51
|
Varshney VK, Gupta RK, Saluja SS, Tyagi I, Mishra PK, Batra VV. Analysis of clinicopathological and immunohistochemical parameters and correlation of outcomes in gastrointestinal stromal tumors. Indian J Cancer 2019; 56:135-143. [PMID: 31062732 DOI: 10.4103/ijc.ijc_352_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. We analyzed the clinicopathological features, resectability, immunohistochemical markers, and various factors predictive of disease recurrence and survival. MATERIALS AND METHODS Retrospective analysis of prospectively maintained database of GIST patients managed from 2005 to 2016 was done. Size, site, malignant potential, nuclear pleomorphism, histopathological variety, immunohistochemical markers, type of surgery, and adjuvant imatinib therapy were analyzed. RESULTS Ninety-two patients with GIST were analyzed. Immunohistochemistry showed positivity for c-kit (82.4%), DOG1 (75%), and PDGFR-α (79%). Among 16 patients with c-kit-negative tumors, 10 patients were positive for DOG1, PDGFR-α, or both. The most common primary site was stomach (44, 47.8%) followed by small bowel (17, 18.5%) and duodenum (14, 15.2%). Of 92 patients, 80 (87%) underwent R0 resection with organ sparing resection in 56 (70%) patients. Seventeen (21.3%) patients showed recurrence at a median follow-up of 6 years. Median and 5-year overall survival (OS) was 36 months (12-120) and 75%, respectively, and 5-year RFS was 81.8%. On univariate analysis, size, mitotic activity, malignant potential, and nuclear pleomorphism were predictors of recurrence. However, on multivariate analysis, only nuclear pleomorphism was significant. CONCLUSIONS GISTs had a wide spectrum of presentation, and immunohistopathological features with organ sparing resection were conceivable in maximum. Nuclear pleomorphism may be considered as an important variable to predict recurrence in addition to malignant potential of tumors.
Collapse
Affiliation(s)
- Vaibhav Kumar Varshney
- Department of Gastrointestinal Surgery, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| | - Rakesh Kumar Gupta
- Department of Pathology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| | - Sundeep Singh Saluja
- Department of Gastrointestinal Surgery, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| | - Ila Tyagi
- Department of Pathology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| | - Pramod Kumar Mishra
- Department of Gastrointestinal Surgery, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| | - Vineeta V Batra
- Department of Pathology, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research, New Delhi, India
| |
Collapse
|
52
|
Ge XY, Lei LW, Ge F, Jiang X. Analysis of risk factors of gastrointestinal stromal tumors in different age groups based on SEER database. Scand J Gastroenterol 2019; 54:480-484. [PMID: 31017491 DOI: 10.1080/00365521.2019.1604798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective: To investigate the risk factors affecting the survival of patients with gastrointestinal stromal tumors (GISTs) in different age groups. Methods: Information on 6089 GIST patients was screened from the Surveillance, Epidemiology, and End Results (SEER) database. Risk factor analysis was performed using a chi-square test (univariate analysis). Survival analysis was performed using the Kaplan-Meier method (log-rank test) and the COX proportional hazard model. p Value < .05 was considered statistically significant. Results: Analyzed statistically to reveal that in addition to tumor size, mitotic index, and primary location, age, gender, race, and surgical treatment also were independent risk factors for GISTs. Gender, race, and location of disease influenced the survival rate of patients, which was higher in the young group (≤60 years old) than the elderly group (>60 years). Risk factors such as primary location, tumor diameter, and mitotic index varied significantly between the different age groups. Conclusions: Age, gender, race, and surgical treatment are independent risk factors that influence the prognosis in patients with GISTs. Some risk factors affecting prognosis are age dependent.
Collapse
Affiliation(s)
- Xin-Yu Ge
- a Department of General Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Li-Wang Lei
- a Department of General Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Fei Ge
- a Department of General Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , PR China
| | - Xian Jiang
- a Department of General Surgery , The First Affiliated Hospital of Harbin Medical University , Harbin , PR China
| |
Collapse
|
53
|
Gastrointestinal stromal tumors (GISTs) with remarkable cystic change: a specific subtype of GISTs with relatively indolent behaviors and favorable prognoses. J Cancer Res Clin Oncol 2019; 145:1559-1568. [PMID: 30923944 PMCID: PMC6527525 DOI: 10.1007/s00432-019-02853-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/24/2019] [Indexed: 12/26/2022]
Abstract
Purpose Gastrointestinal stromal tumors (GISTs) are typically solid neoplasms with small cystic change detected occasionally but in rare instances may present predominantly as cystic lesions. The histopathologic features and prognoses of cystic GISTs (cGISTs) are poorly understood. Methods We herein reviewed 20 cGISTs resected or consulted in our institution from January 1, 2003 to December 31, 2014. Results Of the 20 patients included, the mean age was 61 years and the male-to-female ratio was 9:11. The original locations were the stomach (n = 10, 50%), the small intestine (n = 9, 45%) and the omentum (n = 1, 5%). Indistinct diagnosis or misdiagnosis was established in 15 cases based only on preoperative radiology. Grossly, the cystic component made up the bulk of masses and was filled by dark bloody fluid and necrotic debris in 18 cases. Microscopically, cyst wall was composed of neoplastic spindle (n = 14, 70%)/epithelioid cells (n = 6, 30%) and collagenous fiber, with necrotic debris and granulation tissue lining on the inner surface. cGISTs resembled their solid counterparts in terms of morphology and immunohistology but demonstrated fewer malignant parameters. c-kit or PDGFRα mutations were detected in eleven cases with the remaining being wild type for these two mutations. Although classified as intermediate or high (3 and 17, respectively) risk of recurrence according to modified National Institute of Health criterion, most patients with cGISTs experienced long-term recurrence-free survival without adjuvant imatinib. Conclusions Cystic GISTs is a relatively indolent subset of GISTs with favorable prognoses and adjuvant imatinib should be a prudent consideration.
Collapse
|
54
|
Heinrich MC, Patterson J, Beadling C, Wang Y, Debiec-Rychter M, Dewaele B, Corless CL, Duensing A, Raut CP, Rubin B, Ordog T, van de Rijn M, Call J, Mühlenberg T, Fletcher JA, Bauer S. Genomic aberrations in cell cycle genes predict progression of KIT-mutant gastrointestinal stromal tumors (GISTs). Clin Sarcoma Res 2019; 9:3. [PMID: 30867899 PMCID: PMC6399846 DOI: 10.1186/s13569-019-0112-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Activating mutations of the receptor tyrosine kinase KIT are early events in the development of most gastrointestinal stromal tumors (GISTs). Although GISTs generally remain dependent on oncogenic KIT during tumor progression, KIT mutations alone are insufficient to induce malignant behavior. This is evidenced by KIT-mutant micro-GISTs, which are present in up to one-third of normal individuals, but virtually never progress to malignancy. METHODS We performed whole exome sequencing on 29 tumors obtained from 21 patients with high grade or metastatic KIT-mutant GIST (discovery set). We further validated the frequency and potential prognostic significance of aberrations in CDKN2A/B, RB1, and TP53 in an independent series of 71 patients with primary GIST (validation set). RESULTS Using whole exome sequencing we found significant enrichment of genomic aberrations in cell cycle-associated genes (Fisher's Exact p = 0.001), most commonly affecting CDKN2A/B, RB1, and TP53 in our discovery set. We found a low mutational tumor burden in these 29 advanced GIST samples, a finding with significant implications for the development of immunotherapy for GIST. In addition, we found mutation of spliceosome genes in a minority of cases, implicating dysregulation of splicing as a potential cancer promoting mechanism in GIST. We next assessed the prognostic significance of CDKN2A, RB1 or TP53 mutation/copy loss in an independent cohort of 71 patients with primary GIST. Genetic events (mutation, deletion, and/or LOH) involving at least one of the three genes examined were found in 17% of the very low-risk, 36% of the low-risk, 42% of the intermediate risk, 67% of the high-risk/low mitotic-count, and in 86% of the high-risk/high mitotic-count group. The presence of cell cycle-related events was associated with a significantly shorter relapse-free survival (median 67 months versus not reached; p < 0.0001) and overall survival (Log Rank, p = 0.042). CONCLUSION Our results demonstrate that genomic events targeting cell cycle-related genes are associated with GIST progression to malignant disease. Based on this data, we propose a model for molecular pathogenesis of malignant GIST.
Collapse
Affiliation(s)
- Michael C. Heinrich
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, 3710 SW U.S. Veterans Hospital Road, R&D 19, Portland, OR 97239 USA
| | - Janice Patterson
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, 3710 SW U.S. Veterans Hospital Road, R&D 19, Portland, OR 97239 USA
| | - Carol Beadling
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, 3710 SW U.S. Veterans Hospital Road, R&D 19, Portland, OR 97239 USA
| | - Yuexiang Wang
- Department of Pathology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Maria Debiec-Rychter
- Department of Human Genetics, Katholieke Universiteit Leuven and University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Barbara Dewaele
- Department of Human Genetics, Katholieke Universiteit Leuven and University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Christopher L. Corless
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, 3710 SW U.S. Veterans Hospital Road, R&D 19, Portland, OR 97239 USA
| | - Anette Duensing
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Chandrajit P. Raut
- Department of Surgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA
| | - Brian Rubin
- Department of Molecular Genetics, Cleveland Clinic and Lerner Research Institute, L25, 9500 Euclid Avenue, Cleveland, OH 44195 USA
| | - Tamas Ordog
- Department of Physiology and Biomedical Engineering, Division of Gastroenterology and Hepatology and Center for Individualized Medicine, Mayo Clinic, 200 1st Street SW, Rochester, MN USA
| | - Matt van de Rijn
- Department of Pathology, Stanford University Medical Center, 300 Pasteur Dr., Stanford, CA 94305 USA
| | - Jerry Call
- The Life Raft Group, 155 Route 46 West, Suite 202, Wayne, NJ 07470 USA
| | - Thomas Mühlenberg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
- Germany and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
55
|
Remarkable effects of imatinib in a family with young onset gastrointestinal stromal tumors and cutaneous hyperpigmentation associated with a germline KIT-Trp557Arg mutation: case report and literature overview. Fam Cancer 2019; 17:247-253. [PMID: 28710566 DOI: 10.1007/s10689-017-0024-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) occur mostly sporadically. GISTs associated with a familial syndrome are very rare and are mostly wild type for KIT and platelet-derived growth factor alpha (PDGFRA). To date 35 kindreds and 8 individuals have been described with GISTs associated with germline KIT mutations. This is the third family described with a germline p.Trp557Arg mutation in exon 11 of the KIT gene. The effect of imatinib in patients harboring a germline KIT mutation has been rarely described. Moreover, in some studies imatinib treatment was withheld considering the lack of evidence for efficacy of this treatment in GIST patients harboring a germline KIT mutation. This paper describes a 52-year old patient with a de novo germline p.Trp557Arg mutation with multiple GISTs throughout the gastrointestinal tract and cutaneous hyperpigmentation. Imatinib treatment showed long-term regression of the GISTs and evident pathological response was seen after resection. Remarkably, the hyperpigmentation of the skin also diminished during imatinib treatment. Genetic screening of the family revealed the same mutation in two daughters, both with similar cutaneous hyperpigmentation. One daughter, aged 23, was diagnosed with multiple small intestine GISTs, which were resected. She was treated with adjuvant imatinib which prompted rapid regression of the cutaneous hyperpigmentation. Imatinib treatment in GIST patients harboring a germline KIT mutation shows favorable and long-term responses in both the tumor and the phenotypical hyperpigmentation.
Collapse
|
56
|
Berger EP, Johannes CM, Jergens AE, Allenspach K, Powers BE, Du Y, Mochel JP, Fox LE, Musser ML. Retrospective evaluation of toceranib phosphate (Palladia®) use in the treatment of gastrointestinal stromal tumors of dogs. J Vet Intern Med 2018; 32:2045-2053. [PMID: 30307656 PMCID: PMC6271363 DOI: 10.1111/jvim.15335] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/24/2018] [Accepted: 09/05/2018] [Indexed: 12/20/2022] Open
Abstract
Background Gastrointestinal stromal tumors (GISTs) are uncommon intestinal neoplasms in the dog. Literature regarding adjunctive therapy for GISTs in dogs is sparse. High‐risk GISTs in humans respond to tyrosine kinase inhibition in the adjuvant setting. Objectives To review cases of toceranib phosphate use in dogs with GISTs and provide initial assessment of possible biological activity. A secondary aim was to evaluate patient and tumor characteristics for possible prognostic value. Animals Twenty‐seven dogs with confirmed GISTs based on histopathology and immunohistochemistry treated with toceranib. Methods Retrospective study in which cases of toceranib use in dogs with GIST were solicited using the American College of Veterinary Internal Medicine Oncology and Small Animal Internal Medicine listservs. Results Five of 7 dogs with gross disease experienced clinical benefit (71%; 3 complete responses, 1 partial response, 1 stable disease). These included 2 dogs with durable responses after toceranib discontinuation. Median progression‐free interval (PFI) in dogs with gross disease was 110 weeks (range, 36‐155 weeks). Median PFI in dogs with microscopic disease was 67 weeks (range, 9‐257 weeks). Metastasis at diagnosis (P = 0.04) and high mitotic index (P < 0.001) were associated with shorter PFI in toceranib‐treated dogs. Conclusions and Clinical Importance Biological activity of toceranib is evident in dogs with gross disease. Metastasis of GIST at diagnosis, as well as high tumor mitotic index, was associated with shorter PFI in toceranib‐treated dogs. Larger studies are needed to define postsurgical risk and refine the use of toceranib in dogs with gross and microscopic GIST.
Collapse
Affiliation(s)
- Erika P Berger
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Chad M Johannes
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Albert E Jergens
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Karin Allenspach
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Barbara E Powers
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, Colorado
| | - Yingzhou Du
- Department of Statistics, Iowa State University, Ames, Iowa
| | | | - Leslie E Fox
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Margaret L Musser
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| |
Collapse
|
57
|
You YH, Zhang Y. Primary prostatic extragastrointestinal stromal tumor: a case report and literature review. J Int Med Res 2018; 46:4343-4349. [PMID: 30152251 PMCID: PMC6166354 DOI: 10.1177/0300060518791677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective This study was performed to discuss the characteristics, diagnosis, and
treatment of primary prostatic extragastrointestinal stromal tumor
(EGIST). Methods The case history data of a patient with an EGIST were analyzed and discussed
with a literature review. Results The patient was diagnosed with a pelvic tumor, possibly malignant. We
ascertained the diagnosis by exploratory surgery and pathological biopsy.
The tumor was present in the prostate and infiltrated and pressed against
the anterior rectal wall. Pathological biopsy showed that the tumor
comprised spindle cells, which were also present at the junction of the
tumor and prostate tissue. Immunohistochemically, the tumor cells were
positive for CD117, DOG-1, CD34, and smooth muscle actin and negative for
S100 and desmin; Ki-67LI was about 10%. These results support the diagnosis
of primary prostatic EGIST. Conclusion The rarity and nonspecific clinical manifestation of prostatic EGIST
facilitate misdiagnosis. Diagnosis mainly depends on imaging examination and
characteristic histopathological and immunohistochemical features, and GIST
must be excluded. Surgery is the main treatment method, and imatinib is
suggested for unresectable and malignant EGISTs.
Collapse
Affiliation(s)
- Yong-Hao You
- 1 Department of Surgery, the First Clinical Medical College of Yangtze University, Jingzhou, Hubei Province, China.,2 Department of Surgery, the First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Yi Zhang
- 3 Department of Urology, Huangpi District People's Hospital, Wuhan, Hubei Province, China
| |
Collapse
|
58
|
Ricci R, Giustiniani MC, Gessi M, Lanza P, Castri F, Biondi A, Persiani R, Vecchio FM, Risio M. Telocytes are the physiological counterpart of inflammatory fibroid polyps and PDGFRA-mutant GISTs. J Cell Mol Med 2018; 22:4856-4862. [PMID: 30117724 PMCID: PMC6156396 DOI: 10.1111/jcmm.13748] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/29/2018] [Indexed: 12/23/2022] Open
Abstract
PDGFRA mutations in the gastrointestinal (GI) tract can cause GI stromal tumour (GIST) and inflammatory fibroid polyp (IFP). Hitherto no cell type has been identified as a physiological counterpart of the latter, while interstitial Cajal cells (ICC) are considered the precursor of the former. However, ICC hyperplasia (ICCH), which strongly supports the ICC role in GIST pathogenesis, has been identified in germline KIT-mutant settings but not in PDGFRA-mutant ones, challenging the precursor role of ICC for PDGFRA-driven GISTs. Telocytes are a recently described interstitial cell type, CD34+/PDGFRA+. Formerly considered fibroblasts, they are found in many organs, including the GI tract where they are thought to be involved in neurotransmission. Alongside IFPs and gastric GISTs, GI wall "fibrosis" has been reported in germline PDGFRA-mutants. Taking the opportunity offered by its presence in a germline PDGFRA-mutant individual, we demonstrate that this lesion is sustained by hyperplastic telocytes, constituting the PDGFRA-mutant counterpart of germline KIT mutation-associated ICCH. Moreover, our findings support a pathogenetic relationship between telocyte hyperplasia and both IFPs and PDGFRA-mutant GISTs. We propose the term "telocytoma" for defining IFP, as it conveys both the pathogenetic (neoplastic) and histotypic ("telocytary") essence of this tumour, unlike IFP, which rather evokes an inflammatory-hyperplastic lesion.
Collapse
Affiliation(s)
- Riccardo Ricci
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Maria Cristina Giustiniani
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Marco Gessi
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Paola Lanza
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Federica Castri
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Alberto Biondi
- Department of Surgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Roberto Persiani
- Department of Surgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Fabio M Vecchio
- Department of Pathology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Mauro Risio
- Department of Pathology, Emeritus, IRCC, Candiolo, Italy
| |
Collapse
|
59
|
Roelofs KA, Medlicott S, Henning JW, Weis E. Gastrointestinal Stromal Tumor Metastasis to the Orbit. Ophthalmic Plast Reconstr Surg 2018; 34:e131-e133. [DOI: 10.1097/iop.0000000000001144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
60
|
Mavroeidis L, Metaxa-Mariatou V, Papoudou-Bai A, Lampraki AM, Kostadima L, Tsinokou I, Zarkavelis G, Papadaki A, Petrakis D, Gκoura S, Kampletsas E, Nasioulas G, Batistatou A, Pentheroudakis G. Comprehensive molecular screening by next generation sequencing reveals a distinctive mutational profile of KIT/ PDGFRA genes and novel genomic alterations: results from a 20-year cohort of patients with GIST from north-western Greece. ESMO Open 2018; 3:e000335. [PMID: 29636989 PMCID: PMC5890860 DOI: 10.1136/esmoopen-2018-000335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction Gastrointestinal stromal tumours (GIST) are mesenchymal neoplasms that usually carry an activating mutation in KIT or platelet-derived growth factor receptor alpha (PDGFRA) genes with predictive and prognostic significance. We investigated the extended mutational status of GIST in a patient population of north-western Greece in order to look at geopraphic/genotypic distinctive traits. Patient and methods Clinicopathological and molecular data of 38 patients diagnosed from 1996 to 2016 with GIST in the region of Epirus in Greece were retrospectively assessed. Formalin-fixed paraffin-embedded tumours were successfully analysed for mutations in 54 genes with oncogenic potential. Next generation sequencing was conducted by using the Ion AmpliSeqCancer Hotspot Panel V.2 for DNA analysis (Thermofisher Scientific). Results Among 38 tumours, 24 (63.16%) and seven (18.42%) of the tumours harboured mutations in the KIT and PDGFRA genes, respectively, while seven (18.42%) tumours were negative for either KIT or PDGFRA mutation. No mutations were detected in five (13.16%) cases. Concomitant mutations of BRAF and fibroblast growth factor receptor 3 (FGFR3) genes were observed in two patients with KIT gene mutation. Two patients with KIT/PDGFRA wild-type GIST had mutations in either KRAS or phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) genes. There was no significant survival difference regarding the exonic site of mutation in either KIT or PDGFRA gene. The presence of a mutation in pathway effectors downstream of KIT or PDGFRA, such as BRAF, KRAS or PIK3CA, was associated with poor prognosis. Adverse prognosticators were also high mitotic index and the advanced disease status at diagnosis. Conclusions We report comparable incidence of KIT and PDGFRA mutation in patients with GIST from north-western Greece as compared with cohorts from other regions. Interestingly, we identified rare mutations on RAS, BRAF and PIK3CA genes in patients with poor prognosis.
Collapse
Affiliation(s)
- Leonidas Mavroeidis
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | | | | | | | - Lida Kostadima
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Ilias Tsinokou
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Alexandra Papadaki
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Dimitrios Petrakis
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Stefania Gκoura
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - Eleftherios Kampletsas
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | | | - Anna Batistatou
- Department of Pathology, School of Medicine, Ioannina, Greece
| | - George Pentheroudakis
- Department of Medical Oncology, School of Medicine, Ioannina, Greece.,Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| |
Collapse
|
61
|
Zhu R, Liu F, Grisotti G, Pérez-Irizarry J, Cha CH, Johnson CH, Boffa DJ, Han D, Johung KL, Zhang Y, Khan SA. Distinctive features of gastrointestinal stromal tumors arising from the colon and rectum. J Gastrointest Oncol 2018; 9:231-240. [PMID: 29755761 DOI: 10.21037/jgo.2017.11.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Colon and rectal gastrointestinal stromal tumors (GISTs) are rare and poorly characterized. Because the majority of treatment guidelines for GISTs are extrapolated from tumors of gastric and small bowel origin, our aim was to better characterize the unique clinicopathologic features and prognostic factors of colon and rectal GISTs to guide clinical care. Methods The National Cancer Data Base (NCDB) was queried from 2006 to 2013 for cases of GISTs in the stomach, colon, and rectum. Patient demographics, clinical characteristics, and survival were compared. Results A total of 11,302 gastric GISTs were compared to 398 colon and 393 rectal GISTs. After propensity matching, compared to gastric GISTs, rectal GISTs had improved overall survival (HR =0.695, P=0.0264), while colon GISTs had worse overall survival (HR =1.6, P=0.0005). Surgical treatment for rectal GISTs was more likely to be local excision compared to colonic GISTs (51.1% vs. 8.4%, P<0.0001). Colon and gastric GISTs were less likely to receive systemic therapy compared to rectal GISTs (34.2% vs. 34.0% vs. 55.2%, P<0.0001). Adjuvant systemic therapy conveyed a survival advantage to rectal GISTs (HR =0.47, P=0.042) but not colon GISTs. There was a negative impact of adjuvant therapy on survival for colon GISTs <5 cm (HR =3.41, P=0.032). Conclusions Patients with rectal GISTs live longer than those with colon and gastric GISTs, and adjuvant therapy prolongs their survival. Many patients with colon GISTs are treated with adjuvant therapy despite a detrimental effect on survival. Tumor biology of colon and rectal GISTs needs to be better studied to tailor treatment.
Collapse
Affiliation(s)
- Rebecca Zhu
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Fangfang Liu
- Department of Liver Failure Treatment and Research Center, Beijing 302 Hospital, Beijing 100039, China
| | - Gabriella Grisotti
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | | | - Charles H Cha
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT, USA
| | - Daniel J Boffa
- Department of Surgery, Section of Thoracic Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Dale Han
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Kimberly L Johung
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yawei Zhang
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA.,Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT, USA
| | - Sajid A Khan
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
62
|
Büttner J, Jöhrens K, Klauschen F, Hummel M, Lenze D, Saeger W, Lehmann A. Intratumoral morphological heterogeneity can be an indicator of genetic heterogeneity in colorectal cancer. Exp Mol Pathol 2018; 104:76-81. [DOI: 10.1016/j.yexmp.2018.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/10/2018] [Indexed: 12/24/2022]
|
63
|
Prognostic significance of neutrophil to lymphocyte ratio in patients with gastrointestinal stromal tumors: A meta-analysis. Clin Chim Acta 2018; 477:7-12. [DOI: 10.1016/j.cca.2017.11.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 12/18/2022]
|
64
|
The Effect of Marital Status on Survival of Patients with Gastrointestinal Stromal Tumors: A SEER Database Analysis. Gastroenterol Res Pract 2018; 2018:5740823. [PMID: 29483926 PMCID: PMC5816869 DOI: 10.1155/2018/5740823] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 12/10/2017] [Indexed: 02/06/2023] Open
Abstract
Background Marital status has been reported to be a prognostic factor in multiple malignancies. However, its prognostic value on gastrointestinal stromal tumors (GISTs) have not yet been determined. The objective of the present analysis was to assess the effects of marital status on survival in patients with GISTs. Methods The Surveillance, Epidemiology, and End Results (SEER) database was used to analyze 6195 patients who were diagnosed with GISTs from 2001 to 2014. We also use Kaplan-Meier analysis and Cox regression to analyze the impact of marital status on cancer-specific survival (CSS). Results Patients in the married group had more frequency in white people, more high/moderate grade tumors, and were more likely to receive surgery. Widowed patients had a higher proportion of women, a greater proportion of older patients (>60 years), and more common site of the stomach. Multivariate analysis demonstrated that marital status was an independent prognostic factor for GISTs (P < 0.001). Married patients had better CSS than unmarried patients (P < 0.001). Subgroup analysis suggested that widowed patients had the lowest CSS compared with all other patients. Conclusions Marital status is a prognostic factor for survival in patients with GISTs, and widowed patients are at greater risk of cancer-specific mortality.
Collapse
|
65
|
Neurofibromin C terminus-specific antibody (clone NFC) is a valuable tool for the identification of NF1-inactivated GISTs. Mod Pathol 2018; 31:160-168. [PMID: 28862263 DOI: 10.1038/modpathol.2017.105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 12/13/2022]
Abstract
An increasing body of evidence supports the involvement of NF1 mutations, constitutional or somatic, in the pathogenesis of gastrointestinal stromal tumors (GISTs). Due to the large size of the NF1 locus, the existence of multiple pseudogenes and the wide spectrum of mechanisms of gene inactivation, the analysis of NF1 gene status is still challenging for most laboratories. Here we sought to assess the efficacy of a recently developed neurofibromin-specific antibody (NFC) in detecting NF1-inactivated GISTs. NFC reactivity was analyzed in a series of 98 GISTs. Of these, 29 were 'NF1-associated' (17 with ascertained NF1 mutations and 12 arising in the context of clinically diagnosed Neurofibromatosis type 1 syndrome and thus considered bona fine NF1 inactivated); 38 were 'NF1-unrelated' (either wild-type or carrying non-pathogenic variants of NF1). Thirty-one additional GISTs with no available information on NF1 gene status or with NF1 gene variants of uncertain pathogenic significance were also included in the analysis. Cases were scored as NFC negative when, in the presence of NFC positive internal controls, no cytoplasmic staining was detected in the neoplastic cells. NFC immunoreactivity was lost in 24/29 (83%) NF1-associated GISTs as opposed to only 2/38 (5%) NF1-unrelated GISTs (P=3e-11). NFC staining loss significantly correlated (P=0.007) with the presence of biallelic NF1 inactivation, due essentially to large deletions or truncating mutations. NFC reactivity was instead retained in two cases in which the NF1 alteration was heterozygous and in one case where the pathogenic NF1 variant, although homo/hemizygous, was a missense mutation predicted not to affect neurofibromin half-life. Overall this study provides evidence that NFC is a valuable tool for identifying NF1-inactivated GISTs, thus serving as a surrogate for molecular analysis.
Collapse
|
66
|
Charville GW, Longacre TA. Surgical Pathology of Gastrointestinal Stromal Tumors: Practical Implications of Morphologic and Molecular Heterogeneity for Precision Medicine. Adv Anat Pathol 2017; 24:336-353. [PMID: 28820749 DOI: 10.1097/pap.0000000000000166] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastrointestinal stromal tumor (GIST), the most common mesenchymal neoplasm of the gastrointestinal tract, exhibits diverse histologic and clinical manifestations. With its putative origin in the gastrointestinal pacemaker cell of Cajal, GIST can arise in association with any portion of the tubular gastrointestinal tract. Morphologically, GISTs are classified as spindled or epithelioid, though each of these subtypes encompasses a broad spectrum of microscopic appearances, many of which mimic other histologic entities. Despite this morphologic ambiguity, the diagnosis of GIST is aided in many cases by immunohistochemical detection of KIT (CD117) or DOG1 expression. The natural history of GIST ranges from that of a tumor cured by surgical resection to that of a locally advanced or even widely metastatic, and ultimately fatal, disease. This clinicopathologic heterogeneity is paralleled by an underlying molecular diversity: the majority of GISTs are associated with spontaneous activating mutations in KIT, PDGFRA, or BRAF, while additional subsets are driven by genetic lesions-often inherited-of NF1 or components of the succinate dehydrogenase enzymatic complex. Specific gene mutations correlate with particular anatomic or morphologic characteristics and, in turn, with distinct clinical behaviors. Therefore, prognostication and treatment are increasingly dictated not only by morphologic clues, but also by accompanying molecular genetic features. In this review, we provide a comprehensive description of the heterogenous molecular underpinnings of GIST, including implications for the practicing pathologist with regard to morphologic identification, immunohistochemical diagnosis, and clinical management.
Collapse
|
67
|
Phase IV Study of Sunitinib in Chinese Patients with Imatinib-Resistant or Imatinib-Intolerant Gastrointestinal Stromal Tumors. Oncol Ther 2017. [DOI: 10.1007/s40487-017-0052-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Abstract
Introduction
Sunitinib is approved in China for treatment of gastrointestinal stromal tumors (GIST), after disease progression on, or intolerance to, imatinib. However, available data from prospective clinical trials on its efficacy and safety in Chinese patients is limited. Our objective is to determine the efficacy and safety of sunitinib in Chinese patients with imatinib-resistant/intolerant GIST.
Methods
An open-label, single-arm, multicenter, phase IV study was performed in Chinese patients with imatinib-resistant/intolerant GIST. Sunitinib was administered orally in 6-week cycles of 4 weeks on-treatment (50 mg once daily) and 2 weeks off-treatment. The primary endpoint was progression-free survival (PFS). Tumors were assessed every 6 weeks for the first 24 weeks and every 12 weeks thereafter. Responses were evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0.
Results
A total of 60 patients were enrolled, of whom 59 were treated with sunitinib. All patients were Asian, and mean age was 55.1 years. Median PFS was 46.4 weeks (95% CI 33.6–53.1). An objective response (complete or partial) was observed in 11/58 (19%) patients. Median overall survival was 111.3 weeks (95% CI 75.4–167.1), median time to tumor progression was 47.3 weeks (95% CI 34.1–59.3), and median time to tumor response was 22.6 weeks (95% CI 10.4–57.3). The most common adverse events included leukopenia, fatigue, hand-foot syndrome, and neutropenia. No new safety concerns were identified.
Conclusions
This study confirms that sunitinib is active and well tolerated in Chinese patients with imatinib-resistant/intolerant GIST.
ClinicalTrials.gov identifier
NCT00793871.
Funding
Pfizer Inc, USA.
Collapse
|
68
|
Kang G, Kang Y, Kim KH, Ha SY, Kim JY, Shim YM, Heinrich MC, Kim KM, Corless CL. Gastrointestinal stromal tumours of the oesophagus: a clinicopathological and molecular analysis of 27 cases. Histopathology 2017. [PMID: 28644569 DOI: 10.1111/his.13292] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIMS Gastrointestinal stromal tumours (GISTs) may arise anywhere in the gastrointestinal tract, but are rare in the oesophagus. We describe the clinical, pathological and molecular characteristics of 27 primary oesophageal GISTs, the largest series to date. METHODS AND RESULTS DNA was extracted and exons 9, 11, 13 and 17 of KIT, exons 12, 14 and 18 of PDGFRA and exon 15 of BRAF were amplified and sequenced. Oesophageal GISTs occurred in 14 men and 13 women aged between 22 and 80 years (mean: 56 years). All 27 cases were immunohistochemically positive for KIT, and 92 and 47% co-expressed CD34 or smooth muscle actin, respectively. Fifteen (71% of analysed cases) harboured KIT exon 11 mutations and one case each had a mutation in KIT exon 13 (K642E) or BRAF exon 15 (V600E). Long-term follow-up data (median, 96.5 months) were obtained for 20 cases; two patients had metastases at presentation and seven had developed local recurrence and/or metastasis after surgery. A large tumour size (≥ 10 cm), high mitotic rate (> 5/5 mm2 ), presence of a deletion mutation in KIT exon 11 involving codons 557-558 and a positive microscopic margin were associated with recurrence and metastasis. The KIT mutations identified in oesophageal GISTs are similar to those observed in gastric GISTs. CONCLUSIONS Complete surgical resection with clear margins is recommended, if technically feasible, and genotyping can help to improve diagnosis and further patient management in oesophageal GIST.
Collapse
Affiliation(s)
- Guhyun Kang
- Department of Pathology, Sanggye Paik Hospital, Inje University, Seoul, Korea
| | - Yuna Kang
- Department of Pathology, Dongsan Medical Center, Keimyung University, Daegu, Korea
| | - Kyung-Hee Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sang Yun Ha
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Jung Yeon Kim
- Department of Pathology, Sanggye Paik Hospital, Inje University, Seoul, Korea
| | - Young Mog Shim
- Department of Thoracic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Michael C Heinrich
- Division of Hematology and Medical Oncology, VA Portland Health Care System and Oregon Health and Science University, Portland, OR, USA
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Christopher L Corless
- Department of Pathology and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
69
|
Campanella NC, Scapulatempo-Neto C, Abrahão-Machado LF, Torres De Oliveira AT, Berardinelli GN, Guimarães DP, Reis RM. Lack of microsatellite instability in gastrointestinal stromal tumors. Oncol Lett 2017; 14:5221-5228. [PMID: 29113157 PMCID: PMC5662911 DOI: 10.3892/ol.2017.6884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/09/2017] [Indexed: 12/18/2022] Open
Abstract
The microsatellite instability (MSI) phenotype may constitute an important biomarker for patient response to immunotherapy, particularly to anti-programmed death-1 inhibitors. MSI is a type of genomic instability caused by a defect in DNA mismatch repair (MMR) proteins, which is present mainly in colorectal cancer and its hereditary form, hereditary nonpolyposis colorectal cancer. Gastrointestinal stromal tumor (GIST) development is associated with activating mutations of KIT proto-oncogene receptor tyrosine kinase (KIT) or platelet-derived growth factor receptor α (PDGFRA), which are oncogenes that predict the response to imatinib mesylate. In addition to KIT/PDGFRA mutations, other molecular alterations are important in GIST development. In GISTs, the characterization of the MSI phenotype is scarce and the results are not consensual. The present study aimed to assess MSI in a series of 79 GISTs. The evaluation of MSI was performed by pentaplex polymerase chain reaction comprising five markers, followed by capillary electrophoresis. The expression of MMR proteins was evaluated by immunohistochemistry. Regarding the KIT/PDGFRA/B-Raf proto-oncogene, serine/threonine kinase molecular profile of the 79 GISTs, 83.6% of the tumors possessed KIT mutations, 10.1% had PDGFRA mutations and 6.3% were triple wild-type. The mutated-PDGFRA cases were associated with gastric location and a lower mitotic index compared with KIT-mutated and wild-types, and these patients were more likely to be alive and without cancer. MSI analysis identified 4 cases with instability in one marker, however, additional evaluation of normal tissue and immunohistochemical staining of MMR proteins confirmed their microsatellite-stable nature. The results of the present study indicated that MSI is not involved in GIST tumorigenesis and, therefore, cannot serve as a biomarker to immunotherapy response in GIST.
Collapse
Affiliation(s)
- Nathália C Campanella
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
| | | | | | - Gustavo N Berardinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil.,Department of Endoscopy, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
| | - Rui M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil.,Life and Health Sciences Research Institute, Health Sciences School, University of Minho, Guimarães, Braga 4704-553, Portugal.,Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, University of Minho, Guimarães, Braga 4704-553, Portugal
| |
Collapse
|
70
|
Khalikov DD, Akhmetzyanov FS, Petrov SV. [Clinical and morphological characteristics of gastrointestinal stromal tumors]. Arkh Patol 2017; 79:48-55. [PMID: 28791999 DOI: 10.17116/patol201779448-55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. Despite this, GISTS comprise about 2% in the structure of digestive tract cancers. They are usually localized in the stomach; however, they can be found in the small intestine and more rarely in the colon and esophagus. Although approximately 70% of the GISTs consist predominantly of spindle cells; the epithelioid cell tumors represent 20% of cases; there are also mixed variants. This variability in the morphological structure of GISTs complicates their diagnosis.
Collapse
Affiliation(s)
- D D Khalikov
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia; Republican Clinical Oncology Dispensary, Ministry of Heath of the Republic of Tatarstan, Kazan, Russia
| | - F Sh Akhmetzyanov
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia; Republican Clinical Oncology Dispensary, Ministry of Heath of the Republic of Tatarstan, Kazan, Russia; Volga Branch, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Kazan, Russia
| | - S V Petrov
- Kazan State Medical University, Ministry of Health of the Russian Federation, Kazan, Russia; Republican Clinical Oncology Dispensary, Ministry of Heath of the Republic of Tatarstan, Kazan, Russia; Volga Branch, N.N. Blokhin Russian Cancer Research Center, Ministry of Health of the Russian Federation, Kazan, Russia
| |
Collapse
|
71
|
Liu W, Zeng X, Wu X, He J, Gao J, Shuai X, Wang G, Zhang P, Tao K. Clinicopathologic study of succinate-dehydrogenase-deficient gastrointestinal stromal tumors: A single-institutional experience in China. Medicine (Baltimore) 2017; 96:e7668. [PMID: 28796048 PMCID: PMC5556214 DOI: 10.1097/md.0000000000007668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) that are not driven by kinase mutations, as are most GISTs, often show loss of function of the succinate dehydrogenase (SDH) complex and are considered SDH-deficient GISTs. SDH-deficient GISTs share many distinct characteristics compared with conventional GISTs. However, data regarding these characteristics, particularly among Asian people, are relatively limited. The objective of this study was to characterize the clinicopathologic characteristics, treatment, and prognosis of these uncommon GISTs.This retrospective observational study enrolled 12 patients with SDH-deficient GISTs, who were selected from 335 patients with GIST diagnosed at our institution between October 31, 2013 and October 31, 2016 by succinate dehydrogenase subunit B staining.There were 8 male and 4 female patients, with a median age of 57 years (range, 21-73 years). Ten patients (83.3%) were diagnosed at or after the age of 40 years and represented 7.2% (10/138) of the entire population of elderly patients with gastric GISTs. The tumor size ranged from 3 to 19 cm (median, 7 cm); the primary tumor was multifocal in 6 cases (50%), and tumors had a multinodular or plexiform architecture in 10 cases (83.3%). Ten cases (83.3%) showed pure epithelioid morphology, with the remaining 2 cases (16.7%) showing mixed histologic subtype. Lymph node metastasis was found at the time of primary resection in 50% (3/6) of patients. Four cases (33.3%) had distant metastasis at presentation. Four patients (33.3%) developed disease progression during imatinib treatment after initial resection, but all of these patients regained disease control when the treatment was altered to sunitinib targeted therapy.SDH-deficient GISTs arise exclusively in the stomach and account for approximately 7.4% (12/162) of gastric GISTs. Moreover, those affecting people older than 40 years are not uncommon and sunitinib may work well for cases showing treatment failure with imatinib.
Collapse
Affiliation(s)
| | | | - Xiuli Wu
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun He
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery
| | | | | | | | | |
Collapse
|
72
|
Pai T, Bal M, Shetty O, Gurav M, Ostwal V, Ramaswamy A, Ramadwar M, Desai S. Unraveling the spectrum of KIT mutations in gastrointestinal stromal tumors: An Indian Tertiary Cancer Center Experience. South Asian J Cancer 2017; 6:113-117. [PMID: 28975118 PMCID: PMC5615879 DOI: 10.4103/sajc.sajc_275_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Primary mutations in the KIT gene are the driving force for gastrointestinal stromal tumors (GIST) tumorigenesis. Predictive role of KIT mutation status aids oncologists in patient management. There is a paucity of comprehensive data on the frequency of mutations in the KIT gene in GIST affecting Indian patients. The aims of this study were to determine the frequency and spectrum of molecular alterations affecting the KIT gene and assess their association with clinicopathologic features in a cohort of patients of GIST. MATERIALS AND METHODS Morphological and immunohistochemically confirmed GIST cases (n = 114) accessioned from August 2014-June 2015 were analyzed for mutations in KIT exons 9, 11, 13, and 17 and subjected to Sanger sequencing onto the ABI 3500 Genetic Analyzer. The sequences were analyzed using sequence analysis software: SeqScape® and Chromas Lite. RESULTS KIT mutations were seen in 70% of cases and the majority of KIT mutations involved exon 11 (57%), followed by exon 9 (10%), exon 13 (3%), and exon 17 (1%). Most common exon 11 mutations were in-frame deletions (61.4%) followed by substitution mutations (19.3%). Exon 9 mutations showed identical duplication of Ala-Tyr at codons 502-503. Simultaneous mutations affecting exon 11 and 13 were discovered. Novel variations, namely, p.Q556E (c.1666C>G), p.Q556dup (c.1666_1668dupCAG), p.K558_V559delinsS (c.1672_1677delAAGGTTinsAGT), p.Y503_F504insTY (c.1509_1510insACCTAT), and p.K642R (c.1925A>G) involving exons 11, 9, and 13, respectively, were observed. INTERPRETATION AND CONCLUSIONS First study with complete analysis of all 4 exons of KIT (exons 9, 11, 13, and 17) in Indian GIST patients. Along with well-described KIT mutations, several rare double mutations as well as novel alterations were reported in this series.
Collapse
Affiliation(s)
- Trupti Pai
- Division of Molecular Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Omshree Shetty
- Division of Molecular Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Mamta Gurav
- Division of Molecular Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Mukta Ramadwar
- Department of Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Sangeeta Desai
- Division of Molecular Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
- Department of Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| |
Collapse
|
73
|
Kosemehmetoglu K, Kaygusuz G, Fritchie K, Aydin O, Yapicier O, Coskun O, Karatayli E, Boyacigil S, Guler G, Dervisoglu S, Kuzu I. Clinical and pathological characteristics of gastrointestinal stromal tumor (GIST) metastatic to bone. Virchows Arch 2017; 471:77-90. [PMID: 28488171 DOI: 10.1007/s00428-017-2138-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/11/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022]
Abstract
Our aim in this study was to describe the clinical, morphological, and molecular profile of gastrointestinal stromal tumor (GIST) metastatic to bone. We analyzed the morphological, phenotypic, and molecular characteristics of seven cases, and in addition reviewed 17 cases from literature. Sequence analysis of KIT and PDGFRA genes was possible for six cases. For the GIST cases with bone metastasis, the most common primaries were small intestine (29%), stomach (25%), and rectum (21%). Sites of bone metastases were vertebrae (11), pelvis (8), femur (8), ribs (6), humerus (5), skull (3), scapula (1), and mandible (1). The size ranged from 1.5 to 13 cm (median, 3.8 cm). Bone metastases without involvement of any other organ were seen in 17% of the cases and were solitary in 14 (58%). Adjacent soft tissue involvement was present in nearly half of the patients. Bone metastasis was either manifest at the time of diagnosis (28%) or occurred after a mean period of 4.7 years (3 months-20 years). Morphologically, neoplastic cells were spindle in 67%, epithelioid in 13%, and mixed epithelioid and spindle in 20%. CD117, DOG1, and CD34 were positive in 88, 86, and 85% of the cases, respectively. KIT Exon 11 mutations were the most frequent gene alteration (78%), followed by KIT Exon 13 mutations. Of 17 of the cases with available follow-up information, 7 (41%) patients developed bone metastasis under imatinib therapy. Five patients (29%) died of disease within a mean of 17 months. Bone metastases from GIST are usually found in patients with advanced disease and typically present as lytic masses with occasional soft tissue involvement. We could not identify any KIT or PDGFRA alterations predisposing to bone metastasis.
Collapse
Affiliation(s)
- Kemal Kosemehmetoglu
- Department of Pathology, Hacettepe University School of Medicine, Ankara, Turkey.
| | - Gulsah Kaygusuz
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Karen Fritchie
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ovgu Aydin
- Department of Pathology, Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Ozlem Yapicier
- Department of Pathology, Acıbadem University School of Medicine, Istanbul, Turkey
| | - Oznur Coskun
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | | | - Senay Boyacigil
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | | | - Sergulen Dervisoglu
- Department of Pathology, Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Isinsu Kuzu
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
74
|
Farag S, Somaiah N, Choi H, Heeres B, Wang WL, van Boven H, Nederlof P, Benjamin R, van der Graaf W, Grunhagen D, Boonstra PA, Reyners AK, Gelderblom H, Steeghs N. Clinical characteristics and treatment outcome in a large multicentre observational cohort of PDGFRA exon 18 mutated gastrointestinal stromal tumour patients. Eur J Cancer 2017; 76:76-83. [DOI: 10.1016/j.ejca.2017.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/05/2017] [Accepted: 02/05/2017] [Indexed: 12/11/2022]
|
75
|
Lehmann-Che J, Poirot B, Boyer JC, Evrard A. La génétique somatique des tumeurs solides, un incontournable à l’ère de la médecine de précision. Therapie 2017; 72:217-230. [DOI: 10.1016/j.therap.2016.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 10/20/2022]
|
76
|
Baskin Y, Kocal GC, Kucukzeybek BB, Akbarpour M, Kayacik N, Sagol O, Ellidokuz H, Oztop I. PDGFRA and KIT Mutation Status and Its Association With Clinicopathological Properties, Including DOG1. Oncol Res 2017; 24:41-53. [PMID: 27178821 PMCID: PMC7838738 DOI: 10.3727/096504016x14576297492418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Most of the gastrointestinal stromal tumors (GISTs) have gain-of-function mutations in the KIT gene, which can be used as a prognostic marker for the biological behavior of tumors, predictive marker for the response of tyrosine kinase inhibitors, and diagnostic marker. Researchers have focused on PDGFRA mutations because of both their prognostic and predictive potential and DOG1 positivity for diagnosis on GISTs. The aim of this study is to investigate the effect DOG1, PDGFRA, and KIT mutations on the prediction of the outcome for GIST management. Polymerase chain reaction was performed for KIT gene exons 9, 11, 13, and 17 and PDGFRA gene exons 12 and 18 with the genomic DNA of 46 GIST patients, and amplicons were sequenced in both directions. Immunocytochemical stainings were done by using primary antibodies. Molecular analysis revealed that the KIT mutation was observed in 63% of all cases, while the PDGFRA mutation was observed in 23.9% of cases. Significant relationships were found between age and KIT mutation, tumor location and KIT mutations, and tumor location and PDGFRA mutations (p ≤ 0.05). DOG1 positivity was detected in 65.2% of all GISTs and DOG1-positive cells had a higher KIT mutation ratio than DOG1-negative cells (p ≤ 0.05). KIT gene exon 11 mutations in DOG1-positive cells was higher than DOG1-negative cells (p ≤ 0.05). Conversely, KIT gene exon 13 mutations were higher in DOG1-negative cells than DOG1-positive cells (p ≤ 0.05). In this study, KIT mutation frequency was found similar with the European population; conversely, PDGFRA mutation frequency was similar with an Asian-Chinese-based study. KIT/PDGFRA mutations and tumor location can be used for the prediction of tumor behavior and the management of disease in GISTs. DOG1 positivity might be a candidate marker to support KIT and PDGFRA mutations, due to the higher DOG1 positivity in KIT exon 11 mutant and stomach- and small intestine-localized GISTs.
Collapse
Affiliation(s)
- Yasemin Baskin
- Institute of Oncology, Department of Basic Oncology, Dokuz Eylul University, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Nakajima N, Sato H, Takahashi K, Hasegawa G, Mizuno K, Hashimoto S, Sato Y, Terai S. Muscle layer histopathology and manometry pattern of primary esophageal motility disorders including achalasia. Neurogastroenterol Motil 2017; 29. [PMID: 27699951 DOI: 10.1111/nmo.12968] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/08/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Histopathology of muscularis externa in primary esophageal motility disorders has been characterized previously. We aimed to correlate the results of high-resolution manometry with those of histopathology. METHODS During peroral endoscopic myotomy, peroral esophageal muscle biopsy was performed in patients with primary esophageal motility disorders. Immunohistochemical staining for c-kit was performed to assess the interstitial cells of Cajal (ICCs). Hematoxylin Eosin and Azan-Mallory staining were used to detect muscle atrophy, inflammation, and fibrosis, respectively. KEY RESULTS Slides from 30 patients with the following motility disorders were analyzed: achalasia (type I: 14, type II: 5, type III: 3), one diffuse esophageal spasm (DES), two outflow obstruction (OO), four jackhammer esophagus (JE), and one nutcracker esophagus (NE). ICCs were preserved in high numbers in type III achalasia (n=9.4±1.2 cells/high power field [HPF]), compared to types I (n=3.7±0.3 cells/HPF) and II (n=3.5±1.0 cells/HPF). Moreover, severe fibrosis was only observed in type I achalasia and not in other types of achalasia, OO, or DES. Four of five patients with JE and NE had severe inflammation with eosinophilic infiltration of the esophageal muscle layer (73.8±50.3 eosinophils/HPF) with no epithelial eosinophils. One patient with JE showed a visceral myopathy pattern. CONCLUSIONS & INFERENCES Compared to types I and II, type III achalasia showed preserved ICCs, with variable data regarding DES and OO. In disorders considered as primary esophageal motility disorders, a disease category exists, which shows eosinophilic infiltration in the esophageal muscle layer with no eosinophils in the epithelium.
Collapse
Affiliation(s)
- N Nakajima
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - H Sato
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan.,Division of Gastroenterology and Hepatology, Saiseikai Niigata Daini Hospital, Niigata, Japan
| | - K Takahashi
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - G Hasegawa
- Division of Cellular and Molecular Pathology, Department of Cellular Function, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - K Mizuno
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - S Hashimoto
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Y Sato
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - S Terai
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, Niigata, Japan
| |
Collapse
|
78
|
Ramaswamy A, Bal M, Swami R, Shetty O, Bose S, Pai T, Gurav M, Gupta S, Ostwal V. Early outcomes of exon 11 mutants in GIST treated with standard dose Imatinib. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:134. [PMID: 28462214 PMCID: PMC5395474 DOI: 10.21037/atm.2017.03.31] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The exon 11 KIT mutant gastrointestinal stromal tumors (GIST) is a heterogeneous cohort with variable biological behavior based on different mutational subtypes. METHODS Patients with histologically proven GIST with KIT exon 11 mutations were selected from a prospectively maintained database, and evaluated for clinical characteristics and event free survival (EFS). Patients were divided into mutations upstream to codon 557 (G1), mutations involving codon 557-558 (G2) and mutation downstream to codon 558 (G3). RESULTS A total of 90 patients satisfied the inclusion criteria for study. Substitutions, indels and duplications were seen in 23 patients. Deletions were seen in 67 patients, of which 44 patients had large deletions (>6 base pairs), while 23 has small deletions (<6 base pairs). Complex mutations were seen in 15 patients. G2 mutations were noted in 33 patients, while G1 and G3 mutations were seen 32 and 25 patients respectively. With a median follow-up of 26 months, estimated median EFS for the entire cohort was 55 months. The G2 cohort had an inferior EFS compared to the G1 and G3 cohorts (46 vs. 55 months), but this did not achieve statistical significance (univariate analysis: P=0.075). On multivariate analysis, patients undergoing radical intent surgery vs. no surgery (58 vs. 55 months; P=0.005) and G1 or G3 vs. G2 cohort (P=0.058) showed trend towards improved EFS. CONCLUSIONS In patients with GIST exon 11 codon 557-558 mutation subset there is a trend towards an inferior survival even when treated with imatinib mesylate (IM).
Collapse
Affiliation(s)
- Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Rohit Swami
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Omshree Shetty
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Subhadeep Bose
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Trupti Pai
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Mamta Gurav
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Sudeep Gupta
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| |
Collapse
|
79
|
Correlation of Classic and Molecular Cytogenetic Alterations in Soft-Tissue Sarcomas: Analysis of 46 Tumors With Emphasis on Adipocytic Tumors and Synovial Sarcoma. Appl Immunohistochem Mol Morphol 2017; 25:168-177. [DOI: 10.1097/pai.0000000000000294] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
80
|
Bezawork-Geleta A, Rohlena J, Dong L, Pacak K, Neuzil J. Mitochondrial Complex II: At the Crossroads. Trends Biochem Sci 2017; 42:312-325. [PMID: 28185716 DOI: 10.1016/j.tibs.2017.01.003] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
Abstract
Mitochondrial complex II (CII), also called succinate dehydrogenase (SDH), is a central purveyor of the reprogramming of metabolic and respiratory adaptation in response to various intrinsic and extrinsic stimuli and abnormalities. In this review we discuss recent findings regarding SDH biogenesis, which requires four known assembly factors, and modulation of its enzymatic activity by acetylation, succinylation, phosphorylation, and proteolysis. We further focus on the emerging role of both genetic and epigenetic aberrations leading to SDH dysfunction associated with various clinical manifestations. This review also covers the recent discovery of the role of SDH in inflammation-linked pathologies. Conceivably, SDH is a potential target for several hard-to-treat conditions, including cancer, that remains to be fully exploited.
Collapse
Affiliation(s)
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czechia
| | - Lanfeng Dong
- School of Medical Science, Griffith University, Southport, Australia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Jiri Neuzil
- School of Medical Science, Griffith University, Southport, Australia; Institute of Biotechnology, Czech Academy of Sciences, Prague, Czechia.
| |
Collapse
|
81
|
Lehmann-Che J, Poirot B, Boyer JC, Evrard A. Cancer genomics guide clinical practice in personalized medicine. Therapie 2017; 72:439-451. [PMID: 28258721 DOI: 10.1016/j.therap.2016.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 01/04/2023]
Abstract
Targeted therapies have revolutionized the treatment of many cancers. Widely developed over the last decade, this new concept of precision medicine relies on the use of genomic technologies to analyze tumor samples in order to identify actionable targets and biomarkers of resistance. The goal is to optimize treatment by identifying which therapeutic approach is best for each patient, i.e. the treatment that is effective, has minimal adverse effects, and avoids unnecessary intervention and cost. The purpose of this review is to highlight, using a few seminal examples of therapeutic targets, the important contribution of appropriate analysis of key oncogenes or driver genes in making clinical decisions. Cancer genomics is now an indispensable part of clinical management. Furthermore, the development of next generation sequencing (NGS) will enable exploration of more and more genes of interest, leading to new treatment options for personalized medicine.
Collapse
Affiliation(s)
- Jacqueline Lehmann-Che
- Laboratoire d'oncologie moléculaire, hôpital Saint-Louis, 1, avenue Claude-Vellefaux 75475 Paris cedex 10, France; Unité CNRS UMR7212/U944, équipe de recherche translationnelle en oncologie, bâtiment Jean-Bernard, 75475 Paris, France.
| | - Brigitte Poirot
- Laboratoire d'oncologie moléculaire, hôpital Saint-Louis, 1, avenue Claude-Vellefaux 75475 Paris cedex 10, France; Unité CNRS UMR7212/U944, équipe de recherche translationnelle en oncologie, bâtiment Jean-Bernard, 75475 Paris, France
| | - Jean-Christophe Boyer
- Laboratoire de biochimie, CHU de Nîmes Carémeau, 30029 Nîmes, France; EA 2415, « Aide à la décision médicale personnalisée : aspects méthodologiques » IURC, faculté de médecine de Montpellier, 34093 Montpellier, France
| | - Alexandre Evrard
- Laboratoire de biochimie, CHU de Nîmes Carémeau, 30029 Nîmes, France; Unité Inserm U1194, Institut de recherche en cancérologie de Montpellier (IRCM), 34298 Montpellier, France
| |
Collapse
|
82
|
Takahashi T, Elzawahry A, Mimaki S, Furukawa E, Nakatsuka R, Nakamura H, Nishigaki T, Serada S, Naka T, Hirota S, Shibata T, Tsuchihara K, Nishida T, Kato M. Genomic and transcriptomic analysis of imatinib resistance in gastrointestinal stromal tumors. Genes Chromosomes Cancer 2017; 56:303-313. [PMID: 27997714 PMCID: PMC5324566 DOI: 10.1002/gcc.22438] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/01/2022] Open
Abstract
Gastrointestinal stromal tumors represent the most common mesenchymal tumor of the digestive tract, driven by gain‐of‐function mutations in KIT. Despite its proven benefits, half of the patients treated with imatinib show disease progression within 2 years due to secondary resistance mutations in KIT. It remains unclear how the genomic and transcriptomic features change during the acquisition of imatinib resistance. Here, we performed exome sequencing and microarray transcription analysis for four imatinib‐resistant cell lines and one cell line briefly exposed to imatinib. We also performed exome sequencing of clinical tumor samples. The cell line briefly exposed to imatinib exhibited few single‐nucleotide variants and copy‐number alterations, but showed marked upregulation of genes related to detoxification and downregulation of genes involved in cell cycle progression. Meanwhile, resistant cell lines harbored numerous genomic changes: amplified genes related to detoxification and deleted genes with cyclin‐dependent kinase activity. Some variants in the resistant samples were traced back to the drug‐sensitive samples, indicating the presence of ancestral subpopulations. The subpopulations carried variants associated with cell death. Pre‐existing cancer cells with genetic alterations promoting apoptosis resistance may serve as a basis whereby cancer cells with critical mutations, such as secondary KIT mutations, can establish full imatinib resistance. © 2017 The Authors Genes, Chromosomes and Cancer Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Asmaa Elzawahry
- Department of Bioinformatics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,JST, CREST, 5-3 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| | - Sachiyo Mimaki
- Division of Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Eisaku Furukawa
- Department of Bioinformatics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Rie Nakatsuka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Hiromi Nakamura
- Division of Cancer Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takahiko Nishigaki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2 E2, Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Satoshi Serada
- Laboratory for Immune Signal, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan
| | - Tetsuji Naka
- Laboratory for Immune Signal, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki City, Osaka, 567-0085, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical College, 1-1, Mukogawa-cho, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Katsuya Tsuchihara
- Division of Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toshirou Nishida
- Department of Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Mamoru Kato
- Department of Bioinformatics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,JST, CREST, 5-3 Yonbancho, Chiyoda-ku, Tokyo, 102-0081, Japan
| |
Collapse
|
83
|
Kou Y, Zhao Y, Bao C, Wang Q. Comparison of Gene Expression Profile Between Tumor Tissue and Adjacent Non-tumor Tissue in Patients with Gastric Gastrointestinal Stromal Tumor (GIST). Cell Biochem Biophys 2017; 72:571-8. [PMID: 25586720 DOI: 10.1007/s12013-014-0504-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are defined as spindle cell and/or epithelioid tumors originated from interstitial Cajal cells or precursors in the digestive tract. This study was conducted to identify genes differing in expression between the gastric tumors and the adjacent non-cancerous mucosas in patients with primary gastric GIST. The gene expression profile was determined by using oligonucleotide-based DNA microarrays and further validated by quantitative real-time PCR. The Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis was performed to predict signaling pathways involved in gastric GIST. Our data showed that the expression levels of 957 genes (RAB39B, member RAS oncogene family; VCAN, versican; etc.) were higher and that of 526 genes (CXCL14, chemokine C-X-C motif ligand 14; MTUS1, microtubule-associated tumor suppressor 1; etc.) were lower in the gastric tumor tissues as compared with normal gastric tissues. Results from KEGG pathway analysis revealed that the differentially expressed genes were enriched into 16 signaling transduction pathways, including Hedeghog and Wnt signaling pathways. Our study may provide basis for identification of novel biomarkers associated with primary gastric GIST pathogenesis and for exploration of underlying mechanisms involved in this gastric sarcoma.
Collapse
Affiliation(s)
- Youwei Kou
- Department of Gastrointestinal and Nutriology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Ying Zhao
- Department of Gastrointestinal and Nutriology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Chenhui Bao
- Department of Gastrointestinal and Nutriology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Qiang Wang
- Department of Gastrointestinal and Nutriology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
84
|
De Manzoni G, Marrelli D, Baiocchi GL, Morgagni P, Saragoni L, Degiuli M, Donini A, Fumagalli U, Mazzei MA, Pacelli F, Tomezzoli A, Berselli M, Catalano F, Di Leo A, Framarini M, Giacopuzzi S, Graziosi L, Marchet A, Marini M, Milandri C, Mura G, Orsenigo E, Quagliuolo V, Rausei S, Ricci R, Rosa F, Roviello G, Sansonetti A, Sgroi G, Tiberio GAM, Verlato G, Vindigni C, Rosati R, Roviello F. The Italian Research Group for Gastric Cancer (GIRCG) guidelines for gastric cancer staging and treatment: 2015. Gastric Cancer 2017; 20:20-30. [PMID: 27255288 DOI: 10.1007/s10120-016-0615-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/01/2016] [Indexed: 02/07/2023]
Abstract
This article reports the guidelines for gastric cancer staging and treatment developed by the GIRCG, and contains comprehensive indications for clinical management, including radiological, endoscopic, surgical, pathological, and oncological paths.
Collapse
Affiliation(s)
- Giovanni De Manzoni
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Daniele Marrelli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy.
| | - Gian Luca Baiocchi
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Paolo Morgagni
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Luca Saragoni
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Maurizio Degiuli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Annibale Donini
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Uberto Fumagalli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Maria Antonietta Mazzei
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Fabio Pacelli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Anna Tomezzoli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Mattia Berselli
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Filippo Catalano
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Alberto Di Leo
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Massimo Framarini
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Simone Giacopuzzi
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Luigina Graziosi
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Alberto Marchet
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Mario Marini
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Carlo Milandri
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Gianni Mura
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Elena Orsenigo
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Vittorio Quagliuolo
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Stefano Rausei
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Riccardo Ricci
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Fausto Rosa
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Giandomenico Roviello
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Andrea Sansonetti
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Giovanni Sgroi
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Guido Alberto Massimo Tiberio
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Giuseppe Verlato
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Carla Vindigni
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Riccardo Rosati
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| | - Franco Roviello
- GIRCG Secretary: Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100, Siena, Italy
| |
Collapse
|
85
|
Conventional Risk Stratification Fails to Predict Progression of Succinate Dehydrogenase–deficient Gastrointestinal Stromal Tumors. Am J Surg Pathol 2016; 40:1616-1621. [DOI: 10.1097/pas.0000000000000685] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
86
|
Sugase T, Takahashi T, Ishikawa T, Ichikawa H, Kanda T, Hirota S, Nakajima K, Tanaka K, Miyazaki Y, Makino T, Kurokawa Y, Yamasaki M, Takiguchi S, Wakai T, Mori M, Doki Y. Surgical resection of recurrent gastrointestinal stromal tumor after interruption of long-term nilotinib therapy. Surg Case Rep 2016; 2:137. [PMID: 27864817 PMCID: PMC5116018 DOI: 10.1186/s40792-016-0266-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/15/2016] [Indexed: 01/01/2023] Open
Abstract
Background Nilotinib inhibits the tyrosine kinase activities of ABL1/BCR-ABL1, KIT, and platelet-derived growth factor receptors (PDGFRs). The results of a phase III clinical trial indicated that nilotinib could not be recommended for broad use as first-line therapy for gastrointestinal stromal tumor (GIST). However, some clinical studies have reported the effectiveness of nilotinib. We report here the cases of two patients who underwent surgical resections of nilotinib-resistant lesions after long-term nilotinib administration. Case presentation Two Japanese female patients, aged 66 and 70 years, experienced peritoneal recurrence of intestinal GIST several years after surgery. Both were registered in the ENESTg1 trial and received nilotinib therapy. Although they continued nilotinib administration with a partial response according to the protocol, nilotinib-resistant lesions, which were diagnosed as focally progressive disease, developed and complete surgical resection was performed. Pathological examination revealed that the tumors were composed of viable KIT-positive spindle cells, and the recurrent tumors were diagnosed as nilotinib-resistant GIST. In gene mutation analysis, a secondary KIT gene mutation was detected in one case. Both patients have survived more than 5 years after the first surgery. Conclusions Of patients who were registered in this trial, we have encountered two patients with long-term effects after nilotinib administration. Moreover, secondary mutations in the KIT gene, similar to those involved in resistance to imatinib, might be involved in resistance to nilotinib.
Collapse
Affiliation(s)
- Takahito Sugase
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takashi Ishikawa
- Division of Digestive and General Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tatsuo Kanda
- Department of Surgery, Sanjo General Hospital, Niigata, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
87
|
Toda-Ishii M, Akaike K, Suehara Y, Mukaihara K, Kubota D, Kohsaka S, Okubo T, Mitani K, Mogushi K, Takagi T, Kaneko K, Yao T, Saito T. Clinicopathological effects of protein phosphatase 2, regulatory subunit A, alpha mutations in gastrointestinal stromal tumors. Mod Pathol 2016; 29:1424-1432. [PMID: 27469332 DOI: 10.1038/modpathol.2016.138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 06/13/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
Recently, several studies have reported that dysfunctions in protein phosphatase 2A (PP2A) caused by alterations in protein phosphatase 2 regulatory subunit A, alpha (PPP2R1A) are responsible for tumorigenesis and tumor progression in several types of cancers. The impact of PPP2R1A mutations remains unknown in gastrointestinal stromal tumors (GISTs), although mutations in KIT and PDGFRA, which result in constitutive activation of the receptor tyrosine kinase pathway, are important in GIST tumorigenesis. In this study, we performed mutation analysis of PPP2R1A to examine the frequency of PPP2R1A mutations and their clinicopathological correlation in 94 GIST cases. In addition, we performed an in vitro analysis to investigate the effects of PPP2R1A mutations on cell proliferation and kinase phosphorylation in GIST cells. Seventeen GIST cases (18%) harbored mutations in PPP2R1A. All but one of these 17 cases harbored a KIT, PDGFRA, HRAS, NRAS, or KRAS mutation as the oncogenic driver mutation, and the remaining case was immunohistochemically negative for succinate dehydrogenase B (SDHB). Multivariate analysis showed that larger tumor size, higher mitotic rate, and PPP2R1A mutation are independent prognostic factors for overall survival; however, PPP2R1A mutation was not an independent prognostic factor for disease-free survival. The transduction of GIST cells with mutant PPP2R1A induced an accelerated growth rate via increased phosphorylation of Akt1/2, ERK1/2, and WNK1, a kinase associated with angiogenesis. In addition, the transduction of GIST cells with mutant PPP2R1A caused increased c-kit phosphorylation, suggesting that c-kit is also a target of PP2A, reinforcing the tumorigenic capabilities of c-kit. Furthermore, the transducing GIST cells with wild-type PP2A dephosphorylated mutant c-kit. This study provides a new insight into the biology of GISTs and their phosphatase activity, and activated PP2A could be a therapeutic target in GISTs.
Collapse
Affiliation(s)
- Midori Toda-Ishii
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Keisuke Akaike
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kenta Mukaihara
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Daisuke Kubota
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Shinji Kohsaka
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taketo Okubo
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Keiko Mitani
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kaoru Mogushi
- Center for Genomic and Regenerative Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Tatsuya Takagi
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuo Kaneko
- Department of Orthopaedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Yao
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
88
|
Dracopoli NC, Boguski MS. The Evolution of Oncology Companion Diagnostics from Signal Transduction to Immuno-Oncology. Trends Pharmacol Sci 2016; 38:41-54. [PMID: 27789023 DOI: 10.1016/j.tips.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/08/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
Sixteen oncology drugs have been approved with a companion diagnostic (CDx) test by the FDA. These represent only 9.6% of the 167 oncology drug approvals since 1998, the year the first CDx test for Herceptin was approved. The great majority of CDx tests are for drugs that inhibit signal transduction pathways by either inhibiting the intracellular kinase activity with a small molecule or preventing ligand-induced receptor activation with a monoclonal antibody. In most of these cases, prospective patient selection for the biomarker-positive subpopulation was initiated in or before Phase II. The development of CDx tests for emerging immunotherapies will be more complicated because they are not dependent on driver mutations in the drug target, the mechanism of action is often pleiotropic, and will require both protein and cell-based assays to evaluate the interaction of the tumor with the immune system. Consequently, we will need to develop new biomarker strategies for the development of immunotherapies and to determine whether the optimum strategy is to release a prior checkpoint blockade in patients with a suppressed immune response, or to prime a new immune response to the tumor.
Collapse
Affiliation(s)
| | - Mark S Boguski
- Precision Medicine Network Inc., 1620 Sudbury Road, Concord, MA 01742, USA.
| |
Collapse
|
89
|
Zhang R, Zhao J, Xu J, Liu F, Xu Y, Bu X, Dai C, Song C. Genetic variations in the TERT and CLPTM1L gene region and gastrointestinal stromal tumors risk. Oncotarget 2016; 6:31360-7. [PMID: 26372813 PMCID: PMC4741611 DOI: 10.18632/oncotarget.5153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/27/2015] [Indexed: 12/03/2022] Open
Abstract
Recent studies have suggested polymorphisms in the TERT and CLPTM1L region are associated with carcinogenesis of many distinct cancer types, including gastrointestinal cancers. However, the contribution of polymorphisms in the TERT and CLPTM1L gene region to gastrointestinal stromal tumors (GISTs) risk is still unknown. We tested the six tagSNPs on TERT and CLPTM1L region with GIST risk, using a population-based, two-stage, case-control study in 2,000 subjects. Functional validation was conducted to validate our findings of TERT rs2736098 and explore its influence on relative telomere length (RTL) in GIST cells. It showed that variant rs2736098 was significantly associated with increased risk of GIST (per allele OR = 1.29, 95% CI: 1.14–1.47, P = 7.03 × 10−5). The difference remain significant after Bonferroni correction (P = 7.03 × 10−5 * 6 = 4.2 × 10−4). Real-time PCR showed carriers of genotype CC have the longest RTL, following by carriers of genotype CT, while carriers of genotype TT have the shortest RTL in GIST tissues (P < 0.001). Our data provide evidence to implicate TERT rs2736098 polymorphism as a novel susceptibility factor for GIST risk.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, P.R. China
| | - Jian Zhao
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, P.R. China
| | - Jian Xu
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, P.R. China
| | - Fang Liu
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, P.R. China
| | - Yongqing Xu
- Department of Hepatobiliary and Splenic Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, Liaoning Province, P.R. China
| | - Xianmin Bu
- Department of Hepatobiliary and Splenic Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, Liaoning Province, P.R. China
| | - Chaoliu Dai
- Department of Hepatobiliary and Splenic Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, Liaoning Province, P.R. China
| | - Chun Song
- Department of Colorectal Surgery, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, P.R. China
| |
Collapse
|
90
|
Hallin M, Mudan S, Thway K. Interstitial Cells of Cajal in Deep Esophageal Leiomyoma. Int J Surg Pathol 2016; 25:51-53. [DOI: 10.1177/1066896916660197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are potentially aggressive mesenchymal neoplasms with spindle cell, epithelioid, or mixed morphology. They typically express CD117, DOG1, and CD34 and can be diffusely and strongly positive for h-caldesmon. Leiomyomas are benign smooth muscle neoplasms that can arise in a variety of visceral and soft tissue sites, including the gastrointestinal tract. We illustrate a case of a neoplasm of the gastroesophageal junction that was clinically suspected to be a GIST. Histology showed a tumor composed of ovoid and spindle cells arranged in short intersecting fascicles, which was positive for desmin, smooth muscle actin, and h-caldesmon, with a prominent interspersed subpopulation of CD117- and DOG1-positive elongated or dendritic-like cells. These features were of leiomyoma with entrapped interstitial cells of Cajal (ICC). The recognition of possible entrapment of ICC in leiomyomas as a potential mimic of GIST is important for correct treatment and prognostication.
Collapse
|
91
|
Othman ER, Elgamal DA, Refaiy AM, Abdelaal II, Abdel-Mola AF, Al-Hendy A. Identification and potential role of telocytes in human uterine leiomyoma. Contracept Reprod Med 2016; 1:12. [PMID: 29201401 PMCID: PMC5693520 DOI: 10.1186/s40834-016-0022-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/11/2016] [Indexed: 02/07/2023] Open
Abstract
Background Telocytes are specialized interstitial tissue cell type. Our aim is to characterize telocytes in human uterine leiomyoma (ULM) and its adjacent myometrium (Myo-F) as well as normal myometrium (Myo-N). Methods ULMs and Myo-F tissues were taken from hysterectomy specimens done to treat symptomatic uterine fibroids (N = 20). Myo-N is isolated from hysterectomies done on ULM- free uteri for other benign indications (N = 15). Telocytes were detected using immunohistochemistry to detect c-Kit (CD-117), as a surface marker expressed on telocytes, and electron microscopic examination to identify telocytes characteristic ultrastructure. Cellular count and electron microscopic features of telocytes in each of the studied tissues were compared. Results Telocytes could be detected in ULMs, Myo-F and Myo-N using c-KIT immunostaining. Electron microscopy confirmed the presence of telocytes in the three types of tissues identifying their characteristic features including small triangular or fusiform cell bodies with extensive cellular prolongations. ULM telocytes showed ultrastructural features suggestive of high cellular activities. Cell counts of ULM telocytes (3.35 ± 0.39) were significantly higher (P value = 0.00039) than that of Myo-F (1.39 ± 0.13). Myo-N (2.6 ± 0.36) contained higher telocyte numbers than Myo-F (1.39 ± 0.13), but the difference did not reach statistical significance (P value = 0.19). Conclusions Telocytes are detected in higher numbers and activity in ULMs than Myo-F or Myo-N. In ULMs, telocytes can work as a hormonal sensors for stem cells, provide scaffold for newly formed myocytes, or control important downstream signaling pathways.
Collapse
Affiliation(s)
- Essam R Othman
- OB-GYN Department, Assiut University, Assiut, Egypt.,Center of Excellence of Stem Cells and Regenerative Medicine, Assiut University, Assiut, 71111 Egypt
| | | | | | | | | | - Ayman Al-Hendy
- OB-GYN Department, Medical College of Georgia MCG, Augusta Univerity, Augusta, GA USA
| |
Collapse
|
92
|
Ricci R. Syndromic gastrointestinal stromal tumors. Hered Cancer Clin Pract 2016; 14:15. [PMID: 27437068 PMCID: PMC4950812 DOI: 10.1186/s13053-016-0055-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 06/09/2016] [Indexed: 12/28/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of gastrointestinal tract. They feature heterogeneous triggering mechanisms, implying relevant clinical differences. The vast majority of GISTs are sporadic tumors. Rarely, however, GIST-prone syndromes occur, mostly depending on heritable GIST predisposing molecular defects involving the entire organism. These conditions need to be properly identified in order to plan appropriate diagnostic, prognostic and therapeutic procedures. Clinically, GIST-prone syndromes must be thought of whenever GISTs are multiple and/or associated with accompanying signs peculiar to the background tumorigenic trigger, either in single individuals or in kindreds. Moreover, syndromic GISTs, individually considered, tend to show distinctive features depending on the underlying condition. When applicable, genotyping is usually confirmatory. In GIST-prone conditions, the prognostic features of each GIST, defined according to the criteria routinely applied to sporadic GISTs, combine with the characters proper to the background syndromes, defining peculiar clinical settings which challenge physicians to undertake complex decisions. The latter concern preventive therapy and single tumor therapy, implying possible surgical and molecularly targeted options. In the absence of specific comprehensive guidelines, this review will highlight the traits characteristic of GIST-predisposing syndromes, with particular emphasis on diagnostic, prognostic and therapeutic implications, which can help the clinical management of these rare diseases.
Collapse
Affiliation(s)
- Riccardo Ricci
- Department of Pathology, Università Cattolica del S. Cuore, Largo Agostino Gemelli, 8, I-00168 Rome, Italy
| |
Collapse
|
93
|
Farid M, Ngeow J. Sarcomas Associated With Genetic Cancer Predisposition Syndromes: A Review. Oncologist 2016; 21:1002-13. [PMID: 27401891 DOI: 10.1634/theoncologist.2016-0079] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/15/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED : Sarcomas are rare mesenchymal malignancies that demonstrate great clinical and biological heterogeneity. A variety of sarcomas develop in the context of well-defined heritable cancer predisposition syndromes, associations that are often overlooked, given the rarity and diversity of sarcomas and the equivalent relative infrequency of cancer genetic syndromes. This review describes in detail selected heritable cancer predisposition syndromes that are known to be associated with sarcomas. Beyond the molecular and clinical features that define each syndrome, disparities in clinical presentation, natural history, and treatment of syndrome-associated compared with otherwise histologically identical sporadic sarcomas will be described. The clinical approach to selected sarcoma subsets with a view to identifying possible associations with these syndromes will then be described. Although the treatment of the majority of sarcomas will not differ significantly between sporadic cases and those associated with predisposition syndromes, knowledge of features such as unique anatomic sites of affliction or excess toxicities with particular cytotoxic therapies can facilitate alterations in therapeutic strategies to maximize efficacy and minimize toxicity. In addition, recognition of cancer genetic predisposition syndrome will allow patients and their relatives to undertake appropriate genetic counseling and testing, as well as screening, surveillance, and interventional measures, as needed. Situating sarcomas within the genetic endowment of particular patients-specifically that which confers a higher risk of malignancy-will enable clinicians to better manage the patient as a whole, complementing the great efforts currently routinely undertaken to genomically characterize somatic tumor changes with a view to achieving the dream of personalized medicine. IMPLICATIONS FOR PRACTICE Sarcomas are uncommon malignancies that often occur sporadically but can also arise in the setting of a recognized heritable cancer predisposition syndrome. Identification of such associations when present can facilitate refinement and optimization of treatment strategies for the sarcoma so as to minimize toxicity and maximize efficacy. Discerning genetic predisposition can also facilitate institution of genetic counseling, as well as screening or surveillance schema for both the patient and his or her relatives, if required. Vigilance for these syndromes has the potential to significantly enhance the quality and comprehensiveness of sarcoma clinical management.
Collapse
Affiliation(s)
- Mohamad Farid
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joanne Ngeow
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
94
|
Gasparotto D, Rossi S, Polano M, Tamborini E, Lorenzetto E, Sbaraglia M, Mondello A, Massani M, Lamon S, Bracci R, Mandolesi A, Frate E, Stanzial F, Agaj J, Mazzoleni G, Pilotti S, Gronchi A, Dei Tos AP, Maestro R. Quadruple-Negative GIST Is a Sentinel for Unrecognized Neurofibromatosis Type 1 Syndrome. Clin Cancer Res 2016; 23:273-282. [PMID: 27390349 DOI: 10.1158/1078-0432.ccr-16-0152] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/26/2016] [Accepted: 06/14/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE The majority of gastrointestinal stromal tumors (GIST) are driven by KIT, PDGFRA, or, less commonly, BRAF mutations, and SDH gene inactivation is involved in a limited fraction of gastric lesions. However, about 10% of GISTs are devoid of any of such alterations and are poorly responsive to standard treatments. This study aims to shed light on the molecular drivers of quadruple-negative GISTs. EXPERIMENTAL DESIGN Twenty-two sporadic quadruple-negative GISTs with no prior association with Neurofibromatosis Type 1 syndrome were molecularly profiled for a panel of genes belonging to tyrosine kinase pathways or previously implicated in GISTs. For comparison purposes, 24 GISTs carrying KIT, PDGFRA, or SDH gene mutations were also analyzed. Molecular findings were correlated to clinicopathologic features. RESULTS Most quadruple-negative GISTs featured intestinal localization, with a female predilection. About 60% (13/22) of quadruple-negative tumors carried NF1 pathogenic mutations, often associated with biallelic inactivation. The analysis of normal tissues, available in 11 cases, indicated the constitutional nature of the NF1 mutation in 7 of 11 cases, unveiling an unrecognized Neurofibromatosis Type 1 syndromic condition. Multifocality and a multinodular pattern of growth were common findings in NF1-mutated quadruple-negative GISTs. CONCLUSIONS NF1 gene mutations are frequent in quadruple-negative GISTs and are often constitutional, indicating that a significant fraction of patients with apparently sporadic quadruple-negative GISTs are affected by unrecognized Neurofibromatosis Type 1 syndrome. Hence, a diagnosis of quadruple-negative GIST, especially if multifocal or with a multinodular growth pattern and a nongastric location, should alert the clinician to a possible Neurofibromatosis Type 1 syndromic condition. Clin Cancer Res; 23(1); 273-82. ©2016 AACR.
Collapse
Affiliation(s)
- Daniela Gasparotto
- Experimental Oncology 1, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Sabrina Rossi
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Maurizio Polano
- Experimental Oncology 1, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Elena Tamborini
- Department of Pathology and Molecular Biology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Erica Lorenzetto
- Experimental Oncology 1, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Marta Sbaraglia
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Alessia Mondello
- Experimental Oncology 1, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Marco Massani
- Department of Surgery, Treviso General Hospital, Treviso, Italy
| | - Stefano Lamon
- Department of Oncology, Treviso General Hospital, Treviso, Italy
| | - Raffaella Bracci
- Department of Internal Medicine, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | - Franco Stanzial
- Clinical Genetics Service, Bolzano General Hospital, Bolzano/Bozen, Italy
| | - Jerin Agaj
- Department of Surgery, Vipiteno General Hospital, Vipiteno/Sterzing, Italy
| | - Guido Mazzoleni
- Department of Pathology, Bolzano General Hospital, Bolzano/Bozen, Italy
| | - Silvana Pilotti
- Department of Pathology and Molecular Biology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Angelo Paolo Dei Tos
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Roberta Maestro
- Experimental Oncology 1, CRO Aviano National Cancer Institute, Aviano, Italy.
| |
Collapse
|
95
|
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
|
96
|
Feng F, Tian Y, Liu S, Zheng G, Liu Z, Xu G, Guo M, Lian X, Fan D, Zhang H. Combination of PLR, MLR, MWR, and Tumor Size Could Significantly Increase the Prognostic Value for Gastrointestinal Stromal Tumors. Medicine (Baltimore) 2016; 95:e3248. [PMID: 27057867 PMCID: PMC4998783 DOI: 10.1097/md.0000000000003248] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Systemic inflammation and immune response were associated with prognosis of tumors. However, data was limited due to the relatively low incidence of gastrointestinal stromal tumors (GISTs). The aim of the present study was to investigate the predictive value of preoperative peripheral blood cells in prognosis of GISTs.From September 2008 to July 2015, a total of 274 GIST patients in our department were enrolled in the present study. Clinicopathological features of GISTs were recorded. The association between preoperative peripheral blood cells and prognosis of GISTs were analyzed.Tumor location, tumor size, mitotic index, intratumoral necrosis, and National Institutes of Health (NIH) risk category were associated with prognosis of GISTs. High neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), neutrophil-to-white blood cell ratio (NWR), monocyte-to-white blood cell ratio (MWR) and low lymphocyte-to-white blood cell ratio (LWR) was associated with poor prognosis of GISTs (76.2% vs 83.7%, P = 0.010. 70.5% vs 98.7%, P = 0.000. 65.7% vs 96.4%, P = 0.004. 78.5% vs 82.5%, P = 0.044. 73.5% vs 97.8%, P = 0.004. 76.6% vs 83.6%, P = 0.012, respectively). However, tumor size was the only independent risk factor for prognosis according to multivariate analysis (P = 0.006). Tumor location, tumor size, mitotic index, and NIH risk category were significantly correlated with the above-mentioned parameters (all P < 0.05). The prognosis of GISTs with tumor size >5 cm, high MLR, high PLR, and high MWR was significantly lower than the remnant patients (P = 0.010).The peripheral blood routine test is convenient, reproducible, and inexpensive. High NLR, MLR, PLR, NWR, MWR, and low LWR were associated with poor prognosis of GISTs. The association between the above parameters and prognosis of GISTs may be attributed to their correlation with tumor size, mitotic index, and NIH risk category. The combination of tumor size, MLR, PLR, and MWR could further increase the predictive value of prognosis of GISTs.
Collapse
Affiliation(s)
- Fan Feng
- From the Department of Digestive Surgery (FF, SL, GZ, ZL, GX, MG, XL, DF, HZ), Xijing Hospital, Fourth Military Medical University; and Department of Dermatology (YT), Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Boaz RJ, George AP, Kumar RM, Devasia A. Giant seminal vesicle cyst: an unusual site for a malignant extragastrointestinal stromal tumour. BMJ Case Rep 2016; 2016:bcr-2015-214066. [PMID: 26935954 DOI: 10.1136/bcr-2015-214066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Mesenchymal tumours with clinicopathological and molecular profiles similar to gastrointestinal stromal tumours (GISTs) are, on occasion, found in extragastrointestinal locations. Extra GIST (EGIST) is a singular occurrence in the genitourinary tract. A 30-year-old man, catheterised following urinary retention, was found to have a complex pelvic retrovesical cyst on imaging. At operation, origin from the right seminal vesicle was evident with histopathology confirming a GIST. The patient received adjuvant therapy with tyrosine kinase inhibitor and is currently disease free at 2 years. This is only the second report of an EGIST at this anatomic locale. The current literature presents significant uncertainty in defining the true origin of EGISTs, particularly those in the pelvis. We propose the designation origin indeterminate stromal tumour (OIST), to facilitate disambiguation and advance accurate profiling of EGIST; a subject in evolution.
Collapse
Affiliation(s)
- Ranil Johann Boaz
- Department of Urology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Arun Philip George
- Department of Urology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Ramani Manoj Kumar
- Department of Pathology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Antony Devasia
- Department of Urology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| |
Collapse
|
98
|
Ang C, Maki RG. Contemporary Management of Metastatic Gastrointestinal Stromal Tumors: Systemic and Locoregional Approaches. Oncol Ther 2016; 4:1-16. [PMID: 28261637 PMCID: PMC5315077 DOI: 10.1007/s40487-015-0014-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Indexed: 12/25/2022] Open
Abstract
The treatment of metastatic gastrointestinal stromal tumors (GISTs) changed dramatically with the introduction of imatinib into the therapeutic lexicon in 2001. Over the past 15 years, tyrosine kinase inhibitors in the adjuvant and metastatic settings have remained the standard of care for this disease, though alternate classes of agents and new therapeutic targets are being actively explored in clinical trials. Although data are limited, the use of surgical and non-surgical locoregional techniques for the treatment of GIST metastases has increased and given reports of promising and durable responses. Herein we provide an overview of the contemporary therapeutic landscape of metastatic GIST.
Collapse
Affiliation(s)
- Celina Ang
- Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Robert G Maki
- Division of Hematology/Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
99
|
Asić K. Dominant mechanisms of primary resistance differ from dominant mechanisms of secondary resistance to targeted therapies. Crit Rev Oncol Hematol 2016; 97:178-96. [DOI: 10.1016/j.critrevonc.2015.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 02/07/2023] Open
|
100
|
Noguchi R, Yano H, Gohda Y, Suda R, Igari T, Ohta Y, Yamashita N, Yamaguchi K, Terakado Y, Ikenoue T, Furukawa Y. Molecular profiles of high-grade and low-grade pseudomyxoma peritonei. Cancer Med 2015; 4:1809-16. [PMID: 26475379 PMCID: PMC5123786 DOI: 10.1002/cam4.542] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/18/2015] [Accepted: 08/23/2015] [Indexed: 02/02/2023] Open
Abstract
Pseudomyxoma peritonei (PMP) is a rare disease exhibiting a distinct clinical feature caused by cancerous cells that produce mucinous fluid in the abdominal cavity. PMPs originate most frequently from the appendix and less frequently from the ovary. This disease can range from benign to malignant, and histologically, PMP is classified into two types: disseminated peritoneal adenomucinosis (DPAM) representing the milder phenotype, and peritoneal mucinous adenocarcinomas (PMCA) representing the aggressive phenotype. Although histological classification is clinically useful, the pathogenesis of PMP remains largely unknown. To elucidate the molecular mechanisms underlying PMP, we analyzed 18 PMP tumors comprising 10 DPAMs and 8 PMCAs. DNA was extracted from tumor and matched non-tumorous tissues, and was sequenced using Ion AmpliSeq Cancer Panel containing 50 cancer-related genes. Analysis of the data identified a total of 35 somatic mutations in 10 genes, and all mutations were judged as pathological mutations. Mutations were frequently identified in KRAS (14/18) and GNAS (8/18). Interestingly, TP53 mutations were found in three of the eight PMCAs, but not in the DPAMs. PIK3CA and AKT1 mutations were also identified in two PMCAs, but not in the DPAMs. These results suggested that KRAS and/or GNAS mutations are common genetic features of PMP, and that mutations in TP53 and/or genes related to the PI3K-AKT pathway may render malignant properties to PMP. These findings may be useful for the understanding of tumor characteristics, and facilitate the development of therapeutic strategies.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Clinical Genome ResearchThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Hideaki Yano
- Department of SurgeryNational Center for Global Health and MedicineTokyoJapan
| | - Yoshimasa Gohda
- Department of SurgeryNational Center for Global Health and MedicineTokyoJapan
| | - Ryuichiro Suda
- Department of SurgeryNational Center for Global Health and MedicineTokyoJapan
| | - Toru Igari
- Pathology Division of Clinical LaboratoryNational Center for Global Health and MedicineTokyoJapan
| | - Yasunori Ohta
- Department of Pathology, Research HospitalThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Naohide Yamashita
- Department of Advanced Medical ScienceResearch HospitalThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome ResearchThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yumi Terakado
- Division of Clinical Genome ResearchThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tsuneo Ikenoue
- Division of Clinical Genome ResearchThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yoichi Furukawa
- Division of Clinical Genome ResearchThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| |
Collapse
|