51
|
Pardanani A. Systemic mastocytosis in adults: 2017 update on diagnosis, risk stratification and management. Am J Hematol 2016; 91:1146-1159. [PMID: 27762455 DOI: 10.1002/ajh.24553] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/17/2022]
Abstract
Disease overview:Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extra-cutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. Risk stratification: The 2008 World Health Organization (WHO) classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent (SM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD) and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. MANAGEMENT SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-α (±corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting; there is a role for allogeneic stem cell transplantation in select cases. Investigational drugs: Recent data confirms midostaurin's significant anti-MC activity in patients with advanced SM. Am. J. Hematol. 91:1147-1159, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
52
|
Ke H, Kazi JU, Zhao H, Sun J. Germline mutations of KIT in gastrointestinal stromal tumor (GIST) and mastocytosis. Cell Biosci 2016; 6:55. [PMID: 27777718 PMCID: PMC5070372 DOI: 10.1186/s13578-016-0120-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 10/04/2016] [Indexed: 01/01/2023] Open
Abstract
Somatic mutations of KIT are frequently found in mastocytosis and gastrointestinal stromal tumor (GIST), while germline mutations of KIT are rare, and only found in few cases of familial GIST and mastocytosis. Although ligand-independent activation is the common feature of KIT mutations, the phenotypes mediated by various germline KIT mutations are different. Germline KIT mutations affect different tissues such as interstitial cells of Cajal (ICC), mast cells or melanocytes, and thereby lead to GIST, mastocytosis, or abnormal pigmentation. In this review, we summarize germline KIT mutations in familial mastocytosis and GIST and discuss the possible cellular context dependent transforming activity of KIT mutations.
Collapse
Affiliation(s)
- Hengning Ke
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, No. 1160 Shengli Street, Yinchuan, 750004 People's Republic of China ; Translational Cancer Lab, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Hui Zhao
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, People's Republic of China
| | - Jianmin Sun
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, No. 1160 Shengli Street, Yinchuan, 750004 People's Republic of China ; Division of Translational Cancer Research, Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
53
|
Hole A, Blake D. A review of imatinib mesilate in the treatment of patients with gastrointestinal stromal tumours and chronic myeloid leukaemia. J Oncol Pharm Pract 2016. [DOI: 10.1191/1078155203jp103oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Context. Imatinib mesylate (GlivecÒ, GleevecÒ, Novartis) has made a dramatic impact on the treatment available for both chronic myeloid leukaemia (CML) and gastrointestinal stromal tumours (GISTs). Both are rare diseases that have possibly been unfamiliar to many pharmacists until now. Objective. To provide readers with a structured overview of CML and GISTs and the trial data available to support the role imatinib is playing in the treatment of these diseases. Data sources. Published reviews and primary trial results were sourced via Medline to prepare the review. Articles published up to and including August 2002 were considered. Additionally, pertinent books were used to prepare the text and Novartis kindly supplied information currently only available on file at the company. As imatinib has only been available for a short time, all types and sizes of trials were considered for inclusion. The authors made the final decision as to the relevance and therefore subsequent inclusion of data. Conclusions. Imatinib is a new molecularly targeted therapy for the treatment of CML and GISTs. It has provided an efficacious alternative therapy choice for physicians managing the care of patients suffering from these diseases. In the future there is the possibility that imatinib may demonstrate activity in other disease states characterized by the genetic changes targeted by imatinib. The results of randomized controlled trials in new and existing indications are awaited.
Collapse
Affiliation(s)
- Alison Hole
- WestawayGillis Ltd. (formerly Medicines Information Pharmacist at The Royal Marsden Hospital, UK)
| | | |
Collapse
|
54
|
Capelli L, Petracci E, Quagliuolo V, Saragoni L, Colombo P, Morgagni P, Calistri D, Tomezzoli A, Di Cosmo M, Roviello F, Vindigni C, Coniglio A, Villanacci V, Catarci M, Coppola L, Alfieri S, Ricci R, Capella C, Rausei S, Gulino D, Amadori D, Ulivi P. Gastric GISTs: Analysis of c-Kit, PDGFRA and BRAF mutations in relation to prognosis and clinical pathological characteristics of patients – A GIRCG study. Eur J Surg Oncol 2016; 42:1206-14. [DOI: 10.1016/j.ejso.2016.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/28/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
|
55
|
Chen YC, Liao JW, Hsu WL, Chang SC. Identification of the two KIT isoforms and their expression status in canine hemangiosarcomas. BMC Vet Res 2016; 12:142. [PMID: 27422008 PMCID: PMC4947345 DOI: 10.1186/s12917-016-0772-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/13/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND KIT is a tyrosine kinase growth factor receptor. High expression of KIT has been found in several tumors including canine hemangiosarcoma (HSA). This study investigated the correlation of KIT expression and c-kit sequence mutations in canine HSAs and benign hemangiomas (HAs). RESULTS Immunohistochemistry (IHC) staining confirmed KIT expression in 94.4 % (34/36) of HSAs that was significantly higher than 0 % in HAs (0/16). Sequencing the entire c-kit coding region of HSAs and normal canine cerebellums (NCCs) revealed GNSK-deletion in exon 9. As for exon 9 genotyping by TA-cloning strategy, GNSK-deletion c-kit accounted for 48.6 % (68/140) colonies amplified from12 KIT-positive HSAs, a significantly higher frequency than 14.1 % (9/64) of colonies amplified from six NCCs. CONCLUSIONS Due to the distinct expression pattern revealed by IHC, KIT might be used to distinguish benign or malignant vascular endothelial tumors. Moreover, the high incidence of GNSK-deletion c-kit in canine HSAs implicates KIT isoforms as possibly participating in the tumorigenesis of canine HSAs.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan.,Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan.
| | - Shih-Chieh Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan. .,Veterinary Medical Teaching Hospital, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo-Kuang Road, Taichung, 40227, Taiwan.
| |
Collapse
|
56
|
Jakhetiya A, Garg PK, Prakash G, Sharma J, Pandey R, Pandey D. Targeted therapy of gastrointestinal stromal tumours. World J Gastrointest Surg 2016; 8:345-352. [PMID: 27231512 PMCID: PMC4872062 DOI: 10.4240/wjgs.v8.i5.345] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/07/2016] [Accepted: 03/07/2016] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are mesenchymal neoplasms originating in the gastrointestinal tract, usually in the stomach or the small intestine, and rarely elsewhere in the abdomen. The malignant potential of GISTs is variable ranging from small lesions with a benign behaviour to fatal sarcomas. The majority of the tumours stain positively for the CD-117 (KIT) and discovered on GIST-1 (DOG-1 or anoctamin 1) expression, and they are characterized by the presence of a driver kinase-activating mutation in either KIT or platelet-derived growth factor receptor α. Although surgery is the primary modality of treatment, almost half of the patients have disease recurrence following surgery, which highlights the need for an effective adjuvant therapy. Traditionally, GISTs are considered chemotherapy and radiotherapy resistant. With the advent of targeted therapy (tyrosine kinase inhibitors), there has been a paradigm shift in the management of GISTs in the last decade. We present a comprehensive review of targeted therapy in the management of GISTs.
Collapse
|
57
|
McLoughlin J, Nodit L, Heidel RE, Van Meter S, Macy S, Kestler D. The clinical correlation of phosphatase and tensin homolog on chromosome 10, phosphorylation of AKT to an activated state, and odontogenic ameloblast-associated protein in gastrointestinal stromal tumors. J Surg Res 2016; 202:403-12. [PMID: 27229116 DOI: 10.1016/j.jss.2016.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 11/23/2015] [Accepted: 01/11/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Approximately 15% of gastrointestinal stromal tumors (GISTs) will not respond to tyrosine kinase inhibitors and drug resistance can develop over time. For refractory tumors, additional therapies are needed. Odontogenic ameloblast-associated protein (ODAM) is expressed in some epithelial malignancies and can correlate with clinical outcomes. This study evaluated ODAM and its relationship to phosphatase and tensin homolog on chromosome 10 (PTEN) and phosphorylation of AKT to an activated state (pAKT) in GISTs. MATERIALS AND METHODS Ninety-five distinct tumor specimens from 79 patients were identified. Morphologic features and clinical data were recorded for all tumors. Risk of recurrence was calculated using the Memorial Sloan-Kettering nomogram. Immunohistochemistry was performed using antibodies to ODAM, PTEN, and pAKT. Immunoreactivity was assessed for both cytoplasmic and nuclear expression. Staining patterns were correlated with clinical outcomes. RESULTS Increasing cytoplasmic ODAM staining correlated with a lower recurrence score (P = 0.002), a lower mitotic rate (P = 0.0001), and smaller tumor size (P = 0.038). Increasing pAKT cytoplasmic staining correlated with a higher recurrence score (P = 0.037) and a higher mitotic rate (P = 0.036). ODAM and pAKT expression in the nucleus was associated with tumor origin. PTEN nuclear expression increased with increasing mitotic rate. pAKT expression increased in the cytoplasm and nucleus in high-risk tumors. CONCLUSIONS Risk of recurrence correlated with cytoplasmic expression of ODAM and pAKT, whereas nuclear expression did not predict recurrence. The staining pattern for ODAM and pAKT in the cytoplasm may further clarify the risk of recurrence beyond the available nomograms. The increased expression of pAKT in the cytoplasm and nucleus of high-risk tumors suggests a potential target for systemic therapy.
Collapse
Affiliation(s)
- James McLoughlin
- Department of Surgery, University of Tennessee Medical Center, Knoxville, Tennessee.
| | - Laurentia Nodit
- Department of Pathology, University of Tennessee Medical Center, Knoxville, Tennessee
| | - R Eric Heidel
- Department of Biostatistics, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Stuart Van Meter
- Department of Pathology, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Sallie Macy
- University of Tennessee Medical Center, Graduate School of Medicine, Knoxville, Tennessee
| | - Daniel Kestler
- University of Tennessee Medical Center, Graduate School of Medicine, Knoxville, Tennessee
| |
Collapse
|
58
|
Mastoraki A, Toliaki E, Chrisovergi E, Mastoraki S, Papanikolaou IS, Danias N, Smyrniotis V, Arkadopoulos N. Metastatic Liver Disease Associated with Gastrointestinal Stromal Tumors: Controversies in Diagnostic and Therapeutic Approach. J Gastrointest Cancer 2016; 46:237-42. [PMID: 26163021 DOI: 10.1007/s12029-015-9748-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal lesions of the GI tract. They are considered to originate from neoplastic transformation of either the intestinal pacemaker cells of Cajal or the precursor pluripotential stem cells. The genetic basis of GIST growth is an activating mutation of two receptor tyrosine kinases. Recent epidemiologic studies demonstrate that the GIST prevalence is approximately 20/1000000/year. Although GISTs develop in every part of the GI tract, stomach remains the most common localization. About 80 % of the patients experience tumor recurrence or hepatic metastasis after radical resection. GIST liver metastases are usually multiple, large in diameter, and localized in both lobes. In addition, GISTs are usually completely asymptomatic, discovered incidentally. Symptoms are not typical and depend on the location, size, and aggressiveness of the tumor. DISCUSSION Diagnostic evaluation is based on imaging techniques, such as computed tomography, magnetic resonance imaging, positron emission tomography, and endoscopic ultrasound. Despite recent research on the therapeutic strategies against GISTs, surgical resection appears the only potentially curative approach. For the advanced metastatic disease, imatinib, a tyrosine kinase inhibitor, has been proposed neoadjuvantly with the surgery performed after the adequate reduction of tumor burden. The aim of this review was to evaluate the results of surgical treatment for metastatic GIST with special reference to the extent of its histological spread and to present the recent literature in order to provide an update on the current concepts of advanced surgical management of this entity.
Collapse
Affiliation(s)
- Aikaterini Mastoraki
- 4th Department of Surgery, Medical School, ATTIKON University Hospital, Athens University, 1 Rimini str, 12462, Chaidari, Athens, Greece,
| | | | | | | | | | | | | | | |
Collapse
|
59
|
Wang HC, Li TY, Chao YJ, Hou YC, Hsueh YS, Hsu KH, Shan YS. KIT Exon 11 Codons 557-558 Deletion Mutation Promotes Liver Metastasis Through the CXCL12/CXCR4 Axis in Gastrointestinal Stromal Tumors. Clin Cancer Res 2016; 22:3477-87. [PMID: 26936919 DOI: 10.1158/1078-0432.ccr-15-2748] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/15/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE KIT mutations, the most prevalent genetic event in gastrointestinal stromal tumors (GIST), are associated with malignant features and poor prognosis. Aggressive GISTs possess a high propensity to spread to the liver. This study aimed to explore the role of KIT mutations in GIST liver metastasis. EXPERIMENTAL DESIGN A total of 170 GISTs were used to determine the association between KIT mutations and liver metastasis. Immunohistochemistry was performed to assess the correlation of KIT mutations with CXCR4 and ETV1 expression. Genetic and pharmacologic methods were used to study the regulation of CXCR4 and ETV1 by KIT mutations. RESULTS Codons 557 and 558 in KIT exon 11 were deletion hot spots in GISTs. KIT exon 11 deletions involving codons 557-558 were highly associated with liver metastasis. Overexpression of mutant KIT with exon 11 codons 557-558 deletion (KIT Δ557-558) increased GIST cell motility and liver metastasis. Mechanistically, overexpression of KIT Δ557-558 in GIST cells increased ETV1 and CXCR4 expression. CXCR4 knockdown counteracted KIT Δ557-558-mediated cell migration. Moreover, KIT Δ557-558-induced CXCR4 expression could be abolished by silencing ETV1. The chromatin immunoprecipitation assay showed that ETV1 directly bound to the CXCR4 promoter. After ERK inhibitor PD325901 treatment, the upregulation of ETV1 by KIT Δ557-558 was prevented. In addition, KIT exon 11 codons 557-558 deletion enhanced CXCL12-mediated GIST cell migration and invasion. CONCLUSIONS KIT exon 11 557-558 deletion upregulates CXCR4 through increased binding of ETV1 to the CXCR4 promoter in GIST cells, which thus promotes liver metastasis. These findings highlighted the potential therapeutic targets for metastatic GISTs. Clin Cancer Res; 22(14); 3477-87. ©2016 AACR.
Collapse
Affiliation(s)
- Hao-Chen Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Ying Li
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jui Chao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yuan-Shuo Hsueh
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kai-Hsi Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, Tainan Hospital, Department of Health, Executive Yuan, Tainan, Taiwan.
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan. Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan.
| |
Collapse
|
60
|
Hilal L, Barada K, Mukherji D, Temraz S, Shamseddine A. Gastrointestinal (GI) leiomyosarcoma (LMS) case series and review on diagnosis, management, and prognosis. Med Oncol 2016; 33:20. [PMID: 26786155 DOI: 10.1007/s12032-016-0730-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 01/07/2016] [Indexed: 02/07/2023]
Abstract
This review of 76 gastrointestinal (GI) leiomyosarcoma (LMS) cases that include 11 cases from the American University of Beirut Medical Center represents, to our knowledge, the largest number of combined GI LMS cases reported. The age range of GI LMS is variable, and the presentation is non-specific, making pathological diagnosis essential. LMSs usually lack CD117 and CD 34 mutations and are usually positive for smooth muscle cell markers. The review highlights surgery as the mainstay of treatment with negative margins attained most of the times. Adjuvant chemotherapy is used in around 7-27 % of the cases mainly for small intestinal and colorectal LMS. The relatively small number of patients is a limitation on outcome analysis. However, LMS has a risk of recurrence reaching 39-80 % and secondary metastasis reaching 55-71 % in small intestinal and colorectal cases. In light of the high frequency of recurrence and metastasis, enrolling patients in clinical randomized trials to investigate the role of chemotherapy, radiation therapy, and targeted therapy is required for better control of this rare aggressive GI tumor.
Collapse
Affiliation(s)
- Lara Hilal
- Department of Radiation Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Kassem Barada
- Gastrointestinal Division, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Deborah Mukherji
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Sally Temraz
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Ali Shamseddine
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
- Hematology - Oncology Division, Tumor Registry, P.O.Box: 11-0236, Riad El Solh, Beirut, 110 72020, Lebanon.
| |
Collapse
|
61
|
Kafshdooz T, Mohaddes Ardabili SM, Kafshdooz L, Tabrizi AD, Ghojazadeh M, Gharesouran J, Akbarzadeh A. C-kit Mutations in Endometrial Cancer: Correlation with Tumor Histologic Type. Asian Pac J Cancer Prev 2015; 16:7449-52. [PMID: 26625742 DOI: 10.7314/apjcp.2015.16.17.7449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Endometrial cancer is the fourth most common cancer among women in developed countries. Affected patients may benefit from systemic chemotherapy, alone or in combination with targeted therapies if the disease is clinically diagnosed prior to expansion and metastasis to other organs.The aim of this study was to evaluate the prognostic role of c-kit mutations and comparision with tumor type and grade in human uterine endometrial carcinomas. MATERIALS AND METHODS Seventy five patients with endometrial carcinoma and seventy five normal controls were studied for possible mutations in exon 17 of the c-kit gene using single strand conformational polymorphisms and sequencing. RESULTS c-kit mutation in exon 17 appeared to be significantly different between endometrial carcinoma and normal endometrium. The pattern and frequency of the mutations was also shown to be different between tumors from different stages.
Collapse
|
62
|
Vajravelu BN, Hong KU, Al-Maqtari T, Cao P, Keith MCL, Wysoczynski M, Zhao J, Moore IV JB, Bolli R. C-Kit Promotes Growth and Migration of Human Cardiac Progenitor Cells via the PI3K-AKT and MEK-ERK Pathways. PLoS One 2015; 10:e0140798. [PMID: 26474484 PMCID: PMC4608800 DOI: 10.1371/journal.pone.0140798] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023] Open
Abstract
A recent phase I clinical trial (SCIPIO) has shown that autologous c-kit+ cardiac progenitor cells (CPCs) improve cardiac function and quality of life when transplanted into patients with ischemic heart disease. Although c-kit is widely used as a marker of resident CPCs, its role in the regulation of the cellular characteristics of CPCs remains unknown. We hypothesized that c-kit plays a role in the survival, growth, and migration of CPCs. To test this hypothesis, human CPCs were grown under stress conditions in the presence or absence of SCF, and the effects of SCF-mediated activation of c-kit on CPC survival/growth and migration were measured. SCF treatment led to a significant increase in cell survival and a reduction in cell death under serum depletion conditions. In addition, SCF significantly promoted CPC migration in vitro. Furthermore, the pro-survival and pro-migratory effects of SCF were augmented by c-kit overexpression and abrogated by c-kit inhibition with imatinib. Mechanistically, c-kit activation in CPCs led to activation of the PI3K and the MAPK pathways. With the use of specific inhibitors, we confirmed that the SCF/c-kit-dependent survival and chemotaxis of CPCs are dependent on both pathways. Taken together, our findings suggest that c-kit promotes the survival/growth and migration of human CPCs cultured ex vivo via the activation of PI3K and MAPK pathways. These results imply that the efficiency of CPC homing to the injury site as well as their survival after transplantation may be improved by modulating the activity of c-kit.
Collapse
Affiliation(s)
- Bathri N. Vajravelu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Kyung U. Hong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Tareq Al-Maqtari
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Pengxiao Cao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Matthew C. L. Keith
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - John Zhao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Joseph B. Moore IV
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
- * E-mail:
| |
Collapse
|
63
|
Hayashi Y, Bardsley MR, Toyomasu Y, Milosavljevic S, Gajdos GB, Choi KM, Reid-Lombardo KM, Kendrick ML, Bingener-Casey J, Tang CM, Sicklick JK, Gibbons SJ, Farrugia G, Taguchi T, Gupta A, Rubin BP, Fletcher JA, Ramachandran A, Ordog T. Platelet-Derived Growth Factor Receptor-α Regulates Proliferation of Gastrointestinal Stromal Tumor Cells With Mutations in KIT by Stabilizing ETV1. Gastroenterology 2015; 149:420-32.e16. [PMID: 25865047 PMCID: PMC4516576 DOI: 10.1053/j.gastro.2015.04.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS In gastrointestinal muscles, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) is predominantly expressed by interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor-α (PDGFRA) polypeptide is expressed by so-called fibroblast-like cells. KIT and PDGFRA have been reported to be coexpressed in ICC precursors and gastrointestinal stromal tumors (GISTs), which originate from the ICC lineage. PDGFRA signaling has been proposed to stimulate growth of GISTs that express mutant KIT, but the effects and mechanisms of selective blockade of PDGFRA are unclear. We investigated whether inhibiting PDGFRA could reduce proliferation of GIST cells with mutant KIT via effects on the KIT-dependent transcription factor ETV1. METHODS We studied 53 gastric, small intestinal, rectal, or abdominal GISTs collected immediately after surgery or archived as fixed blocks at the Mayo Clinic and University of California, San Diego. In human GIST cells carrying imatinib-sensitive and imatinib-resistant mutations in KIT, PDGFRA was reduced by RNA interference (knockdown) or inhibited with crenolanib besylate (a selective inhibitor of PDGFRA and PDGFRB). Mouse ICC precursors were retrovirally transduced to overexpress wild-type Kit. Cell proliferation was analyzed by methyltetrazolium, 5-ethynyl-2'-deoxyuridine incorporation, and Ki-67 immunofluorescence assays; we also analyzed growth of xenograft tumors in mice. Gastric ICC and ICC precursors, and their PDGFRA(+) subsets, were analyzed by flow cytometry and immunohistochemistry in wild-type, Kit(+/copGFP), Pdgfra(+/eGFP), and NOD/ShiLtJ mice. Immunoblots were used to quantify protein expression and phosphorylation. RESULTS KIT and PDGFRA were coexpressed in 3%-5% of mouse ICC, 35%-44% of ICC precursors, and most human GIST samples and cell lines. PDGFRA knockdown or inhibition with crenolanib efficiently reduced proliferation of imatinib-sensitive and imatinib-resistant KIT(+)ETV1(+)PDGFRA(+) GIST cells (50% maximal inhibitory concentration = 5-32 nM), but not of cells lacking KIT, ETV1, or PDGFRA (50% maximal inhibitory concentration >230 nM). Crenolanib inhibited phosphorylation of PDGFRA and PDGFRB, but not KIT. However, Kit overexpression sensitized mouse ICC precursors to crenolanib. ETV1 knockdown reduced KIT expression and GIST proliferation. Crenolanib down-regulated ETV1 by inhibiting extracellular-signal-regulated kinase (ERK)-dependent stabilization of ETV1 protein and also reduced expression of KIT and PDGFRA. CONCLUSIONS In KIT-mutant GIST, inhibition of PDGFRA disrupts a KIT-ERK-ETV1-KIT signaling loop by inhibiting ERK activation. The PDGFRA inhibitor crenolanib might be used to treat patients with imatinib-resistant, KIT-mutant GIST.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Srdjan Milosavljevic
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Kyoung Moo Choi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Chih-Min Tang
- Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, California
| | - Jason K. Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, California
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Takahiro Taguchi
- Division of Human Health and Medical Science, Graduate School of Kuroshio Science, Kochi University, Kochi, Japan
| | - Anu Gupta
- Departments of Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Brian P. Rubin
- Departments of Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Tamas Ordog
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
64
|
Abstract
Approximately 85-90% of adult gastrointestinal stromal tumors (GISTs) harbor KIT and PDGFRA mutations. The remaining cases, including the majority of pediatric GISTs, lack these mutations, and have been designated as KIT/PDGFRA wild-type (WT) GISTs. Nearly 15% of WT GISTs harbor BRAF mutations, while others arise in patients with type I neurofibromatosis. Recent work has confirmed that 20-40% of KIT/PDGFRA WT GISTs show loss of function of succinate dehydrogenase complex. Less than 5% of GISTs lack known molecular alterations ("quadruple-negative" GISTs). Thus, it is important to consider genotyping these tumors to help better define their clinical behavior and therapy.
Collapse
Affiliation(s)
- Deepa T Patil
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, L-25, Cleveland, OH 44195, USA.
| | - Brian P Rubin
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, L-25, Cleveland, OH 44195, USA; Department of Molecular Genetics, Cleveland Clinic and Lerner Research Institute, 9500 Euclid Avenue, L-25, Cleveland, OH 44195, USA
| |
Collapse
|
65
|
Harlan LC, Eisenstein J, Russell MC, Stevens JL, Cardona K. Gastrointestinal stromal tumors: treatment patterns of a population-based sample. J Surg Oncol 2015; 111:702-7. [PMID: 25900896 DOI: 10.1002/jso.23879] [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] [Received: 10/20/2014] [Revised: 11/26/2014] [Accepted: 12/04/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The National Cancer Institute (NCI) annually confirms therapy with treating physicians on a sample of patients diagnosed with a specific cancer. METHODS Using the NCI Patterns of Care data, treatment patterns were examined on a population-based sample of patients diagnosed with gastrointestinal stromal tumors (GIST) in 2008. RESULTS A random sample of 323 of 405 GIST patients registered in SEER was selected. Most patients had gastric GISTs, were ≥ 65 years, white, had private insurance, and treated in a hospital with a residency program. Surgery was primarily performed in patients with non-metastatic disease (94%), in which: 26, 12, and 36% were at low, intermediate, and high-risk of recurrence, respectively. Amongst low-risk patients, ∼ 30% received adjuvant therapy. Amongst patients at higher risk, 26-40% did not receive adjuvant therapy. Imatinib was the most common targeted therapy administered. On multivariate analysis, age and risk-group were associated with receipt of adjuvant targeted therapy. CONCLUSIONS Our study shows that in 2008, the majority of patients diagnosed with GIST received appropriate surgical and adjuvant therapies. However, a considerable subset may have been overtreated and undertreated. Future studies identifying factors that impact the delivery of adjuvant therapy should be conducted.
Collapse
Affiliation(s)
- Linda C Harlan
- Applied Research Program, National Cancer Institute, Bethesda, Maryland
| | | | | | | | | |
Collapse
|
66
|
Amagai Y, Matsuda A, Jung K, Oida K, Jang H, Ishizaka S, Matsuda H, Tanaka A. A point mutation in the extracellular domain of KIT promotes tumorigenesis of mast cells via ligand-independent auto-dimerization. Sci Rep 2015; 5:9775. [PMID: 25965812 PMCID: PMC4428273 DOI: 10.1038/srep09775] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/19/2015] [Indexed: 12/11/2022] Open
Abstract
Mutations in the juxtamembrane and tyrosine kinase domains of the KIT receptor have been implicated in several cancers and are known to promote tumorigenesis. However, the pathophysiological manifestations of mutations in the extracellular domain remain unknown. In this study, we examined the impact of a mutation in the extracellular domain of KIT on mast cell tumorigenesis. A KIT mutant with an Asn508Ile variation (N508I) in the extracellular domain derived from a canine mast cell tumor was introduced into IC-2 cells. The IC-2(N508I) cells proliferated in a cytokine-independent manner and showed KIT auto-phosphorylation. Subcutaneous injection of IC-2(N508I) cells into the dorsal area of immunodeficient BALB/c-nu/nu mice resulted in the formation of solid tumors, but tumor progression was abrogated by treatment with a tyrosine kinase inhibitor (STI571). In addition, the N508I mutant KIT protein dimerized in the absence of the natural ligand, stem cell factor. Structure modeling indicates that the increased hydrophobicity of the mutant led to the stabilization of KIT dimers. These results suggest that this extracellular domain mutation confers a ligand-independent tumorigenic phenotype to mast cells by KIT auto-dimerization that is STI571-sensitive. This is the first report demonstrating the tumorigenic potential of a mutation in the extracellular domain of KIT.
Collapse
Affiliation(s)
- Yosuke Amagai
- 1] Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan [2] Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akira Matsuda
- Laboratories of Veterinary Molecular Pathology and Therapeutics
| | - Kyungsook Jung
- 1] Comparative Animal Medicine, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan [2] Eco-friendly Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeonbuk, Korea
| | - Kumiko Oida
- Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hyosun Jang
- Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Saori Ishizaka
- Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroshi Matsuda
- 1] Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan [2] Laboratories of Veterinary Molecular Pathology and Therapeutics
| | - Akane Tanaka
- 1] Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan [2] Comparative Animal Medicine, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
67
|
Thys A, Vandenberghe P, Hague P, Klein OD, Erneux C, Vanderwinden JM. Hyperplasia of interstitial cells of cajal in sprouty homolog 4 deficient mice. PLoS One 2015; 10:e0124861. [PMID: 25923139 PMCID: PMC4414615 DOI: 10.1371/journal.pone.0124861] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/11/2015] [Indexed: 12/28/2022] Open
Abstract
Gastrointestinal stromal tumors, which are thought to derive from interstitial cells of Cajal or their precursors, often harbor an oncogenic mutation of the KIT receptor tyrosine kinase. Sprouty homolog 4, a known negative regulator of ERK pathway, has been identified in the interstitial cells of Cajal in the KitK641E murine model of gastrointestinal stromal tumors. Sprouty homolog 4 was upregulated both at the mRNA and protein level in these cells, suggesting that Sprouty homolog 4 is downstream of oncogenic KIT activation and potentially engaged in the negative feedback loop of ERK activation in this model. Here, we used KitK641E heterozygous and Sprouty homolog 4 knock out animals to quantify interstitial cells of Cajal in situ, using quantitative immunofluorescence for the receptor tyrosine kinase Kit and for phosphodiesterase 3a (PDE3A). In the antrum of Sprouty homolog 4 knock out mice, hyperplasia of interstitial cells of Cajal was reminiscent of the KitK641E heterozygous mice antrum. Additionally, the density of interstitial cells of Cajal was higher in the colon of adult Sprouty homolog 4 knock out mice than in WT littermates, although hyperplasia seemed more severe in KitK641E heterozygous mice. Functional transit studies also show similarities between Sprouty homolog 4 knock out and KitK641E heterozygous mice, as the total transit time in 9 month old animals was significantly increased in both genotypes compared to WT littermates. We concluded that the lack of Sprouty homolog 4 expression leads to hyperplasia of the interstitial cells of Cajal and is functionally associated with a delayed transit time.
Collapse
Affiliation(s)
- An Thys
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Vandenberghe
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Perrine Hague
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, California, United States of America
- Department of Pediatrics and Institute for Human genetics, University of California, San Francisco, California, United States of America
| | - Christophe Erneux
- IRIBHM, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail:
| |
Collapse
|
68
|
Hameed M, Corless C, George S, Hornick JL, Kakar S, Lazar AJ, Tang L. Template for Reporting Results of Biomarker Testing of Specimens From Patients With Gastrointestinal Stromal Tumors. Arch Pathol Lab Med 2015; 139:1271-5. [DOI: 10.5858/arpa.2014-0578-cp] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meera Hameed
- From Surgical Pathology, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York (Drs Hameed and Tang); the Department of Pathology, Oregon Health & Science University, Portland (Dr Corless); the Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (Dr George); the Department of Pathology, Brigham and Women's Hospital, Boston (Dr Ho
| | | | | | | | | | | | | |
Collapse
|
69
|
Pardanani A. Systemic mastocytosis in adults: 2015 update on diagnosis, risk stratification, and management. Am J Hematol 2015; 90:250-62. [PMID: 25688753 DOI: 10.1002/ajh.23931] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
DISEASE OVERVIEW Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extracutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. RISK STRATIFICATION The 2008 World Health Organization classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent SM (ISM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD), and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. MANAGEMENT SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-α (+/-corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal, and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting; there is a role for allogeneic stem cell transplantation in select cases. Investigational Drugs: Recent data confirms midostaurin's significant anti-MC activity in patients with advanced SM.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology; Mayo Clinic; Rochester Minnesota
- Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
70
|
Rubió-Casadevall J, Martinez-Trufero J, Garcia-Albeniz X, Calabuig S, Lopez-Pousa A, Del Muro JG, Fra J, Redondo A, Lainez N, Poveda A, Valverde C, De Juan A, Sevilla I, Casado A, Andres R, Cruz J, Martin-Broto J, Maurel J. Role of surgery in patients with recurrent, metastatic, or unresectable locally advanced gastrointestinal stromal tumors sensitive to imatinib: a retrospective analysis of the Spanish Group for Research on Sarcoma (GEIS). Ann Surg Oncol 2015; 22:2948-57. [PMID: 25608769 DOI: 10.1245/s10434-014-4360-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recurrent, metastatic, and locally advanced gastrointestinal stromal tumors (GISTs) can be treated successfully with imatinib mesylate. Surgery for residual disease has been suggested for nonrefractory metastatic GISTs to reduce the probability of resistant recurrent clones, although no randomized Phase III trial has been performed to answer the question about its benefit. We carried out an analysis of the outcome of patients with recurrent unresectable locally advanced or metastatic imatinib-sensitive priamary GIST in 14 institutions in Spain. We compared two cohorts: treated or not treated with surgery after partial response or stabilization by imatinib. PATIENTS AND METHODS Data were obtained from the online GIST registry of the Spanish Group for Research in Sarcomas. Selected patients were then divided into two groups: group A, treated initially only with imatinib, and group B, treated additionally with metastasectomy. Baseline characteristics between groups were compared, and univariate and multivariate analysis for progression-free survival and overall survival (OS) were performed. RESULTS Analysis was undertaken in 171 patients considered nonrefractory to imatinib. The median follow-up time was 56.6 months. Focusing on OS, the Eastern Cooperative Oncology Group performance status different than 0, extent of disease limited to one metastatic organ, and comparison between groups A or B achieved statistical difference in the multivariate analysis. Median survival was 59.9 months in group A and 87.6 months in group B. CONCLUSIONS Based in its benefit in OS, our study supports surgery of metastatic disease in GIST patients who respond to imatinib therapy.
Collapse
Affiliation(s)
- Jordi Rubió-Casadevall
- Department of Medical Oncology, Institut Català d'Oncologia de Girona, Hospital Josep Trueta, Girona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Tornillo L. Gastrointestinal stromal tumor - an evolving concept. Front Med (Lausanne) 2014; 1:43. [PMID: 25593916 PMCID: PMC4291900 DOI: 10.3389/fmed.2014.00043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors of the gastrointestinal tract. The discovery that these tumors, formerly thought of smooth muscle origin, are indeed better characterized by specific activating mutation in genes coding for the receptor tyrosine kinases (RTKs) CKIT and PDGFRA and that these mutations are strongly predictive for the response to targeted therapy with RTK inhibitors has made GISTs the typical example of the integration of basic molecular knowledge in the daily clinical activity. The information on the mutational status of these tumors is essential to predict (and subsequently to plan) the therapy. As resistant cases are frequently wild type, other possible oncogenic events, defining other "entities," have been discovered (e.g., succinil dehydrogenase mutation/dysregulation, insuline growth factor expression, and mutations in the RAS-RAF-MAPK pathway). The classification of disease must nowadays rely on the integration of the clinico-morphological characteristics with the molecular data.
Collapse
Affiliation(s)
- Luigi Tornillo
- Institute of Pathology, University of Basel , Basel , Switzerland
| |
Collapse
|
72
|
Macleod AC, Klug LR, Patterson J, Griffith DJ, Beadling C, Town A, Heinrich MC. Combination therapy for KIT-mutant mast cells: targeting constitutive NFAT and KIT activity. Mol Cancer Ther 2014; 13:2840-51. [PMID: 25253785 DOI: 10.1158/1535-7163.mct-13-0830] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Resistant KIT mutations have hindered the development of KIT kinase inhibitors for treatment of patients with systemic mastocytosis. The goal of this research was to characterize the synergistic effects of a novel combination therapy involving inhibition of KIT and calcineurin phosphatase, a nuclear factor of activated T cells (NFAT) regulator, using a panel of KIT-mutant mast cell lines. The effects of monotherapy or combination therapy on the cellular viability/survival of KIT-mutant mast cells were evaluated. In addition, NFAT-dependent transcriptional activity was monitored in a representative cell line to evaluate the mechanisms responsible for the efficacy of combination therapy. Finally, shRNA was used to stably knockdown calcineurin expression to confirm the role of calcineurin in the observed synergy. The combination of a KIT inhibitor and a calcineurin phosphatase inhibitor (CNPI) synergized to reduce cell viability and induce apoptosis in six distinct KIT-mutant mast cell lines. Both KIT inhibitors and CNPIs were found to decrease NFAT-dependent transcriptional activity. NFAT-specific inhibitors induced similar synergistic apoptosis induction as CNPIs when combined with a KIT inhibitor. Notably, NFAT was constitutively active in each KIT-mutant cell line tested. Knockdown of calcineurin subunit PPP3R1 sensitized cells to KIT inhibition and increased NFAT phosphorylation and cytoplasmic localization. Constitutive activation of NFAT appears to represent a novel and targetable characteristic of KIT-mutant mast cell disease. Our studies suggest that combining KIT inhibition with NFAT inhibition might represent a new treatment strategy for mast cell disease.
Collapse
Affiliation(s)
- Alison C Macleod
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Lillian R Klug
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Janice Patterson
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Diana J Griffith
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Carol Beadling
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Ajia Town
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon
| | - Michael C Heinrich
- Portland VA Medical Center, Portland, Oregon. OHSU Knight Cancer Institute, Portland, Oregon.
| |
Collapse
|
73
|
Analysis of gene expression profiles of soft tissue sarcoma using a combination of knowledge-based filtering with integration of multiple statistics. PLoS One 2014; 9:e106801. [PMID: 25188299 PMCID: PMC4154757 DOI: 10.1371/journal.pone.0106801] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 08/01/2014] [Indexed: 12/21/2022] Open
Abstract
The diagnosis and treatment of soft tissue sarcomas (STS) have been difficult. Of the diverse histological subtypes, undifferentiated pleomorphic sarcoma (UPS) is particularly difficult to diagnose accurately, and its classification per se is still controversial. Recent advances in genomic technologies provide an excellent way to address such problems. However, it is often difficult, if not impossible, to identify definitive disease-associated genes using genome-wide analysis alone, primarily because of multiple testing problems. In the present study, we analyzed microarray data from 88 STS patients using a combination method that used knowledge-based filtering and a simulation based on the integration of multiple statistics to reduce multiple testing problems. We identified 25 genes, including hypoxia-related genes (e.g., MIF, SCD1, P4HA1, ENO1, and STAT1) and cell cycle- and DNA repair-related genes (e.g., TACC3, PRDX1, PRKDC, and H2AFY). These genes showed significant differential expression among histological subtypes, including UPS, and showed associations with overall survival. STAT1 showed a strong association with overall survival in UPS patients (logrank p = 1.84 × 10(-6) and adjusted p value 2.99 × 10(-3) after the permutation test). According to the literature, the 25 genes selected are useful not only as markers of differential diagnosis but also as prognostic/predictive markers and/or therapeutic targets for STS. Our combination method can identify genes that are potential prognostic/predictive factors and/or therapeutic targets in STS and possibly in other cancers. These disease-associated genes deserve further preclinical and clinical validation.
Collapse
|
74
|
Zhu Y, Wang Y, Guan B, Rao Q, Wang J, Ma H, Zhang Z, Zhou X. C-kit and PDGFRA gene mutations in triple negative breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:4280-4285. [PMID: 25120810 PMCID: PMC4129045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/20/2014] [Indexed: 06/03/2023]
Abstract
In this study, we evaluated C-kit immunohistochemical expression and C-kit and platelet derived growth factor receptor A (PDGFRA) gene mutations in triple negative breast cancer. 171 cases were analyzed by immunohistochemical staining for the expression of C-kit and 45 cases, including 10 C-kit negative cases and 35 C-kit positive cases, were performed for C-kit gene mutations in exons 9, 11, 13 and 17 and PDGFRA gene mutations in exons 12 and 18. C-kit expression was detected in 42.1% of triple negative breast cancers. Only 1 activating mutation was detected in exon 11 of C-kit gene in 1 case. No activating mutations were found in the other 44 cases. C-kit expression is a frequent finding in triple negative breast cancers; 1 activating mutation which was also found in gastrointestinal stromal tumors was detected; a few cases might benefit from imatinib.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210003, China
| | - Bing Guan
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| | - Qiu Rao
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| | - Jiandong Wang
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| | - Henghui Ma
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| | - Zhihong Zhang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, China
| | - Xiaojun Zhou
- Department of Pathology, Clinical School of Medical College of Nanjing University Nanjing Jinling HospitalNanjing 210002, China
| |
Collapse
|
75
|
Abstract
Imatinib mesylate (Gleevec, Glivec [Novartis, Basel, Switzerland], formerly referred to as STI571 or CGP57148B) represents the paradigm of a new class of anticancer agents, so-called small molecules. They have a high selectivity against a specific molecular target known to be the cause for the establishment and maintenance of the malignant phenotype. Imatinib is a rationally designed oral signal transduction inhibitor that specifically targets several protein tyrosine kinases, Abl, Arg (Abl-related gene), the stem cell factor receptor (c-KIT), platelet-derived growth factor receptor (PDGF-R), and their oncogenic forms, most notably BCR-ABL. Imatinib has been shown to have remarkable clinical activity in patients with chronic myeloid leukemia (CML) and malignant gastrointestinal stroma tumors (GIST) leading to its approval for treatment of these diseases. Treatment with imatinib is generally well tolerated with a low incidence of severe side effects. The most common adverse events include mild to moderate edema, muscle cramps, diarrhea, nausea, skin rashes, and myelosuppression. Several mechanisms of resistance have been identified. Clonal evolution, amplification, or overexpression of BCR-ABL as well as mutations in the catalytic domain, P-loop, and other mutations have been demonstrated to play a role in primary and secondary resistance to imatinib, respectively. Understanding of the underlying mechanisms of resistance has led to the development of new second- and third-generation tyrosine kinase inhibitors (see chapters on dasatinib, nilotinib, bosutinib, and ponatinib).
Collapse
Affiliation(s)
- Cornelius F Waller
- Department of Hematology and Oncology, University of Freiburg Medical Center, Hugstetter Street 55, 79106, Freiburg, Germany,
| |
Collapse
|
76
|
Corless CL. Gastrointestinal stromal tumors: what do we know now? Mod Pathol 2014; 27 Suppl 1:S1-16. [PMID: 24384849 DOI: 10.1038/modpathol.2013.173] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/15/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the GI tract, arising from the interstitial cells of Cajal, primarily in the stomach and small intestine. They manifest a wide range of morphologies, from spindle cell to epithelioid, but are immunopositive for KIT (CD117) and/or DOG1 in essentially all cases. Although most tumors are localized at presentation, up to half will recur in the abdomen or spread to the liver. The growth of most GISTs is driven by oncogenic mutations in either of two receptor tyrosine kinases: KIT (75% of cases) or PDGFRA (10%). Treatment with tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, and regorafenib is effective in controlling unresectable disease; however, drug resistance caused by secondary KIT or PDGFRA mutations eventually develops in 90% of cases. Adjuvant therapy with imatinib is commonly used to reduce the likelihood of disease recurrence after primary surgery, and for this reason assessing the prognosis of newly resected tumors is one of the most important roles for pathologists. Approximately 15% of GISTs are negative for mutations in KIT and PDGFRA. Recent studies of these so-called wild-type GISTs have uncovered a number of other oncogenic drivers, including mutations in neurofibromatosis type I, RAS genes, BRAF, and subunits of the succinate dehydrogenase complex. Routine genotyping is strongly recommended for optimal management of GISTs, as the type and dose of TKI used for treatment is dependent on the mutation identified.
Collapse
Affiliation(s)
- Christopher L Corless
- Department of Pathology (L471) and Knight Diagnostic Laboratories, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
77
|
Al-Zaid T, Somaiah N, Lazar AJ. Targeted therapies for sarcomas: new roles for the pathologist. Histopathology 2013; 64:119-33. [DOI: 10.1111/his.12297] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 09/24/2013] [Indexed: 12/23/2022]
Affiliation(s)
- Tariq Al-Zaid
- Department of Pathology; King Faisal Specialist Hospital and Research Center; Riyadh Saudi Arabia
| | - Neeta Somaiah
- Sarcoma Research Center; The University of Texas MD Anderson Cancer Center; Houston TX USA
- Departments of Sarcoma Medical Oncology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Alexander J Lazar
- Sarcoma Research Center; The University of Texas MD Anderson Cancer Center; Houston TX USA
- Departments of Pathology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| |
Collapse
|
78
|
Doyle LA, Hornick JL. Gastrointestinal stromal tumours: from KIT to succinate dehydrogenase. Histopathology 2013; 64:53-67. [DOI: 10.1111/his.12302] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Leona A Doyle
- Department of Pathology; Brigham and Women's Hospital ; Harvard Medical School; Boston MA USA
| | - Jason L Hornick
- Department of Pathology; Brigham and Women's Hospital ; Harvard Medical School; Boston MA USA
| |
Collapse
|
79
|
Ma D, Stence AA, Bossler AB, Hackman JR, Bellizzi AM. Identification of KIT activating mutations in paediatric solitary mastocytoma. Histopathology 2013; 64:218-25. [PMID: 24128084 DOI: 10.1111/his.12212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 06/19/2013] [Indexed: 10/26/2022]
Abstract
AIMS Mastocytosis is an abnormal mast cell proliferation involving one or more organs, in particular the skin and bone marrow. In children, disease is usually limited to the skin, with three distinct clinical presentations: urticaria pigmentosa, diffuse cutaneous mastocytosis and solitary mastocytoma. Although the KIT D816V mutation is typically found in adult-onset mastocytosis, it is less commonly seen in childhood-onset mastocytosis, and the frequency of KIT mutations in paediatric solitary mastocytoma is poorly documented. METHODS AND RESULTS In this study we analysed KIT exons 8, 9, 11, 13 and 17 in nine cases of paediatric solitary mastocytoma using a laboratory-developed Sanger sequencing assay. A KIT mutation was identified in six cases (67%), including three with the D816V mutation typical of adult-onset disease, and another three with an internal tandem duplication (p.A502_Y503dup) in exon 9, previously described in gastrointestinal stromal tumour. CONCLUSIONS Paediatric solitary mastocytoma is frequently associated with KIT activating mutations, in keeping with a clonal process. KIT mutational status appears insufficient to explain the divergent biology of childhood and adult-onset disease.
Collapse
Affiliation(s)
- Deqin Ma
- Department of Pathology, University of Iowa Hospitals and Clinics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|
80
|
Hayashi Y, Asuzu DT, Gibbons SJ, Aarsvold KH, Bardsley MR, Lomberk GA, Mathison AJ, Kendrick ML, Shen KR, Taguchi T, Gupta A, Rubin BP, Fletcher JA, Farrugia G, Urrutia RA, Ordog T. Membrane-to-nucleus signaling links insulin-like growth factor-1- and stem cell factor-activated pathways. PLoS One 2013; 8:e76822. [PMID: 24116170 PMCID: PMC3792098 DOI: 10.1371/journal.pone.0076822] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/29/2013] [Indexed: 11/29/2022] Open
Abstract
Stem cell factor (mouse: Kitl, human: KITLG) and insulin-like growth factor-1 (IGF1), acting via KIT and IGF1 receptor (IGF1R), respectively, are critical for the development and integrity of several tissues. Autocrine/paracrine KITLG-KIT and IGF1-IGF1R signaling are also activated in several cancers including gastrointestinal stromal tumors (GIST), the most common sarcoma. In murine gastric muscles, IGF1 promotes Kitl-dependent development of interstitial cells of Cajal (ICC), the non-neoplastic counterpart of GIST, suggesting cooperation between these pathways. Here, we report a novel mechanism linking IGF1-IGF1R and KITLG-KIT signaling in both normal and neoplastic cells. In murine gastric muscles, the microenvironment for ICC and GIST, human hepatic stellate cells (LX-2), a model for cancer niches, and GIST cells, IGF1 stimulated Kitl/KITLG protein and mRNA expression and promoter activity by activating several signaling pathways including AKT-mediated glycogen synthase kinase-3β inhibition (GSK3i). GSK3i alone also stimulated Kitl/KITLG expression without activating mitogenic pathways. Both IGF1 and GSK3i induced chromatin-level changes favoring transcriptional activation at the Kitl promoter including increased histone H3/H4 acetylation and H3 lysine (K) 4 methylation, reduced H3K9 and H3K27 methylation and reduced occupancy by the H3K27 methyltransferase EZH2. By pharmacological or RNA interference-mediated inhibition of chromatin modifiers we demonstrated that these changes have the predicted impact on KITLG expression. KITLG knock-down and immunoneutralization inhibited the proliferation of GIST cells expressing wild-type KIT, signifying oncogenic autocrine/paracrine KITLG-KIT signaling. We conclude that membrane-to-nucleus signaling involving GSK3i establishes a previously unrecognized link between the IGF1-IGF1R and KITLG-KIT pathways, which is active in both physiologic and oncogenic contexts and can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - David T. Asuzu
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kirsten H. Aarsvold
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gwen A. Lomberk
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Angela J. Mathison
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michael L. Kendrick
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - K. Robert Shen
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Takahiro Taguchi
- Division of Human Health and Medical Science, Graduate School of Kuroshio Science, Kochi University, Nankoku, Kochi, Japan
| | - Anu Gupta
- Departments of Anatomic Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Brian P. Rubin
- Departments of Anatomic Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Raul A. Urrutia
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tamas Ordog
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
81
|
Bello DM, Ariyan CE, Carvajal RD. Melanoma Mutagenesis and Aberrant Cell Signaling. Cancer Control 2013; 20:261-81. [DOI: 10.1177/107327481302000404] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Danielle M. Bello
- Department of Surgery Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Charlotte E. Ariyan
- Department of Surgery Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Richard D. Carvajal
- Department of Medical Oncology Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
82
|
Malik L, Hemmings C, Beshay V, Fox S, Yip D. Metastatic gastrointestinal stromal tumour of the ileum with dual primary c-KIT missence mutations. Pathology 2013; 45:604-606. [PMID: 24018806 DOI: 10.1097/pat.0b013e3283653792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Laeeq Malik
- *Medical Oncology Unit, The Canberra Hospital, Garran †ANU Medical School, Australian National University, Acton, ACT ‡School of Surgery, University of Western Australia §St John of God Pathology, Subiaco, WA ||Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | | | | | | | | |
Collapse
|
83
|
Barnett CM, Corless CL, Heinrich MC. Gastrointestinal stromal tumors: molecular markers and genetic subtypes. Hematol Oncol Clin North Am 2013; 27:871-88. [PMID: 24093165 DOI: 10.1016/j.hoc.2013.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutation-activated signaling from the KIT and PDGFRA kinases has been successfully targeted in gastrointestinal stromal tumors (GISTs), with subtle differences between the mutations serving to refine prognosis and more precisely tailor therapy. There is a growing understanding of the molecular drivers of GISTs lacking mutations in KIT or PDGFRA, so called wild-type GISTs, further aiding in management decisions. This article provides an overview of all the known molecular subtypes of GIST and provides information about clinical correlates, treatment, and prognosis depending on the subtype.
Collapse
Affiliation(s)
- Christine M Barnett
- Hematology and Medical Oncology, Division of Hematology/Oncology, Portland VA Medical Center, OHSU Knight Cancer Institute, Oregon Health & Science University, Mail Code L586, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | |
Collapse
|
84
|
Abstract
Gastrointestinal stromal tumours (GISTs) are mesenchymal neoplasms that arise in the gastrointestinal tract, usually in the stomach or the small intestine and rarely elsewhere in the abdomen. They can occur at any age, the median age being 60-65 years, and typically cause bleeding, anaemia, and pain. GISTs have variable malignant potential, ranging from small lesions with a benign behaviour to fatal sarcomas. Most tumours stain positively for the mast/stem cell growth factor receptor KIT and anoctamin 1 and harbour a kinase-activating mutation in either KIT or PDGFRA. Tumours without such mutations could have alterations in genes of the succinate dehydrogenase complex or in BRAF, or rarely RAS family genes. About 60% of patients are cured by surgery. Adjuvant treatment with imatinib is recommended for patients with a substantial risk of recurrence, if the tumour has an imatinib-sensitive mutation. Tyrosine kinase inhibitors substantially improve survival in advanced disease, but secondary drug resistance is common.
Collapse
Affiliation(s)
- Heikki Joensuu
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| | | | | |
Collapse
|
85
|
Linch M, Claus J, Benson C. Update on imatinib for gastrointestinal stromal tumors: duration of treatment. Onco Targets Ther 2013; 6:1011-23. [PMID: 23935374 PMCID: PMC3735340 DOI: 10.2147/ott.s31260] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common sarcoma of the gastrointestinal tract, with transformation typically driven by activating mutations of c-KIT and less commonly platelet-derived growth factor receptor alpha (PDGFRA). Successful targeting of c-KIT and PDGFRA with imatinib, a tyrosine kinase inhibitor (TKI), has had a major impact in advanced GIST and as an adjuvant and neoadjuvant treatment. If treatment with imatinib fails, further lines of TKI therapy have a role, but disease response is usually only measured in months, so strategies to maximize the benefit from imatinib are paramount. Here, we provide an overview of the structure and signaling of c-KIT coupled with a review of the clinical trials of imatinib in GIST. In doing so, we make recommendations about the duration of imatinib therapy and suggest how best to utilize imatinib in order to improve patient outcomes in the future.
Collapse
Affiliation(s)
- Mark Linch
- Sarcoma Unit, Royal Marsden Hospital, United Kingdom ; Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | | | | |
Collapse
|
86
|
KIT gene mutation analysis in solid tumours: biology, clincial applications and trends in diagnostic reporting. Pathology 2013; 45:127-37. [PMID: 23277171 DOI: 10.1097/pat.0b013e32835c7645] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gain-of-function mutations involving c-kit protein, a cell-surface transmembrane receptor for stem cell factor, have been identified as a key oncogenic driver in a variety of solid tumours. Coupled with the development of tyrosine kinase inhibitors such as imatinib, c-kit has emerged as a viable drug target in what seems to be a validated therapeutic concept. This review will focus on gastrointestinal stromal tumours and melanomas, two types of solid tumours most closely associated with KIT gene mutations. The biology of KIT mutations in both conditions, as well as the value of KIT mutation testing in predicting disease and treatment outcomes are discussed. Since initial response to imatinib is largely influenced by mutation status, genotyping these tumours serves to facilitate personalised oncology. We also summarise our experience with diagnostic reporting of KIT mutation analysis over a period of 3 years, and briefly survey future developments in treatment, which indeed look very promising.
Collapse
|
87
|
Abstract
PURPOSE OF REVIEW Malignant melanoma is the most frequent cause of mortality in skin cancer. Unlike Caucasians, mucosal and acral melanomas account for approximately 65% of all melanomas in Chinese. Genetic mutations of c-kit are detected in mucosal and acral melanomas, which thus can be regarded as molecular targets for treatments. This review describes the recent proceedings in the c-kit-targeted molecular therapy of melanoma, on the basis of our experiences in Chinese melanoma patients. RECENT FINDINGS Somatic mutations within c-kit gene have been detected in 10.8% of Chinese melanoma patients. Imatinib, a selective inhibitor targeting Abl as well as c-kit and the platelet-derived growth factor receptor, has been tested for the efficacy and toxicities in metastatic melanoma patients, suggesting that imatinib may increase the progression-free survival and overall survival in selected melanoma patients harboring mutations in c-kit gene. The current status of c-kit mutation and the standard for selection of imatinib-sensitive patients have been tentatively established. SUMMARY C-kit is a proto-oncogene that can serve as a potential target for molecular therapy of metastatic melanoma. Imatinib is effective and well tolerated in the treatment of metastatic melanoma patients. In selected subsets of melanoma patients harboring genetic alterations of c-kit, imatinib may be a promising agent.
Collapse
|
88
|
Pardanani A. Systemic mastocytosis in adults: 2013 update on diagnosis, risk stratification, and management. Am J Hematol 2013; 88:612-24. [PMID: 23720340 DOI: 10.1002/ajh.23459] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/04/2013] [Indexed: 12/20/2022]
Abstract
DISEASE OVERVIEW Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extracutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. RISK STRATIFICATION The 2008 World Health Organization (WHO) classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent (SM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD) and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. MANAGEMENT SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-α (±corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal, and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting. INVESTIGATIONAL DRUGS Dasatinib's in vitro anti- KITD816V activity has not translated into significant therapeutic activity in most SM patients. In contrast, recently updated data confirms Midostaurin's significant anti-MC activity in patients with advanced SM.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
89
|
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumor of the gastrointestinal tract. Soon after GIST was recognized as a tumor driven by a KIT or platelet-derived growth factor receptor mutation, it became the first solid tumor target for tyrosine kinase inhibitor therapies. More recently, alternative molecular mechanisms for GIST pathogenesis have been discovered. These are related to deficiencies in the succinate dehydrogenase complex, NF1-gene alterations in connection with neurofibromatosis type 1 tumor syndrome, and mutational activation of the BRAF oncogene in very rare cases.
Collapse
Affiliation(s)
- Markku Miettinen
- Laboratory of Pathology, NCI/NIH, 9000 Rockville Pike, Building 10, Rm. 2B50, Bethesda, Maryland 20892,
| | - Jerzy Lasota
- Laboratory of Pathology, NCI/NIH, 9000 Rockville Pike, Building 10, Rm. 2B50, Bethesda, Maryland 20892,
| |
Collapse
|
90
|
Extracellular domain c-kit mutation with duplication of Ser501Ala502 found in gastrointestinal stromal tumors is more imatinib- and nilotinib-sensitive than that with duplication of Ala502Tyr503. J Transl Med 2013; 93:502-7. [PMID: 23459373 DOI: 10.1038/labinvest.2013.43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The great majority of gastrointestinal stromal tumors (GISTs) have gain-of-function mutations of the c-kit gene, which encodes KIT receptor tyrosine kinase. Most of the mutations are located at exon 11, but some are at exon 9 or at other exons. Mutation types at exon 11 vary, while most mutations at exon 9 are a particular duplication of Ala502Tyr503 (KIT-Dup-Ala502Tyr503). Recently a duplication of Ser501Ala502 (KIT-Dup-Ser501Ala502) at exon 9 has been reported in two cases of pediatric mastocytosis and one case of adult mast cell leukemia. Although KIT-Dup-Ser501Ala502 had not been reported in GISTs, we found two GIST cases possessing the mutation in 45 GIST cases with exon 9 c-kit gene mutations, among a total of approximately 500 GIST cases examined. In this report, we briefly summarize clinicopathological findings of the two cases, and characterize the biology of the mutation. When autophosphorylation of KIT-Dup-Ser501Ala502 was examined by transient transfection of c-kit cDNA with Dup-Ser501Ala502 into CHO-K1 cells, KIT-Dup-Ser501Ala502 was ligand-independently activating. The inhibitory effect of selective tyrosine kinase inhibitors, imatinib and nilotinib, on KIT-Dup-Ser501Ala502 was examined and compared with that of KIT-Dup-Ala502Tyr503. Imatinib efficiently inhibited constitutive activation of KIT-Dup-Ser501Ala502 at a concentration of 0.1 μM, whereas it inhibited that of KIT-Dup-Ala502Tyr503 at a concentration of 10 μM. Constitutive activation of KIT-Dup-Ser502Ala503 was not inhibited by nilotinib even at a concentration of 10 μM but that of KIT-Dup-Ala501Tyr502 was almost completely inhibited at a concentration of 1 μM. The results suggest that imatinib and nilotinib could be more effective on GISTs with KIT-Dup-Ser501Ala502 than those with KIT-Dup-Ala502Tyr503. In fact, a patient with KIT-Dup-Ser501Ala502 showed long-term stable disease with administration of the usual dose of 400 mg imatinib. Although mutation sites of KIT-Dup-Ser501Ala502 and KIT-Dup-Ala502Tyr503 are closely located, imatinib- and nilotinib-sensitive KIT-Dup-Ser501Ala502 are distinguishable from KIT-Dup-Ala502Tyr503.
Collapse
|
91
|
Yushak M, Kluger HM, Sznol M. Advances in the systemic treatment of metastatic melanoma. ONCOLOGY (WILLISTON PARK, N.Y.) 2013; 27:374-81. [PMID: 25184258 PMCID: PMC6092183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Prior to 2011, the only commercially available agents commonly used to treat metastatic melanoma-including dacarbazine, temozolomide (Temodar), fotemustine, carboplatin, paclitaxel, and interleukin-2-demonstrated limited efficacy, and no study involving these agents had shown an improvement in overall survival. The standard of care for the treatment of metastatic melanoma was radically changed by the subsequent approval of two agents, ipilimumab (Yervoy) and vemurafenib (Zelboraf), both of which improved survival in randomized phase III trials. Within the relatively short time that ipilimumab and vemurafenib have been commercially available, phase II data for the investigational agents nivolumab and MK-3475, for the combination of dabrafenib and trametinib, and for adoptive cell therapy strongly suggest even further improvements in treatment outcomes. Within this rich context of effective agents, the challenge for clinicians and investigators will be to develop predictive biomarkers of response, the optimal sequence of therapy for individual patients, and effective combinations. An additional challenge will be to find the appropriate venue and populations to test promising new agents arising from substantial advances in our understanding of molecular alterations in melanoma cells, of mechanisms of resistance to current agents, and of tumor-host immune interactions.
Collapse
Affiliation(s)
- Melinda Yushak
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT
| | - Harriet M. Kluger
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT
| | - Mario Sznol
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
92
|
Kara T, Serinsoz E, Arpaci RB, Gubur O, Orekici G, Ata A, Colak T, Arican A. Contribution of DOG1 expression to the diagnosis of gastrointestinal stromal tumors. Pathol Res Pract 2013; 209:413-7. [PMID: 23722018 DOI: 10.1016/j.prp.2013.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 03/11/2013] [Accepted: 04/16/2013] [Indexed: 12/25/2022]
Abstract
Gastrointestinal stromal tumors (GIST) are the most common mesenchymal tumors of the gastrointestinal tract, and the majority contain KIT or PDGFRA-activating mutations. However, up to 10% of GISTs are c-kit-negative. Antibodies with increased sensitivity and specificity for the detection of c-kit-negative GIST cases may be of value, especially because some of these cases may also benefit from tyrosine kinase inhibitor therapy. Hematoxylin and Eosin sections of 33 GISTs were re-examined in order to define histopathological criteria used in risk assessment of these tumors. Immunohistochemistry with a panel of antibodies [c-kit, DOG1 (discovered on GIST 1), CD34, smooth muscle actin (SMA), Desmin, S100 and Ki67] was performed on 5μm-thick paraffin sections of all tumors. Statistical analysis of immunohistochemical studies showed that DOG1 and CD117 were the most sensitive and specific antibodies in the diagnosis of GISTs. Other antibodies were unhelpful in confirming a diagnosis of GIST, but were particularly useful in the differential diagnosis. Reactivity for DOG1 may aid in the diagnosis of GISTs, which fail to express c-kit antigen, and lead to appropriate treatment with imatinib mesylate, an inhibitor of the KIT tyrosine kinase.
Collapse
Affiliation(s)
- Tuba Kara
- Mersin University, Medical School, Department of Pathology, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Imatinib and Dasatinib Inhibit Hemangiosarcoma and Implicate PDGFR-β and Src in Tumor Growth. Transl Oncol 2013; 6:158-68. [PMID: 23544168 DOI: 10.1593/tlo.12307] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 01/24/2013] [Accepted: 01/28/2013] [Indexed: 12/15/2022] Open
Abstract
Hemangiosarcoma, a natural model of human angiosarcoma, is an aggressive vascular tumor diagnosed commonly in dogs. The documented expression of several receptor tyrosine kinases (RTKs) by these tumors makes them attractive targets for therapeutic intervention using tyrosine kinase inhibitors (TKIs). However, we possess limited knowledge of the effects of TKIs on hemangiosarcoma as well as other soft tissue sarcomas. We report here on the use of the TKIs imatinib and dasatinib in canine hemangiosarcoma and their effects on platelet-derived growth factor receptor β (PDGFR-β) and Src inhibition. Both TKIs reduced cell viability, but dasatinib was markedly more potent in this regard, mediating cytotoxic effects orders of magnitude greater than imatinib. Dasatinib also inhibited the phosphorylation of the shared PDGFR-β target at a concentration approximately 1000 times less than that needed by imatinib and effectively blocked Src phosphorylation. Both inhibitors augmented the response to doxorubicin, suggesting that clinical responses likely will be improved using both drugs in combination; however, dasatinib was significantly (P < .05) more effective in this context. Despite the higher concentrations needed in cell-based assays, imatinib significantly inhibited tumor growth (P < .05) in a tumor xenograft model, highlighting that disruption of PDGFR-β/PDGF signaling may be important in targeting the angiogenic nature of these tumors. Treatment of a dog with spontaneously occurring hemangiosarcoma established that clinically achievable doses of dasatinib may be realized in dogs and provides a means to investigate the effect of TKIs on soft tissue sarcomas in a large animal model.
Collapse
|
94
|
Gastrointestinal Stromal Tumours: Review of 150 Cases from a Single Centre. Indian J Surg 2013; 77:505-10. [PMID: 26730054 DOI: 10.1007/s12262-013-0899-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 03/11/2013] [Indexed: 02/08/2023] Open
Abstract
Gastrointestinal stromal tumours (GIST) are the most common mesenchymal tumours of the gastrointestinal tract. They constitute a significant percentage ranging from 1-2 % of all the gastrointestinal neoplasms [11]. Knowledge on the molecular biology and behavior of these tumours is still not very clear. The clinicopathologial features are variable and surgical resection with chemotherapy is the main modality of treatment. We have retrospectively analyzed the clinicopathological features, treatment and prognosis of 150 patients managed in the Department of Surgery. Retrospective review of the records of 150 patients diagnosed with gastrointestinal stromal tumours and managed during the period January 2006 to December 2011. Clinicopathological features, immunohistochemistry, mitotic index, surgical resection adjuvant chemotherapy and survival analyzed. One hundred and fifty patients diagnosed with GIST and treated were reviewed. Ninety five of them were males (63.3 %). The tumour was most commonly seen during the fourth and fifth decades of life. Abdominal pain (52 %), intestinal bleeding (40 %) and abdominal mass (25 %) were the common clinical symptoms. Sixty percent of the tumours (90/150) were located in the stomach followed by small bowel (20 %) and duodenum (14.6 %). One hundred and thirty-five patients underwent excision of the tumour and five patients had multi organ resection of the adjacent organs like spleen, tail of the pancreas and kidney. Fifteen patients (10 %) received neoadjuvant Imatinib for down staging of the tumour prior to surgery. The tumour size ranged from 1 to 34 cm. One third of the tumours (42/150) belonged to the high-grade category. KIT protein (CD117) was positive in 90 %( 135/150), while CD34 was positive in 50 % (74/150) of tumours. Majority of the patients with high and intermediate-risk category received adjuvant Imatinib (65/77). Seventeen patients (11.3 %) developed recurrence of the tumour on follow-up and rest of the patients had stable disease. Eight of the 15 patients (53 %) who had advanced disease developed recurrence of the disease over 6 months to 1 year. Fifteen patients died on follow-up between 2 and 5 years. Gastrointestinal tumours are the most common non epithelial tumour of the GIT. GISTS are found to show a male preponderance and are common during the fourth and fifth decades. Abdominal pain and intestinal bleeding are the most common clinical presentation. Most of the tumours were located in the stomach. Surgical resection is the best modality of treatment for operable lesions. Tyrosine kinase receptor (KIT) inhibitor like imatinib is used for adjuvant treatment. Regular follow-up with ultra sonogram or computed tomogram helps in diagnosing disease recurrence.
Collapse
|
95
|
Hayes S, Yuzbasiyan-Gurkan V, Gregory-Bryson E, Kiupel M. Classification of canine nonangiogenic, nonlymphogenic, gastrointestinal sarcomas based on microscopic, immunohistochemical, and molecular characteristics. Vet Pathol 2013; 50:779-88. [PMID: 23456969 DOI: 10.1177/0300985813478211] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Canine nonangiogenic, nonlymphogenic, gastrointestinal sarcomas have been previously diagnosed as gastrointestinal stromal tumors (GIST), leiomyosarcomas, or nonspecified spindle cell sarcomas, but diagnostic criteria for each entity are poorly defined. We propose a classification for canine nonangiogenic, nonlymphogenic, gastrointestinal sarcomas based on microscopic, immunohistochemical, and molecular characteristics. Applying the classification to 40 canine nonangiogenic, nonlymphogenic, gastrointestinal sarcomas documented its diagnostic and prognostic value. Eighteen (45%) sarcomas were classified as GIST based on positive KIT immunoreactivity. All GISTs were positive for vimentin, 14 (78%) were positive for S-100, and 6 (33%) were positive for smooth muscle actin (SMA). In contrast to their human counterparts, canine GISTs occurred mainly in the small intestine (67%) but commonly metastasized (5/18) to liver, lymph nodes, and omentum. Six GISTs had an activated KIT mutation in exon 11 of c-Kit, but no mutations were detected in exons 8, 9, 13, and 17. Twelve (30%) sarcomas were classified as leiomyosarcomas based on positive labeling for SMA and negative labeling for KIT. Four of these neoplasms were well differentiated leiomyosarcomas characterized by weak to no labeling for vimentin, and 8 were poorly differentiated leiomyosarcomas characterized by strong labeling for vimentin. None of the leiomyosarcomas metastasized, but poorly differentiated leiomyosarcomas had a higher risk of local invasion. Ten (25%) sarcomas were classified as non-GIST/nonleiomyosarcomas that were negative for KIT and SMA but positive for vimentin and either S-100 and/or PGP 9.5. These neoplasms most likely represent sarcomas of neurogenic differentiation resembling Schwann cells or perineurial or endoneurial fibroblasts, respectively.
Collapse
Affiliation(s)
- S Hayes
- Diagnostic Center for Population and Animal Health, College of Veterinary Medicine, Michigan State University, Lansing, MI 48910, USA
| | | | | | | |
Collapse
|
96
|
A novel germline SDHB mutation in a gastrointestinal stromal tumor patient without bona fide features of the Carney-Stratakis dyad. Fam Cancer 2012; 11:189-94. [PMID: 22160509 DOI: 10.1007/s10689-011-9499-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchyme neoplasms of the gastrointestinal tract. Gain-of-function somatic mutations of the KIT or PDGFRA genes represent the most prevalent molecular alterations in GISTs. In Carney-Stratakis dyad, patients portray germline mutations of the succinate dehydrogenase subunits B (SDHB), C (SDHC) and D (SDHD) and develop multifocal GISTs and multicentric paragangliomas (PGLs). We herein report a novel germline SDHB mutation (c.T282A--Ile44Asn) occurring in a 26 years-old patient diagnosed with a spindle cell intermediate risk GIST that did not present KIT/PDGFRA/BRAF gene mutations. Further analyses revealed loss of the wild-type SDHB allele and complete loss of SDHB expression in the tumor tissue. After genetic screening of other family members, we detected in the patient's mother a SDHB mutation without any clinical/laboratorial evidence of GIST or PGL. Altogether, our findings (germline SDHB mutation with absence of PGL in the index case and of GIST and/or PGL in his mother) raise the possibility that this familiar setting corresponds to an incomplete phenotype of the Carney-Stratakis dyad.
Collapse
|
97
|
Hou XW, Bai CG, Liu XH, Qiu C, Huang L, Xu JJ, Ma DAL. Expression of stem cell factor in gastrointestinal stromal tumors: Implications for proliferation and imatinib resistance. Oncol Lett 2012; 5:552-558. [PMID: 23420128 PMCID: PMC3572958 DOI: 10.3892/ol.2012.1019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/31/2012] [Indexed: 01/13/2023] Open
Abstract
KIT autophosphorylation caused by mutation of KIT is considered to be a critical mechanism for the oncogenesis of gastrointestinal stromal tumors (GISTs). However, little is known regarding whether stem cell factor (SCF), the KIT ligand, is able to induce the proliferation of GIST cells by activating the wild-type KIT receptor in GISTs. Imatinib, a tyrosine kinase inhibitor, has been demonstrated to be effective as treatment for the majority of GISTs. However, primary resistance to imatinib in GISTs with wild-type KIT and acquired resistance in GISTs with mutant KIT are becoming increasingly significant problems. The aims of this study were to detect the expression and function of SCF in 68 GIST samples, and to explore the relationship between SCF activity and imatinib resistance using immunohistochemical staining and western blot analysis. Results showed abundant expression of SCF in GISTs and demonstrated that SCF is capable of enhancing GIST cell proliferation. Similar to its ineffectiveness in wild-type GISTs, imatinib also failed to inhibit SCF-induced KIT activation in GISTs with mutant KIT. We also found increased SCF expression in GIST cells treated with imatinib. Overall, our results indicated that SCF-induced KIT activation is a novel essential pathway for the proliferation of GISTs. Imatinib was not able to inhibit the activity of SCF, while it promoted the expression of SCF, which may have contributed to acquired imatinib resistance.
Collapse
Affiliation(s)
- Xiao-Wei Hou
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433; ; Department of Oncology, 401 Hospital of PLA, Qingdao, Shandong 266071
| | | | | | | | | | | | | |
Collapse
|
98
|
Chu PY, Pan SL, Liu CH, Lee J, Yeh LS, Liao AT. KIT gene exon 11 mutations in canine malignant melanoma. Vet J 2012; 196:226-30. [PMID: 23069279 DOI: 10.1016/j.tvjl.2012.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 08/24/2012] [Accepted: 09/11/2012] [Indexed: 12/17/2022]
Abstract
The proto-oncogene KIT encodes a receptor tyrosine kinase which has been shown to be upregulated in canine melanomas. KIT mutations lead to constitutive phosphorylation and activation of KIT in the absence of ligand binding. The presence of KIT mutations and KIT protein expression was examined in a cohort of 49 dogs with canine malignant melanoma. An exon 11 synonymous nucleotide 1743C→T mutation was identified in five cases in which one also harbored a L579P mutation. Tumors that harbored the KIT exon 11 mutation(s) correlated significantly with disease recurrence (P = 0.05). All 36 melanomas available for immunohistochemical analysis showed either weak (16 cases, 44.4%) or strong (20 cases, 55.6%) expression of the KIT protein. The five KIT mutation carriers were all strongly positive for KIT by immunohistochemical staining. These findings suggest that a subset of canine malignant melanomas harbors a KIT exon 11 mutation.
Collapse
Affiliation(s)
- Pei-Yi Chu
- Department of Pathology, St. Martin De Porres Hospital, Chiayi 60069, Taiwan
| | | | | | | | | | | |
Collapse
|
99
|
Chen YH, Liu KH, Yeh CN, Hsu JT, Liu YY, Tsai CY, Chiu CT, Jan YY, Yeh TS. Laparoscopic resection of gastrointestinal stromal tumors: safe, efficient, and comparable oncologic outcomes. J Laparoendosc Adv Surg Tech A 2012; 22:758-763. [PMID: 22957924 DOI: 10.1089/lap.2012.0115] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Surgery of gastrointestinal stromal tumors (GISTs) has been modified, and laparoscopic resection of GIST has gained improvement and roles. PATIENTS AND METHODS We retrospectively reviewed clinical data and oncological outcomes of our GIST patients who underwent laparoscopic surgery and traditional open surgery. In total, 227 pathologically diagnosed GIST cases were retrospectively reviewed in Chang Gung Memorial Hospital at Linkou, Taipei, Taiwan, between 2005 and 2010. We excluded those with tumor size >5 cm, biopsy-only, combined other operation, endoscopic mucosal resection, tumor located in the duodenum, colon-rectum, esophagocardiac junction, omentum, pelvic area, or retroperitoneum, or metastasis when operated on and those diagnosed as other disease after immunohistologic examination of GIST. Fifty-eight cases were enrolled, including 16 patients in the laparoscopic surgery group (LSG) and 42 patients in the open surgery group (OSG). The patients' demography, perioperative, pathologic result, and oncology result were recorded and analyzed. RESULTS Both groups showed no difference in clinical demography, tumor size, and locations. LSG patients showed fewer days to resume diet, shorter postoperative hospital stays, and less use of patient-controlled analgesia. The postoperative morbidity in LSG and OSG was 6.3% and 19%, respectively. The median follow-up time was 32.73 months in LSG and 39.75 months in OSG. Recurrence or metastasis was observed in 3 patients (1 in LSG and 2 in OSG). The recurrence rate between LSG and OSG showed no significant difference. CONCLUSIONS Laparoscopic surgery was technically feasible for GIST of no more than 5 cm located at the stomach and small bowel. In the current study, we demonstrated that LSG patients benefited from fewer days to resume diet (5 versus 5.71 days), shorter postoperative stays (8 versus 9.07 days), and less patient-controlled analgesia use (6.7% versus 90.9%) during the perioperative period with the same short-term oncology result compared with OSG patients.
Collapse
Affiliation(s)
- Yu-Hsien Chen
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Thumar J, Giesen E, Kluger HM. Drug targets and predictive biomarkers in the management of metastatic melanoma. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2012; 5:139-48. [PMID: 23226069 PMCID: PMC3513235 DOI: 10.2147/pgpm.s25100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Indexed: 12/05/2022]
Abstract
Melanoma is the leading cause of fatal skin cancer, and in the past few decades, there has been an increase in the incidence of and mortality from metastatic melanoma. Until recently, the therapeutic options for treatment of metastatic melanoma were limited. The approval of ipilimumab (an anti-CTLA-4 antibody) and vemurafenib (mutant B-RAFV600E kinase inhibitor) by the Federal Drug Administration has led to a new era in melanoma treatment, and additional promising drugs and drug combinations are currently being investigated. As the choices of treatment for melanoma have expanded, the need to identify predictive biomarkers to tailor treatment strategies to individual tumor or immune system characteristics has become necessary. Such strategies have the potential of maximizing antitumor effect while minimizing toxicity and improving clinical benefit. In this article, we review the currently approved targeted therapies in melanoma and discuss the future of personalized therapy for this disease.
Collapse
Affiliation(s)
- Jaykumar Thumar
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|