101
|
Merten L, Agaimy A, Moskalev EA, Giedl J, Kayser C, Geddert H, Schaefer IM, Cameron S, Werner M, Ströbel P, Hartmann A, Haller F. Inactivating Mutations of RB1 and TP53 Correlate With Sarcomatous Histomorphology and Metastasis/Recurrence in Gastrointestinal Stromal Tumors. Am J Clin Pathol 2016; 146:718-726. [PMID: 28028119 DOI: 10.1093/ajcp/aqw193] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVES Loss-of-function mutations in TP53 and CDKN2A have been found at varying frequencies in gastrointestinal stromal tumors (GISTs), while no mutations of RB1 have been reported to date. The aim of the current study was to determine the mutation frequency of TP53, RB1, and CDKN2A in GISTs. METHODS A cohort of 83 primary untreated GISTs was analyzed for mutations in TP53, RB1, and CDKN2A by massive parallel sequencing. Tumors with mutations in TP53 and RB1 were analyzed by fluorescence in situ hybridization for the corresponding gene loci. RESULTS Two GISTs harbored inactivating mutations in RB1, and two other GISTs displayed inactivating mutations in TP53 All four tumors were KIT mutant high-risk tumors with highly cellular sarcomatous histomorphology and variable combinations of plump spindle cells to epithelioid highly atypical cells and high mitotic activity. Three of these patients developed recurrent or metastatic disease, while the fourth patient showed tumor rupture intraoperatively. The combined overall frequency of TP53 and RB1 mutations was 13% considering high-risk or malignant GISTs. CONCLUSIONS TP53 and RB1 mutations seem to be restricted to high-risk/malignant GISTs and occur at an equal although relatively low frequency.
Collapse
Affiliation(s)
- Larissa Merten
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Abbas Agaimy
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Evgeny A Moskalev
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Giedl
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Claudia Kayser
- Institute of Pathology, Albert-Ludwigs University, Freiburg, Germany
| | - Helene Geddert
- Institute of Pathology, St. Vincentius Hospital, Karlsruhe, Germany
| | - Inga-Marie Schaefer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and
| | - Silke Cameron
- Clinic for Gastroenterology and Gastrointestinal Oncology
| | - Martin Werner
- Institute of Pathology, Albert-Ludwigs University, Freiburg, Germany
| | - Philip Ströbel
- Institute of Pathology, Georg August University, Göttingen, Germany
| | - Arndt Hartmann
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Florian Haller
- From the Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
102
|
Abstract
The first 15 years of management of gastrointestinal stromal tumor (GIST) have led to 3 lines of therapy for metastatic disease: imatinib, sunitinib, and regorafenib. In the adjuvant setting, imatinib is usually given for 3 years postoperatively to patients with higher-risk primary tumors that are completely resected. In this review, issues regarding GIST adjuvant therapy are discussed. It is hoped this review will help the reader understand the present standard of care to improve upon it in years to come.
Collapse
|
103
|
Capelli L, Petracci E, Quagliuolo V, Saragoni L, Colombo P, Morgagni P, Calistri D, Tomezzoli A, Di Cosmo M, Roviello F, Vindigni C, Coniglio A, Villanacci V, Catarci M, Coppola L, Alfieri S, Ricci R, Capella C, Rausei S, Gulino D, Amadori D, Ulivi P. Gastric GISTs: Analysis of c-Kit, PDGFRA and BRAF mutations in relation to prognosis and clinical pathological characteristics of patients – A GIRCG study. Eur J Surg Oncol 2016; 42:1206-14. [DOI: 10.1016/j.ejso.2016.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/28/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
|
104
|
Chetty R, Serra S. Molecular and morphological correlation in gastrointestinal stromal tumours (GISTs): an update and primer. J Clin Pathol 2016; 69:754-60. [PMID: 27317811 DOI: 10.1136/jclinpath-2016-203807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/19/2022]
Abstract
Gastrointestinal stromal tumours (GISTs) are a commonly encountered tumour in routine practice. In the main, the morphology of spindle, epithelioid or mixed are well recognised along with mutations of c-kit However, there are other genes that are mutated resulting in characteristic clinicopathological correlations. GISTs harbouring platelet-derived growth factor receptor α (PDGFRα) gene mutations lead to a typical morphological constellation of findings: gastric and omental location, gross tumour that is cystic and haemorrhagic, composed of epithelioid, plasmacytoid cells exhibiting pleomorphism, low mitotic count and containing characteristic giant cells with peripherally placed nuclei. These cells are set in a myxoid stroma containing several mast cells. In addition, perivascular/intratumoural hyalinisation is often seen. These tumours are CD117 and DOG-1 positive. GISTs with SDH mutations are multinodular/bilobed/dumb-bell shape tumour masses with mucosal ulceration and histologically characterised by fibrous bands around and within nodules of epithelioid or mixed epithelioid/spindle cells. Lymphovascular invasion with lymph node metastases are usual. Immunohistochemically, the GISTs are CD117, DOG-1 positive, SDHA negative (if SDHA mutated), SDHA positive (if SDHA intact) and SDHB negative. BRAF and NF-1 mutated GISTs do not have any characteristic morphological features.
Collapse
Affiliation(s)
- Runjan Chetty
- Departments of Pathology, Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Stefano Serra
- Departments of Pathology, Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
105
|
Jiang Z, Zhang J, Li Z, Liu Y, Wang D, Han G. A meta-analysis of prognostic value of KIT mutation status in gastrointestinal stromal tumors. Onco Targets Ther 2016; 9:3387-98. [PMID: 27350754 PMCID: PMC4902249 DOI: 10.2147/ott.s101858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Numerous types of KIT mutations have been reported in gastrointestinal stromal tumors (GISTs); however, controversy still exists regarding their clinicopathological significance. In this study, we reviewed the publicly available literature to assess the data by a meta-analysis to characterize KIT mutations and different types of KIT mutations in prognostic prediction in patients with GISTs. Twenty-eight studies that included 4,449 patients were identified and analyzed. We found that KIT mutation status was closely correlated with size of tumors and different mitosis indexes, but not with tumor location. KIT mutation was also observed to be significantly correlated with tumor recurrence, metastasis, as well as the overall survival of patients. Interestingly, there was higher risk of progression in KIT exon 9-mutated patients than in exon 11-mutated patients. Five-year relapse-free survival (RFS) rate was significantly higher in KIT exon 11-deleted patients than in those with other types of KIT exon 11 mutations. In addition, RFS for 5 years was significantly worse in patients bearing KIT codon 557–558 deletions than in those bearing other KIT exon 11 deletions. Our results strongly support the hypothesis that KIT mutation status is another evaluable factor for prognosis prediction in GISTs.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jian Zhang
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zhi Li
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yingjun Liu
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Daohai Wang
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Guangsen Han
- Department of General Surgery, Affiliated Tumor Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
106
|
Ronellenfitsch U, Henzler T, Menge F, Dimitrakopoulou-Strauss A, Hohenberger P. [Advanced gastrointestinal stromal tumors : What role does surgery currently play in multimodal concepts?]. Chirurg 2016; 87:389-97. [PMID: 27080051 DOI: 10.1007/s00104-016-0180-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GIST) have an incidence of 1-2/100,000 and thus constitute the most common mesenchymal neoplasm of the digestive tract. Their specific tumor biology with mutations in the protooncogenes c-KIT and PDGFR α acting as drivers of tumor growth facilitate targeted therapy with tyrosine kinase inhibitors. In this context, there are several specific indications for surgery in patients with advanced GIST. OBJECTIVE This article discusses the importance of surgery within multimodal therapeutic concepts for advanced GIST. MATERIAL AND METHODS The results of a selective literature search including own studies and case reports are presented. RESULTS For large GIST at unfavorable anatomical locations, which are not amenable to organ-sparing resection, neoadjuvant imatinib therapy is the standard upfront treatment prior to surgery in the case of imatinib-sensitive mutations in the c-KIT protooncogene. This usually reduces the extent of resection without increasing perioperative morbidity. In the metastatic setting, surgery can constitute a significant part of multimodal therapy in patients with a generalized response to drug therapy by resection of residual tumor masses, although there are no prospective studies to prove a beneficial effect on overall survival. In patients with focal progression on anti-proliferative therapy, local therapeutic measures can make an important contribution to multimodal tumor control. In patients with generalized progression, an operation should only be performed in highly selected cases with the goal of symptom control. Local ablative therapies, such as radiofrequency ablation (RFA), irreversible electroporation (IRE) and selective internal radiotherapy (SIRT) are a therapeutic option particularly for liver metastases. CONCLUSION Surgery plays an important role in the multimodal therapy of advanced GIST particularly in the neoadjuvant setting. Its role is more limited in metastatic stages where systemic treatment represents the frontline therapeutic approach.
Collapse
Affiliation(s)
- U Ronellenfitsch
- Sektion Spezielle Chirurgische Onkologie und Thoraxchirurgie, Chirurgische Klinik, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer 1-3, 68135, Mannheim, Deutschland
| | - T Henzler
- Institut für Klinische Radiologie und Nuklearmedizin, Universitätsmedizin Mannheim, Mannheim, Deutschland
| | - F Menge
- Sektion Spezielle Chirurgische Onkologie und Thoraxchirurgie, Chirurgische Klinik, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer 1-3, 68135, Mannheim, Deutschland
| | - A Dimitrakopoulou-Strauss
- Klinische Kooperationseinheit Nuklearmedizin, Deutsches Krebsforschungszentrum (DKFZ) Heidelberg, Heidelberg, Deutschland
| | - P Hohenberger
- Sektion Spezielle Chirurgische Onkologie und Thoraxchirurgie, Chirurgische Klinik, Universitätsmedizin Mannheim, Theodor-Kutzer-Ufer 1-3, 68135, Mannheim, Deutschland.
| |
Collapse
|
107
|
Gastric GIST with chondroid differentiation presented with gastric abscess – A case report and literature review. Arab J Gastroenterol 2016; 17:56-9. [DOI: 10.1016/j.ajg.2015.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 08/08/2014] [Accepted: 07/01/2015] [Indexed: 11/21/2022]
|
108
|
Tatangelo F, Cantile M, Collina F, Belli A, DE Franciscis S, Bianco F, Botti G. Gastric schwannoma misdiagnosed as GIST: A case report with immunohistochemical and molecular study. Oncol Lett 2016; 11:2497-2501. [PMID: 27073505 DOI: 10.3892/ol.2016.4281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Schwannomas are tumors derived from Schwann cells. Generally, they are benign and their typical site of origin is the subcutaneous tissue of the distal extremities or of the head and neck region. Gastrointestinal localization of schwannomas is extremely rare, and the stomach is the prevalent site. The present study describes the case of a gastric schwannoma in a 61-year-old male who underwent subtotal gastrectomy following a clinical diagnosis of a gastrointestinal stromal tumor (GIST). A histological, immunohistochemical and molecular study was performed to exclude the misdiagnosis of GIST. The histomorphological features of the lesion and absence of c-Kit and PDGFRA mutations indicated the diagnosis of gastric schwannoma.
Collapse
Affiliation(s)
- Fabiana Tatangelo
- Pathology Unit, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Monica Cantile
- Pathology Unit, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Francesca Collina
- Pathology Unit, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Andrea Belli
- Department of General and Hepato-Pancreato-Biliary Surgery, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Silvia DE Franciscis
- Department of General and Hepato-Pancreato-Biliary Surgery, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Franco Bianco
- Department of General and Hepato-Pancreato-Biliary Surgery, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| | - Gerardo Botti
- Pathology Unit, National Cancer Institute 'G. Pascale' Foundation, Naples, Campania 80131, Italy
| |
Collapse
|
109
|
Rizzo FM, Palmirotta R, Marzullo A, Resta N, Cives M, Tucci M, Silvestris F. Parallelism of DOG1 expression with recurrence risk in gastrointestinal stromal tumors bearing KIT or PDGFRA mutations. BMC Cancer 2016; 16:87. [PMID: 26867653 PMCID: PMC4750215 DOI: 10.1186/s12885-016-2111-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 02/03/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are characterized by mutations of KIT (v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog) or PDGFRA (platelet-derived growth factor receptor α) that may be efficiently targeted by tyrosine kinase inhibitors (TKI). Notwithstanding the early responsiveness to TKI, the majority of GISTs progress, imposing the need for alternative therapeutic strategies. DOG1 (discovered on GIST-1) shows a higher sensitivity as a diagnostic marker than KIT, however its prognostic role has been little investigated. METHODS We evaluated DOG1 expression by immunohistochemistry (IHC) in 59 patients with GISTs, and correlated its levels with clinical and pathological features as well as mutational status. Kaplan-Meier analysis was also applied to assess correlations of the staining score with patient recurrence-free survival (RFS). RESULTS DOG1 was expressed in 66% of CD117(+) GISTs and highly associated with tumor size and the rate of wild-type tumors. Kaplan-Meier survival analysis showed that a strong DOG1 expression demonstrated by IHC correlated with a worse 2-year RFS rate, suggesting its potential ability to predict GISTs with poor prognosis. CONCLUSIONS These findings suggest a prognostic role for DOG1, as well as its potential for inclusion in the criteria for risk stratification.
Collapse
Affiliation(s)
- Francesca Maria Rizzo
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Piazza Giulio Cesare, 11-70124, Bari, Italy.
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Piazza Giulio Cesare, 11-70124, Bari, Italy.
- University San Raffaele Rome, Interinstitutional Multidisciplinary BioBank (BioBIM), IRCCS San Raffaele Pisana, Rome, Italy.
| | - Andrea Marzullo
- Department of Pathology, University of Bari "A. Moro", Bari, Italy.
| | - Nicoletta Resta
- Division of Medical Genetics, Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Bari, Italy.
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Piazza Giulio Cesare, 11-70124, Bari, Italy.
| | - Marco Tucci
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Piazza Giulio Cesare, 11-70124, Bari, Italy.
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Piazza Giulio Cesare, 11-70124, Bari, Italy.
| |
Collapse
|
110
|
Olsen L, Åkesson CP, Aleksandersen M, Boysen P, Press CM, Drouet F, Storset AK, Espenes A. NCR1+ cells appear early in GALT development of the ovine foetus and acquire a c-kit+ phenotype towards the end of gestation. Vet Immunol Immunopathol 2016; 169:79-84. [PMID: 26827843 DOI: 10.1016/j.vetimm.2015.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/17/2022]
Abstract
The amount, distribution and phenotype of ovine NCR1+ cells were investigated during developing GALT from day 70 of gestation. Antibodies against CD3 and CD79 were used to identify the compartments of GALT, and the localization of NCR1+ cells were correlated within these structures. Markers CD34 and c-kit, in addition to Ki67, were used to investigate possible origin and the stage of development of the NCR1+ cells. NCR1+ cells were present as single cells in the subepithelial tissue as early as 70 days of gestation, and were predominantly present in the T cell rich IFAs and domes as these intestinal wall compartments developed. While NCR1+ cells proliferated more intensively at mid-gestation (70-104 days), the number of NCR1+ cells also expressing c-kit, increased at the end of gestation. In conclusion, NCR1+ cells appeared early in T cell areas of the gut and displayed a phenotype consistent with intermediate stages of cNK cells and/or a subpopulation of ILC22.
Collapse
Affiliation(s)
- Line Olsen
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Caroline Piercey Åkesson
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Mona Aleksandersen
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Preben Boysen
- Department of Food Safety & Infection Biology, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.
| | - Charles McL Press
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| | - Françoise Drouet
- Institut National de la Recherche Agronomique, UMR1282, Infectiologie et Santé Publique, Laboratoire Apicomplexes et Immunité Muqueuse, Nouzilly, France.
| | - Anne K Storset
- Department of Food Safety & Infection Biology, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.
| | - Arild Espenes
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
111
|
Reichardt P, Demetri GD, Gelderblom H, Rutkowski P, Im SA, Gupta S, Kang YK, Schöffski P, Schuette J, Soulières D, Blay JY, Goldstein D, Fly K, Huang X, Corsaro M, Lechuga MJ, Martini JF, Heinrich MC. Correlation of KIT and PDGFRA mutational status with clinical benefit in patients with gastrointestinal stromal tumor treated with sunitinib in a worldwide treatment-use trial. BMC Cancer 2016; 16:22. [PMID: 26772734 PMCID: PMC4714485 DOI: 10.1186/s12885-016-2051-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/06/2016] [Indexed: 01/15/2023] Open
Abstract
Background Several small studies indicated that the genotype of KIT or platelet-derived growth factor receptor-α (PDGFRA) contributes in part to the level of clinical effectiveness of sunitinib in gastrointestinal stromal tumor (GIST) patients. This study aimed to correlate KIT and PDGFRA mutational status with clinical outcome metrics (progression-free survival [PFS], overall survival [OS], objective response rate [ORR]) in a larger international patient population. Methods This is a non-interventional, retrospective analysis in patients with imatinib-resistant or intolerant GIST who were treated in a worldwide, open-label treatment-use study (Study 1036; NCT00094029) in which sunitinib was administered at a starting dose of 50 mg/day on a 4-week-on, 2-week-off schedule. Molecular status was obtained in local laboratories with tumor samples obtained either pre-imatinib, post-imatinib/pre-sunitinib, or post-sunitinib treatment, and all available data were used in the analyses regardless of collection time. The primary analysis compared PFS in patients with primary KIT exon 11 versus exon 9 mutations (using a 2-sided log-rank test) and secondary analyses compared OS (using the same test) and ORR (using a 2-sided Pearson χ2 test) in the same molecular subgroups. Results Of the 1124 sunitinib-treated patients in the treatment-use study, 230 (20 %) were included in this analysis, and baseline characteristics were similar between the two study populations. Median PFS was 7.1 months. A significantly better PFS was observed in patients with a primary mutation in KIT exon 9 (n = 42) compared to those with a primary mutation in exon 11 (n = 143; hazard ratio = 0.59; 95 % confidence interval, 0.39–0.89; P = 0.011), with median PFS times of 12.3 and 7.0 months, respectively. Similarly, longer OS and higher ORR were observed in patients with a primary KIT mutation in exon 9 versus exon 11. The data available were limited to investigate the effects of additional KIT or PDGFRA mutations on the efficacy of sunitinib treatment. Conclusions This large retrospective analysis confirms the prognostic significance of KIT mutation status in patients with GIST. This analysis also confirms the effectiveness of sunitinib as a post-imatinib therapy, regardless of mutational status. Trial registration NCT01459757. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2051-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany.
| | - George D Demetri
- Ludwig Center at Harvard and Dana-Farber Cancer Institute, Boston, MA, USA.
| | | | - Piotr Rutkowski
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | | | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, and Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium.
| | | | - Denis Soulières
- Centre Hospitalier de l'Université de Montreal, Montreal, QC, Canada.
| | - Jean-Yves Blay
- Centre Léon Bérard, Université Claude Bernard, Lyon, France.
| | | | | | | | | | | | | | - Michael C Heinrich
- VA Portland Health Care System and Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
112
|
Feng F, Tian Y, Liu Z, Xu G, Liu S, Guo M, Lian X, Fan D, Zhang H. Clinicopathologic Features and Clinical Outcomes of Esophageal Gastrointestinal Stromal Tumor: Evaluation of a Pooled Case Series. Medicine (Baltimore) 2016; 95:e2446. [PMID: 26765432 PMCID: PMC4718258 DOI: 10.1097/md.0000000000002446] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinicopathologic features and clinical outcomes of gastrointestinal stromal tumors (GISTs) in esophagus are limited, because of the relatively rare incidence of esophageal GISTs. Therefore, the aim of the current study was to investigate the clinicopathologic features and clinical outcomes of esophageal GISTs, and to investigate the potential factors that may predict prognosis.Esophageal GIST cases were obtained from our center and from case reports and clinical studies extracted from MEDLINE. Clinicopathologic features and survivals were analyzed and compared with gastric GISTs from our center.The most common location was lower esophagus (86.84%), followed by middle and upper esophagus (11.40% and 1.76%). The majority of esophageal GISTs were classified as high-risk category (70.83%). Mitotic index was correlated with histologic type, mutational status, and tumor size. The 5-year disease-free survival and disease-specific survival were 65.1% and 65.9%, respectively. Tumor size, mitotic index, and National Institutes of Health risk classification were associated with prognosis of esophageal GISTs. Only tumor size, however, was the independent risk factor for the prognosis of esophageal GISTs. In comparison to gastric GISTs, the distribution of tumor size, histologic type, and National Institutes of Health risk classification were significantly different between esophageal GISTs and gastric GISTs. The disease-free survival and disease-specific survival of esophageal GISTs were significantly lower than that of gastric GISTs.The most common location for esophageal GISTs was lower esophagus, and most of the esophageal GISTs are high-risk category. Tumor size was the independent risk factor for the prognosis of esophageal GISTs. Esophageal GISTs differ significantly from gastric GISTs in respect to clinicopathologic features. The prognosis of esophageal GISTs was worse than that of gastric GISTs.
Collapse
Affiliation(s)
- Fan Feng
- From the Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University (FF, ZL, GX, SL, MG, XL, DF, HZ) and Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China (YT)
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Ahmad A, Libbey NP, Somasundar P, Katz SC. Gastrointestinal stromal tumour metastatic to the epididymis. BMJ Case Rep 2015; 2015:bcr-2015-211555. [PMID: 26400593 DOI: 10.1136/bcr-2015-211555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Gastrointestinal stromal tumours (GIST) are mesenchymal neoplasms with a propensity to metastasise to the liver and peritoneal cavity. Since the advent of tyrosine kinase inhibitors, outcomes for patients with metastatic GIST have improved dramatically. Secondary to the longevity in survival, patients may develop metastatic disease in very unusual locations, which poses significant diagnostic dilemmas and management challenges. We report a case of a patient with GIST who presented with an epididymal metastasis manifesting as a scrotal mass. Resistance to targeted medical therapies continues to pose a challenge, and our case highlights the importance of a multidisciplinary approach in such patients, including long-term follow-up.
Collapse
Affiliation(s)
- Ali Ahmad
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Norman Peter Libbey
- Department of Pathology, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Ponnandai Somasundar
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, Rhode Island, USA
| |
Collapse
|
114
|
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
|
115
|
Chen EC, Karl TA, Kalisky T, Gupta SK, O'Brien CA, Longacre TA, van de Rijn M, Quake SR, Clarke MF, Rothenberg ME. KIT Signaling Promotes Growth of Colon Xenograft Tumors in Mice and Is Up-Regulated in a Subset of Human Colon Cancers. Gastroenterology 2015; 149:705-17.e2. [PMID: 26026391 PMCID: PMC4550533 DOI: 10.1053/j.gastro.2015.05.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 05/12/2015] [Accepted: 05/19/2015] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Receptor tyrosine kinase (RTK) inhibitors have advanced colon cancer treatment. We investigated the role of the RTK KIT in development of human colon cancer. METHODS An array of 137 patient-derived colon tumors and their associated xenografts were analyzed by immunohistochemistry to measure levels of KIT and its ligand KITLG. KIT and/or KITLG was stably knocked down by expression of small hairpin RNAs from lentiviral vectors in DLD1, HT29, LS174T, and COLO320 DM colon cancer cell lines, and in UM-COLON#8 and POP77 xenografts; cells transduced with only vector were used as controls. Cells were analyzed by real-time quantitative reverse transcription polymerase chain reaction, single-cell gene expression analysis, flow cytometry, and immunohistochemical, immunoblot, and functional assays. Xenograft tumors were grown from control and KIT-knockdown DLD1 and UM-COLON#8 cells in immunocompromised mice and compared. Some mice were given the RTK inhibitor imatinib after injection of cancer cells; tumor growth was measured based on bioluminescence. We assessed tumorigenicity using limiting dilution analysis. RESULTS KIT and KITLG were expressed heterogeneously by a subset of human colon tumors. Knockdown of KIT decreased proliferation of colon cancer cell lines and growth of xenograft tumors in mice compared with control cells. KIT knockdown cells had increased expression of enterocyte markers, decreased expression of cycling genes, and, unexpectedly, increased expression of LGR5 associated genes. No activating mutations in KIT were detected in DLD1, POP77, or UM-COLON#8 cells. However, KITLG-knockdown DLD1 cells formed smaller xenograft tumors than control cells. Gene expression analysis of single CD44(+) cells indicated that KIT can promote growth via KITLG autocrine and/or paracrine signaling. Imatinib inhibited growth of KIT(+) colon cancer organoids in culture and growth of xenograft tumors in mice. Cancer cells with endogenous KIT expression were more tumorigenic in mice. CONCLUSIONS KIT and KITLG are expressed by a subset of human colon tumors. KIT signaling promotes growth of colon cancer cells and organoids in culture and xenograft tumors in mice via its ligand, KITLG, in an autocrine or paracrine manner. Patients with KIT-expressing colon tumors can benefit from KIT RTK inhibitors.
Collapse
Affiliation(s)
- Evan C Chen
- Stanford School of Medicine, Stanford, California
| | - Taylor A Karl
- Division of Gastroenterology and Hepatology, Stanford School of Medicine, Stanford, California
| | - Tomer Kalisky
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel
| | - Santosh K Gupta
- Department of Pathology, Stanford School of Medicine, California
| | | | - Teri A Longacre
- Department of Pathology, Stanford School of Medicine, California
| | - Matt van de Rijn
- Department of Pathology, Stanford School of Medicine, California
| | - Stephen R Quake
- Department of Bioengineering, Stanford School of Medicine, Stanford, California; Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Michael F Clarke
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, California
| | - Michael E Rothenberg
- Division of Gastroenterology and Hepatology, Stanford School of Medicine, Stanford, California; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, California.
| |
Collapse
|
116
|
Cananzi FCM, Belgaumkar A, Lorenzi B, Mudan S. Liver surgery in the multidisciplinary management of gastrointestinal stromal tumour. ANZ J Surg 2015; 84:937-42. [PMID: 25444423 DOI: 10.1111/ans.12199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2013] [Indexed: 01/23/2023]
Abstract
BACKGROUND After the introduction of tyrosine kinase inhibitors (TKIs), the role of surgical resection in treating liver metastasis from gastrointestinal stromal tumour (GIST) is unclear. In this study, we evaluated the outcome of patients treated with TKIs followed by surgery for metastatic GIST. METHODS Eleven patients who underwent liver resection after downsizing TKIs therapy for metastatic GIST from 2006 until 2010 were reviewed. RESULTS One and 2-year overall survival (OS) rates were 80.8% and 70.7%. All patients with an initially resectable tumour were still alive without recurrence. Patients operated on clinical response had a better outcome (1-year and 2-year OS rate of 100%) than those operated on disease progression (1-year and 2-year OS rates of 60% and 40%; P = 0.043). No deaths were observed among patients who achieved an R0 resection (R0 versus R1/R2, P = 0.001). CONCLUSION R0 resection and clinical response to TKI are predictors of survival. Surgical resection should be performed as soon as feasible in responder patients. In poor responders, surgery may not add any survival benefit, except in localized progressive disease. In resectable metastatic liver disease, preoperative TKIs or upfront surgery followed by adjuvant therapy could be considered. Larger studies are needed to determine the optimum approach in patients with metastatic GIST.
Collapse
|
117
|
Expression of CD117, DOG-1, and IGF-1R in gastrointestinal stromal tumours - an analysis of 70 cases from 2004 to 2010. GASTROENTEROLOGY REVIEW 2015; 11:115-22. [PMID: 27350839 PMCID: PMC4916232 DOI: 10.5114/pg.2015.52587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/08/2015] [Indexed: 12/12/2022]
Abstract
Introduction Determination of the type of mutations in gastrointestinal stromal tumours (GIST) plays a major role in assessing the risk of progression of the disease, and also allows determination of the clinical management and treatment. More accurate GIST diagnosis is possible by using simultaneously various types of antibodies to immunohistochemistry methods in routine procedures. Aim To evaluate the expression of CD117, DOG-1, and IGF-1R in patients with gastrointestinal stromal tumours, and analysis of the impact of the examined protein expression on patient survival with emphasis on specific recognition and prognostication of these tumours. Material and methods The protein expression was analyzed in 70 patients who had undergone surgical treatment for mesenchymal tumours of the gastrointestinal tract, using the immunohistochemical method. Results Positive expression of CD117, DOG-1, and IGF1R included 95.71%, 88.57% and 11.43% of study GISTs, respectively. Statistical analysis showed positive significant correlation between DOG-1 expression and histological type of tumour (p = 0.024). Analysis of overall survival curves of 70 GIST patients according to expression of CD117, DOG-1, and IGF1R did not show a tendency towards longer survival of patients with positive expression (p > 0.05). Conclusions Predictive factors determining the survival time of patients are strongly associated with morphological features of tumours. A thorough analysis of each case plays a key role in predicting survival time of patients and may be a clue in targeting the therapeutic procedure.
Collapse
|
118
|
Yoo C, Ryu MH, Jo J, Park I, Ryoo BY, Kang YK. Efficacy of Imatinib in Patients with Platelet-Derived Growth Factor Receptor Alpha-Mutated Gastrointestinal Stromal Tumors. Cancer Res Treat 2015; 48:546-52. [PMID: 26130666 PMCID: PMC4843750 DOI: 10.4143/crt.2015.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/12/2015] [Indexed: 12/11/2022] Open
Abstract
Purpose The incidence of gastrointestinal stromal tumors (GISTs) harboring platelet-derived growth factor receptor alpha (PDGFRA) mutations is low, therefore further investigation of the efficacy of imatinib in this subgroup was needed. Materials and Methods Patients with PDGFRA-mutant GISTs who received imatinib as primary therapy for advanced disease between January 2000 and June 2012 were identified from the GIST registry of Asan Medical Center, Seoul, Korea. Results KIT and PDGFRA genotyping in 823 patients identified 18 patients (2%) with PDGFRA mutations who were treated with first-line imatinib. Exon 18 D842V substitution, non-D842V exon 18 mutations, and exon 12 mutations were detected in nine (50%), four (22%), and five (28%) patients, respectively. Objective response rate differed significantly between patients with the D842V mutation and those with non-D842V mutations (0% [0/5] vs. 71% [5/7], p=0.03). In all patients, median progression-free survival (PFS) and overall survival (OS) was 24.8 months (95% confidence interval [CI], 0.0 to 57.2) and 51.2 months (95% CI, 37.1 to 65.3), respectively. Significantly, poorer PFS was observed for patients with D842V-mutant GISTs than those with non-D842V PDGFRA-mutant GISTs: median 3.8 months (95% CI, 1.4 to 6.3) versus 29.5 months (95% CI, 18.3 to 40.7) (p < 0.001). Patients with the D842V mutation had poorer OS than those with non-D842V PDGFRA mutations: median 25.2 months (95% CI, 12.7 to 37.8) versus 59.8 months (95% CI, 43.0 to 76.5) (p=0.02). Conclusion Imatinib is active against non-D842V PDGFRA-mutant GISTs, whereas GISTs harboring the D842V mutation are primarily resistant to imatinib.
Collapse
Affiliation(s)
- Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jungmin Jo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
119
|
Theodoropoulos DG. Gastrointestinal stromal tumors of the colon and rectum. SEMINARS IN COLON AND RECTAL SURGERY 2015. [DOI: 10.1053/j.scrs.2015.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
120
|
Szarek E, Ball ER, Imperiale A, Tsokos M, Faucz FR, Giubellino A, Moussallieh FM, Namer IJ, Abu-Asab MS, Pacak K, Taïeb D, Carney JA, Stratakis CA. Carney triad, SDH-deficient tumors, and Sdhb+/- mice share abnormal mitochondria. Endocr Relat Cancer 2015; 22:345-52. [PMID: 25808178 PMCID: PMC4433412 DOI: 10.1530/erc-15-0069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2015] [Indexed: 12/20/2022]
Abstract
Carney triad (CTr) describes the association of paragangliomas (PGL), pulmonary chondromas, and gastrointestinal (GI) stromal tumors (GISTs) with a variety of other lesions, including pheochromocytomas and adrenocortical tumors. The gene(s) that cause CTr remain(s) unknown. PGL and GISTs may be caused by loss-of-function mutations in succinate dehydrogenase (SDH) (a condition known as Carney-Stratakis syndrome (CSS)). Mitochondrial structure and function are abnormal in tissues that carry SDH defects, but they have not been studied in CTr. For the present study, we examined mitochondrial structure in human tumors and GI tissue (GIT) of mice with SDH deficiency. Tissues from 16 CTr tumors (n=12), those with isolated GIST (n=1), and those with CSS caused by SDHC (n=1) and SDHD (n=2) mutations were studied by electron microscopy (EM). Samples of GIT from mice with a heterozygous deletion in Sdhb (Sdhb(+) (/-), n=4) were also studied by EM. CTr patients presented with mostly epithelioid GISTs that were characterized by plump cells containing a centrally located, round nucleus and prominent nucleoli; these changes were almost identical to those seen in the GISTs of patients with SDH. In tumor cells from patients, regardless of diagnosis or tumor type, cytoplasm contained an increased number of mitochondria with a 'hypoxic' phenotype: mitochondria were devoid of cristae, exhibited structural abnormalities, and were of variable size. Occasionally, mitochondria were small and round; rarely, they were thin and elongated with tubular cristae. Many mitochondria exhibited amorphous fluffy material with membranous whorls or cystic structures. A similar mitochondrial hypoxic phenotype was seen in Sdhb(+) (/-) mice. We concluded that tissues from SDH-deficient tumors, those from mouse GIT, and those from CTr tumors shared identical abnormalities in mitochondrial structure and other features. Thus, the still-elusive CTr defect(s) is(are) likely to affect mitochondrial function, just like germline SDH-deficiency does.
Collapse
Affiliation(s)
- Eva Szarek
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Evan R Ball
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Alessio Imperiale
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of Immunolo
| | - Maria Tsokos
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Fabio R Faucz
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Alessio Giubellino
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - François-Marie Moussallieh
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of Immunolo
| | - Izzie-Jacques Namer
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of Immunolo
| | - Mones S Abu-Asab
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Karel Pacak
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - David Taïeb
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of Immunolo
| | - J Aidan Carney
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics (SEGEN)Program on Developmental Endocrinology and Genetics (PDEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, Maryland 20892, USADepartment of Biophysics and Nuclear MedicineUniversity Hospitals of Strasbourg, Strasbourg, FranceFaculty of MedicineIcube UMR 7357 University of Strasbourg/CNRS and FMTS, Strasbourg, FranceLaboratory of PathologyNational Cancer Institute (NCI), NIH, Bethesda, Maryland 20892, USASection on Medical Neuroendocrinology (SMN)Program on Reproductive and Adult Endocrinology (PRAE), NICHD, NIH, Bethesda, Maryland 20892, USASection of Immunopathology and Laboratory of ImmunologyNational Eye Institute, U.S. National Institutes of Health, Bethesda, Maryland 20892, USADepartment of Nuclear MedicineLa Timone University Hospital, CERIMED, 264, Rue Saint-Pierre, 13385 Marseille Cedex 5, FranceInstitut Paoli-CalmettesInserm UMR1068 Marseille Cancerology Research Center, Marseille, FranceEmeritus Staff CenterMayo Clinic Rochester, 200 First Street Southwest, Rochester, Minnesota 55905, USA
| |
Collapse
|
121
|
Zhu JQ, Ou WB. Therapeutic targets in gastrointestinal stromal tumors. World J Transl Med 2015; 4:25-37. [DOI: 10.5528/wjtm.v4.i1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 09/14/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common type of mesenchymal tumor of the gastrointestinal tract. The tumorigenesis of GISTs is driven by gain-of-function mutations in KIT or platelet-derived growth factor receptor α (PDGFRA), resulting in constitutive activation of the tyrosine kinase and its downstream signaling pathways. Oncogenic KIT or PDGFRA mutations are compelling therapeutic targets for the treatment of GISTs, and the KIT/PDGFRA inhibitor imatinib is the standard of care for patients with metastatic GISTs. However, most GIST patients develop clinical resistance to imatinib and other tyrosine kinase inhibitors. Five mechanisms of resistance have been characterized: (1) acquisition of a secondary point mutation in KIT or PDGFRA; (2) genomic amplification of KIT; (3) activation of an alternative receptor tyrosine kinase; (4) loss of KIT oncoprotein expression; and (5) wild-type GIST. Currently, sunitinib is used as a second-line treatment for patients after imatinib failure, and regorafenib has been approved for patients whose disease is progressing on both imatinib and sunitinib. Phase II/III trials are currently in progress to evaluate novel inhibitors and immunotherapies targeting KIT, its downstream effectors such as phosphatidylinositol 3-kinase, protein kinase B and mammalian target of rapamycin, heat shock protein 90, and histone deacetylase inhibitor. Other candidate targets have been identified, including ETV1, AXL, insulin-like growth factor 1 receptor, KRAS, FAS receptor, protein kinase c theta, ANO1 (DOG1), CDC37, and aurora kinase A. These candidates warrant clinical evaluation as novel therapeutic targets in GIST.
Collapse
|
122
|
Hameed M, Corless C, George S, Hornick JL, Kakar S, Lazar AJ, Tang L. Template for Reporting Results of Biomarker Testing of Specimens From Patients With Gastrointestinal Stromal Tumors. Arch Pathol Lab Med 2015; 139:1271-5. [DOI: 10.5858/arpa.2014-0578-cp] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meera Hameed
- From Surgical Pathology, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York (Drs Hameed and Tang); the Department of Pathology, Oregon Health & Science University, Portland (Dr Corless); the Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (Dr George); the Department of Pathology, Brigham and Women's Hospital, Boston (Dr Ho
| | | | | | | | | | | | | |
Collapse
|
123
|
Jones DH, Caracciolo JT, Hodul PJ, Strosberg JR, Coppola D, Bui MM. Familial Gastrointestinal Stromal Tumor Syndrome: Report of 2 Cases with KIT Exon 11 Mutation. Cancer Control 2015; 22:102-8. [DOI: 10.1177/107327481502200113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Derek H. Jones
- University of South Florida Morsani College of Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jamie T. Caracciolo
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Pamela J. Hodul
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jonathan R. Strosberg
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Domenico Coppola
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Marilyn M. Bui
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Sarcoma, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
124
|
Ahmad F, Lad P, Bhatia S, Das BR. Molecular spectrum of c-KIT and PDGFRA gene mutations in gastro intestinal stromal tumor: determination of frequency, distribution pattern and identification of novel mutations in Indian patients. Med Oncol 2015; 32:424. [PMID: 25481675 DOI: 10.1007/s12032-014-0424-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 11/29/2014] [Indexed: 01/08/2023]
Abstract
KIT and PDGFRA gene mutations are the major genetic alterations seen in gastrointestinal stromal tumors (GISTs) and are being used clinically for predicting response to imatinib therapy. In the current study, we set out to explore the frequency and distribution pattern of c-KIT (exons 9, 11 and 13) and PDGFRA (exons 12 and 18) by direct sequencing in a series of 70 Indian GIST cases. Overall, 27 (38.5 %) and 4 (5.7 %) of the cases had c-KIT and PDGFRA mutations, respectively. Majority of KIT mutations involved exon 11 (85.7 %), followed by exon 9 (14.3 %), while none showed exon 13 mutation. Most exon 9 mutations showed Ala503-Tyr504 duplication, while one had novel point mutation at codon 476 (S476G). In contrast to exon 9 mutations, most exon 11 mutations were in-frame deletions (79 %, 19/24), predominantly at codons 550-560, while remaining exon 11 mutant cases were point mutations at codons 559, 560, 568, 573 and 575. Interestingly, P573T, Q556_V560delinsH, Q575H and Q575_P577 were novel variations observed in exon 11. The PDGFRA mutations were seen mostly in exon 18, which showed point mutation at codon 842 (D842V), while exon 12 showed a novel indel variation (V561_H570delinsT). No significant correlation between c-KIT/PDGFRA mutations and clinicopathological data was observed. In conclusion, this study highlights the frequency and distribution pattern of c-KIT/PDGFRA mutation in Indian cohort. The current study identified novel variations that added new insights into the genetic heterogeneity of GIST patients. Furthermore, this is the first study to report the presence of PDGFRA mutation from Indian subcontinent.
Collapse
Affiliation(s)
- Firoz Ahmad
- Research and Development, SRL Ltd, Plot No. 1, Prime Square Building, S. V. Road Goregaon (W), Mumbai, 400062, India
| | | | | | | |
Collapse
|
125
|
Abstract
In anatomic pathology, the current state encompassing the pre-analytic processes of tissue collection, handling, examination, preparation, processing, and storage are largely uncontrolled, inconsistently performed, and/or not standardized according to the sound scientific data. Pre-analytic defects result in nearly three-quarters of the problems in laboratory diagnostics. This is evident in quality surveys from well-respected institutions that document high miss rates in the required basics of information related to patient and tissue identity, let alone parameters documenting quality aspects related to the surgical specimen and its preservation. This talk will describe the historical approach to tissue processing and identify gaps from worldwide observations in current laboratory practices. It will also offer potential methodological and technological solutions and process improvements that laboratories may consider in serving the ultimate users of pathology information: the clinician and the patient. It illustrates the need for scientifically validated specimen guidelines and a performance based, standardized and documented "chain of custody" of the pre-analytical steps from the patient's body through fixation. For thought leaders and professional standard setters, opportunities for optimizing molecular studies exist in specimen collection, transfer, grossing, fixation, and decalcification protocols. In this evolving era of molecular profiling and personalized therapeutic decision-making, a well-reasoned and coordinated focus on pre-analytic processes that optimizes specimens for subsequent testing will result in: Improved specimen quality for molecular testing Improved accuracy of diagnostic and molecular test results Reduced Turnaroundtimes for same-day diagnosis Enhanced satisfaction of clinicians and patients.
Collapse
Affiliation(s)
- Franco Visinoni
- Milestone Srl, Via Fatebenefratelli 1/5, 24010, Sorisole Bergamo, Italy,
| |
Collapse
|
126
|
Oncogenic Kit signals on endolysosomes and endoplasmic reticulum are essential for neoplastic mast cell proliferation. Nat Commun 2014; 5:5715. [PMID: 25493654 PMCID: PMC4284665 DOI: 10.1038/ncomms6715] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/30/2014] [Indexed: 11/08/2022] Open
Abstract
Kit is a receptor-type tyrosine kinase found on the plasma membrane. It can transform mast cells through activating mutations. Here, we show that a mutant Kit from neoplastic mast cells from mice, Kit(D814Y), is permanently active and allows cells to proliferate autonomously. It does so by activating two signalling pathways from different intracellular compartments. Mutant Kit from the cell surface accumulates on endolysosomes through clathrin-mediated endocytosis, which requires Kit's kinase activity. Kit(D814Y) is constitutively associated with phosphatidylinositol 3-kinase, but the complex activates Akt only on the cytoplasmic surface of endolysosomes. It resists destruction because it is under-ubiquitinated. Kit(D814Y) also appears in the endoplasmic reticulum soon after biosynthesis, and there, can activate STAT5 aberrantly. These mechanisms of oncogenic signalling are also seen in rat and human mast cell leukemia cells. Thus, oncogenic Kit signalling occurs from different intracellular compartments, and the mutation acts by altering Kit trafficking as well as activation.
Collapse
|
127
|
Hsueh YS, Chang HH, Chiang NJ, Yen CC, Li CF, Chen LT. MTOR inhibition enhances NVP-AUY922-induced autophagy-mediated KIT degradation and cytotoxicity in imatinib-resistant gastrointestinal stromal tumors. Oncotarget 2014; 5:11723-11736. [PMID: 25375091 PMCID: PMC4294368 DOI: 10.18632/oncotarget.2607] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/21/2014] [Indexed: 01/16/2023] Open
Abstract
Our previous study demonstrated NVP-AUY922, a HSP90AA1 inhibitor, could enhance mutant KIT degradation in gastrointestinal stromal tumor (GIST) cells through both proteasome- and autophagy-mediated pathways. Herein, we showed rapamycin, a MTOR inhibitor and autophagy inducer, could reduce total and phospho-KIT expression levels and enhance apoptosis in imatinib-resistant GIST cells. The involvement of autophagy in rapamycin-induced KIT downregulation was further confirmed by co-localization of KIT and autophagosome, and partial recovery of KIT expression level by either siRNA-mediated BECN1 and ATG5 silencing or autophagy inhibitors after rapamycin. Rapamycin and NVP-AUY922 synergistically inhibited GIST cells growth in vitro. The combination of low-dose NVP-AUY922 with rapamycin had comparable effects on reducing KIT expression, increasing MAP1LC3B puncta and tumor necrosis, and inhibiting tumor growth as high-dose NVP-AUY922 did in GIST430 xenograft model. Our results suggest the addition of a MTOR inhibitor may reduce NVP-AUY922 dose requirement and potentially improve its therapeutic index in mutant KIT-expressing GISTs.
Collapse
Affiliation(s)
- Yuan-Shuo Hsueh
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nai-Jung Chiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chueh-Chuan Yen
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chien-Feng Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Pathology, Chi-Mei Foundation Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwann
| |
Collapse
|
128
|
Tornillo L. Gastrointestinal stromal tumor - an evolving concept. Front Med (Lausanne) 2014; 1:43. [PMID: 25593916 PMCID: PMC4291900 DOI: 10.3389/fmed.2014.00043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors of the gastrointestinal tract. The discovery that these tumors, formerly thought of smooth muscle origin, are indeed better characterized by specific activating mutation in genes coding for the receptor tyrosine kinases (RTKs) CKIT and PDGFRA and that these mutations are strongly predictive for the response to targeted therapy with RTK inhibitors has made GISTs the typical example of the integration of basic molecular knowledge in the daily clinical activity. The information on the mutational status of these tumors is essential to predict (and subsequently to plan) the therapy. As resistant cases are frequently wild type, other possible oncogenic events, defining other "entities," have been discovered (e.g., succinil dehydrogenase mutation/dysregulation, insuline growth factor expression, and mutations in the RAS-RAF-MAPK pathway). The classification of disease must nowadays rely on the integration of the clinico-morphological characteristics with the molecular data.
Collapse
Affiliation(s)
- Luigi Tornillo
- Institute of Pathology, University of Basel , Basel , Switzerland
| |
Collapse
|
129
|
Jové M, Mora J, Sanjuan X, Rodriguez E, Robledo M, Farran L, Garcia del Muro X. Simultaneous KIT mutation and succinate dehydrogenase (SDH) deficiency in a patient with a gastrointestinal stromal tumour and Carney-Stratakis syndrome: a case report. Histopathology 2014; 65:712-7. [DOI: 10.1111/his.12506] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Maria Jové
- Department of Medical Oncology; Institut Català d'Oncologia; L'Hospitalet de Llobregat; Barcelona Spain
| | - Jaume Mora
- Department of Pediatric Oncology; Hospital Sant Joan de Déu (HSJD); Barcelona Spain
| | - Xavier Sanjuan
- Department of Pathology; Hospital Universitari de Bellvitge; L'Hospitalet de Llobregat; Barcelona Spain
| | - Eva Rodriguez
- Department of Pediatric Oncology; Hospital Sant Joan de Déu (HSJD); Barcelona Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer, Group Leader, Human Cancer Genetics Programme; Spanish National Cancer Centre (CNIO); Madrid Spain
| | - Leandre Farran
- Department of Surgery; Hospital Universitari de Bellvitge; L'Hospitalet de Llobregat; Barcelona Spain
| | - Xavier Garcia del Muro
- Department of Medical Oncology; Institut Català d'Oncologia; L'Hospitalet de Llobregat; Barcelona Spain
| |
Collapse
|
130
|
Imaging and clinicopathologic features of esophageal gastrointestinal stromal tumors. AJR Am J Roentgenol 2014; 203:306-14. [PMID: 25055264 DOI: 10.2214/ajr.13.11841] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The purpose of this article is to describe the imaging and clinicopathologic characteristics of esophageal gastrointestinal stromal tumors (GISTs) and to emphasize the features that differentiate esophageal GISTs from esophageal leiomyomas. MATERIALS AND METHODS A pathology database search identified all surgically resected or biopsied esophageal GISTs, esophageal leiomyomas, and esophageal leiomyosarcomas from 1994 to 2012. Esophageal GISTs were included only if imaging studies (including CT, fluoroscopic, or (18)F-FDG PET/CT scans) and clinical data were available. RESULTS Nineteen esophageal mesenchymal tumors were identified, including eight esophageal GISTs (42%), 10 esophageal leiomyomas (53%), and one esophageal leiomyosarcoma (5%). Four patients (50%) with esophageal GIST had symptoms, including dysphagia in three (38%), cough in one (13%), and chest pain in one (13%). One esophageal GIST appeared on barium study as a smooth submucosal mass. All esophageal GISTs appeared on CT as well-marginated predominantly distal lesions, isoattenuating to muscle, that moderately enhanced after IV contrast agent administration. Compared with esophageal leiomyomas, esophageal GISTs tended to be more distal, larger, and more heterogeneous and showed greater IV enhancement on CT. All esophageal GISTs showed marked avidity (mean maximum standardized uptake value, 16) on PET scans. All esophageal GISTs were positive for c-KIT (a cell-surface transmembrane tyrosine kinase also known as CD117) and CD34. On histopathology, six esophageal GISTs (75%) were of the spindle pattern and two (25%) were of a mixed spindle and epithelioid pattern. Five esophageal GISTs had exon 11 mutations (with imatinib sensitivity). Clinical outcome correlated with treatment strategy (resection plus adjuvant therapy or resection alone) rather than risk stratification. CONCLUSION Esophageal GISTs are unusual but clinically important mesenchymal neoplasms. Although esophageal GISTs and esophageal leiomyomas had overlapping imaging features, esophageal GISTs tended to be more distal, larger, more heterogeneous, and more enhancing on CT and were markedly FDG avid on PET. Given their malignant potential, esophageal GISTs should be included in the differential diagnosis of intramural esophageal neoplasms.
Collapse
|
131
|
Poveda A, del Muro XG, López-Guerrero JA, Martínez V, Romero I, Valverde C, Cubedo R, Martín-Broto J. GEIS 2013 guidelines for gastrointestinal sarcomas (GIST). Cancer Chemother Pharmacol 2014; 74:883-98. [PMID: 25193432 PMCID: PMC4209233 DOI: 10.1007/s00280-014-2547-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/19/2014] [Indexed: 02/06/2023]
Abstract
Gastrointestinal stromal tumors (GIST) are the most common mesenchymal soft tissue sarcoma of the gastrointestinal tract. Correct diagnosis with thorough use of pathologic and molecular tools of GIST mutations has been of the foremost importance. GIST are usually (95 %) KIT positive and harbor frequent KIT or platelet-derived growth factor receptor α-activating mutations. This deep molecular understanding has allowed the correct classification into risk groups with implications regarding prognosis, essential use in the development of targeted therapies and even response prediction to this drugs. Treatment has been evolving and an update to include lessons learned from recent trials in advanced disease as well as controversies in the adjuvant setting that are changing daily practice, is reviewed here. An effort from the Spanish Group for Sarcoma Research with investigators from the group has been undertaken to launch this third version of the GIST guidelines and provide a practical means for the different disciplines that treat this complex disease.
Collapse
Affiliation(s)
- Andrés Poveda
- Instituto Valenciano de Oncología, Calle del Profesor Beltrán Bàguena, 8, 46009, Valencia, Spain,
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Miettinen M, Lasota J. Succinate dehydrogenase deficient gastrointestinal stromal tumors (GISTs) - a review. Int J Biochem Cell Biol 2014; 53:514-9. [PMID: 24886695 DOI: 10.1016/j.biocel.2014.05.033] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/20/2014] [Accepted: 05/21/2014] [Indexed: 02/07/2023]
Abstract
Loss of function of the succinate dehydrogenase complex characterizes a rare group of human tumors including some gastrointestinal stromal tumors, paragangliomas, renal carcinomas, and pituitary adenomas, and these can all be characterized as SDH-deficient tumors. Approximately 7.5% of gastric gastrointestinal stromal tumors are SDH-deficient and not driven by KIT/PDGFRA mutations, as are most other GISTs. The occurrence of SDH-deficient GISTs is restricted to stomach, and they typically occur in children and young adults representing a spectrum of clinical behavior from indolent to progressive. Slow progression is a common feature even after metastatic spread has taken place, and many patients live years with metastases. SDH-deficient GISTs have characteristic morphologic features including multinodular gastric wall involvement, often multiple separate tumors, common lymphovascular invasion, and occasional lymph node metastases. Diagnostic is the loss of succinate dehydrogenase subunit B (SDHB) from the tumor cells and this can be practically assessed by immunohistochemistry. SDHA is lost in cases associated with SDHA mutations. Approximately half of the patients have SDH subunit gene mutations, often germline and most commonly A (30%), and B, C or D (together 20%), with both alleles inactivated in the tumor cells according to the classic tumor suppressor gene model. Half of the cases are not associated with SDH-mutations and epigenetic silencing of the SDH complex is the possible pathogenesis. Extensive genomic methylation has been observed in these tumors, which is in contrast with other GISTs. SDH-loss causes succinate accumulation and activation of pseudohypoxia signaling via overexpression of HIF-proteins. Activation of insulin-like growth factor 1-signaling is also typical of these tumors. SDH-deficient GISTs are a unique group of GISTs with an energy metabolism defect as the key oncogenic mechanism. This article is part of a Directed Issue entitled: Rare Cancers.
Collapse
Affiliation(s)
- Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
133
|
Jour G, Scarborough JD, Jones RL, Loggers E, Pollack SM, Pritchard CC, Hoch BL. Molecular profiling of soft tissue sarcomas using next-generation sequencing: a pilot study toward precision therapeutics. Hum Pathol 2014; 45:1563-71. [PMID: 24908143 DOI: 10.1016/j.humpath.2014.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/04/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022]
Abstract
Next-generation sequencing (NGS) can provide in-depth detection of numerous gene alterations. To date, there are very few reports describing the use of this technique in soft tissue sarcomas. Herein, we aim to test the utility of NGS in identifying targetable mutations in these tumors. NGS was performed using a clinically validated multiplexed gene sequencing panel interrogating the full coding sequence of 194 cancer-related genes. A custom bioinformatics pipeline was developed to detect all classes of mutations directly from the NGS data, including single-nucleotide variants, small insertions and deletions, copy number variation, and complex structural variations. Twenty-five soft tissue sarcomas were analyzed; 18 of these patients had metastatic disease and 7 primary locally advanced tumors. Targetable mutations for which clinical trials are available were identified in 60% of the cases. MAP2K4, AURKA, AURKB, and c-MYC amplification were recurrent events in leiomyosarcomas. Frequent non-targetable variants included copy losses of the TP53 (24%), PTEN (16%), and CDKN2A (20%). Additional frameshift mutations, deletion mutations, and single-nucleotide variants involving numerous genes, including RB1, NOTCH1, PIK3CA, PDGFRB, EPHA5, KDM6A, NF1, and FLT4 genes, were also identified. NGS is useful in identifying targetable mutations in soft tissue sarcomas that can serve as a rationale for inclusion of patients with advanced disease in ongoing clinical trials and allow for better risk stratification.
Collapse
Affiliation(s)
- George Jour
- University of Washington Medical Center, Department of Anatomic Pathology, Seattle, WA, 98195, USA.
| | - John D Scarborough
- University of Washington Medical Center, Department of Laboratory Medicine Seattle, WA, 98195, USA
| | - Robin L Jones
- University of Washington Medical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, 98195, USA; University of Washington Medical Center Division of Medical Oncology, Seattle, WA, 98195, USA
| | - Elizabeth Loggers
- University of Washington Medical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, 98195, USA; University of Washington Medical Center Division of Medical Oncology, Seattle, WA, 98195, USA
| | - Seth M Pollack
- University of Washington Medical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, 98195, USA; University of Washington Medical Center Division of Medical Oncology, Seattle, WA, 98195, USA
| | - Colin C Pritchard
- University of Washington Medical Center, Department of Laboratory Medicine Seattle, WA, 98195, USA; University of Washington, Department of Genome Sciences, University of Washington, Seattle, WA, 98195,USA
| | - Benjamin L Hoch
- University of Washington Medical Center, Department of Anatomic Pathology, Seattle, WA, 98195, USA
| |
Collapse
|
134
|
Bednarski BK, Araujo DM, Yi M, Torres KE, Lazar A, Trent JC, Cormier JN, Pisters PWT, Lev DC, Pollock RE, Feig BW, Hunt KK. Analysis of prognostic factors impacting oncologic outcomes after neoadjuvant tyrosine kinase inhibitor therapy for gastrointestinal stromal tumors. Ann Surg Oncol 2014; 21:2499-505. [PMID: 24639192 DOI: 10.1245/s10434-014-3632-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Management of gastrointestinal stromal tumors (GISTs) has been transformed with tyrosine kinase inhibitors (TKIs). While data on optimal duration of adjuvant imatinib remains elusive, guidelines for administration of neoadjuvant TKIs remain unknown. METHODS Under an institutional review board-approved protocol, patients at our institution with a diagnosis of GIST treated with neoadjuvant TKIs and surgical resection were identified. Clinical and pathologic characteristics were obtained from medical records. RESULTS Ninety-three patients underwent surgical resection after neoadjuvant TKI therapy; 41 had primary and 52 had recurrent/metastatic GIST. Median follow-up was 2.4 years. Median duration of neoadjuvant therapy was 315 (range 3-1,611) days for primary and 537 (range 4-3,257) days for recurrent/metastatic GIST (p = 0.001). Two-year, recurrence-free survival (RFS) was 85 and 44 % for primary and recurrent/metastatic disease, respectively, whereas 2-year overall survival (OS) was 97 % for primary and 73 % for recurrent/metastatic GIST. For primary GIST, duration of neoadjuvant therapy >365 days (p = 0.02) was associated with higher risk of recurrence on univariate analysis, whereas none of the clinicopathologic factors impacted OS. For recurrent/metastatic disease, disease progression was associated with a shorter OS (p = 0.001), but no factors were found to impact RFS. Lastly, when examining all patients, KIT mutations (p = 0.03) and multivisceral resection (p = 0.011) predicted shorter RFS. CONCLUSIONS Neoadjuvant TKIs can be effectively used for the treatment of primary and recurrent/metastatic GIST. While duration of neoadjuvant therapy, KIT mutation status, and the need for multivisceral resection can help to predict higher risk for recurrence, progression on neoadjuvant TKIs can aid in selection of patients with recurrent/metastatic disease for surgical resection.
Collapse
Affiliation(s)
- Brian K Bednarski
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Dubbink HJ, Deans ZC, Tops BBJ, van Kemenade FJ, Koljenović S, van Krieken HJM, Blokx WAM, Dinjens WNM, Groenen PJTA. Next generation diagnostic molecular pathology: critical appraisal of quality assurance in Europe. Mol Oncol 2014; 8:830-9. [PMID: 24704265 DOI: 10.1016/j.molonc.2014.03.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/10/2014] [Indexed: 01/08/2023] Open
Abstract
Tumor evaluation in pathology is more and more based on a combination of traditional histopathology and molecular analysis. Due to the rapid development of new cancer treatments that specifically target aberrant proteins present in tumor cells, treatment decisions are increasingly based on the molecular features of the tumor. Not only the number of patients eligible for targeted precision medicine, but also the number of molecular targets per patient and tumor type is rising. Diagnostic molecular pathology, the discipline that determines the molecular aberrations present in tumors for diagnostic, prognostic or predictive purposes, is faced with true challenges. The laboratories have to meet the need of comprehensive molecular testing using only limited amount of tumor tissue, mostly fixed in formalin and embedded in paraffin (FFPE), in short turnaround time. Choices must be made for analytical methods that provide accurate, reliable and cost-effective results. Validation of the test procedures and results is essential. In addition, participation and good performance in internal (IQA) and external quality assurance (EQA) schemes is mandatory. In this review, we critically evaluate the validation procedure for comprehensive molecular tests as well as the organization of quality assurance and assessment of competence of diagnostic molecular pathology laboratories within Europe.
Collapse
Affiliation(s)
- Hendrikus J Dubbink
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Zandra C Deans
- UK NEQAS for Molecular Genetics, Department of Laboratory Medicine, The Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Bastiaan B J Tops
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Folkert J van Kemenade
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Koljenović
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Han J M van Krieken
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Willeke A M Blokx
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands
| | - Winand N M Dinjens
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Patricia J T A Groenen
- Department of Pathology, Radboud University Medical Center, PO Box 9101, Nijmegen, The Netherlands.
| |
Collapse
|
136
|
High-resolution melting analysis is a sensitive diagnostic tool to detect imatinib-resistant and imatinib-sensitive PDGFRA exon 18 mutations in gastrointestinal stromal tumors. Hum Pathol 2014; 45:573-82. [DOI: 10.1016/j.humpath.2013.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/10/2013] [Accepted: 10/19/2013] [Indexed: 01/01/2023]
|
137
|
Virgilio E, Mercantini P, Tarantino G, Socciarelli F, Pilozzi E, Uccini S, Caterino S, Ziparo V. Gastrointestinal Stromal Tumors With De Novo Anaplastic Dedifferentiation: Considerations on a Little-Known Neoplastic Metamorphosis. Int J Surg Pathol 2014; 22:385. [PMID: 24487042 DOI: 10.1177/1066896914521288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
138
|
Bridge JA. The role of cytogenetics and molecular diagnostics in the diagnosis of soft-tissue tumors. Mod Pathol 2014; 27 Suppl 1:S80-97. [PMID: 24384855 DOI: 10.1038/modpathol.2013.179] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 07/19/2013] [Indexed: 12/11/2022]
Abstract
Soft-tissue sarcomas are rare, comprising <1% of all cancer diagnoses. Yet the diversity of histological subtypes is impressive with >100 benign and malignant soft-tissue tumor entities defined. Not infrequently, these neoplasms exhibit overlapping clinicopathologic features posing significant challenges in rendering a definitive diagnosis and optimal therapy. Advances in cytogenetic and molecular science have led to the discovery of genetic events in soft-tissue tumors that have not only enriched our understanding of the underlying biology of these neoplasms but have also proven to be powerful diagnostic adjuncts and/or indicators of molecular targeted therapy. In particular, many soft-tissue tumors are characterized by recurrent chromosomal rearrangements that produce specific gene fusions. For pathologists, identification of these fusions as well as other characteristic mutational alterations aids in precise subclassification. This review will address known recurrent or tumor-specific genetic events in soft-tissue tumors and discuss the molecular approaches commonly used in clinical practice to identify them. Emphasis is placed on the role of molecular pathology in the management of soft-tissue tumors. Familiarity with these genetic events provides important ancillary testing for pathologists to include in their diagnostic armamentarium.
Collapse
Affiliation(s)
- Julia A Bridge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
139
|
Affiliation(s)
- Mark R Wick
- Departments of Pathology, University of Virginia Health System, Charlottesville, VA.
| | | | | |
Collapse
|
140
|
Gastrointestinal stromal tumors with KIT exon 9 mutations: Update on genotype-phenotype correlation and validation of a high-resolution melting assay for mutational testing. Am J Surg Pathol 2013; 37:1648-59. [PMID: 24061512 DOI: 10.1097/pas.0b013e3182986b88] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
KIT exon 9 mutations in gastrointestinal stromal tumors (GISTs) are highly relevant and have direct therapeutic implications. In this context, we established and validated a fast and sensitive high-resolution melting assay. Analyzing 126 primary and 18 metastatic KIT exon 9-mutated cases from our registry, we demonstrate that the mutational spectrum of exon 9 is broader than previously thought and describe 3 novel mutations. Including these cases and the common p.A502_Y503dup mutation, we provide a comprehensive list of all known KIT exon 9 mutations according to the Human Genome Variation Society nomenclature. Two of the newly described mutations were associated with an aggressive phenotype and tumor progression while being treated with 400 mg imatinib, indicating that also GIST with rare exon 9 mutations could be treated with increased imatinib dosage. On the basis of >1500 GISTs from our registry, we have determined the frequency of KIT exon 9 mutations to be 9.2% among all GISTs and 22.5% among small-bowel cases. We describe for the first time that nearly 20% of exon 9-mutated GIST occur in the stomach or rectum. Furthermore, we provide first evidence that exon 9-mutated GISTs metastasize significantly more often to the peritoneum than to the liver. Performing extensive statistical analyses on data from our registry and from the literature, we demonstrate that KIT exon 9 mutations are neither associated with intermediate-risk/high-risk status nor overrepresented among metastatic lesions. Thus, we conclude that exon 9 mutations per se do not have prognostic relevance.
Collapse
|
141
|
Abstract
The study of sarcoma pathology is a rapidly evolving field. The continued refinement of classic diagnostic techniques in conjunction with the molecular diagnostics has resulted in an abundance of data regarding this diverse and rare group of tumors. We anticipate that cutting edge technology including next generation sequencing will continue to further our understanding of saromagenesis and enable more precise classification and diagnosis of sarcomas in the future.
Collapse
|
142
|
Cheng JM, Tirumani SH, Shinagare AB, Jagannathan JP, Hornick JL, Raut CP, Ramaiya NH. MDCT of primary, locally recurrent, and metastatic duodenal gastrointestinal stromal tumours (GISTs): a single institution study of 25 patients with review of literature. Clin Radiol 2013; 69:137-44. [PMID: 24161459 DOI: 10.1016/j.crad.2013.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/22/2013] [Indexed: 12/12/2022]
Abstract
AIM To describe the multidetector computed tomography (MDCT) features of primary, locally recurrent, and metastatic duodenal gastrointestinal stromal tumours (GISTs). MATERIALS AND METHODS In this institutional review board-approved, Health Insurance Portability and Accountability Act of 1996 (HIPAA)-compliant, retrospective study, 25 patients [13 men, 12 women; mean age 56 years (34-74 years)] with histopathologically confirmed duodenal GISTs seen at Dana Farber Cancer Institute and Brigham and Women's Hospital from December 1999 to October 2009 were identified. The MDCT of primary tumours in six patients and follow-up imaging in all the 25 patients was reviewed by two radiologists in consensus. Electronic medical records were reviewed to document the clinical characteristics and management. RESULTS The mean size of the primary tumour was 3.7 cm (range 2.5-5.6 cm). Three of six primary tumours were in the second and third portions of the duodenum, one in the third portion, one in the third and fourth portions, and one in the fourth portion. Three of six of the tumours were exophytic, two were both exophytic and intraluminal, and one was intramural. The tumours were well-circumscribed, round or oval masses, with few lobulations, and were either homogeneously hyper-enhancing or heterogeneously isodense at MDCT. None of the tumours had necrosis, haemorrhage, calcification, or loco regional lymphadenopathy on imaging. Sixteen of 25 (64%) patients developed metastatic disease, the most common sites being liver (14/16; 87.5%) and peritoneum (5/16; 31%). CONCLUSION Duodenal GISTs are well-circumscribed, round or oval masses, and occur in the second through fourth portions of the duodenum, without lymphadenopathy or duodenal obstruction. Duodenal GISTS metastasize frequently to the liver and peritoneum.
Collapse
Affiliation(s)
- J M Cheng
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - S H Tirumani
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - A B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J P Jagannathan
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - C P Raut
- Department of Surgery, Division of Surgical Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - N H Ramaiya
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
143
|
Maes RK, Langohr IM, Wise AG, Smedley RC, Thaiwong T, Kiupel M. Beyond H&E: integration of nucleic acid-based analyses into diagnostic pathology. Vet Pathol 2013; 51:238-56. [PMID: 24129897 DOI: 10.1177/0300985813505878] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Veterinary pathology of infectious, particularly viral, and neoplastic diseases has advanced significantly with the advent of newer molecular methodologies that can detect nucleic acid of infectious agents within microscopic lesions, differentiate neoplastic from nonneoplastic cells, or determine the suitability of a targeted therapy by detecting specific mutations in certain cancers. Polymerase chain reaction-based amplification of DNA or RNA and in situ hybridization are currently the most commonly used methods for nucleic acid detection. In contrast, the main methodology used for protein detection within microscopic lesions is immunohistochemistry. Other methods that allow for analysis of nucleic acids within a particular cell type or individual cells, such as laser capture microdissection, are also available in some laboratories. This review gives an overview of the factors that influence the accurate analysis of nucleic acids in formalin-fixed tissues, as well as of different approaches to detect such targets.
Collapse
Affiliation(s)
- R K Maes
- College of Veterinary Medicine, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA. and
| | | | | | | | | | | |
Collapse
|
144
|
Yang J, Feng F, Li M, Sun L, Hong L, Cai L, Wang W, Xu G, Zhang H. Surgical resection should be taken into consideration for the treatment of small gastric gastrointestinal stromal tumors. World J Surg Oncol 2013; 11:273. [PMID: 24119564 PMCID: PMC3852122 DOI: 10.1186/1477-7819-11-273] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/27/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The National Comprehensive Cancer Network (NCCN) recommends conservative follow-up for gastric gastrointestinal stromal tumors (GISTs) less than 2 cm. The aim of the present study was to investigate the clinical and pathological features of small gastric GISTs, re-evaluate the risk potential, and discuss the treatment strategy of small gastric GISTs. METHODS In this retrospective study, 63 cases of small gastric GISTs (less than 2 cm) were resected surgically from May 2010 to March 2013 in our department. Clinicopathological factors were collected and the malignant potential of small gastric GISTs was analyzed. RESULTS The mitotic index of 14 out of 63 cases (22.22%) exceeded 5. The malignant potential of small gastric GISTs was related to tumor location (P = 0.0218). The mitotic index of 4 out of 8 GISTs (50%) located in gastric cardia exceeded 5, 8 out 28 GISTs (28.57%) located in the gastric fundus exceeded 5, and only 2 out of 27 GISTs (7.41%) located in the gastric body exceeded 5. We also discovered a good consistency between mitotic index and Ki-67 expression of small gastric GISTs. CONCLUSIONS Gastric GISTs less than 2 cm also have malignant potential. Thus, we recommended surgical resection of all small gastric GISTs once diagnosed.
Collapse
Affiliation(s)
- Jianjun Yang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, the Fourth Military Medical University, 127 West Changle Road, 710032, Xi'an, Shaanxi, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Small gastrointestinal stromal tumor in the stomach: identification of precursor for clinical gastrointestinal stromal tumor using c-kit and α-smooth muscle actin expression. Hum Pathol 2013; 44:2628-35. [PMID: 24119563 DOI: 10.1016/j.humpath.2013.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/16/2013] [Accepted: 07/17/2013] [Indexed: 01/09/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the digestive tract. To find precursors for clinical GISTs of the stomach, small gastric stromal tumors of less than 3 cm were collected and examined immunohistochemically with analysis of the KIT mutation. Sixty-eight of 74 lesions were classified into 4 representative groups according to the expression of c-kit and α-smooth muscle actin (αSMA): group A, c-kit diffusely positive and αSMA negative (18 cases); group B, c-kit diffusely positive and αSMA focally positive (13); group C, c-kit focally positive and αSMA diffusely positive (27); and group D, c-kit negative and αSMA diffusely positive (10). Of the 4 groups, groups A and B of c-kit diffuse expression showed higher cellularity and labeling indices of p27(Kip1) and Ki-67 than did groups C and D of diffuse αSMA expression. Incidence of KIT exon 11 mutation in groups A and B was 86% (25/29), whereas that in groups C and D was 0% (0/20). Small gastric stromal tumors with c-kit diffuse expression were considered precursors for clinical GIST because they were significantly different from c-kit focally positive or negative tumors. The mutation of KIT is considered as an early event in tumorigenesis of GIST.
Collapse
|
146
|
Kang YK, Yoo C, Ryoo BY, Lee JJ, Tan E, Park I, Park JH, Choi YJ, Jo J, Ryu JS, Ryu MH. Phase II study of dovitinib in patients with metastatic and/or unresectable gastrointestinal stromal tumours after failure of imatinib and sunitinib. Br J Cancer 2013; 109:2309-15. [PMID: 24084771 PMCID: PMC3817332 DOI: 10.1038/bjc.2013.594] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/27/2013] [Accepted: 09/09/2013] [Indexed: 12/24/2022] Open
Abstract
Background: This prospective, phase II trial evaluated the efficacy and safety of dovitinib in patients with metastatic and/or unresectable gastrointestinal stromal tumours (GISTs) after failure of at least imatinib and sunitinib. Methods: Patients received oral dovitinib, 500 mg once daily, for 5 consecutive days, followed by a 2-day rest, every 28 days. The primary endpoint was disease control rate (DCR; objective response+stable disease (SD)) at 24 weeks, assessed by computed tomography (CT) scan according to RECIST v1.0. Metabolic response was evaluated by positron emission tomography (PET)–CT scans performed at baseline and after 4 weeks of treatment. Results: Between September 2011 and April 2012, 30 patients were enroled. DCR at 24 weeks by RECIST v1.0 was 13% and one patient (3%) had a partial response. Based on the European Organization for Research and Treatment of Cancer PET response criteria, four patients (13%) had a metabolic partial response after 4 weeks of treatment. At a median follow-up of 8.3 months (range, 6.3–12.2 months), median progression-free survival (PFS) was 3.6 months (95% confidence interval (CI), 3.5–3.7 months) and median overall survival was 9.7 months (95% CI, 6.0–13.4 months). Metabolic progressive disease at Week 4 was significantly associated with shorter PFS (P=0.03). Grade 3/4 adverse events included asthenia (20%), neutropenia (13%), thrombocytopenia (10%), and hypertriglyceridaemia (10%). Most toxicities were manageable by dose modification. Conclusion: Dovitinib showed modest antitumour activity with manageable toxicities in heavily pretreated patients with advanced GISTs.
Collapse
Affiliation(s)
- Y-K Kang
- Department of Oncology and Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Wong NACS, Wingate J, Colling R. A study of α5 chain of collagen IV, caldesmon, placental alkaline phosphatase and smoothelin as immunohistochemical markers of gastrointestinal smooth muscle neoplasms. J Clin Pathol 2013; 67:105-11. [DOI: 10.1136/jclinpath-2013-201797] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
148
|
Cananzi FCM, Judson I, Lorenzi B, Benson C, Mudan S. Multidisciplinary care of gastrointestinal stromal tumour: a review and a proposal for a pre-treatment classification. Eur J Surg Oncol 2013; 39:1171-8. [PMID: 24063969 DOI: 10.1016/j.ejso.2013.08.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 07/31/2013] [Accepted: 08/28/2013] [Indexed: 12/15/2022] Open
Abstract
The introduction of receptor tyrosine kinase inhibitors (TKIs) has revolutionized the management of gastrointestinal stromal tumour (GIST). Strong evidence supports the use of imatinib as first-line treatment in metastatic or unresectable tumours and its efficacy in the post-operative adjuvant setting has been confirmed by phase III trials. There are a number of reports concerning the administration of imatinib in the pre-operative setting, however, the heterogeneity of the terminology used and the indications for pre-operative treatment make it difficult to determine the true value of pre-operative imatinib. Larger studies, or a phase III trial could be helpful but patient accrual and standardization of care could be difficult. We propose a pre-treatment classification of GIST in order to facilitate the comparison and collection of data from different institutions, and overcome the difficulties related to accrual. Moreover, in the current era of multidisciplinary treatment of GIST, an appropriate classification is mandatory to properly design clinical trials and plan stage-adapted treatment.
Collapse
Affiliation(s)
- F C M Cananzi
- Department of Surgery, The Royal Marsden, Fulham Road, London SW3 6JJ, UK.
| | | | | | | | | |
Collapse
|
149
|
Wang C, Jin MS, Zou YB, Gao JN, Li XB, Peng F, Wang HY, Wu ZD, Wang YP, Duan XM. Diagnostic significance of DOG-1 and PKC-θ expression and c-Kit/PDGFRA mutations in gastrointestinal stromal tumours. Scand J Gastroenterol 2013; 48:1055-65. [PMID: 23862765 DOI: 10.3109/00365521.2013.816770] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To investigate discovered on gastrointestinal stromal tumor (GIST)-1 (DOG-1) and protein kinase C-θ (PKC-θ) expression in a series of GISTs and determine the sensitivity, specificity, and diagnostic value of these two antigens. METHODS Immnunohistochemistry (IHC) was used to detect CD117, DOG-1, PKC-θ, CD34, Ki-67, α-smooth muscle actin (SMA), S100, and Desmin expression in 147 GISTs and 51 non-GISTs. c-Kit gene (exons 9, 11, 13, and 17) and platelet-derived growth factor receptor-alpha (PDGFRA) gene (exons 12 and 18) mutations were also detected. RESULTS About 94.5% GISTs were CD117 positive, 96% were DOG-1 positive, and 90.5% were PKC-θ positive. DOG-1 had a specificity of 100%, while CD117 and PKC-θ had a specificity of 90% and 80%, respectively. There was no significant difference between DOG-1 and PKC-θ expressions when compared to CD117 expression. In 30 out of 42 (71.5%) GISTs, a c-Kit gene mutation was found, and in 3 out of 42 cases (7%), PDGFRA was mutated. Wild-type c-Kit/PDGFRA genes accounted for 21.5% (9/42). Most c-Kit gene mutations were found to be located at exon 11, mainly as in-frame deletions. Mutations in exon 9 were all missense mutations. Most PDGFRA gene mutations were found in exon 18, codon 842. c-Kit gene mutations in exons 13 and 17, and the PDGFRA gene mutation in exon 12 were not detected. CONCLUSIONS Compared to CD117, DOG-1 is a biomarker with higher sensitivity and specificity. The combination of CD117 and DOG-1 can be used to improve the diagnosis of GIST. Although PKC-θ has a lower specificity than DOG-1, it can be a useful biomarker, especially in CD117(-) and/or DOG-1(-) cases.
Collapse
Affiliation(s)
- Chao Wang
- Pathological Diagnosis Center, First Hospital of Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Abstract
Gastrointestinal stromal tumors (GISTs) represent a rare group of neoplasms of the digestive tract deriving from the mesenchyme. Giant GISTs (over 10 cm in diameter) represent only 20 % of all cases and are associated with a high risk of malignancy. We present the case of a giant GIST of the jejunum successfully treated by surgical resection and adjuvant therapy with imatinib.
Collapse
|