1
|
Karki B, Munir M, Parajuli A, Santhosh S, Singh A. Gastrointestinal Stromal Tumor (GIST) in a Young Adult: A Rare Presentation and a Multidisciplinary Approach. Cureus 2024; 16:e68001. [PMID: 39347296 PMCID: PMC11428075 DOI: 10.7759/cureus.68001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
A gastrointestinal stromal tumor is a rare gastrointestinal tumor of mesenchymal origin. We present a rare case of a 32-year-old male patient with a history of iron deficiency anemia who presented with nocturnal cramping abdominal pain, nausea, non-bloody vomiting, loss of appetite, and weight loss. The patient had no significant family history of cancer. Prior imaging showed gastric distension with chronic inflammatory changes, but a scheduled esophagogastroduodenoscopy (EGD) was not done due to loss of follow-up. On admission, the patient was tachycardic with anemia. An abdominal CT scan showed new areas of gastric wall thickening with edematous wall thickening at the gastric cardia. An EGD revealed deep gastric ulcers with elevated edges with central necrosis, raising concerns for malignancy. A biopsy of the gastric cardia confirmed a high-grade stromal tumor with the aid of a DOG1 test; the gastric cardia was KIT-positive in immunohistochemical testing but negative for Helicobacter pylori. The tumor was staged at pT3N0. He was treated with surgical resection with negative margins and imatinib therapy. Postoperative surveillance showed no evidence of malignancy and the patient experienced a positive response to treatment with stable hemoglobin levels and significant weight gain.
Collapse
Affiliation(s)
- Bibek Karki
- Internal Medicine, Hurley Medical Center - Michigan State University, Flint, USA
| | - Mahpara Munir
- Internal Medicine, Hurley Medical Center - Michigan State University, Flint, USA
| | - Abinash Parajuli
- Internal Medicine, Hurley Medical Center - Michigan State University, Flint, USA
| | - Sisira Santhosh
- Internal Medicine and Pediatrics, Hurley Medical Center - Michigan State University, Flint, USA
| | - Adiraj Singh
- Internal Medicine and Pediatrics, Hurley Medical Center - Michigan State University, Flint, USA
| |
Collapse
|
2
|
Klein-Rodewald T, Micklich K, Sanz-Moreno A, Tost M, Calzada-Wack J, Adler T, Klaften M, Sabrautzki S, Aigner B, Kraiger M, Gailus-Durner V, Fuchs H, Gründer A, Pahl H, Wolf E, Hrabe de Angelis M, Rathkolb B, Rozman J, Puk O, Schrewe A, Schulz H, Adamski J, Busch DH, Esposito I, Wurst W, Stoeger C, Gründer A, Pahl H, Wolf E, Hrabe de Angelis M, Rathkolb B. New C3H Kit N824K/WT cancer mouse model develops late-onset malignant mammary tumors with high penetrance. Sci Rep 2022; 12:19793. [PMID: 36396684 PMCID: PMC9671887 DOI: 10.1038/s41598-022-23218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Gastro-intestinal stromal tumors and acute myeloid leukemia induced by activating stem cell factor receptor tyrosine kinase (KIT) mutations are highly malignant. Less clear is the role of KIT mutations in the context of breast cancer. Treatment success of KIT-induced cancers is still unsatisfactory because of primary or secondary resistance to therapy. Mouse models offer essential platforms for studies on molecular disease mechanisms in basic cancer research. In the course of the Munich N-ethyl-N-nitrosourea (ENU) mutagenesis program a mouse line with inherited polycythemia was established. It carries a base-pair exchange in the Kit gene leading to an amino acid exchange at position 824 in the activation loop of KIT. This KIT variant corresponds to the N822K mutation found in human cancers, which is associated with imatinib-resistance. C3H KitN824K/WT mice develop hyperplasia of interstitial cells of Cajal and retention of ingesta in the cecum. In contrast to previous Kit-mutant models, we observe a benign course of gastrointestinal pathology associated with prolonged survival. Female mutants develop mammary carcinomas at late onset and subsequent lung metastasis. The disease model complements existing oncology research platforms. It allows for addressing the role of KIT mutations in breast cancer and identifying genetic and environmental modifiers of disease progression.
Collapse
Affiliation(s)
- Tanja Klein-Rodewald
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kateryna Micklich
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Adrián Sanz-Moreno
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Monica Tost
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Julia Calzada-Wack
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thure Adler
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Klaften
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,Present Address: amcure GmbH, Herrman-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Sibylle Sabrautzki
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.4567.00000 0004 0483 2525Research Unit Comparative Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Bernhard Aigner
- grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Kraiger
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Valerie Gailus-Durner
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Albert Gründer
- grid.7708.80000 0000 9428 7911Section of Molecular Hematology, Department of Hematology/Oncology, Universitäts Klinikum Freiburg, Freiburg, Germany
| | - Heike Pahl
- grid.7708.80000 0000 9428 7911Section of Molecular Hematology, Department of Hematology/Oncology, Universitäts Klinikum Freiburg, Freiburg, Germany
| | - Eckhard Wolf
- grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Hrabe de Angelis
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany ,grid.6936.a0000000123222966Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Birgit Rathkolb
- grid.4567.00000 0004 0483 2525Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.5252.00000 0004 1936 973XInstitute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany ,grid.452622.5German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Cohen-Gogo S, Kanwar N, Shaikh F, Baertschiger RM, Shlien A, Malkin D, Putra J, Coblentz A, Villani A, Gupta AA, Morgenstern DA. Response to Alpelisib in an Adolescent With PIK3CA-Mutated Metastatic Gastrointestinal Stromal Tumor. JCO Precis Oncol 2022; 6:e2200105. [PMID: 35917491 DOI: 10.1200/po.22.00105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Sarah Cohen-Gogo
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Nisha Kanwar
- Division of Pathology and Laboratory Medicine, The Hospital for Sick Children, Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, Canada
| | - Furqan Shaikh
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Reto M Baertschiger
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Adam Shlien
- Division of Pathology and Laboratory Medicine, The Hospital for Sick Children, Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, Canada
| | - David Malkin
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Juan Putra
- Department of Pathology, Boston Children's Hospital, Boston, MA
| | - Ailish Coblentz
- Diagnostic Imaging, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anita Villani
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Abha A Gupta
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Daniel A Morgenstern
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Liu TT, Li CF, Tan KT, Jan YH, Lee PH, Huang CH, Yu SC, Tsao CF, Wang JC, Huang HY. Characterization of Aberrations in DNA Damage Repair Pathways in Gastrointestinal Stromal Tumors: The Clinicopathologic Relevance of γH2AX and 53BP1 in Correlation with Heterozygous Deletions of CHEK2, BRCA2, and RB1. Cancers (Basel) 2022; 14:1787. [PMID: 35406559 PMCID: PMC8997382 DOI: 10.3390/cancers14071787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Genetic aberrations involving DNA damage repair (DDR) remain underexplored in gastrointestinal stromal tumors (GISTs). We characterized DDR abnormalities using targeted next-generation sequencing and multiplex ligation-dependent probe amplification, and performed immunofluorescence (IF) and immunohistochemistry (IHC) analyses of γH2AX and 53BP1. Consistent with IF-validated nuclear co-localization, γH2AX and 53BP1 showed robust correlations in expression levels, as did both biomarkers between IF and IHC. Without recurrent pathogenic single-nucleotide variants, heterozygous deletions (HetDels) frequently targeted DNA damage-sensing genes, with CHEK2-HetDel being the most prevalent. Despite their chromosomal proximity, BRCA2 and RB1 were occasionally hit by HetDels and were seldom co-deleted. HetDels of CHEK2 and BRCA2 showed a preference for older age groups, while RB1-HetDel predominated in the non-gastric, high-risk, and 53BP1-overexpressing GISTs. Higher risk levels were consistently related to γ-H2AX or 53BP1 overexpression (all p < 0.01) in two validation cohorts, while only 53BP1 overexpression was associated with the deletion of KIT exon 11 (KITex11-del) among genotyped GISTs. Low expressers of dual biomarkers were shown by univariate analysis to have longer disease-free survival (p = 0.031). However, higher risk levels, epithelioid histology, and KITex11-del retained prognostic independence. Conclusively, IHC is a useful surrogate of laborious IF in the combined assessment of 53BP1 and γ-H2AX to identify potential DDR-defective GISTs, which were frequently aberrated by HetDels and a harbinger of progression.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung 833, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan 710, Taiwan;
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan 710, Taiwan
| | - Kien-Thiam Tan
- Department of Medical Informatic, ACT Genomics Co., Ltd., Taipei 100, Taiwan; (K.-T.T.); (Y.-H.J.)
| | - Yi-Hua Jan
- Department of Medical Informatic, ACT Genomics Co., Ltd., Taipei 100, Taiwan; (K.-T.T.); (Y.-H.J.)
| | - Pei-Hang Lee
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
| | - Chih-Hao Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
| | - Shih-Chen Yu
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
| | - Cheng-Feng Tsao
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Jui-Chu Wang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (T.-T.L.); (P.-H.L.); (C.-H.H.); (S.-C.Y.); (J.-C.W.)
| |
Collapse
|
5
|
Sun KH(M, Wong YT(H, Cheung KM(C, Yuen C(M, Chan YT(T, Lai WY(J, Chao C(D, Fan WS(K, Chow YK(K, Law MF, Tam HC(T. Update on Molecular Diagnosis in Extranodal NK/T-Cell Lymphoma and Its Role in the Era of Personalized Medicine. Diagnostics (Basel) 2022; 12:diagnostics12020409. [PMID: 35204500 PMCID: PMC8871212 DOI: 10.3390/diagnostics12020409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK)/T-cell lymphoma (NKTCL) is an aggressive malignancy with unique epidemiological, histological, molecular, and clinical characteristics. It occurs in two pathological forms, namely, extranodal NKTCL (ENKTCL) and aggressive NK leukemia, according to the latest World Health Organization (WHO) classification. Epstein–Barr virus (EBV) infection has long been proposed as the major etiology of lymphomagenesis. The adoption of high-throughput sequencing has allowed us to gain more insight into the molecular mechanisms of ENKTCL, which largely involve chromosome deletion and aberrations in Janus kinase (JAK)-signal transducer and activator of transcription (STAT), programmed cell death protein-1 (PD-1)/PD-ligand 1 (PD-L1) pathways, as well as mutations in tumor suppressor genes. The molecular findings could potentially influence the traditional chemoradiotherapy approach, which is known to be associated with significant toxicity. This article will review the latest molecular findings in NKTCL and recent advances in the field of molecular diagnosis in NKTCL. Issues of quality control and technical difficulties will also be discussed, along with future prospects in the molecular diagnosis and treatment of NKTCL.
Collapse
Affiliation(s)
- Ka-Hei (Murphy) Sun
- Division of Hematopathology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Hong Kong; (K.-H.S.); (C.Y.)
| | | | - Ka-Man (Carmen) Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Carmen (Michelle) Yuen
- Division of Hematopathology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Hong Kong; (K.-H.S.); (C.Y.)
| | - Yun-Tat (Ted) Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Wing-Yan (Jennifer) Lai
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Chun (David) Chao
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Wing-Sum (Katie) Fan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Yuen-Kiu (Karen) Chow
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Man-Fai Law
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
- Correspondence:
| | - Ho-Chi (Tommy) Tam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| |
Collapse
|
6
|
Hsu JY, Seligson ND, Hays JL, Miles WO, Chen JL. Clinical Utility of CDK4/6 Inhibitors in Sarcoma: Successes and Future Challenges. JCO Precis Oncol 2022; 6:e2100211. [PMID: 35108033 PMCID: PMC8820917 DOI: 10.1200/po.21.00211] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/08/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Soft tissue and bone sarcomas are rare malignancies that exhibit significant pathologic and molecular heterogeneity. Deregulation of the CDKN2A-CCND-CDK4/6-retinoblastoma 1 (Rb) pathway is frequently observed in about 25% of unselected sarcomas and is pathognomonic for specific sarcoma subtypes. This genomic specificity has fueled the clinical evaluation of selective CDK4/6 inhibitors in sarcomas. Here, we highlight successes, opportunities, and future challenges for using CDK4/6 inhibitors to treat sarcoma. MATERIALS AND METHODS This review summarizes the current evidence for the use of CDK4/6 inhibitors in sarcoma while identifying molecular rationale and predictive biomarkers that provide the foundation for targeting the CDK4/6 pathway in sarcoma. A systematic review was performed of articles indexed in the PubMed database and the National Institutes of Health Clinical Trials Registry (ClinicalTrials.gov). For each sarcoma subtype, we discuss the preclinical rationale, case reports, and available clinical trials data. RESULTS Despite promising clinical outcomes in a subset of sarcomas, resistance to CDK4/6 inhibitors results in highly heterogeneous clinical outcomes. Current clinical data support the use of CDK4/6 inhibitors in subsets of sarcoma primarily driven by CDK4/6 deregulation. When dysregulation of the Rb pathway is a secondary driver of sarcoma, combination therapy with CDK4/6 inhibition may be an option. Developing strategies to identify responders and the mechanisms that drive resistance is important to maximize the clinical utility of these drugs in patients with sarcoma. Potential biomarkers that indicate CDK4/6 inhibitor sensitivity in sarcoma include CDK4, CCND, CCNE, RB1, E2F1, and CDKN2A. CONCLUSION CDK4/6 inhibitors represent a major breakthrough for targeted cancer treatment. CDK4/6 inhibitor use in sarcoma has led to limited, but significant, early clinical success. Targeted future clinical research will be key to unlocking the potential of CDK4/6 inhibition in sarcoma.
Collapse
Affiliation(s)
- Jocelyn Y. Hsu
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Nathan D. Seligson
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Department of Pharmacotherapy and Translational Research, University of Florida, Jacksonville, FL
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Nemours Children's Specialty Care, Jacksonville, FL
| | - John L. Hays
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH
| | - Wayne O. Miles
- Department of Molecular Genetics, The Ohio State University, Columbus, OH
| | - James L. Chen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Division of Bioinformatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH
| |
Collapse
|
7
|
Ordulu Z, Chai H, Peng G, McDonald AG, De Nictolis M, Garcia-Fernandez E, Hardisson D, Prat J, Li P, Hui P, Oliva E, Buza N. Molecular and clinicopathologic characterization of intravenous leiomyomatosis. Mod Pathol 2020; 33:1844-1860. [PMID: 32341498 PMCID: PMC7483566 DOI: 10.1038/s41379-020-0546-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
Intravenous leiomyomatosis (IVL) is an unusual uterine smooth muscle proliferation that can be associated with aggressive clinical behavior despite a histologically benign appearance. It has some overlapping molecular characteristics with both uterine leiomyoma and leiomyosarcoma based on limited genetic data. In this study, we assessed the clinical and morphological characteristics of 28 IVL and their correlation with molecular features and protein expression, using array comparative genomic hybridization (aCGH) and Cyclin D1, p16, phosphorylated-Rb, SMARCB1, SOX10, CAIX, SDHB and FH immunohistochemistry. The most common morphologies were cellular (n = 15), usual (n = 11), and vascular (n = 5; including 3 cellular IVL showing both vascular and cellular features). Among the immunohistochemical findings, the most striking was that all IVL showed differential expression of either p16 or Cyclin D1 in comparison to surrounding nonneoplastic tissue. Cytoplasmic phosphorylated-Rb was present in all but one IVL with hyalinization. SMARCB1, FH, and SDHB were retained; S0X10 and CAIX were not expressed. The most common genetic alterations involved 1p (39%), 22q (36%), 2q (29%), 1q (25%), 13q (21%), and 14q (21%). Hierarchical clustering analysis of recurrent aberrations revealed three molecular groups: Groups 1 (29%) and 2 (18%) with associated del(22q), and Group 3 (18%) with del(10q). The remaining IVL had nonspecific or no alterations by aCGH. Genomic index scores were calculated for all cases and showed no significant difference between the 14 IVL associated with aggressive clinical behavior (extrauterine extension or recurrence) and those without (median scores 5.15 vs 3.5). Among the 5 IVL associated with recurrence, 4 had a vascular morphology and 3 had alterations of 8q. Recurrent chromosome alterations detected herein overlap with those observed in the spectrum of uterine smooth muscle tumors and involve genes implicated in mesenchymal tumors at different sites with distinct morphological features.
Collapse
Affiliation(s)
- Zehra Ordulu
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Hongyan Chai
- Department of Genetics, Yale University School of Medicine; New Haven, CT
| | - Gang Peng
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
| | - Anna G McDonald
- Department of Pathology, Wake Forest Baptist Medical Center, Winston Salem, NC
| | | | - Eugenia Garcia-Fernandez
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, and Faculty of Medicine, Universidad Autónoma de Madrid, Spain
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, and Faculty of Medicine, Universidad Autónoma de Madrid, Spain
| | - Jaime Prat
- Department of Pathology, Hospital de la Sta Creu i Sant Pau, Barcelona, Spain
| | - Peining Li
- Department of Genetics, Yale University School of Medicine; New Haven, CT
| | - Pei Hui
- Department of Pathology, Yale University School of Medicine; New Haven, CT
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Natalia Buza
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Wang L, Li LR, Zhang L, Wang JW. The landscape of new drugs in extranodal NK/T-cell lymphoma. Cancer Treat Rev 2020; 89:102065. [PMID: 32653806 DOI: 10.1016/j.ctrv.2020.102065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
To date, much progress has been made in early-stage extranodal NK/T-cell lymphoma (ENKTCL), and risk-adapted therapy with radiotherapy (RT) alone for the low-risk group and RT combined with asparaginase-based chemotherapy (CT) for the high-risk group yields favorable outcomes. However, optimal treatment strategies have not been defined yet for advanced-stage ENKTCL. Historically, ENKTCL responded poorly to conventional anthracycline-based chemotherapy probably because of inherent multidrug resistance (MDR). The fact that ENKTCL cells lack asparagine synthetase (ASNS) activity warranted the use of L-asparaginase or pegaspargase as frontline chemotherapies. Even though, due to high mortality of the disease, approximately 50% patients failing the frontline therapy arrived at dismal clinical outcomes with a median progression-free survival (PFS) less than 8 months. As distinctive molecular and biological subgroups are increasingly discovered within the disease entity of ENKTCL, novel targeted therapies and immunotherapy are of the urgent need for those heterogeneous subgroups. In this review, we sought to summarize the preclinical and clinical results of 6 categories of promising targeted therapy and immunotherapy for the treatment of ENKTCL, including monoclonal antibodies, immune checkpoint inhibitors, small-molecular inhibitors, epigenetic therapy, immunomodulatory drugs, and adoptive T-cell therapy, and these might change the landscape of treatment for ENKTCL in the near future.
Collapse
Affiliation(s)
- Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing Tongren Hospital, Beijing 100730, China.
| | - Lin-Rong Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing 100032, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing 100730, China
| | - Jing-Wen Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
9
|
Ravegnini G, Serrano C, Simeon V, Sammarini G, Nannini M, Roversi E, Urbini M, Ferrè F, Ricci R, Tarantino G, Pantaleo MA, Hrelia P, Angelini S. The rs17084733 variant in the KIT 3' UTR disrupts a miR-221/222 binding site in gastrointestinal stromal tumour: a sponge-like mechanism conferring disease susceptibility. Epigenetics 2019; 14:545-557. [PMID: 30983504 PMCID: PMC6557610 DOI: 10.1080/15592294.2019.1595997] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Several miRNAs are dysregulated in gastrointestinal stromal tumours (GIST), and miR-221/222 appear to have a prominent role in GIST biology. Therefore, we investigated the role of DNA variants located in miR-221/222 precursor sequences and their target KIT 3'UTR. Ninety-five polymorphisms were analysed in 115 GIST cases and 88 healthy controls. KIT 3'UTR rs17084733 and pri-miR-222 rs75246947 were found significantly associated with GIST susceptibility. Specifically, KIT rs17084733 A allele was more common in GIST, particularly in KIT wild-type (WT) patients (Padj = 0.017). rs17084733 variant is located within one of the three miR-221/222 binding sites in the KIT 3'UTR, resulting in a mismatch in this seed region. Conversely, KIT mRNA levels were lower in patients carrying the variant allele, except for KIT mutant GIST. Luciferase assay data in GIST cells, generated using a construct containing all the three miR-221/222 binding sites, are consistent with KIT mRNA levels in GIST patients. Reporter assay data, generated using a construct containing only the site encompassing rs17084733, confirmed that this is a functional variant disrupting the miR-221/222 binding site. In conclusion, this is the first study investigating the role of SNPs on miR-221/222 precursor sequences and their binding region on KIT 3'UTR in GIST. We identified the KIT variant rs17084733 as a possible novel genetic biomarker for risk of developing KIT-WT GIST. Moreover, our findings suggest the role of one of the three miR-221/222 binding sites on KIT 3'UTR as endogenous sponge, soaking up and subtracting miR-221/222 to the other two sites characterized by a higher affinity.
Collapse
Affiliation(s)
- Gloria Ravegnini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - César Serrano
- b Medical Oncology Department , Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital , Barcelona , Spain
| | - Vittorio Simeon
- c Medical Statistics Unit , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Giulia Sammarini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Margherita Nannini
- d Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| | - Erica Roversi
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Milena Urbini
- e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Fabrizio Ferrè
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Riccardo Ricci
- f UOC di Anatomia Patologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.,g Department of Pathology , Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Tarantino
- e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Maria A Pantaleo
- d Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy.,e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Patrizia Hrelia
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Sabrina Angelini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| |
Collapse
|
10
|
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
|
11
|
Joglekar-Javadekar M, Van Laere S, Bourne M, Moalwi M, Finetti P, Vermeulen PB, Birnbaum D, Dirix LY, Ueno N, Carter M, Rains J, Ramachandran A, Bertucci F, van Golen KL. Characterization and Targeting of Platelet-Derived Growth Factor Receptor alpha (PDGFRA) in Inflammatory Breast Cancer (IBC). Neoplasia 2017; 19:564-573. [PMID: 28609680 PMCID: PMC5470553 DOI: 10.1016/j.neo.2017.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/05/2023] Open
Abstract
PURPOSE: Inflammatory breast cancer (IBC) is arguably the deadliest form of breast cancer due to its rapid onset and highly invasive nature. IBC carries 5- and 10-year disease-free survival rates of ~45% and <20%, respectively. Multiple studies demonstrate that in comparison with conventional breast cancer, IBC has a unique molecular identity. Here, we have identified platelet-derived growth factor receptor alpha (PDGFRA) as being uniquely expressed and active in IBC patient tumor cells. EXPERIMENTAL DESIGN: Here we focus on characterizing and targeting PDGFRA in IBC. Using gene expression, we analyzed IBC patient samples and compared them with non-IBC patient samples. Further, using IBC cells in culture, we determined the effect of small molecules inhibitors in both in vitro and in vivo assays. RESULTS: In IBC patients, we show more frequent PDGFRA activation signature than non-IBC samples. In addition, the PDGFRA activation signature is associated with shorter metastasis-free survival in both uni- and multivariate analyses. We also demonstrate that IBC cells express active PDGFRA. Finally, we show that PDGFRA targeting by crenolanib (CP-868-596), but not imatinib (STI571), two small molecule inhibitors, interferes with IBC cell growth and emboli formation in vitro and tumor growth in vivo. CONCLUSIONS: Our data suggest that PDGFRA may be a promising target for therapy in IBC.
Collapse
Affiliation(s)
- Madhura Joglekar-Javadekar
- The Laboratory for Cytoskeletal Physiology, Department of Biological Sciences, The University of Delaware, Newark, DE; The Center for Translational Cancer Research, The University of Delaware, Newark, DE
| | - Steven Van Laere
- Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Michael Bourne
- The Laboratory for Cytoskeletal Physiology, Department of Biological Sciences, The University of Delaware, Newark, DE; The Center for Translational Cancer Research, The University of Delaware, Newark, DE
| | - Manal Moalwi
- The Laboratory for Cytoskeletal Physiology, Department of Biological Sciences, The University of Delaware, Newark, DE; The Center for Translational Cancer Research, The University of Delaware, Newark, DE
| | - Pascal Finetti
- Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Peter B Vermeulen
- Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Daniel Birnbaum
- Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Luc Y Dirix
- Center for Oncological Research, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Naoto Ueno
- Breast Cancer Translational Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Breast Medical Oncology, Morgan Welch Inflammatory Breast Cancer Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Francois Bertucci
- Department of Molecular Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes (IPC), Marseille, France
| | - Kenneth L van Golen
- The Laboratory for Cytoskeletal Physiology, Department of Biological Sciences, The University of Delaware, Newark, DE; The Center for Translational Cancer Research, The University of Delaware, Newark, DE; The Helen F. Graham Cancer Center, Newark, DE.
| |
Collapse
|
12
|
Zhou J, Xu J, Jiang G, Ma Y, Qi J, Li W, Zhang D. Gastrointestinal stromal tumor with a PDGFRA mutation masquerading as gastric plexiform fibromyxoma: A comparative clinicopathological study of two cases. Oncol Lett 2016; 13:887-892. [PMID: 28356974 PMCID: PMC5351284 DOI: 10.3892/ol.2016.5486] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/26/2016] [Indexed: 12/13/2022] Open
Abstract
Gastric plexiform fibromyxoma (PF) is a rare mesenchymal tumor with a histologically distinctive multinodular pattern, dissimilar to conventional gastrointestinal stromal tumor (GIST). The current study presents one case of gastric PF, and one case of GIST with a platelet-derived growth factor receptor α (PDGFRA) mutation mimicking PF, and discusses their differential diagnoses. The two patients were a 51-year-old male with PF and a 47-year-old female with GIST, each of whom presented with an occupying lesion in the gastric antrum. Histologically, the two cases shared a rare and approximately unanimous morphological pattern of a prominent multinodular and plexiform figuration in the gastric wall, including mucoid matrix, short spindle cells and small caliber vascular elements, and areas of stromal tumor cells exhibited an epithelioid appearance. Immunohistochemistry revealed that the PF tumor cells were positive for smooth muscle actin (SMA), but negative for mast/stem cell growth factor receptor (KIT), GIST-1 (DOG1), cluster of differentiation (CD) 34, S-100, desmin and cytokeratin AE1/AE3. The case of GIST expressed KIT and DOG1, but was negative for SMA, CD34, S-100, desmin and AE1/AE3. In addition, the GIST case, which was observed to harbor a D842V mutation in exon 18 of PDGFRA, was demonstrated to be genetically distinct from PF. The cases presented in the current study were uncommon in that GIST exhibited a plexiform appearance that mimicked the histology of the rare PF tumor; therefore, GIST must be considered and discounted first when determining a differential diagnosis for a gastrointestinal mesenchymal neoplasm.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jingjing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yihui Ma
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jingwen Qi
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dandan Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
13
|
Belinsky MG, Rink L, Cai KQ, Capuzzi SJ, Hoang Y, Chien J, Godwin AK, von Mehren M. Somatic loss of function mutations in neurofibromin 1 and MYC associated factor X genes identified by exome-wide sequencing in a wild-type GIST case. BMC Cancer 2015; 15:887. [PMID: 26555092 PMCID: PMC4641358 DOI: 10.1186/s12885-015-1872-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/30/2015] [Indexed: 12/25/2022] Open
Abstract
Background Approximately 10–15 % of gastrointestinal stromal tumors (GISTs) lack gain of function mutations in the KIT and platelet-derived growth factor receptor alpha (PDGFRA) genes. An alternate mechanism of oncogenesis through loss of function of the succinate-dehydrogenase (SDH) enzyme complex has been identified for a subset of these “wild type” GISTs. Methods Paired tumor and normal DNA from an SDH-intact wild-type GIST case was subjected to whole exome sequencing to identify the pathogenic mechanism(s) in this tumor. Selected findings were further investigated in panels of GIST tumors through Sanger DNA sequencing, quantitative real-time PCR, and immunohistochemical approaches. Results A hemizygous frameshift mutation (p.His2261Leufs*4), in the neurofibromin 1 (NF1) gene was identified in the patient’s GIST; however, no germline NF1 mutation was found. A somatic frameshift mutation (p.Lys54Argfs*31) in the MYC associated factor X (MAX) gene was also identified. Immunohistochemical analysis for MAX on a large panel of GISTs identified loss of MAX expression in the MAX-mutated GIST and in a subset of mainly KIT-mutated tumors. Conclusion This study suggests that inactivating NF1 mutations outside the context of neurofibromatosis may be the oncogenic mechanism for a subset of sporadic GIST. In addition, loss of function mutation of the MAX gene was identified for the first time in GIST, and a broader role for MAX in GIST progression was suggested. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1872-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin G Belinsky
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA.
| | - Lori Rink
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA.
| | - Kathy Q Cai
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Stephen J Capuzzi
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA. .,Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, USA.
| | - Yen Hoang
- Department of Bioinformatics and Biosystems Technology, University of Applied Sciences Wildau, Wildau, Germany. .,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Jeremy Chien
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Margaret von Mehren
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA.
| |
Collapse
|
14
|
Nannini M, Ravegnini G, Angelini S, Astolfi A, Biasco G, Pantaleo MA. miRNA profiling in gastrointestinal stromal tumors: implication as diagnostic and prognostic markers. Epigenomics 2015; 7:1033-49. [PMID: 26447534 DOI: 10.2217/epi.15.52] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are a class of short noncoding RNAs, that play a relevant role in multiple biological processes, such as differentiation, proliferation and apoptosis. Gastrointestinal stromal tumors (GIST) are considered as a paradigm of molecular biology in solid tumors worldwide, and after the discovery of specific alterations in the KIT and PDGFRA genes, they have emerged from anonymity to become a model for targeted therapy. Epigenetics have an emerging and relevant role in different steps of GIST biology such as tumorigenesis, disease progression, prognosis and drug resistance. The aim of the present review was to summarize the current evidence about the role of microRNAs in GIST, including their potential application as well as their limits.
Collapse
Affiliation(s)
- Margherita Nannini
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy & Biotechnology, FaBit; University of Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy & Biotechnology, FaBit; University of Bologna, Italy
| | - Annalisa Astolfi
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Guido Biasco
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,"Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Maria A Pantaleo
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,"Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| |
Collapse
|
15
|
Pessetto ZY, Ma Y, Hirst JJ, von Mehren M, Weir SJ, Godwin AK. Drug repurposing identifies a synergistic combination therapy with imatinib mesylate for gastrointestinal stromal tumor. Mol Cancer Ther 2014; 13:2276-87. [PMID: 25122069 DOI: 10.1158/1535-7163.mct-14-0043] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gastrointestinal stromal tumor (GIST) is a rare and therefore often neglected disease. Introduction of the kinase inhibitor imatinib mesylate radically improved the clinical response of patients with GIST; however, its effects are often short-lived, with GISTs demonstrating a median time-to-progression of approximately two years. Although many investigational drugs, approved first for other cancers, have been subsequently evaluated for the management of GIST, few have greatly affected the overall survival of patients with advanced disease. We employed a novel, focused, drug-repurposing effort for GIST, including imatinib mesylate-resistant GIST, evaluating a large library of FDA-approved drugs regardless of current indication. As a result of the drug-repurposing screen, we identified eight FDA-approved drugs, including fludarabine phosphate (F-AMP), that showed synergy with and/or overcame resistance to imatinib mesylate. F-AMP induces DNA damage, Annexin V, and caspase-3/7 activities as the cytotoxic effects on GIST cells, including imatinib mesylate-resistant GIST cells. F-AMP and imatinib mesylate combination treatment showed greater inhibition of GIST cell proliferation when compared with imatinib mesylate and F-AMP alone. Successful in vivo experiments confirmed the combination of imatinib mesylate with F-AMP enhanced the antitumor effects compared with imatinib mesylate alone. Our results identified F-AMP as a promising, repurposed drug therapy for the treatment of GISTs, with potential to be administered in combination with imatinib mesylate or for treatment of imatinib mesylate-refractory tumors.
Collapse
Affiliation(s)
- Ziyan Y Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Ma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jeff J Hirst
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Scott J Weir
- Department of Pharmacology, Toxicology and Therapeutics, Kansas City, Kansas. Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, Kansas. University of Kansas Cancer Center, Kansas City, Kansas
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas. University of Kansas Cancer Center, Kansas City, Kansas.
| |
Collapse
|
16
|
Patruno R, Marech I, Zizzo N, Ammendola M, Nardulli P, Gadaleta C, Introna M, Capriuolo G, Rubini RA, Ribatti D, Gadaleta CD, Ranieri G. c-Kit expression, angiogenesis, and grading in canine mast cell tumour: a unique model to study c-Kit driven human malignancies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:730246. [PMID: 24900982 PMCID: PMC4036613 DOI: 10.1155/2014/730246] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/18/2014] [Indexed: 12/31/2022]
Abstract
Canine cutaneous mast cell tumour (CMCT) is a c-Kit driven tumour sharing similar c-Kit aberrations found in human gastrointestinal stromal tumour. CMCT is classified into three forms: well- (G1), intermediately (G2) (more benign diseases), and poorly (G3) differentiated (malignant) forms. We assess a correlation between c-Kit status, grading, and angiogenesis in CMCTs to explore their potential significance in humans. C-Kit receptor (c-KitR) expression, microvascular density (MVD), and mast cell granulated and degranulated status density (MCGD and MCDD, resp.) were analyzed in 97 CMCTs, by means of histochemistry, immunohistochemistry double staining, and image analysis system. Data showed that predominantly diffuse cytoplasmic- and predominantly focal paranuclear- (Golgi-like) c-Kit protein (PDC-c-Kit and PFP-c-Kit, resp.) expression correlate with high MVD, G3 histopathological grade, and MCDD. Moreover, predominant cell membrane-c-KitR (PCM-c-KitR) expression status correlates with low MVD, G1-G2 histopathological grade, and MCGD. These findings underline the key role of c-Kit in the biopathology of canine MCTs, indicating a link between aberrant c-Kit expression, increased angiogenesis, and higher histopathological grade. CMCT seems to be a model to study contributions of c-Kit activated MCs in tumour angiogenesis and to evaluate the inhibition of MCs activation by means of c-Kit tyrosine kinase inhibitors, currently translated in humans.
Collapse
Affiliation(s)
- Rosa Patruno
- Animal Health Unit, Department of Prevention, ASL BAT, Via Andria 176, 70051 Barletta, Italy
| | - Ilaria Marech
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Via Orazio Flacco 65, 70124 Bari, Italy
| | - Nicola Zizzo
- Chair of Pathology, University of Bari, Via Casamassima, 70010 Valenzano, Italy
| | - Michele Ammendola
- Chair of Clinical Surgery, University of Catanzaro, Via Europa, 88100 Germaneto, Italy
| | - Patrizia Nardulli
- Pharmacy Unit, National Cancer Research Centre, “Giovanni Paolo II”, Via Orazio Flacco 65, 70100 Bari, Italy
| | - Claudia Gadaleta
- Chair of Pathology, University of Bari, Via Casamassima, 70010 Valenzano, Italy
| | - Marcello Introna
- Chair of Pathology, University of Bari, Via Casamassima, 70010 Valenzano, Italy
| | - Gennaro Capriuolo
- Animal Health Unit, Department of Prevention, ASL BAT, Via Andria 176, 70051 Barletta, Italy
| | - Rosa Angela Rubini
- Animal Health Unit, Department of Prevention, ASL BAT, Via Andria 176, 70051 Barletta, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari, Piazzale Giulio Cesare 11, 70124 Bari, Italy
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Via Orazio Flacco 65, 70124 Bari, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, Via Orazio Flacco 65, 70124 Bari, Italy
| |
Collapse
|
17
|
Yamaguchi Y, Takenobu H, Ohira M, Nakazawa A, Yoshida S, Akita N, Shimozato O, Iwama A, Nakagawara A, Kamijo T. Novel 1p tumour suppressor Dnmt1-associated protein 1 regulates MYCN/ataxia telangiectasia mutated/p53 pathway. Eur J Cancer 2014; 50:1555-65. [PMID: 24559687 DOI: 10.1016/j.ejca.2014.01.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/27/2014] [Indexed: 12/11/2022]
Abstract
Neuroblastoma (NB) is a paediatric solid tumour which originates from sympathetic nervous tissues. Deletions in chromosome 1p are frequently found in unfavourable NBs and are correlated with v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN) amplification; however, it remains to be elucidated how the 1p loss contributes to MYCN-related oncogenic processes in NB. In this study, we identified the role of Dnmt1-associated protein 1 (DMAP1), coded on chromosome 1p34, in the processes. We studied the expression and function of DMAP1 in NB and found that low-level expression of DMAP1 related to poor prognosis, unfavourable histology and 1p Loss of heterozygosity (LOH) of primary NB samples. Intriguingly, DMAP1 induced ataxia telangiectasia mutated (ATM) phosphorylation and focus formation in the presence of a DNA damage reagent, doxorubicin. By DMAP1 expression in NB and fibroblasts, p53 was activated in an ATM-dependent manner and p53-downstream pro-apoptotic Bcl-2 family molecules were induced at the mRNA level, resulting in p53-induced apoptotic death. BAX and p21(Cip1/Waf1) promoter activity dependent on p53 was clearly up-regulated by DMAP1. Further, MYCN transduction in MYCN single-copy NB cells accelerated doxorubicin (Doxo)-induced apoptotic cell death; MYCN is implicated in DMAP1 protein stabilisation and ATM phosphorylation in these situations. DMAP1 knockdown attenuated MYCN-dependent ATM phosphorylation and NB cell apoptosis. Together, DMAP1 appears to be a new candidate for a 1p tumour suppressor and its reduction contributes to NB tumourigenesis via inhibition of MYCN-related ATM/p53 pathway activation.
Collapse
Affiliation(s)
- Yohko Yamaguchi
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan
| | - Hisanori Takenobu
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan
| | - Miki Ohira
- Laboratory of Cancer Genomics, Chiba Cancer Center Research Institute, Japan
| | - Atsuko Nakazawa
- Department of Pathology, National Center for Child Health and Development, Japan
| | - Sayaka Yoshida
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan
| | - Nobuhiro Akita
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan
| | - Osamu Shimozato
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Nakagawara
- Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Japan
| | - Takehiko Kamijo
- Division of Biochemistry and Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Japan.
| |
Collapse
|
18
|
Schaefer IM, Delfs C, Cameron S, Gunawan B, Agaimy A, Ghadimi BM, Haller F. Chromosomal aberrations in primary PDGFRA-mutated gastrointestinal stromal tumors. Hum Pathol 2013; 45:85-97. [PMID: 24157063 DOI: 10.1016/j.humpath.2013.05.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 05/21/2013] [Accepted: 05/21/2013] [Indexed: 01/08/2023]
Abstract
Approximately 15% of gastrointestinal stromal tumors (GISTs) harbor mutations in the platelet-derived growth factor receptor α (PDGFRA) gene. Chromosomal aberrations play a crucial role in tumor progression and correlate with clinical behavior. Imbalances, particularly in PDGFRA-mutated GISTs, have not yet been evaluated in larger series. We analyzed 53 PDGFRA-mutated GISTs (including 2 with corresponding metastases) for chromosomal imbalances by conventional comparative genomic hybridization and compared them with a historical collective of 122 KIT-mutated GISTs. PDGFRA exon 18 mutations (91% of cases) and exon 12 mutations (9% of cases) correlated significantly with gastric and intestinal sites, respectively. The most common aberrations were identical to those found in KIT-mutated GISTs, with -14q in 70%, -1p in 28%, and -22q in 17% of cases. Overall, there were significantly fewer chromosomal aberrations compared with KIT-mutated GISTs, with a mean of 2.8 (0.6 gains, 2.1 losses) aberrations per tumor. There was a statistically significant association of more than 5 chromosomal imbalances with intermediate/high-risk categories. Regarding specific chromosomal aberrations, -9p, -13q, and -22q correlated with intermediate/high risk, and -1p and +8q with poorer survival, although progression occurred in only 2 cases. Altogether, PDGFRA-mutated GISTs display the same chromosomal aberrations as KIT-mutated GISTs, although they have a lower degree of chromosomal instability in line with their generally favorable outcome.
Collapse
Affiliation(s)
- Inga-Marie Schaefer
- Department of Pathology, University Medical Center Göttingen, Robert-Koch-Straße 40, Göttingen D-37075, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Wang X, Zhou CX, Yan JW, Hou JQ, Chen SB, Ou TM, Gu LQ, Huang ZS, Tan JH. Synthesis and Evaluation of Quinazolone Derivatives as a New Class of c-KIT G-Quadruplex Binding Ligands. ACS Med Chem Lett 2013; 4:909-14. [PMID: 24900584 DOI: 10.1021/ml400271y] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 08/13/2013] [Indexed: 01/12/2023] Open
Abstract
The c-KIT G-quadruplex structures are a novel class of attractive targets for the treatment of gastrointestinal stromal tumor (GIST). Herein, a series of new quinazolone derivatives with the expansion of unfused aromatic ring system were designed and synthesized. Subsequent biophysical studies demonstrated that the derivatives with adaptive scaffold could effectively bind to and stabilize c-KIT G-quadruplexes with good selectivity against duplex DNA. More importantly, these ligands further inhibited the transcription and expression of c-KIT gene and exhibited significant cytotoxicity on the GIST cell line HGC-27. Overall, these quinazolone derivatives represent a new class of promising c-KIT G-quadruplex ligands. The experimental results have also reinforced the idea of inhibition of c-KIT expression through targeting c-KIT G-quadruplex DNA.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chen-Xi Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Wu Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Qiang Hou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shuo-Bin Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tian-Miao Ou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Lian-Quan Gu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhi-Shu Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jia-Heng Tan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
20
|
Lee EJ, Kang G, Kang SW, Jang KT, Lee J, Park JO, Park CK, Sohn TS, Kim S, Kim KM. GSTT1 copy number gain and ZNF overexpression are predictors of poor response to imatinib in gastrointestinal stromal tumors. PLoS One 2013; 8:e77219. [PMID: 24124608 PMCID: PMC3790698 DOI: 10.1371/journal.pone.0077219] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 09/01/2013] [Indexed: 01/25/2023] Open
Abstract
Oncogenic mutations in gastrointestinal stromal tumors (GISTs) predict prognosis and therapeutic responses to imatinib. In wild-type GISTs, the tumor-initiating events are still unknown, and wild-type GISTs are resistant to imatinib therapy. We performed an association study between copy number alterations (CNAs) identified from array CGH and gene expression analyses results for four wild-type GISTs and an imatinib-resistant PDGFRA D842V mutant GIST, and compared the results to those obtained from 27 GISTs with KIT mutations. All wild-type GISTs had multiple CNAs, and CNAs in 1p and 22q that harbor the SDHB and GSTT1 genes, respectively, correlated well with expression levels of these genes. mRNA expression levels of all SDH gene subunits were significantly lower (P≤0.041), whereas mRNA expression levels of VEGF (P=0.025), IGF1R (P=0.026), and ZNFs (P<0.05) were significantly higher in GISTs with wild-type/PDGFRA D842V mutations than GISTs with KIT mutations. qRT-PCR validation of the GSTT1 results in this cohort and 11 additional malignant GISTs showed a significant increase in the frequency of GSTT1 CN gain and increased mRNA expression of GSTT1 in wild-type/PDGFRA D842V GISTs than KIT-mutant GISTs (P=0.033). Surprisingly, all four malignant GISTs with KIT exon 11 deletion mutations with primary resistance to imatinib had an increased GSTT1 CN and mRNA expression level of GSTT1. Increased mRNA expression of GSTT1 and ZNF could be predictors of a poor response to imatinib. Our integrative approach reveals that for patients with wild-type (or imatinib-resistant) GISTs, attempts to target VEGFRs and IGF1R may be reasonable options.
Collapse
Affiliation(s)
- Eui Jin Lee
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Guhyun Kang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pathology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Shin Woo Kang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Mathematics, Korea University, Seoul, Korea
| | - Kee-Taek Jang
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeeyun Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheol Keun Park
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Sung Sohn
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
21
|
Yang H, Shen C, Zhang B, Chen H, Chen Z, Chen J. Expression and clinicopathological significance of CD9 in gastrointestinal stromal tumor. J Korean Med Sci 2013; 28:1443-8. [PMID: 24133347 PMCID: PMC3792597 DOI: 10.3346/jkms.2013.28.10.1443] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/12/2013] [Indexed: 02/06/2023] Open
Abstract
This study investigated the expression and clinicopathological significance of CD9 in gastrointestinal stromal tumor (GIST). Immunohistochemistry staining for CD9 was performed on tumor tissues from 74 GIST patients. The correlation with clinicopathological features, risk classification and prognosis was analyzed. CD9-positive staining comprised 59.5% (44/74) of the GIST patients. The CD9-positive expression rate of the sample was significantly associated with diameter (P = 0.028), mitotic counts (P = 0.035), risk classification (P = 0.018) and three-year recurrence-free survival (RFS) (P < 0.001). Cox proportional hazards regression (HR = 0.352; P = 0.015) showed that CD9 is an independent factor for post-operative RFS. The subgroup analysis showed that CD9 expression in gastric stromal tumor (GST) is significantly associated with diameter (P = 0.031), risk classification (P = 0.023) and three-year RFS (P = 0.001). The Cox proportional hazards regression (HR = 0.104; P = 0.006) also showed that CD9 is an independent factor for RFS of GST. However, CD9 expression does not have a statistically significant correlation with clinicopathological features, risk classification, and prognosis in non-GST. In conclusion, CD9 expression in GIST appears to be associated with the recurrence and/or metastasis of GIST patients, especially in GST, which may indicate the important role of CD9 in the malignant biological behavior and prognosis of GST.
Collapse
Affiliation(s)
- Hongxin Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Gastrointestinal Surgery of the Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou Province, China
| | - Chaoyong Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haining Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhixin Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiaping Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
22
|
Wang L, Wei C. Spectroscopic and Biological Studies of Phenanthroline Compounds: Selective Recognition of Gene-Promoter G-Quadruplex DNAs Preferred over Duplex DNA. Chem Biodivers 2013; 10:1154-64. [DOI: 10.1002/cbdv.201200341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Indexed: 11/06/2022]
|
23
|
Belinsky MG, Rink L, von Mehren M. Succinate dehydrogenase deficiency in pediatric and adult gastrointestinal stromal tumors. Front Oncol 2013; 3:117. [PMID: 23730622 PMCID: PMC3656383 DOI: 10.3389/fonc.2013.00117] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/26/2013] [Indexed: 12/18/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) in adults are generally driven by somatic gain-of-function mutations in KIT or PDGFRA, and biological therapies targeted to these receptor tyrosine kinases comprise part of the treatment regimen for metastatic and inoperable GISTs. A minority (10-15%) of GISTs in adults, along with ∼85% of pediatric GISTs, lacks oncogenic mutations in KIT and PDGFRA. Not surprisingly these wild type (WT) GISTs respond poorly to kinase inhibitor therapy. A subset of WT GISTs shares a set of distinguishing clinical and pathological features, and a flurry of recent reports has convincingly demonstrated shared molecular characteristics. These GISTs have a distinct transcriptional profile including over-expression of the insulin-like growth factor-1 receptor, and exhibit deficiency in the succinate dehydrogenase (SDH) enzyme complex. The latter is often but not always linked to bi-allelic inactivation of SDH subunit genes, particularly SDHA. This review will summarize the molecular, pathological, and clinical connections that link this group of SDH-deficient neoplasms, and offer a view toward understanding the underlying biology of the disease and the therapeutic challenges implicit to this biology.
Collapse
Affiliation(s)
- Martin G. Belinsky
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| | - Lori Rink
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| | - Margaret von Mehren
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| |
Collapse
|
24
|
Pessetto ZY, Weir SJ, Sethi G, Broward MA, Godwin AK. Drug repurposing for gastrointestinal stromal tumor. Mol Cancer Ther 2013; 12:1299-309. [PMID: 23657945 DOI: 10.1158/1535-7163.mct-12-0968] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite significant treatment advances over the past decade, metastatic gastrointestinal stromal tumor (GIST) remains largely incurable. Rare diseases, such as GIST, individually affect small groups of patients but collectively are estimated to affect 25 to 30 million people in the United States alone. Given the costs associated with the discovery, development, and registration of new drugs, orphan diseases such as GIST are often not pursued by mainstream pharmaceutical companies. As a result, "drug repurposing" or "repositioning," has emerged as an alternative to the traditional drug development process. In this study, we screened 796 U.S. Food and Drug Administration (FDA)-approved drugs and found that two of these compounds, auranofin (Ridaura) and fludarabine phosphate, effectively and selectively inhibited the proliferation of GISTs, including imatinib-resistant cells. One of the most notable drug hits, auranofin, an oral, gold-containing agent approved by the FDA in 1985 for the treatment of rheumatoid arthritis, was found to inhibit thioredoxin reductase activity and induce reactive oxygen species (ROS) production, leading to dramatic inhibition of GIST cell growth and viability. Importantly, the anticancer activity associated with auranofin was independent of imatinib-resistant status, but was closely related to the endogenous and inducible levels of ROS. Coupled with the fact that auranofin has an established safety profile in patients, these findings suggest for the first time that auranofin may have clinical benefit for patients with GIST, particularly in those suffering from imatinib-resistant and recurrent forms of this disease.
Collapse
Affiliation(s)
- Ziyan Y Pessetto
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | | | |
Collapse
|
25
|
Huang Y, de Leval L, Gaulard P. Molecular underpinning of extranodal NK/T-cell lymphoma. Best Pract Res Clin Haematol 2013; 26:57-74. [DOI: 10.1016/j.beha.2013.04.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
26
|
Prolonged Therapy with Imatinib Mesylate before Surgery for Advanced Gastrointestinal Stromal Tumor Results of a Phase II Trial. Int J Surg Oncol 2012; 2012:761576. [PMID: 23316352 PMCID: PMC3534224 DOI: 10.1155/2012/761576] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Revised: 11/15/2012] [Accepted: 11/15/2012] [Indexed: 01/13/2023] Open
Abstract
Purpose. Proven efficacy of imatinib mesylate in gastrointestinal stromal tumour (GIST) has led to its use in advanced disease and, more recently, in adjuvant and neoadjuvant settings. The purpose of this study was to evaluate the optimal neoadjuvant imatinib duration to reduce the morbidity of surgery and increase the possibility of resection completeness in advanced tumours. Patients and Method. Patients with advanced GIST were enrolled into a registered open-label multicenter trial and received imatinib daily for a maximum of 12 months, followed by en bloc resection. Data were prospectively collected regarding tumour assessment, response rate, surgical characteristics, recurrence, and survival. Results. Fourteen patients with advanced GIST were enrolled. According to RECIST criteria, 6 patients had partial response and 8 had stable disease. The overall tumour size reduction was 25% (0–62.5%), and there was no tumour progression. Eleven patients underwent tumour resection, and all had R0 resection. After a median followup of 48 months, 4-year OS and DFS were 100% and 64%, respectively. Conclusion. This prospective trial showed that one year of neoadjuvant imatinib in advanced GIST is safe and associated with high rate of complete microscopic resection. It is not associated with increased resistance, progression, or complication rates.
Collapse
|
27
|
Wilmanns C, Steinhauer S, Großmann J, Schmitt-Gräff A, Ruf G. Cooperate concept of metastasis: site-specific requirement of activated differentiation and dynamic deterioration. Cancer Metastasis Rev 2012; 31:269-76. [DOI: 10.1007/s10555-012-9350-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Birner P, Beer A, Vinatzer U, Stary S, Höftberger R, Nirtl N, Wrba F, Streubel B, Schoppmann SF. MAPKAP kinase 2 overexpression influences prognosis in gastrointestinal stromal tumors and associates with copy number variations on chromosome 1 and expression of p38 MAP kinase and ETV1. Clin Cancer Res 2012; 18:1879-87. [PMID: 22351694 DOI: 10.1158/1078-0432.ccr-11-2364] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE ETV1 has been proposed to be activated by KIT mutations in gastrointestinal stromal tumors (GIST). The aim of the study was to evaluate the clinical role of ETV1 and associated proteins in GIST. EXPERIMENTAL DESIGN Expressions of ETV1, MAPKAP kinase 2 (MAPKAPK2), phosphorylated p38 MAP kinase (pp38), phosphorylated MSK1 (pMSK1), phosphorylated RSK1, COP1, and KIT protein were determined immunohistochemically in 139 GISTs. Sequence analysis of KIT, PDGFRA, and MAPKAPK2 and FISHs of ETV1 as well as chromosomes 1 and 7 were done. RESULTS Prominent ETV1 expression was seen in 50% of GISTs, but no correlation with clinical outcome was found. Correlation of ETV1 expression and KIT mutation was seen in 60% of cases. MAPKAPK2 overexpression (n = 62/44.6%) correlated with pp38 expression (P = 0.021, χ(2) test) and alterations of chromosome 1 (n = 17, P = 0.024, χ(2) test). In one of 20 sequenced cases with high MAKAPK2 expression, a putative damaging MAPKAPK2 gene mutation was found. All relapsing GISTs with very low/low risk according to Fletcher showed high MAPKAPK2 and KIT expression. MAPKAPK2 overexpression was an independent prognostic factor for disease-free survival (P = 0.006, Cox regression). CONCLUSION ETV1 is not universally overexpressed in GIST and seems to also be induced by pathways other than KIT mutation. Nevertheless, its clinical relevance is low. Overexpression of ETV1 inhibitor MAPKAPK2 is associated with shorter survival in GIST, indicating a clinically relevant role of this gene not reported previously. Patients with low-risk GISTs showing MAPKAPK2 overexpression might profit from early adjuvant tyrosine kinase inhibitor therapy.
Collapse
Affiliation(s)
- Peter Birner
- Departments of Surgery and Obstetrics and Gynecology, Clinical Institute of Pathology, Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mrowiec S, Jabłońska B, Liszka L, Pająk J, Leidgens M, Szydło R, Sandecka A, Lampe P. Prognostic factors for survival post surgery for patients with gastrointestinal stromal tumors. ACTA ACUST UNITED AC 2011; 48:3-9. [PMID: 22179138 DOI: 10.1159/000334172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 08/15/2011] [Indexed: 01/18/2023]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms located in the alimentary tract. Our aim was to assess the influence of prognostic factors on survival in patients surgically treated for GISTs. STUDY One hundred and five patients treated between January 1989 and December 2008 were available for study. A retrospective analysis of prognostic factors (age, gender, mitotic index, tumor location, tumor size, risk of malignant behavior, and coexisting other neoplasm) was performed. Univariate and multivariate survival analyses were undertaken. RESULTS Univariate analyses revealed the importance of patient gender (p = 0.007), disease location (p = 0.055), mitotic index (p = 0.054) and coexistence with other neoplasms (p = 0.004). However, multivariate analysis showed 3 independently statistically significant factors: coexistence with other neoplasm (RR = 3.53, p = 0.004), male gender (RR = 2.60, p = 0.011) and mitotic index ≥10/50 HPF, (RR = 2.60, p = 0.042). CONCLUSIONS Our study has shown that male gender, a high mitotic index ≥10/50 HPF, and coexistence with other malignant neoplasms were independent poor prognostic factors in patients with GIST. The presence of middle or lower gut disease location leads to an increased risk of mortality when compared with the upper gut.
Collapse
Affiliation(s)
- Sławomir Mrowiec
- Department of Digestive Tract Surgery, University Hospital of the Medical University of Silesia, Katowice, Poland
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Namgung H. Gastrointestinal stromal tumor with KIT mutation in neurofibromatosis type 1. JOURNAL OF THE KOREAN SURGICAL SOCIETY 2011; 81:276-80. [PMID: 22111084 PMCID: PMC3219854 DOI: 10.4174/jkss.2011.81.4.276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/19/2011] [Accepted: 02/07/2011] [Indexed: 11/30/2022]
Abstract
Multiple jejunalgastrointestinal stromal tumors (GISTs) were found in a 52-year-old woman with a history of neurofibromatosis type 1. These tumors were composed of interlacing fascicles of uniform spindle cells with eosinophilic cytoplasm. Immunohistochemically, the tumor cells were positive for CD117, CD34 and negative for S-100, smooth muscle actin. Molecular analysis for activating mutations of KIT and PDGFRA was performed in two tumors. Contrary to sporadic GISTs, the NF1-associated GISTs are characterized by rare mutations of KIT or PDGFRA. But, one missense point mutation (Trp557Gly) was identified in KIT exon 11 of the extramural portion of the largest tumor in this case. The intramural portion of the largest tumor and the other tumor had wild type KIT and PDGFRA.
Collapse
Affiliation(s)
- Hwan Namgung
- Department of Surgery, Dankook University College of Medicine, Cheonan, Korea
| |
Collapse
|
31
|
Lee H, Kim A, Ahn IS, Joo SW, Lee SY, Yoon KA, Lee K. Colorimetric detection of c-Kit mutations using electrostatic attraction induced aggregation of peptide nucleic acid modified gold nanoparticles. Chem Commun (Camb) 2011; 47:11477-9. [PMID: 21952354 DOI: 10.1039/c1cc15280g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a colorimetric detection of c-Kit mutations using selective aggregation of the peptide nucleic acid modified gold nanoparticles that is caused by electrostatic attraction.
Collapse
Affiliation(s)
- Hosub Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, 120-749 Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
Nannini M, Biasco G, Maleddu A, Pantaleo MA. New molecular targets beyond KIT and PDGFRA in gastrointestinal stromal tumors: present and future. Expert Opin Ther Targets 2011; 15:803-15. [DOI: 10.1517/14728222.2011.566215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
Gonzalez RS, Carlson G, Page AJ, Cohen C. Gastrointestinal stromal tumor markers in cutaneous melanomas: relationship to prognostic factors and outcome. Am J Clin Pathol 2011; 136:74-80. [PMID: 21685034 DOI: 10.1309/ajcp9khd7dchwlmo] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Melanoma expresses c-kit, a gastrointestinal stromal tumor marker, but has not been extensively evaluated for protein kinase C θ (PKCθ) or DOG1, and these stains have not been correlated with prognostic factors. We immunostained 62 primary cutaneous and 15 metastatic melanomas for polyclonal c-kit (pc-kit), monoclonal c-kit (mc-kit), PKCθ, and DOG1 and correlated results with prognostic parameters and survival. Of the cutaneous melanomas, 34 (55%) stained for pc-kit, 30 (48%) for mc-kit, 11 (18%) for PKCθ, and 2 (3%) for DOG1. The Breslow depth was 1.00 mm or less in 21 (68%) of 31 pc-kit+ cutaneous melanomas compared with 7 (27%) of 26 pc-kit- melanomas (P = .002). The pc-kit+ melanomas had less nodal disease (1/31 [3%] vs 9/25 [36%]; P = .001) and local recurrence (1/33 [3%] vs 6/27 [22%]; P = .021) but no statistically significant difference in the rate of distant metastases (13/32 [41%] vs 14/27 [52%]; P = .388) or survival (10/34 [29%] vs 16/39 [41%]; P = .301). We found that pc-kit correlates better with prognostic parameters than does mc-kit.
Collapse
|
34
|
Tian Z, Shen J, Wang F, Xiao P, Yang J, Lei H, Kazlauskas A, Kohane IS, Wu E. Cambogin is preferentially cytotoxic to cells expressing PDGFR. PLoS One 2011; 6:e21370. [PMID: 21712951 PMCID: PMC3119700 DOI: 10.1371/journal.pone.0021370] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Accepted: 05/25/2011] [Indexed: 02/05/2023] Open
Abstract
Platelet-derived growth factor receptors (PDGFRs) have been implicated in a wide array of human malignancies, including medulloblastoma (MB), the most common brain tumor of childhood. Although significant progress in MB biology and therapeutics has been achieved during the past decades, MB remains a horrible challenge to the physicians and researchers. Therefore, novel inhibitors targeting PDGFR signaling pathway may offer great promise for the treatment of MB. In the present study, we investigated the cytotoxicity and mechanisms of cambogin in Daoy MB cells. Our results show that cambogin triggers significant S phase cell cycle arrest and apoptosis via down regulation of cyclin A and E, and activation of caspases. More importantly, further mechanistic studies demonstrated that cambogin inhibits PDGFR signaling in Daoy and genetically defined mouse embryo fibroblast (MEF) cell lines. These results suggest that cambogin is preferentially cytotoxic to cells expressing PDGFR. Our findings may provide a novel approach by targeting PDGFR signaling against MB.
Collapse
Affiliation(s)
- Ze Tian
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Informatics Program, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (ZT); (EW)
| | - Jie Shen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fengfei Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, United States of America
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Junshan Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hetian Lei
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Andrius Kazlauskas
- Department of Ophthalmology, Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Isaac S. Kohane
- Informatics Program, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, United States of America
- * E-mail: (ZT); (EW)
| |
Collapse
|
35
|
Setoguchi T, Kikuchi H, Yamamoto M, Baba M, Ohta M, Kamiya K, Tanaka T, Baba S, Goto-Inoue N, Setou M, Sasaki T, Mori H, Sugimura H, Konno H. Microarray analysis identifies versican and CD9 as potent prognostic markers in gastric gastrointestinal stromal tumors. Cancer Sci 2011; 102:883-889. [PMID: 21244575 PMCID: PMC11159159 DOI: 10.1111/j.1349-7006.2011.01872.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 01/06/2011] [Accepted: 01/08/2011] [Indexed: 01/08/2023] Open
Abstract
Although the main cause of gastrointestinal stromal tumor (GIST) is gain-of-function mutations in the c-kit gene in the interstitial cells of Cajal, concomitant genetic or epigenetic changes other than c-kit appear to occur in the development of metastasis. We sought to identify the genes involved in the metastatic process of gastric GIST. Microarray analysis was performed to compare gene expressions between three gastric GIST and four metastatic liver GIST. Expression levels were higher for 165 genes and lower for 146 genes in metastatic liver GIST. The upregulation of five oncogenes and downregulation of four tumor suppressor genes including versican and CD9 were confirmed by quantitative reverse transcriptional PCR. Immunohistochemistry in 117 GIST revealed that protein levels of versican and CD9 were higher and lower, respectively, in metastatic GIST. High expression of versican and low expression of CD9 in 104 primary gastric GIST correlated with poor disease-free survival (P = 0.0078 and P = 0.0018). In addition to the c-kit gene mutation, genetic or epigenetic changes other than c-kit play important roles in the metastatic process. In particular, versican and CD9 are potential prognostic markers in gastric GIST.
Collapse
Affiliation(s)
- Tomohiko Setoguchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Renkonen S, Häyry V, Heikkilä P, Leivo I, Haglund C, Mäkitie AA, Hagström J. Stem cell-related proteins C-KIT, C-MYC and BMI-1 in juvenile nasopharyngeal angiofibroma--do they have a role? Virchows Arch 2010; 458:189-95. [PMID: 21113618 DOI: 10.1007/s00428-010-1010-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 11/03/2010] [Accepted: 11/07/2010] [Indexed: 01/11/2023]
Abstract
Juvenile nasopharyngeal angiofibroma (JNA) is a highly vascular tumour, occurring almost exclusively in adolescent males. Histogenesis of JNA remains unclear, two optional theories proposing either fibrous or vascular tissue as the tissue of origin. Stem cell-related proteins have been discussed to possibly participate in the growth of these tumours. In our study, we reviewed retrospective clinicopathological data of 26 JNA patients. By immunohistochemistry, we investigated the cellular distribution and expression levels of stem cell-related proteins C-KIT, C-MYC and BMI-1 and their correlation with cell and vessel density of the tumour. Contrary to earlier reports, we detected C-KIT expression in addition to stromal cells also in endothelial cells. The C-KIT expression was more dominant in slit vessels than large vessels. A significant correlation was found between endothelial immunoexpression of C-KIT and cellular density of the tumour. C-MYC and BMI-1 expression was detected in stromal cells only. Due to our finding of C-KIT expression in both stromal and endothelial cells and the strong correlation between the endothelial C-KIT expression and cellular density, we suggest that, besides the stromal tissue, the vascular component might take part in the neoplastic growth of JNA.
Collapse
Affiliation(s)
- Suvi Renkonen
- Department of Otorhinolaryngology, Head and Neck Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
38
|
Itoh M, Goto A, Wakasugi H, Yoshida Y, Matsunaga Y, Fujii K, Suzuki K, Yonezawa K, Abe T, Arimura Y, Shinomura Y. Anorectal melanoma with a KIT-activating mutation, which is a target for tyrosine kinase inhibitor. Int J Clin Oncol 2010; 16:428-34. [DOI: 10.1007/s10147-010-0139-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 10/05/2010] [Indexed: 01/31/2023]
|
39
|
Pappou EP, Ahuja N. The role of oncogenes in gastrointestinal cancer. GASTROINTESTINAL CANCER RESEARCH : GCR 2010:S2-S15. [PMID: 21472044 PMCID: PMC3047044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 07/22/2010] [Indexed: 05/30/2023]
Abstract
Oncogene research over the last century has been one of the major advances in understanding the molecular biology of malignant disease. Oncogenes are a structurally and functionally heterogeneous group of genes, whose protein products act pleiotropically and affect multiple complex regulatory cascades within the cell. They regulate cell proliferation, growth, and differentiation, as well as control of the cell cycle and apoptosis. The products of oncogenes include growth factors, growth factor receptors, signal transducers, transcription factors, and apoptosis regulators, as well as chromatin remodelers. Several distinct mechanisms have been described for the conversion of proto-oncogenes to active oncogenes. Quantitative forms of oncogene activation include multiplication (gene amplification) or translocation to an active chromatin domain that brings a growth-regulatory gene under the control of a different promoter, causing inappropriate expression of the gene. Qualitative forms include either point mutations or the production of a novel product from a chimeric gene. Further understanding of the molecular mechanisms by which oncogenes regulate normal development and tumorigenesis may lead to novel concepts in the diagnosis and treatment of cancer in humans. In this review, we focus on the role of selected oncogenes in gastrointestinal cancer.
Collapse
|
40
|
Wang TB, Huang WS, Lin WH, Shi HP, Dong WG. Inhibition of KIT RNAi mediated with adenovirus in gastrointestinal stromal tumor xenograft. World J Gastroenterol 2010; 16:5122-9. [PMID: 20976851 PMCID: PMC2965291 DOI: 10.3748/wjg.v16.i40.5122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate a therapeutic method for gastrointestinal stromal tumor (GIST) based on KIT RNA interference (RNAi) with AdMax adenovirus.
METHODS: KIT short hairpin RNA (shRNA), whose lateral sides were decorated with restriction endonuclease sequences, was designed. T4 DNA ligase catalyzed the joint of the KIT shRNA and the green fluorescent protein-containing PDC316-EGFP-U6 to form PDC316-EGFP-U6-KIT. Homologous recombination of AdEGFP-U6-KIT was performed with the AdMax system. Heterotopically transplanted GISTs were established in nude mice. AdEGFP-U6-KIT was intratumorally injected. The volume, inhibition ratio of tumor and CD117 expression of GIST graft tumor in nude mice were compared between test and control groups.
RESULTS: The length of KIT shRNA was determined to be about 50bp by agarose electrophoresis. Gene sequencing detected the designed KIT RNAi sequence in PDC316-EGFP-U6-KIT. After transfection with AdEGFP-U6-KIT, 293 cells displayed green fluorescence. The physical and infective titers of AdEGFP-U6-KIT were 5 × 1011 viral particles/mL and 5.67 × 107 plaque forming units/mL, respectively. The mean volume of the grafted tumor was significantly smaller in test mice than in control mice (75.3 ± 22.9 mm3vs 988.6 ± 30.5 mm3, t = -18.132, P < 0.05). The inhibition ratio of the tumors was 59.6% in the test group. CD117 positive expression was evident in two cases (20%) in the test group and 10 cases (100%) in the control group (χ2 = 10.2083, P < 0.005).
CONCLUSION: AdEGFP-U6-KIT is successfully constructed, and KIT RNAi mediated with Admax vector system can effectively inhibit the expression of the KIT gene and the growth of GIST in nude mice.
Collapse
|
41
|
Igarashi S, Suzuki H, Niinuma T, Shimizu H, Nojima M, Iwaki H, Nobuoka T, Nishida T, Miyazaki Y, Takamaru H, Yamamoto E, Yamamoto H, Tokino T, Hasegawa T, Hirata K, Imai K, Toyota M, Shinomura Y. A novel correlation between LINE-1 hypomethylation and the malignancy of gastrointestinal stromal tumors. Clin Cancer Res 2010; 16:5114-23. [PMID: 20978145 DOI: 10.1158/1078-0432.ccr-10-0581] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Gastrointestinal stromal tumors (GIST) are the most important mesenchymal tumors of the gastrointestinal tract. The vast majority of GISTs exhibit activating mutations of KIT or PDGFRA, but epigenetic alteration of GISTs is largely unknown. In this study, we aimed to clarify the involvement of DNA methylation in GIST malignancy. EXPERIMENTAL DESIGN A total of 106 GIST specimens were studied. Levels of LINE-1 methylation were analyzed using bisulfite pyrosequencing. In addition, methylation of three other repetitive sequences (Alu Yb8, Satellite-α, and NBL2) was similarly analyzed, and CpG island hypermethylation was analyzed using MethyLight. Array-based comparative genomic hybridization (array CGH) was carried out in 25 GIST specimens. RESULTS LINE-1 hypomethylation was significantly correlated with risk, and high-risk GISTs exhibited significantly lower levels of LINE-1 methylation than low-risk (61.3% versus 53.2%; P = 0.001) or intermediate-risk GISTs (60.8% versus 53.2%; P = 0.002). Hypomethylation of Satellite-α and NBL2 was also observed in high-risk GISTs. By contrast, promoter hypermethylation was relatively infrequent (CDH1, 11.2%; MLH1, 9.8%; SFRP1, 1.2%; SFRP2, 11.0%; CHFR, 9.8%; APC, 6.1%; CDKN2A, 0%; RASSF1A, 0%; RASSF2, 0%) and did not correlate with LINE-1 methylation or risk. Array CGH analysis revealed a significant correlation between LINE-1 hypomethylation and chromosomal aberrations. CONCLUSIONS Our data suggest that LINE-1 hypomethylation correlates significantly with the aggressiveness of GISTs and that LINE-1 methylation could be a useful marker for risk assessment. Hypomethylation may increase the malignant potential of GISTs by inducing accumulation of chromosomal aberrations.
Collapse
Affiliation(s)
- Shinichi Igarashi
- First Department of Internal Medicine, Sapporo Medical University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schulz M, Schreyer A, Glöckner S, Schölmerich J, Zuber-Jerger I. 69-jähriger Patient mit chronischen Bauchschmerzen und Anämie bei Neurofibromatose. Internist (Berl) 2010; 52:81-2, 84-6, 88. [DOI: 10.1007/s00108-010-2653-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
43
|
Zhang Y, Cao H, Wang M, Zhao WY, Shen ZY, Shen DP, Ni XZ, Wu ZY, Shen YY, Song YY. Loss of chromosome 9p21 and decreased p16 expression correlate with malignant gastrointestinal stromal tumor. World J Gastroenterol 2010; 16:4716-24. [PMID: 20872974 PMCID: PMC2951524 DOI: 10.3748/wjg.v16.i37.4716] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate loss of heterozygosity (LOH) of chromosome 9p21 and the prognostic relevance of p16 expression in gastrointestinal stromal tumor (GIST).
METHODS: Fifty-one GIST patients (30 men and 21 women; median age 59 years; range 29-80 years) treated surgically within a 10-year period were grouped by aggressive behavior risk (17 with very low and low, 14 intermediate, and 20 high risk). GISTs were characterized immunohistochemically and evaluated for LOH of 9p21 by microsatellite analysis at D9S1751, D9S1846, D9S942, and D9S1748. LOH of 9p21 and immunohistochemical expression of p16 protein encoded at 9p21 were correlated with clinicopathological parameters, and the prognostic significance of p16 alterations was evaluated.
RESULTS: Thirty-one (63.3%) cases showed LOH with at least one microsatellite marker. LOH frequency was 37.0% at D9S1751, 37.5% at D9S1846, 42.1% at D9S942, and 24.2% at D9S1748. There was a higher LOH frequency of D9S942 in high-risk than in non-high-risk tumors (P < 0.05, χ2 = 4.47). Gender, age, tumor size and site were not correlated with allelic loss. Ninety percent (18/20) of the GIST patients in the high risk group showed LOH with at least one of the 9p21 markers, while 57.1% (8/14) in the intermediate risk group and 33.3% (5/15) in the very low and low risk groups, respectively (P < 0.05, χ2 = 12.16). Eight (28.5%) of 31 patients with LOH and 1 (5.6%) of 18 patients without LOH died of the disease during the follow-up period. Loss of p16 protein expression occurred in 41.2%, but in 60% of the high risk group and 23.5% of the very low and low risk groups (P < 0.05, χ2 = 4.98). p16 loss was associated with poor prognosis (P < 0.05, χ2 = 4.18): the 3- and 5-year overall survival rates were 84.8% and 70.8% for p16-negative and 100% and 92.0% for p16-positive patients, respectively.
CONCLUSION: LOH at 9p21 appears to play an important role in GIST progression; decreased p16 expression in GIST is highly predictive of poor outcome.
Collapse
|
44
|
Ylipää A, Hunt KK, Yang J, Lazar AJF, Torres KE, Lev DC, Nykter M, Pollock RE, Trent J, Zhang W. Integrative genomic characterization and a genomic staging system for gastrointestinal stromal tumors. Cancer 2010; 117:380-9. [PMID: 20818650 DOI: 10.1002/cncr.25594] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 07/15/2010] [Accepted: 07/26/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) historically were grouped with leiomyosarcomas (LMSs) based on their morphologic similarities; however, recently, GIST was established unequivocally as a distinct type of sarcoma based on its molecular features and response to imatinib treatment. METHODS To gain further insight into the genomic differences between GISTs and LMSs, the authors mapped gene copy number aberrations (CNAs) in 42 GISTs and 30 LMSs and integrated the results with gene expression profiles. RESULTS Distinct patterns of CNAs were revealed between GISTs and LMSs. Losses in 1p, 14q, 15q, and 22q were significantly more frequent in GISTs than in LMSs (P < .001); whereas losses in chromosomes 10 and 16 and gains in 1q, 14q, and 15q (P < .001) were more common in LMSs. By integrating CNAs with gene expression data and clinical information, the authors identified several clinically relevant CNAs that were prognostic of survival in patients with GIST. Furthermore, GISTs were categorized into 4 groups according to an accumulating pattern of genetic alterations. Many key cellular pathways were expressed differently in the 4 groups, and the patients in each group had increasingly worse prognoses as the extent of genomic alterations increased. CONCLUSIONS Based on the current findings, the authors proposed a new tumor-progression genetic staging system termed genomic instability stage to complement the current prognostic predictive system based on tumor size, mitotic index, and v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) mutation.
Collapse
Affiliation(s)
- Antti Ylipää
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Astolfi A, Nannini M, Pantaleo MA, Di Battista M, Heinrich MC, Santini D, Catena F, Corless CL, Maleddu A, Saponara M, Lolli C, Di Scioscio V, Formica S, Biasco G. A molecular portrait of gastrointestinal stromal tumors: an integrative analysis of gene expression profiling and high-resolution genomic copy number. J Transl Med 2010; 90:1285-94. [PMID: 20548289 DOI: 10.1038/labinvest.2010.110] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In addition to KIT and PDGFRA mutations, sequential accumulation of other genetic events is involved in the development and progression of gastrointestinal stromal tumors (GISTs). Until recently, the significance of these other alterations has not been thoroughly investigated. We report the first study that integrates gene expression profiling and high-resolution genomic copy number analyses in GIST. Fresh tissue specimens from 25 patients with GIST were collected, and gene expression profiling and high-resolution genomic copy number analyses were performed, using Affymetrix U133Plus and SNP array 6.0. We found that all 21 mutant GIST patients showed both macroscopic cytogenetic alterations and cryptic microdeletions or amplifications, whereas 75% (three of four) of wild-type patients with GIST did not show genomic imbalances. The most frequently observed chromosomal alterations in patients with mutant GIST included 14q complete or partial deletion (17 of 25), 1p deletion (14 of 25) and 22q deletion (10 of 25). Genetic targets of the chromosomal aberrations were selected by integrated analysis of copy number and gene expression data. We detected the involvement of known oncogenes and tumor suppressors including KRAS in chr 12p amplification and KIF1B, PPM1A, NF2 in chr 1p, 14q and 22p deletions, respectively. The genomic segment most frequently altered in mutated samples was the 14q23.1 region, which contains potentially novel tumor suppressors, including DAAM1, RTN1 and DACT1. siRNA-mediated RTN1 downregulation showed evidence for the potential role in GIST pathogenesis. The combination of gene expression profiling and high-resolution genomic copy number analysis offers a detailed molecular portrait of GISTs, providing an essential comprehensive knowledge necessary to guide the discovery of novel target genes involved in tumor development and progression.
Collapse
Affiliation(s)
- Annalisa Astolfi
- Interdepartmental Centre for Cancer Research G. Prodi, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dorn J, Spatz H, Schmieder M, Barth TF, Blatz A, Henne-Bruns D, Knippschild U, Kramer K. Cyclin H expression is increased in GIST with very-high risk of malignancy. BMC Cancer 2010; 10:350. [PMID: 20598140 PMCID: PMC2916921 DOI: 10.1186/1471-2407-10-350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 07/02/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Risk estimation of gastrointestinal stromal tumours (GIST) is based on tumour size and mitotic rate according to the National Institutes of Health consensus classification. The indication for adjuvant treatment of patients with high risk GIST after R0 resection with small molecule inhibitors is still a controversial issue, since these patients represent a highly heterogeneous population. Therefore, additional prognostic indicators are needed. Here, we evaluated the prognostic value of cyclin H expression in GIST. METHODS In order to identify prognostic factors of GIST we evaluated a single centre cohort of ninety-five GIST patients. First, GISTs were classified with regard to tumour size, mitotic rate and localisation according to the NIH consensus and to three additional suggested risk classifications. Second, Cyclin H expression was analysed. RESULTS Of ninety-five patients with GIST (53 female/42 male; median age: 66.78a; range 17-94a) risk classification revealed: 42% high risk, 20% intermediate risk, 23% low risk and 15% very low risk GIST. In patients with high risk GIST, the expression of cyclin H was highly predictive for reduced disease-specific survival (p = 0.038). A combination of cyclin H expression level and high risk classification yielded the strongest prognostic indicator for disease-specific and disease-free survival (p < or = 0.001). Moreover, in patients with tumour recurrence and/or metastases, cyclin H positivity was significantly associated with reduced disease-specific survival (p = 0.016) regardless of risk-classification. CONCLUSION Our data suggest that, in addition to high risk classification, cyclin H expression might be an indicator for "very-high risk" GIST.
Collapse
Affiliation(s)
- Julian Dorn
- Clinic of General-, Visceral- and Transplantation-Surgery, University Hospital Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Silva M, Veiga I, Ribeiro FR, Vieira J, Pinto C, Pinheiro M, Mesquita B, Santos C, Soares M, Dinis J, Santos L, Lopes P, Afonso M, Lopes C, Teixeira MR. Chromosome copy number changes carry prognostic information independent of KIT/PDGFRA point mutations in gastrointestinal stromal tumors. BMC Med 2010; 8:26. [PMID: 20470368 PMCID: PMC2876987 DOI: 10.1186/1741-7015-8-26] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 05/14/2010] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Oncogenic point mutations in KIT or PDGFRA are recognized as the primary events responsible for the pathogenesis of most gastrointestinal stromal tumors (GIST), but additional genomic alterations are frequent and presumably required for tumor progression. The relative contribution of such alterations for the biology and clinical behavior of GIST, however, remains elusive. METHODS In the present study, somatic mutations in KIT and PDGFRA were evaluated by direct sequencing analysis in a consecutive series of 80 GIST patients. For a subset of 29 tumors, comparative genomic hybridization was additionally used to screen for chromosome copy number aberrations. Genotype and genomic findings were cross-tabulated and compared with available clinical and follow-up data. RESULTS We report an overall mutation frequency of 87.5%, with 76.25% of the tumors showing alterations in KIT and 11.25% in PDGFRA. Secondary KIT mutations were additionally found in two of four samples obtained after imatinib treatment. Chromosomal imbalances were detected in 25 out of 29 tumors (86%), namely losses at 14q (88% of abnormal cases), 22q (44%), 1p (44%), and 15q (36%), and gains at 1q (16%) and 12q (20%). In addition to clinico-pathological high-risk groups, patients with KIT mutations, genomic complexity, genomic gains and deletions at either 1p or 22q showed a significantly shorter disease-free survival. Furthermore, genomic complexity was the best predictor of disease progression in multivariate analysis. CONCLUSIONS In addition to KIT/PDGFRA mutational status, our findings indicate that secondary chromosomal changes contribute significantly to tumor development and progression of GIST and that genomic complexity carries independent prognostic value that complements clinico-pathological and genotype information.
Collapse
Affiliation(s)
- Mara Silva
- Department of Genetics, Portuguese Oncology Institute - Porto, Rua Dr, António Bernardino Almeida, 4200-072 Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Choi HJ, Lee H, Kim H, Kwon JE, Kang HJ, You KT, Rhee H, Noh SH, Paik YK, Hyung WJ, Kim H. MicroRNA expression profile of gastrointestinal stromal tumors is distinguished by 14q loss and anatomic site. Int J Cancer 2010; 126:1640-50. [PMID: 19795448 DOI: 10.1002/ijc.24897] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs are known to regulate gene expression. Although unique microRNA expression profiles have been reported in several tumors, little is known about microRNA expression profiles in GISTs. To evaluate the relationship between microRNA expression and clinicopathologic findings of GISTs, we analyzed the microRNA expression profiles of GISTs. We used fresh frozen tissues from 20 GISTs and analyzed KIT and PDGFRA mutations and chromosomal loss status. MicroRNA expression was analyzed using a microRNA chip containing 470 microRNAs. Using unsupervised hierarchical clustering analysis, we found four distinct microRNA expression patterns in our 20 GISTs. Six GISTs that did not have 14q loss formed a separate cluster. In the 14 GISTs with 14q loss, 5 small bowel GISTs formed a separate cluster and the remaining 9 GISTs could be divided into two groups according to frequent chromosomal losses and tumor risk. We found 73 microRNAs that were significantly down-regulated in the GISTs with 14q loss; 38 of these microRNAs are encoded on 14q. We also found many microRNAs that were down-regulated in small bowel and high-risk group GISTs. Most of the microRNAs down-regulated in the high-risk group and small bowel GISTs are known to be involved in tumor progression, specifically by stimulating mitogen-activated protein kinase (MAPK) and the cell cycle. The microRNA expression patterns of GISTs are closely related to the status of 14q loss, anatomic site, and tumor risk. These findings suggest that microRNA expression patterns can differentiate several subsets of GISTs.
Collapse
Affiliation(s)
- Hee-Jung Choi
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Gastrointestinal stromal tumours (GIST) are the most common mesenchymal neoplasm of the gastrointestinal tract. Rectum localisation is infrequent for these neoplasms, accounting for about 5% of all cases. Distant metastases of GIST are also rare. We present a patient with special features: the tumour is localised in rectum and it has an uncommon metastatic site, the skull, implying a complex differential diagnosis approach.
Collapse
|
50
|
Combined treatment strategies in gastrointestinal stromal tumors (GISTs) after imatinib and sunitinib therapy. Cancer Treat Rev 2009; 36:63-8. [PMID: 19914780 DOI: 10.1016/j.ctrv.2009.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 10/14/2009] [Accepted: 10/19/2009] [Indexed: 12/14/2022]
Abstract
Resistance to tyrosine-kinase inhibitors remains an open issue in the treatment of patients with gastrointestinal stromal tumors. The complex biology of disease in the multi-resistant setting has led a progressively growing urgency and interest in development combined or integrated therapies. This mini-review outlines the rationale for developing new combined therapeutic approaches, and describes the state of the art of the various potential strategies and the promising research perspectives.
Collapse
|