncology, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| | | | | | - Jean-Yves Blay
- Department of medical oncology, Centre Leon Berard, 28 rue Laennec, Lyon, France.
| |
Collapse
|
104
|
Sporadic diffuse segmental interstitial cell of Cajal hyperplasia harbouring two gastric gastrointestinal stromal tumours (GIST) mimicking hereditary GIST syndromes. Int J Surg Case Rep 2015; 16:202-5. [PMID: 26521201 PMCID: PMC4643472 DOI: 10.1016/j.ijscr.2015.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 01/28/2023] Open
Abstract
Symptomatic gastric exophytic GIST diagnosed by computerized tomography. Pathology of resected specimen revealed segmental diffuse hyperplasia of the Cajal Cells with multifocal GISTs. Similarity to familiar GIST syndromes, excluded by detection of a somatic c-KIT mutation in exon 11 and inexistence of clinical features of NF-1.
Introduction Gastrointestinal stromal tumours (GISTs) are thought to derive from or differentiate towards the interstitial cells of Cajal (ICC) as most demonstrate a similar immunoprofile: CD117+, CD34+ and DOG1+. ICC hyperplasia refers to KIT-expressing microscopic spindle cell proliferations involving the myenteric plexus. Case report 74 year-old male presented with a 5-year history of heartburn and dysphagia. Imaging revealed a 4 cm GIST in the gastric fundus. Pathology of the resected specimen revealed diffuse segmental ICC hyperplasia harbouring two macroscopic GISTs and a ‘tumorlet’. A mutation in c-KIT exon 11 was detected in both the solid and the diffuse components. Discussion ICC hyperplasia can occur either as a sporadic focal lesion or in a syndromic setting, known to predispose to multiple GIST tumours at different sites. The majority of cases of sporadic ICC hyperplasia previously reported were of localised type. The hereditary form is mostly caused by germline mutations in c-KIT and PDGFRA or in patients with NF-1 andpresents as a diffuse hyperplasia, usually with a confluent, nodular or multifocal growth pattern. Conclusion We describe a diffuse form of sporadic ICC hyperplasia harbouring multifocal GISTs, mimicking diffuse ICC hyperplasia in hereditary GIST syndromes. Detection of somatic c-KIT exon 11 mutation ruled out a hereditary disorder.
Collapse
|
105
|
Clinicopathological significance of c-KIT mutation in gastrointestinal stromal tumors: a systematic review and meta-analysis. Sci Rep 2015; 5:13718. [PMID: 26349547 PMCID: PMC4642566 DOI: 10.1038/srep13718] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/03/2015] [Indexed: 12/28/2022] Open
Abstract
Many types of KIT mutations have been observed in gastrointestinal stromal tumors (GISTs), but their prognostic and predictive significance are still unclear. A meta-analysis and literature review were conducted to estimate the contribution of KIT mutations in prognostic parameters and clinic-pathological significance of GISTs. A total of 18 relevant articles from PubMed, EMBASE and Web of Science databases were included in this study. The frequency of KIT mutation was significantly increased in the GIST patients with higher mitosis (≥5/50 high-power fields (HPFs) and larger size (≥5 cm) of tumors than in those with lower MI (≤5/50HPFs) and smaller size (≤5 cm) of GISTs respectively. The rate of KIT mutation was not significantly changed between GISTs in stomachs and in small intestines. KIT mutational status has prognostic significance for patients’ outcome. GIST patients with KIT exon 9 mutations have higher risk of progression than those with exon 11 mutations. 5 year relapse-free survival (RFS) rate was significantly higher in patients with KIT exon 11 deletion than in those with other type of KIT exon 11 mutations. The deletion involving KIT exon 11, particularly codons 557–558, is a valuable predictor of prognosis for patients with GISTs.
Collapse
|
106
|
Spitaleri G, Biffi R, Barberis M, Fumagalli C, Toffalorio F, Catania C, Noberasco C, Lazzari C, de Marinis F, De Pas T. Inactivity of imatinib in gastrointestinal stromal tumors (GISTs) harboring a KIT activation-loop domain mutation (exon 17 mutation pN822K). Onco Targets Ther 2015; 8:1997-2003. [PMID: 26316776 PMCID: PMC4548749 DOI: 10.2147/ott.s81558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of gastrointestinal stromal tumors (GISTs) is largely driven by mutations in the KIT and PDGFRα genes. Imatinib mesylate is an oral small molecular tyrosine kinase inhibitor that mainly targets abl, c-KIT, and PDGFRα. Imatinib achieves disease control in approximately 70%–85% of patients with advanced GIST, and the median progression-free survival is 20–24 months. The efficacy of imatinib correlates with tumor kinase mutational status (exon 11 mutations mainly), and some mutations are known to be responsible for primary and secondary imatinib resistance. Beyond these, there are many other mutations that are considered rare and are associated with unknown clinical behavior. In the literature, there are poor and inconsistent data about the inhibitor sensitivity of mutations occurring in the activation-loop domain encoded by exon 17. In this article, we focus on a case of a patient suffering from GIST, harboring an extremely rare KIT activation-loop domain mutation (exon 17 mutation pN822K) treated with imatinib. A review of the literature is also presented.
Collapse
Affiliation(s)
- Gianluca Spitaleri
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Roberto Biffi
- Division of Abdominal Surgery, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Massimo Barberis
- Division of Pathology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Caterina Fumagalli
- Division of Pathology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Francesca Toffalorio
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Chiara Catania
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Cristina Noberasco
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Chiara Lazzari
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Filippo de Marinis
- Division of Chest Medical Oncology, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| | - Tommaso De Pas
- Oncology Unit of Thymic cancer, Rare Tumors and Sarcomas, European Institute of Oncology, Milan, Italy
| |
Collapse
|
107
|
Blay JY, Shen L, Kang YK, Rutkowski P, Qin S, Nosov D, Wan D, Trent J, Srimuninnimit V, Pápai Z, Le Cesne A, Novick S, Taningco L, Mo S, Green S, Reichardt P, Demetri GD. Nilotinib versus imatinib as first-line therapy for patients with unresectable or metastatic gastrointestinal stromal tumours (ENESTg1): a randomised phase 3 trial. Lancet Oncol 2015; 16:550-60. [PMID: 25882987 DOI: 10.1016/s1470-2045(15)70105-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Nilotinib inhibits the tyrosine kinase activity of ABL1/BCR-ABL1 and KIT, platelet-derived growth factor receptors (PDGFRs), and the discoidin domain receptor. Gain-of-function mutations in KIT or PDGFRα are key drivers in most gastrointestinal stromal tumours (GISTs). This trial was designed to test the efficacy and safety of nilotinib versus imatinib as first-line therapy for patients with advanced GISTs. METHODS In this randomised, open-label, multicentre, phase 3 trial (ENESTg1), participants from academic centres were aged 18 years or older and had previously untreated, histologically confirmed, metastatic or unresectable GISTs. Patients were stratified by previous adjuvant therapy and randomly assigned (1:1) via a randomisation list to receive oral imatinib 400 mg once daily or oral nilotinib 400 mg twice daily. The primary endpoint was centrally reviewed progression-free survival. Efficacy endpoints were assessed by intention-to-treat. This trial is registered with ClinicalTrials.gov, number NCT00785785. FINDINGS Because the futility boundary was crossed at a preplanned interim analysis, trial accrual terminated in April, 2011. Between March 16, 2009, and April 21, 2011, 647 patients were enrolled; of whom 324 were allocated nilotinib and 320 were allocated imatinib. At final analysis of the core study (data cutoff, October, 2012), 2-year progression-free survival was higher in the imatinib group (59·2% [95% CI 50·9-66·5]) than in the nilotinib group (51·6% [43·0-59·5]; hazard ratio 1·47 [95% CI 1·10-1·95]). In the imatinib group, the most common grade 3-4 adverse events were hypophosphataemia (19 [6%]), anaemia (17 [5%]), abdominal pain (13; 4%), and elevated lipase level (15; 5%), and in the nilotinib group were anaemia (18; 6%), elevated lipase level (15; 5%), elevated alanine aminotransferase concentration (12; 4%), and abdominal pain (11; 3%). The most common serious adverse event in both groups was abdominal pain (11 [4%] in the imatinib group, 14 [4%] in the nilotinib group). INTERPRETATION Nilotinib cannot be recommended for broad use to treat first-line GIST. However, future studies might identify patient subsets for whom first-line nilotinib could be of clinical benefit. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Jean-Yves Blay
- Centre Léon-Bérard, University Claude Bernard Lyon I, Lyon, France.
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GastrointestinaI Oncology, Peking University Cancer Hospital and Institute, Haidian, Beijing, China
| | - Yoon-Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Shukui Qin
- PLA Cancer Center of Nanjing Bayi Hospital, Nanjing, Jiangsu, China
| | - Dmitry Nosov
- Department of Clinical Pharmacology and Chemotherapy, Blokhin Cancer Research Center, Moscow, Russia
| | - Desen Wan
- Department of Colorectal Surgery, Cancer Center, Sun Yat-sen University, State Key Laboratory in Southern China, Guangzhou, China
| | - Jonathan Trent
- Sylvester Cancer Center, University of Miami, Miami, Florida, USA
| | | | - Zsuzsanna Pápai
- Military Hospital-State Health Centre Oncology Department, Budapest, Hungary
| | - Axel Le Cesne
- Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Steven Novick
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Lilia Taningco
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Shuyuan Mo
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Steven Green
- Novartis Pharma AG, Postfach, Basel, Switzerland
| | - Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - George D Demetri
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, and Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
108
|
Dailey DD, Ehrhart EJ, Duval DL, Bass T, Powers BE. DOG1 is a sensitive and specific immunohistochemical marker for diagnosis of canine gastrointestinal stromal tumors. J Vet Diagn Invest 2015; 27:268-77. [DOI: 10.1177/1040638715578878] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) and leiomyosarcomas are histologically similar primary neoplasms commonly occurring in the gastrointestinal tract of dogs and humans. Immunohistochemical staining (IHC) is needed to differentiate between these 2 entities and positive reactivity for KIT (cluster of differentiation [CD]117) is regarded as the gold standard for diagnosis of canine GIST. Studies estimate 5–10% of human GISTs stain negative or only weakly positive for KIT and have identified DOG1 (discovered on gastrointestinal stromal tumors protein 1) as a highly sensitive and specific marker for human GISTs. The purpose of this study was to evaluate immunoreactivity of a commercially available DOG1 antibody for use in diagnosis of canine GISTs. Fifty-five primary mesenchymal gastrointestinal tumors with histologic features consistent with GIST or leiomyosarcoma were evaluated via IHC for KIT, DOG1, and desmin. A subset of tumors was additionally evaluated for reactivity for smooth muscle actin (SMA). Thirty-three tumors (60%) were diagnosed as GIST based on positive immunoreactivity for KIT or DOG1 regardless of reactivity for desmin or SMA. Most GISTs (32/33, 97.0%) had similar staining for both KIT and DOG1. DOG1 expression was identified in 2 tumors (1 study tumor and 1 additional tumor) negative for KIT and desmin that had histologic features consistent with KIT-negative, platelet-derived growth factor receptor-alpha (PDGFRA)-mutant human GISTs. Our results suggest that DOG1 has improved specificity and sensitivity to that of KIT for differentiating between canine GISTs and leiomyosarcomas. Inclusion of both DOG1 and KIT IHC in diagnostic panels will improve the accuracy of canine GIST diagnosis.
Collapse
Affiliation(s)
- Deanna D. Dailey
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - E. J. Ehrhart
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Dawn L. Duval
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Todd Bass
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - B. E. Powers
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
109
|
Ricci R, Chiarello G, Attili F, Fuccio L, Alfieri S, Persiani R, Di Pietro S, Martini M, Costamagna G, Larocca LM, Larghi A. Endoscopic ultrasound-guided fine needle tissue acquisition biopsy samples do not allow a reliable proliferation assessment of gastrointestinal stromal tumours. Dig Liver Dis 2015; 47:291-5. [PMID: 25596931 DOI: 10.1016/j.dld.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Current prognostication of gastrointestinal stromal tumours is validated on/applies to resected tumours, mainly because surgery is recommended whenever possible. However, pre-treatment prognostication is increasingly warranted, considering the follow-up strategy recently admitted for expectedly low-risk tumours and the possible distinctive molecular features/spontaneous regression of some small cases. AIMS To investigate whether endoscopic ultrasound-guided fine-needle tissue acquisition-biopsies reflect prognosticators of resected gastrointestinal stromal tumours, for possibly developing reliable pre-treatment prognostic criteria. METHODS The applicability/reliability of mitotic index/5mm(2) and MIB1 proliferative index/1000 cells were tested in 35 endoscopic ultrasound-guided fine-needle tissue acquisition-biopsies diagnosed as gastrointestinal stromal tumour, subsequently resected without intervening therapy, consecutively collected in thirty months. Size and mitotic/proliferative indexes were compared with resection specimens. The feasibility of bioptic genotyping was also tested. RESULTS 35 patients were studied (45.7% males; mean age 61.6 years, range 26-83 years). Mitotic/proliferative indexes were determinable in 68.6%/88.6% of biopsies, respectively; they were nevertheless underestimated, as happened with endoscopic ultrasound-determined tumour size. Bioptic genotyping revealed reliable. CONCLUSIONS Endoscopic ultrasound-guided fine-needle tissue acquisition does not reliably reflect gastrointestinal stromal tumours' proliferation and size. Alternative parameters should be validated for a pre-surgical prognostic classification. Considering the emerging potentially prognostic genetic markers in gastrointestinal stromal tumours, the reliability of bioptic genotyping is a promising result.
Collapse
Affiliation(s)
- Riccardo Ricci
- Department of Pathology, Catholic University, Rome, Italy.
| | - Gaia Chiarello
- Department of Pathology, Catholic University, Rome, Italy
| | - Fabia Attili
- Digestive Endoscopy Unit, Catholic University, Rome, Italy
| | - Lorenzo Fuccio
- Gastroenterology Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | - Alberto Larghi
- Digestive Endoscopy Unit, Catholic University, Rome, Italy
| |
Collapse
|
110
|
Abstract
Myxoid soft tissue tumors form a heterogeneous group. Their biological potential encompasses the whole spectrum from benign to highly malignant. The present article focuses on myxoid tumors of the deep soft tissues: myxofibrosarcoma, low-grade fibromyxoid sarcoma, myxoma, myxoid liposarcoma, extraskeletal myxoid chondrosarcoma and nodular fasciitis. The last two decades have brought into practice multiple powerful tools that support pathologists in making precise diagnoses, even on small incisional biopsies: detection of fusion transcripts by rt-PCR, detection of chromosomal fusion or breakpoint by FISH, detection of point mutations by PCR and expression of specific markers by immunohistochemistry. Conventional morphology remains the mainstay of diagnosis, and it is essential to obtain adequate clinical and radiological information before interpreting small incisional biopsies. The present article is a summary of morphologic features used to diagnose the most common tumors of the deep soft tissues.
Collapse
Affiliation(s)
- Sébastien Labonté
- CHU de Québec/L'Hôtel-Dieu de Québec, 11, Côte-du-Palais, Québec, QC, G1R 2J6, Canada.
| |
Collapse
|
111
|
Yamamoto H, Oda Y. Gastrointestinal stromal tumor: recent advances in pathology and genetics. Pathol Int 2014; 65:9-18. [PMID: 25414046 DOI: 10.1111/pin.12230] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022]
Abstract
The discovery of KIT gene mutation in gastrointestinal stromal tumor (GIST) has provided a paradigm shift in the classification, diagnosis and molecular-targeted therapy of gastrointestinal mesenchymal tumors. There is growing evidence of phenotype-genotype (KIT, platelet-derived growth factor receptor-alpha, succinate dehydrogenase or other driver gene mutation) and genotype-therapeutic (sensitivity to imatinib) correlations in GIST. Risk stratification based on mitotic counts, tumor size and rupture is useful for the prognostication and management of patients with GIST. Blood vessel invasion is a strong indicator of liver metastasis in GIST. In addition, novel biomarkers such as cell-cycle regulators, microRNAs and their targets have been discovered by using high throughput molecular analyses. In contrast, leiomyosarcoma of the gastrointestinal tract has become a very rare entity in the 'KIT' era, and its molecular pathogenetic mechanism is unclear. Recent studies have revealed a wide spectrum of cytological atypia, mitotic counts and biological behavior of gastrointestinal smooth muscle tumors, suggesting the necessity of establishing the criteria for malignancy. Collectively, both classical histopathological procedures and modern molecular investigations are indispensable for the evolution of diagnosis and treatment of GIST and mimics.
Collapse
Affiliation(s)
- Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University and Division of Pathology, Kyushu University Hospital, Fukuoka, Japan
| | | |
Collapse
|
112
|
Huang YQ. Advances in research of gastrointestinal stromal tumors. Shijie Huaren Xiaohua Zazhi 2014; 22:1633-1641. [DOI: 10.11569/wcjd.v22.i12.1633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract, arising from the interstitial cells of Cajal (ICCs), primarily in the stomach and small intestine. The growth of most GISTs is driven by the mutations of genes encoding oncogenic receptor tyrosine kinase KIT or platelet derived growth factor receptor alpha (PDGFRα). The pathogenesis of GISTs may involve ICCs, microRNAs (miRNAs), signaling pathways, DNA methylation, and KIT or PDGFRα gene mutations. This article systematically describes the advances in research of GISTs in terms of clinical features, imaging characteristics, endoscopic features, histopathological features, diagnosis and therapies.
Collapse
|