1
|
Muramatsu S, Ogura K, Morizane C, Satake T, Toda Y, Iwata S, Kobayashi E, Higashi T, Ikeuchi M, Kawai A. Statistics of Visceral Sarcoma in Japan: Report From the Population-Based National Cancer Registry (NCR) in Japan. J Surg Oncol 2025; 131:274-284. [PMID: 39257202 DOI: 10.1002/jso.27867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/18/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND AND OBJECTIVES Sarcomas developing in the visceral organs are extremely rare, with no previous reports to describe their national epidemiology. We analyzed Japanese domestic statistics for visceral sarcoma, using the National Cancer Registry (NCR) in Japan, a population-based database launched in 2016. METHODS We identified 3245 cases of visceral sarcomas in the NCR dated 2016-2019 to analyze demographic and disease information, initial diagnostic process, volume and type of the hospitals, treatment, and prognosis. RESULTS Visceral sarcoma shows a higher prevalence in the older generation (60+ years), with a significant male predominance (p = 0.006). Leiomyosarcomas occurred frequently in the gastrointestinal tract (N = 240; 39.5%), and angiosarcomas in the liver, gall bladder, pancreas, and spleen (N = 244; 43.9%). Visceral sarcomas were often treated in facilities of lower volume without specific adjuvant treatments (p < 0.001). The cumulative 3-year overall survival was 44.8%, and several factors such as surgery or absence of chemotherapy positively affected survival. CONCLUSIONS This is the first nationwide study in Japan to analyze the inclusive epidemiology of visceral sarcomas. Visceral sarcomas are characterized by senior and male predominance with relatively poor prognosis, often managed in nonspecialized facilities and rarely with adjuvant therapies. Several histologic subtypes had the propensity to develop in specific organs.
Collapse
Affiliation(s)
- Shudai Muramatsu
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
- Department of Orthopaedic Surgery, Kochi Medical School, Kochi, Nankoku, Japan
| | - Koichi Ogura
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Tomoyuki Satake
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Chiba, Kashiwa, Japan
| | - Yu Toda
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Shintaro Iwata
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Eisuke Kobayashi
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Takahiro Higashi
- Division of Health Services Research, Center for Cancer Control and Information Services, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| | - Masahiko Ikeuchi
- Department of Orthopaedic Surgery, Kochi Medical School, Kochi, Nankoku, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Tsukiji, Japan
| |
Collapse
|
2
|
Ali RH, Alsaber AR, Mohanty AK, Alnajjar A, Mohammed EMA, Alateeqi M, Jama H, Almarzooq A, Benobaid N, Alqallaf Z, Ahmed AA, Bahzad S, Alkandari M. Molecular Profiling of KIT/PDGFRA-Mutant and Wild-Type Gastrointestinal Stromal Tumors (GISTs) with Clinicopathological Correlation: An 18-Year Experience at a Tertiary Center in Kuwait. Cancers (Basel) 2024; 16:2907. [PMID: 39199677 PMCID: PMC11352935 DOI: 10.3390/cancers16162907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
In gastrointestinal stromal tumors (GISTs), identifying prototypical mutations in the KIT/PDGFRA oncogenes, or in rare alternate genes, is essential for prognostication and predicting response to tyrosine kinase inhibitors. Conversely, wild-type GISTs (WT-GIST), which lack known mutations, have limited treatment options. Data on the mutational landscape of GISTs and their impact on disease progression are very limited in Kuwait. Using a targeted next-generation sequencing panel, we investigated the spectrum and frequency of KIT, PDGFRA, and RAS-pathway-related mutations in 95 out of 200 GISTs diagnosed at Kuwait Cancer Center from 2005 to 2023 and assessed their correlation with clinicopathological parameters. Among the 200 tumors (median age 55 years; 15-91), 54% originated in the stomach, 33% in the small bowel, 7% in the colorectum, 1.5% in the peritoneum, and 4.5% had an unknown primary site. Of the 95 molecularly profiled cases, 88% had a mutation: KIT (61%), PDGFRA (25%), NF1 (2%), and one NTRK1 rearrangement. Ten WT-GISTs were identified (stomach = 6, small bowel = 2, and colorectum = 2). WT-GISTs tended to be smaller (median 4.0 cm; 0.5-8.0) (p = 0.018), with mitosis ≤5/5 mm2, and were of lower risk (p = 0.019). KIT mutations were an adverse indicator of disease progression (p = 0.049), while wild-type status did not significantly impact progression (p = 0.934). The genetic landscape in this cohort mirrors that of global studies, but regional collaborations are needed to correlate outcomes with genetic variants.
Collapse
Affiliation(s)
- Rola H. Ali
- Department of Pathology, College of Medicine, Kuwait University, Safat 13110, Kuwait
- Histopathology Laboratory, Sabah Hospital, Sabah Medical District, Safat 13001, Kuwait
| | - Ahmad R. Alsaber
- Department of Management, College of Business and Economics, American University of Kuwait, Safat 13034, Kuwait;
| | - Asit K. Mohanty
- Department of Medical Oncology, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (A.K.M.); (A.A.)
| | - Abdulsalam Alnajjar
- Department of Medical Oncology, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (A.K.M.); (A.A.)
| | - Eiman M. A. Mohammed
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Mona Alateeqi
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Hiba Jama
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Ammar Almarzooq
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Noelle Benobaid
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Zainab Alqallaf
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Amir A. Ahmed
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Shakir Bahzad
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Mohammad Alkandari
- Histopathology Laboratory, Farwaniya Hospital, Sabah Al Nasser Area 92426, Kuwait;
| |
Collapse
|
3
|
Elgeyoushy F, Qari AS, Al-Refai MA, Taweek TH, Alharbi HH. Huge Intra-abdominal Retroperitoneal Spindle Cell Mass in a Young Adult: A Report of a Rare Case and Literature Review. Cureus 2024; 16:e67822. [PMID: 39328665 PMCID: PMC11424222 DOI: 10.7759/cureus.67822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
GI stromal tumors (GISTs) are the most common mesenchymal neoplasms in the GI tract. GISTs are malignancies that typically originate in the digestive system, most often in the stomach and small intestine. Histopathological classification identifies three types of GISTs: spindle cell, epithelioid, and mixed. We present a case of a huge intra-abdominal retroperitoneal mass in a 23-year-old female with no notable medical or surgical history. She experienced dysphagia and early satiety for one year but did not seek medical attention until presenting at our clinic. Her abdomen was distended, soft, and firm, with a huge non-tender mass. Abdominal CT revealed a substantial left retroperitoneal soft tissue lesion measuring 17 × 12 × 21 cm, causing a significant mass effect. An exploratory laparotomy via a thoracoabdominal approach was performed. A definitive diagnosis required surgical intervention or tissue biopsy. This case highlights the rarity of spindle cell neoplasms and underscores the importance of maintaining a high index of suspicion for such tumors in young adults.
Collapse
|
4
|
Li C, Wang Q, Jiang KW, Ye YJ. Hallmarks and novel insights for gastrointestinal stromal tumors: A bibliometric analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107079. [PMID: 37826966 DOI: 10.1016/j.ejso.2023.107079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Due to the increasing recognition of gastrointestinal stromal tumor (GIST), novel insights have appeared in both preclinical and clinical research and begun to reshape the field. This study aims to map the research landscape through bibliometric analysis and provide a brief overview for the future of the GIST field. METHODS We searched the Web of Science Core Collection without publication data restrictions for GISTs and performed a bibliometric analysis with CiteSpace and VOSviewer software. RESULTS In sum, 5,911 of 13,776 records were included, and these studies were published in 948 journals and written by 24,965 authors from 4,633 institutions in 100 countries. Referring to published reviews and bibliometric analysis, we classified the future trends in four groups. In epidemiological study, precise incidence and clinicopathological features in different regions and races might become potential hotspots. Novel therapy, such as drugs, modified strategies, radioligand therapy, was persistent hotspots in GIST fields, and ctDNA-guided diagnosis, monitoring, and treatment might meet future clinical needs. The debate over serosa surgery vs. mucosa surgery will remain active for a long time in GIST surgery, and function reserve surgery, biology-based surgery will play an important role in future. Moreover, rare GIST type, like NF-1-associated GIST, Carney triads and SDH mutant GIST, need more studies in pathogenesis and genetic mutation to provide appropriate treatment for this orphan GIST patients. CONCLUSIONS Potential hotspots in future GIST trends might involve epidemiology, agents, resection therapy and rare type GIST, moreover, researchers could pay more attention in these four fields.
Collapse
Affiliation(s)
- Chen Li
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China
| | - Quan Wang
- Ambulatory Surgery Center, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China
| | - Ke-Wei Jiang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China.
| | - Ying-Jiang Ye
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
5
|
Arita A, Takahashi T, Nakajima K, Kurokawa Y, Hirota S, Nishida T, Yamashita K, Saito T, Tanaka K, Makino T, Yamasaki M, Kawai K, Motoyama Y, Morii E, Eguchi H, Doki Y. Surgery for multiple gastric gastrointestinal stromal tumors and large esophageal diverticulum related to germline mutation of the KIT gene: a case report. Surg Case Rep 2023; 9:183. [PMID: 37870660 PMCID: PMC10593636 DOI: 10.1186/s40792-023-01766-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Familial gastrointestinal stromal tumors (GISTs) are mesenchymal tumors of the digestive tract caused by germline gain-of-function mutations in the KIT gene or platelet-derived growth factor receptor alpha gene (PDGFRA). These mutations cause not only multiple GISTs but also diffuse hyperplasia of interstitial cells of Cajal (ICCs), which is related to esophageal motility disorder. CASE PRESENTATION A 53-year-old man was referred to our hospital because of anemia and dysphagia. Fifteen years earlier, he had undergone a laparoscopic partial gastrectomy for multiple gastric GISTs with a germline mutation in exon 17 of the KIT gene. An upper gastrointestinal endoscopy revealed that the patient had multiple gastric GISTs and a large esophageal diverticulum directly above the esophagogastric junction. The largest gastric tumor was 7 cm, with a delle that might cause bleeding. Because the patient presented with dysphagia, we performed video-assisted thoracic esophagectomy and laparoscopic-assisted proximal gastrectomy simultaneously. The patient had survived without metastasis for 4 years after surgery and dysphagia had improved. CONCLUSIONS This is the first report of successful laparoscopic-thoracoscopic surgery for a patient with familial gastric GISTs accompanied with a large esophageal diverticulum.
Collapse
Affiliation(s)
- Asami Arita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Seiichi Hirota
- Department of Pathology, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya City, Hyogo 663-8501 Japan
| | - Toshirou Nishida
- Department of Surgery, Japan Community Healthcare Organization, Osaka Hospital, 4-2-78, Fukushima, Fukushima-Ku, Osaka 553-0003 Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Kunihiko Kawai
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Yuichi Motoyama
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita City, Osaka 565-0871 Japan
| |
Collapse
|
6
|
Pardanani A. Systemic mastocytosis in adults: 2023 update on diagnosis, risk stratification and management. Am J Hematol 2023; 98:1097-1116. [PMID: 37309222 DOI: 10.1002/ajh.26962] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/14/2023]
Abstract
OVERVIEW Systemic mastocytosis (SM) results from clonal proliferation of mast cells (MC) in extracutaneous organs. DIAGNOSIS The major criterion is presence of multifocal MC clusters in the bone marrow and/or extracutaneous organs. Minor diagnostic criteria include elevated serum tryptase level, MC CD25/CD2/CD30 expression, and presence of activating KIT mutations. RISK STRATIFICATION Establishing SM subtype as per the International Consensus Classification/World Health Organization classification systems is an important first step. Patients either have indolent/smoldering SM (ISM/SSM) or advanced SM, including aggressive SM (ASM), SM with associated myeloid neoplasm (SM-AMN), and mast cell leukemia. Identification of poor-risk mutations (i.e., ASXL1, RUNX1, SRSF2, NRAS) further refines the risk stratification. Several risk models are available to help assign prognosis in SM patients. MANAGEMENT Treatment goals for ISM patients are primarily directed toward anaphylaxis prevention/symptom control/osteoporosis treatment. Patients with advanced SM frequently need MC cytoreductive therapy to reverse disease-related organ dysfunction. Tyrosine kinase inhibitors (TKI) (midostaurin, avapritinib) have changed the treatment landscape in SM. While deep biochemical, histological and molecular responses have been documented with avapritinib treatment, its efficacy as monotherapy against a multimutated AMN disease component in SM-AMN patients remains unclear. Cladribine continues to have a role for MC debulking, whereas interferon-α has a diminishing role in the TKI era. Treatment of SM-AMN primarily targets the AMN component, particularly if an aggressive disease such as acute leukemia is present. Allogeneic stem cell transplant has a role in such patients. Imatinib has a therapeutic role only in the rare patient with an imatinib-sensitive KIT mutation.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
He W, Xu L, Ding J, Song L, Yang W, Klooster I, Pilco-Janeta DF, Serrano C, Fang H, Jiang G, Wang X, Yu J, Ou WB. Co-targeting of ACK1 and KIT triggers additive anti-proliferative and -migration effects in imatinib-resistant gastrointestinal stromal tumors. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166690. [PMID: 36921738 DOI: 10.1016/j.bbadis.2023.166690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
Most gastrointestinal stromal tumors (GIST) harbor mutated receptor tyrosine kinase (RTK) KIT/PDGFRA, which provides an attractive therapeutic target. However, a majority of GISTs ultimately develop resistance to KIT/PDGFRA inhibitor imatinib, multiple therapeutic targets will be identified as a reasonable strategy in imatinib-resistant GISTs. Biological mechanisms of non-RTK activated CDC42 associated kinase 1 (ACK1) are still unclear, which has been found to be activated in GISTs. In the current report, ACK1 overexpression is demonstrated in GIST cell lines and biopsies. RNA-seq analysis and immunoblotting show that ACK1 expression is dependent on imatinib treatment time in GIST-T1 cell line. The colocalization/complex of KIT and ACK1 in GIST cells are observed, and ACK1 activation is in a partially KIT and CDC42 dependent manner. Treatment with a specific ACK1 inhibitor AIM-100 or ACK1 siRNA, mildly suppresses cell viability, but markedly inhibits cell migration in imatinib sensitive and in imatinib resistant GIST cell lines, which is associated with inactivation of PI3K/AKT/mTOR and RAF/MAPK signaling pathways, and inhibition of epithelial-mesenchymal transition, evidencing upregulation of E-cadherin and downregulation of ZEB1, N-cadherin, vimentin, snail, and/or β-catenin after treatment with AIM-100 or ACK1/CDC42 shRNAs. Combination inhibition of ACK1 and KIT results in additive effects of anti-proliferation and pro-apoptosis as well as cell cycle arrest, and inhibition of invasiveness and migration in vitro and in vivo, compared to either intervention alone through dephosphorylation of KIT downstream intermediates (AKT, S6, and MAPK). Our data suggest that co-targeting of ACK1 and KIT might be a novel therapeutic strategy in imatinib-resistant GIST.
Collapse
Affiliation(s)
- Wangzhen He
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Liangliang Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Jiongyan Ding
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Li Song
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Weili Yang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Isabella Klooster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Daniel F Pilco-Janeta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Sarcoma Translational Research Laboratory, Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | - César Serrano
- Sarcoma Translational Research Laboratory, Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | - Hongming Fang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Guojun Jiang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyan Wang
- Department of Oncology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jiren Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Treggiari E, Giantin M, Ferro S, Romanelli G. Canine gastrointestinal stromal tumours treated with surgery and imatinib mesylate: three cases (2018-2020). J Small Anim Pract 2023; 64:161-167. [PMID: 36335646 DOI: 10.1111/jsap.13572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Gastrointestinal stromal tumours (GISTs) are described in dogs and are histologically diagnosed with the aid of immunohistochemistry to allow differentiation from leiomyomas/leiomyosarcomas. These tumours express c-kit and in some cases could harbour mutations in KIT coding gene. MATERIALS AND METHODS Dogs with a diagnosis of GIST previously confirmed with histopathology and immunohistochemistry were considered for inclusion. Medical records were reviewed for clinical signs at presentation, results of diagnostic tests, tumour location and treatment. To be included, patients had to undergo staging procedures and treatment with imatinib alone or in combination with surgery. Immunohistochemistry and KIT mutational analysis were performed assessing all included cases. RESULTS Three cases were included. All cases underwent staging procedures and surgical excision. Tumours were located in the stomach (two cases) or caecum (one case). KIT mutational status was assessed and the presence of a 54-base pair deletion in exon 11 was identified in one case. Following surgery, imatinib was used to treat recurrent, metastatic or residual disease and resulted in complete response and stable disease in the macroscopic setting and no evidence of recurrence in the microscopic setting. Follow-up time was 890, 120 and 352 days, respectively. CLINICAL SIGNIFICANCE Surgical and medical treatment resulted in a positive outcome in these cases of canine GIST. Imatinib treatment was well tolerated and resulted in a measurable response and a low spectrum of toxicities. Further studies on the tolerability and efficacy of imatinib in solid tumours and GIST are warranted to define its effectiveness and safety.
Collapse
Affiliation(s)
- E Treggiari
- Centro Specialistico Veterinario, Milan, Italy
| | - M Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - S Ferro
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - G Romanelli
- Centro Specialistico Veterinario, Milan, Italy
| |
Collapse
|
9
|
Khosroyani HM, Klug LR, Heinrich MC. TKI Treatment Sequencing in Advanced Gastrointestinal Stromal Tumors. Drugs 2023; 83:55-73. [PMID: 36607590 PMCID: PMC10029090 DOI: 10.1007/s40265-022-01820-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/07/2023]
Abstract
Prior to the early 2000s, patients with advanced gastrointestinal stromal tumors (GIST) had very poor prognoses owing to a lack of effective therapies. The development of tyrosine kinase inhibitors at the turn of the century significantly improved the overall survival for patients with GIST. The resounding success of imatinib in the first clinical trial of a tyrosine kinase inhibitor to treat GIST led to its approval for first-line therapy for advanced GIST; this study was open to all comers and not restricted to any GIST subtype(s). The trials that led to the approvals of second-, third-, and fourth-line therapy for advanced GIST were also open to all patients with advanced/metastatic GIST. Only in retrospect do we realize the role that the molecular subtypes played in the results observed in these studies. In this review, we discuss the studies that led to the US Food and Drug Administration approval of imatinib (first line), sunitinib (second line), regorafenib (third line), and ripretinib (fourth line) for advanced KIT-mutant GIST. In addition, we review how information about GIST molecular subtypes has been used to accelerate the approval of other targeted therapies for non-KIT mutant GIST, leading to the approval of five additional drugs indicated for the treatment of specific GIST molecular subtypes. We also discuss how our understanding of the molecular subtypes will play a role in the next generation of therapeutic approaches for treating advanced GIST.
Collapse
Affiliation(s)
- Homma M Khosroyani
- Portland VA Health Care System and Knight Cancer Institute, Oregon Health & Science University, R&D-19, 3710 SW US Veterans Hospital Road, Portland, OR, 97239, USA
| | - Lillian R Klug
- Portland VA Health Care System and Knight Cancer Institute, Oregon Health & Science University, R&D-19, 3710 SW US Veterans Hospital Road, Portland, OR, 97239, USA
| | - Michael C Heinrich
- Portland VA Health Care System and Knight Cancer Institute, Oregon Health & Science University, R&D-19, 3710 SW US Veterans Hospital Road, Portland, OR, 97239, USA.
| |
Collapse
|
10
|
Sheikh E, Tran T, Vranic S, Levy A, Bonfil RD. Role and significance of c-KIT receptor tyrosine kinase in cancer: A review. Bosn J Basic Med Sci 2022; 22:683-698. [PMID: 35490363 PMCID: PMC9519160 DOI: 10.17305/bjbms.2021.7399] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
c-kit is a classical proto-oncogene that encodes a receptor tyrosine kinase (RTK) that responds to stem cell factor (SCF). C-KIT signaling is a critical regulator of cell proliferation, survival, and migration and is implicated in several physiological processes, including pigmentation, hematopoiesis and gut movement. Accumulating evidence suggests that dysregulated c-KIT function, caused by either overexpression or mutations in c-kit, promotes tumor development and progression in various human cancers. In this review, we discuss the most important structural and biological features of c-KIT, as well as insights into the activation of intracellular signaling pathways following SCF binding to this RTK. We then illustrate how different c-kit alterations are associated with specific human cancers and describe recent studies that highlight the contribution of c-KIT to cancer stemness, epithelial-mesenchymal transition and progression to metastatic disease in different experimental models. The impact of tyrosine kinase inhibitors in treating c-KIT-positive tumors and limitations due to their propensity to develop drug resistance are summarized. Finally, we appraise the potential of novel therapeutic approaches targeting c-KIT more selectively while minimizing toxicity to normal tissue.
Collapse
Affiliation(s)
- Emana Sheikh
- OMS-III, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - Tony Tran
- OMS-III, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - Semir Vranic
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Arkene Levy
- Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - R. Daniel Bonfil
- Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| |
Collapse
|
11
|
A Gastroenterologist's Approach to the Diagnosis and Management of Gastrointestinal Stromal Tumors. Gastroenterol Clin North Am 2022; 51:609-624. [PMID: 36153113 DOI: 10.1016/j.gtc.2022.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal (GI) tract. These tumors have been shown to harbor oncogenic mutations of the c-kit tyrosine kinase receptor or platelet-derived growth factor receptor alpha (PDGFRA). Immunohistochemical analysis of GISTs allows for the differentiation of these tumors from other mesenchymal tumors of the GI tract such as leiomyomas and leiomyosarcomas. All GISTs have the potential to behave in a malignant fashion. Tumor location, size, and mitotic index are factors used to predict the risk of malignant behavior. Endoscopy and endoscopic ultrasound play a critical role in the diagnosis of GISTs and can yield important information to further risk-stratify tumors and determine management. This article provides a gastroenterologist's perspective on the diagnosis and management of GISTs.
Collapse
|
12
|
Foo T, Goldstein D, Segelov E, Shapiro J, Pavlakis N, Desai J, Yip D, Zalcberg J, Price TJ, Nagrial A, Chantrill L, Burge M, Karapetis CS, Tebbutt N, Roy AC. The Management of Unresectable, Advanced Gastrointestinal Stromal Tumours. Target Oncol 2022; 17:95-110. [PMID: 35290591 PMCID: PMC8995292 DOI: 10.1007/s11523-022-00869-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common gastrointestinal tract mesenchymal tumours. Tyrosine kinase inhibitors (TKIs) have transformed the management of advanced GIST. Imatinib was the first TKI to gain approval as management for patients with advanced GIST, establishing a new standard of care. Since then, as a result of several trials including the GRID and INVICTUS studies, we now have five lines of approved targeted therapy, including imatinib, sunitinib, regorafenib, ripretinib and avapritinib for the treatment of unresectable, advanced GISTs. In this review, the Australasian Gastrointestinal Trials Group (AGITG) provide an overview of the key trials that have changed clinical practice, discuss the molecular drivers of GISTs, the importance of molecular testing and directing therapy according to molecular targets, as well as future strategies in the management of advanced GISTs.
Collapse
Affiliation(s)
- Tiffany Foo
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia
| | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, University of NSW, Sydney, NSW, Australia
| | - Eva Segelov
- Department of Medical Oncology, School of Clinical Sciences, Monash University and Monash Health, Melbourne, VIC, Australia
| | - Jeremy Shapiro
- Cabrini Health, Monash University, Melbourne, VIC, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Desmond Yip
- Department of Medical Oncology, Canberra Region Cancer Centre, The Canberra Hospital, Canberra, ACT, Australia
| | - John Zalcberg
- Alfred Health, Monash University, Melbourne, VIC, Australia
| | - Timothy J Price
- The Queen Elizabeth Hospital/University of Adelaide, Adelaide, SA, Australia
| | - Adnan Nagrial
- Department of Medical Oncology, Westmead and Blacktown Hospitals, University of Sydney, Sydney, NSW, Australia
| | - Lorraine Chantrill
- Department of Medical Oncology, Wollongong Hospital, Illawarra Shoalhaven Local Health District, Illawarra, NSW, Australia
| | - Matt Burge
- Department of Cancer Care Services, Royal Brisbane Hospital, University of Queensland, Herston, QLD, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia
| | - Niall Tebbutt
- Department of Medical Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, VIC, Australia
| | - Amitesh C Roy
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
13
|
Johansson G, Berndsen M, Lindskog S, Österlund T, Fagman H, Muth A, Ståhlberg A. Monitoring Circulating Tumor DNA During Surgical Treatment in Patients with Gastrointestinal Stromal Tumors. Mol Cancer Ther 2021; 20:2568-2576. [PMID: 34552011 PMCID: PMC9398151 DOI: 10.1158/1535-7163.mct-21-0403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023]
Abstract
The majority of patients diagnosed with advanced gastrointestinal stromal tumors (GISTs) are successfully treated with a combination of surgery and tyrosine kinase inhibitors (TKIs). However, it remains challenging to monitor treatment efficacy and identify relapse early. Here, we utilized a sequencing strategy based on molecular barcodes and developed a GIST-specific panel to monitor tumor-specific and TKI resistance mutations in cell-free DNA and applied the approach to patients undergoing surgical treatment. Thirty-two patients with GISTs were included, and 161 blood plasma samples were collected and analyzed at routine visits before and after surgery and at the beginning, during, and after surgery. Patients were included regardless of their risk category. Our GIST-specific sequencing approach allowed detection of tumor-specific mutations and TKI resistance mutations with mutant allele frequency < 0.1%. Circulating tumor DNA (ctDNA) was detected in at least one timepoint in nine of 32 patients, ranging from 0.04% to 93% in mutant allele frequency. High-risk patients were more often ctDNA positive than other risk groups (P < 0.05). Patients with detectable ctDNA also displayed higher tumor cell proliferation rates (P < 0.01) and larger tumor sizes (P < 0.01). All patients who were ctDNA positive during surgery became negative after surgery. Finally, in two patients who progressed on TKI treatment, we detected multiple resistance mutations. Our data show that ctDNA may become a clinically useful biomarker in monitoring treatment efficacy in patients with high-risk GISTs and can assist in treatment decision making.
Collapse
Affiliation(s)
- Gustav Johansson
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marta Berndsen
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stefan Lindskog
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, Halland Regional Hospital Varberg, Region Halland, Varberg, Sweden
| | - Tobias Österlund
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Fagman
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andreas Muth
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Corresponding Authors: Anders Ståhlberg, Sahlgrenska Center for Cancer Research, University of Gothenburg, Box 425, Gothenburg 405 30, Sweden. E-mail: ; and Andreas Muth, Department of Surgery, Sahlgrenska University Hospital, Blå stråket 5, 413 45 Gothenburg, Sweden. E-mail:
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden.,Corresponding Authors: Anders Ståhlberg, Sahlgrenska Center for Cancer Research, University of Gothenburg, Box 425, Gothenburg 405 30, Sweden. E-mail: ; and Andreas Muth, Department of Surgery, Sahlgrenska University Hospital, Blå stråket 5, 413 45 Gothenburg, Sweden. E-mail:
| |
Collapse
|
14
|
Dermawan JK, Rubin BP. Molecular Pathogenesis of Gastrointestinal Stromal Tumor: A Paradigm for Personalized Medicine. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:323-344. [PMID: 34736340 DOI: 10.1146/annurev-pathol-042220-021510] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Over the past three to four decades, the molecular pathogenesis of gastrointestinal stromal tumors (GISTs) has been elucidated in great detail. In this review, we discuss the biological genesis of GISTs, identification of the various primary activating driver mutations (focusing on KIT and PDGFRA), oncogene addiction and targeted therapies with imatinib and other tyrosine kinase inhibitors, and the subsequent characterization of the various mechanisms of drug resistance. We illustrate how GIST has become a quintessential paradigm for personalized medicine. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Josephine K Dermawan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA; ,
| | - Brian P Rubin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA; ,
| |
Collapse
|
15
|
Pathania S, Pentikäinen OT, Singh PK. A holistic view on c-Kit in cancer: Structure, signaling, pathophysiology and its inhibitors. Biochim Biophys Acta Rev Cancer 2021; 1876:188631. [PMID: 34606974 DOI: 10.1016/j.bbcan.2021.188631] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022]
Abstract
Receptor tyrosine kinases play an important role in many cellular processes, and their dysregulation leads to diseases, most importantly cancer. One such receptor tyrosine kinase is c-Kit, a type-III receptor tyrosine kinase, which is involved in various intracellular signaling pathways. The role of different mutant isoforms of c-Kit has been established in several types of cancers. Accordingly, promising c-Kit inhibition results have been reported for the treatment of different cancers (e.g., gastrointestinal stromal tumors, melanoma, acute myeloid leukemia, and other tumors). Therefore, lots of effort has been put to target c-Kit for the treatment of cancer. Here, we provide a comprehensive compilation to provide an insight into c-Kit inhibitor discovery. This compilation provides key information regarding the structure, signaling pathways related to c-Kit, and, more importantly, pharmacophores, binding modes, and SAR analysis for almost all small-molecule heterocycles reported for their c-Kit inhibitory activity. This work could be used as a guide in understanding the basic requirements for targeting c-Kit, and how the selectivity and efficacy of the molecules have been achieved till today.
Collapse
Affiliation(s)
- Shelly Pathania
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur G.T. Road, Moga 142001, Punjab, India
| | - Olli T Pentikäinen
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20520 Turku, Finland
| | - Pankaj Kumar Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20520 Turku, Finland.
| |
Collapse
|
16
|
Mutation Status and Immunohistochemical Correlation of EGFR Mutations in Gastrointestinal Stromal Tumors. Balkan J Med Genet 2021; 24:67-72. [PMID: 34447661 PMCID: PMC8366477 DOI: 10.2478/bjmg-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Being one of the leading causes of cancer deaths worldwide and their resistance to conventional treatment methods, made gastrointestinal stromal tumors (GISTs) one of the hot topics in medical research areas in the past decade. To investigate molecular alterations underlying the tumor is of great importance to be able to develop new, targeted treatment options. In this study, GIST samples obtained from 40 Turkish patients were analyzed for actionable epidermal growth factor receptor (EGFR) mutations that are related to treatment regimes in non small cell lung cancer (NSCLC) to understand whether EGFR expression is altered in GISTs. Established alterations in EGFR can make the use of tyrosine kinase inhibitors possible, which are currently used in cancer therapy, especially in NSCLC. Our results indicated that EGFR mutations are rare in GISTs. Further research is needed to sequence whole coding regions of the gene to investigate new actionable mutations in EGFR in an increased sample size.
Collapse
|
17
|
Preseptal cellulitis, intraocular inflammatory reaction and corneal persistent epithelial defect as side effects of avapritinib. Anticancer Drugs 2021; 32:1127-1130. [PMID: 34282744 DOI: 10.1097/cad.0000000000001123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Avapritinib is a tyrosine kinase inhibitor currently being investigated on clinical trials for the treatment of unresectable or metastatic gastrointestinal stromal tumour (GIST). It has been recently approved by the Food and Drug Administration and the European Medicines Agency for the treatment of unresectable or metastatic GIST harbouring PDGFRa Exon 18 mutation and by the European Medicines Agency for the treatments of unresectable or metastatic GIST harbouring the PDGFRa D842V mutation. We report a clinical case of a 76-year-old female, diagnosed with a stage IV GIST, treated with avapritinib 300 mg once daily. through compassionate use who experienced an intraocular side effect not previously reported avapritinib. She developed preseptal cellulitis on her right eye following 2 months of treatment with avapritinib and, subsequently evolved to an intraocular inflammatory reaction and persistent corneal epithelial defect. The treatment with avapritinib was stopped and the patient received corticosteroid and corneal regenerating agents. The symptoms resolved within 1 month and the patient has remained on stable disease at two subsequent adjusted avapritinib doses (100 mg once daily) for over 1 year.
Collapse
|
18
|
Klug LR, Corless CL, Heinrich MC. Inhibition of KIT Tyrosine Kinase Activity: Two Decades After the First Approval. J Clin Oncol 2021; 39:1674-1686. [PMID: 33797935 DOI: 10.1200/jco.20.03245] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lillian R Klug
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| | - Christopher L Corless
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Department of Pathology, Oregon Health & Science University, Portland, OR
| | - Michael C Heinrich
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| |
Collapse
|
19
|
Pardanani A. Systemic mastocytosis in adults: 2021 Update on diagnosis, risk stratification and management. Am J Hematol 2021; 96:508-525. [PMID: 33524167 DOI: 10.1002/ajh.26118] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
OVERVIEW Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in extra-cutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of spindled MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC CD25 expression, and presence of KITD816V mutation. RISK STRATIFICATION Establishing SM subtype as per the World Health Organization classification system is an important first step. Broadly, patients either have indolent/smoldering SM (ISM/SSM) or advanced SM, the latter includes aggressive SM (ASM), SM with associated hematological neoplasm (SM-AHN), and mast cell leukemia (MCL). Identification of poor-risk mutations (ie, ASXL1, RUNX1, SRSF2, NRAS) further refines the risk stratification. Recently, clinical and hybrid clinical-molecular risk models have been developed to more accurately assign prognosis in SM patients. MANAGEMENT Treatment goals for ISM patients are primarily directed towards anaphylaxis prevention/symptom control/osteoporosis treatment. Patients with advanced SM frequently need MC cytoreductive therapy to ameliorate disease-related organ dysfunction. High response rates have been seen with small-molecule inhibitors that target mutant-KIT, including midostaurin (Food and Drug Administration approved) or avapritinib (investigational). Other options for MC cytoreduction include cladribine or interferon-α, although head-to-head comparisons are lacking. Treatment of SM-AHN primarily targets the AHN component, particularly if an aggressive disease such as acute myeloid leukemia is present. Allogeneic stem cell transplant can be considered in such patients, or in those with relapsed/refractory advanced SM. Imatinib has a limited therapeutic role in SM; effective cytoreduction is limited to those with imatinib-sensitive KIT mutations.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota
| |
Collapse
|
20
|
Schaefer IM, Lundberg MZ, Demicco EG, Przybyl J, Matusiak M, Chibon F, Ingram DR, Hornick JL, Wang WL, Bauer S, Baker LH, Lazar AJ, van de Rijn M, Mariño-Enríquez A, Fletcher JA. Relationships between highly recurrent tumor suppressor alterations in 489 leiomyosarcomas. Cancer 2021; 127:2666-2673. [PMID: 33788262 DOI: 10.1002/cncr.33542] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Leiomyosarcoma (LMS) is the most common soft tissue and uterine sarcoma, but no standard therapy is available for recurrent or metastatic LMS. TP53, p16/RB1, and PI3K/mTOR pathway dysregulations are recurrent events, and some LMS express estrogen receptor (ER) and/or progesterone receptor (PR). To characterize relationships between these pathway perturbations, the authors evaluated protein expression in soft tissue and uterine nonprimary leiomyosarcoma (np-LMS), including local recurrences and distant metastases. METHODS TP53, RB1, p16, and PTEN expression aberrations were determined by immunohistochemistry (IHC) in tissue microarrays (TMAs) from 227 np-LMS and a comparison group of 262 primary leiomyosarcomas (p-LMS). Thirty-five of the np-LMS had a matched p-LMS specimen in the TMAs. Correlative studies included differentiation scoring, ER and PR IHC, and CDKN2A/p16 fluorescence in situ hybridization. RESULTS Dysregulation of TP53, p16/RB1, and PTEN was demonstrated in 90%, 95%, and 41% of np-LMS, respectively. PTEN inactivation was more common in soft tissue np-LMS than uterine np-LMS (55% vs 31%; P = .0005). Moderate-strong ER expression was more common in uterine np-LMS than soft tissue np-LMS (50% vs 7%; P < .0001). Co-inactivation of TP53 and RB1 was found in 81% of np-LMS and was common in both soft tissue and uterine np-LMS (90% and 74%, respectively). RB1, p16, and PTEN aberrations were nearly always conserved in p-LMS and np-LMS from the same patients. CONCLUSIONS These studies show that nearly all np-LMS have TP53 and/or RB1 aberrations. Therefore, therapies targeting cell cycle and DNA damage checkpoint vulnerabilities should be prioritized for evaluations in LMS.
Collapse
Affiliation(s)
- Inga-Marie Schaefer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Meijun Z Lundberg
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joanna Przybyl
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Magdalena Matusiak
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Frédéric Chibon
- The Institut national de la santé et de la recherche médicale (INSERM) U1037, Cancer Research Center of Toulouse, Department of Pathology, Claudius Régaud Institute, IUCT-Oncopole, Toulouse, France
| | - Davis R Ingram
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Duisburg-Essen Medical School, Essen, Germany.,Partner Site Essen and German Cancer Consortium, Heidelberg, Germany
| | - Laurence H Baker
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Adrian Mariño-Enríquez
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Liu Y, Fan Y, Wu S. Developments in research on interstitial Cajal-like cells in the biliary tract. Expert Rev Gastroenterol Hepatol 2021; 15:159-164. [PMID: 32933347 DOI: 10.1080/17474124.2021.1823214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/10/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Interstitial cells of Cajal (ICCs) are a special type of interstitial cells located in the gastrointestinal tract muscles. They are closely related to smooth muscle cells and neurons, participate in gastrointestinal motility and nerve signal transmission, and are pacemaker cells for gastrointestinal electrical activity. Research interest in ICCs has continuously grown since they were first discovered in 1893. Later, researchers discovered that they are also present in other organs, including the biliary tract, urethra, bladder, etc.; these cells were named interstitial Cajal-like cells (ICLCs), and attempts have been made to explain their relationships with certain diseases. AREAS COVERED This review paper summarizes the morphology, identification, classification, function, and distribution of ICLCs in the biliary tract and their relationship to biliary tract diseases. EXPERT OPINION Based on the function and distribution of ICLCs in the biliary tract system, ICLCs will provide a more reliable theoretical basis for the mechanisms of pathogenesis of and treatments for biliary tract diseases.
Collapse
Affiliation(s)
- Yingyu Liu
- Department of General Surgery, Shengjing Hospital of China Medical University , Shenyang, China
| | - Ying Fan
- Department of General Surgery, Shengjing Hospital of China Medical University , Shenyang, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University , Shenyang, China
| |
Collapse
|
22
|
Immunohistochemistry in Bone and Soft Tissue Tumours. Sarcoma 2021. [DOI: 10.1007/978-981-15-9414-4_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
23
|
Papke DJ, Hornick JL. Recent developments in gastroesophageal mesenchymal tumours. Histopathology 2020; 78:171-186. [PMID: 33382494 DOI: 10.1111/his.14164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022]
Abstract
The pathologist's approach to gastroesophageal mesenchymal tumours has changed dramatically during the last 25 years. In particular, gastrointestinal stromal tumour (GIST) has evolved from a wastebasket mesenchymal tumour category to a precisely defined entity with an increasingly detailed genetic subclassification. This subclassification has brought gastrointestinal mesenchymal neoplasia into the realm of precision medicine, with specific treatments optimised for particular genetic subtypes. Molecular genetic data have also greatly improved our understanding of oesophageal mesenchymal tumours, including the discovery that so-called 'giant fibrovascular polyps' in fact represent a clinically distinctive presentation of well-differentiated liposarcoma. Here, we will focus on gastroesophageal mesenchymal tumours for which there have been recent developments in classification, molecular genetics or tumour biology: granular cell tumour, 'giant fibrovascular polyp'/well-differentiated liposarcoma, plexiform fibromyxoma, gastroblastoma and, of course, GIST.
Collapse
Affiliation(s)
- David J Papke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Arshad J, Roberts A, Ahmed J, Cotta J, Pico BA, Kwon D, Trent JC. Utility of Circulating Tumor DNA in the Management of Patients With GI Stromal Tumor: Analysis of 243 Patients. JCO Precis Oncol 2020; 4:66-73. [DOI: 10.1200/po.19.00253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE GI stromal tumor (GIST) is the most common sarcoma of the GI tract. Management of patients with GIST is determined by KIT, PDGFRA, or other genomic alterations. Tissue-based next-generation sequencing (NGS) analysis is the standard approach for diagnosis, prognosis, and treatment selection. However, circulating tumor DNA (ctDNA)–based NGS is a novel and noninvasive alternative. METHODS ctDNA sequencing results were evaluated in blood samples from 243 de-identified patients within the Guardant360 database. Under an approved institutional review board protocol, a retrospective analysis was performed on 45 single-institution patients. RESULTS Of 243 patients, 114 (47%) were women, and the median age was 59 years (range, 17-90 years). Patients with no alterations and variations of uncertain significance were excluded. Of the 162 patients with known pathogenic mutations, KIT was the most common (56%), followed by NF (7%), PDGFRA (6%), PI3KCA (6%), KRAS (5%), and others (6%). Most tumors harbored an actionable KIT or PDGFRA mutation. Our institutional cohort (n = 45) had 16 (35%) KIT exon 11 mutations, 3 (6%) KIT exon 9 mutations, and 1 (2%) PDGFRA mutation detected on ctDNA. Resistance mutations were observed in KIT exon 17 (8 patients), exon 13 (3 patients), and in both (3 patients). Our comparison of ctDNA with tissue NGS revealed a positive predictive value (PPV) of 100%. Failure of concordance was observed in patients with localized or low disease burden. From the time of ctDNA testing, the median overall survival was not reached, whereas the median progression-free survival was 7 months. CONCLUSION ctDNA provides a rapid, noninvasive analysis of current mutations with a high PPV for patients with metastatic GIST. ctDNA-based testing may help to define the optimal choice of therapy on the basis of resistance mutations and should be studied prospectively.
Collapse
Affiliation(s)
- Junaid Arshad
- University of Miami, Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL
| | | | | | - Jared Cotta
- University of Miami, Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL
| | - Brian A. Pico
- University of Miami, Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL
| | - Deukoo Kwon
- University of Miami, Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL
| | - Jonathan C. Trent
- University of Miami, Miller School of Medicine/Sylvester Comprehensive Cancer Center, Miami, FL
| |
Collapse
|
25
|
Astolfi A, Indio V, Nannini M, Saponara M, Schipani A, De Leo A, Altimari A, Vincenzi B, Comandini D, Grignani G, Secchiero P, Urbini M, Pantaleo MA. Targeted Deep Sequencing Uncovers Cryptic KIT Mutations in KIT/PDGFRA/SDH/RAS-P Wild-Type GIST. Front Oncol 2020; 10:504. [PMID: 32391261 PMCID: PMC7188756 DOI: 10.3389/fonc.2020.00504] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Gastrointestinal stromal tumors (GIST) are known to carry oncogenic KIT or PDGFRA mutations, or less commonly SDH or NF1 gene inactivation, with very rare cases harboring mutant BRAF or RAS alleles. Approximately 10% of GISTs are devoid of any of such mutations and are characterized by very limited therapeutic opportunities and poor response to standard treatments. Methods: Twenty-six sporadic KIT/PDGFRA/SDH/RAS-pathway wild type GIST were profiled for the molecular status of genes frequently altered in GIST by a targeted next generation sequencing (NGS) approach. Molecular findings were validated by alternative amplicon-based targeted sequencing, immunohistochemistry, gene expression profiling and Sanger sequencing. Results: Three patients harboring NF1 inactivating mutations were identified and excluded from further analysis. Intriguingly, five patients carried cryptic KIT alterations, mainly represented by low-allele-fraction mutations (12–16% allele ratio). These mutations were confirmed by another targeted NGS approaches and supported by CD117 immuno-staining, gene expression profiling, Sanger sequencing, with peak signals at the level of background noise, and by the patients' clinical course assessment. Conclusion: This study indicates that ~20% patients diagnosed with a KIT/PDGFRA/SDH/RAS-pathway wild-type GIST are bona-fide carriers of pathogenic KIT mutations, thus expected to be eligible for and responsive to the various therapeutic lines of TK-inhibitors in use for KIT/PDGFRA-mutant GIST. The centralization for a second level molecular analysis of GIST samples diagnosed as wild-type for KIT and PDGFRA is once again strongly recommended.
Collapse
Affiliation(s)
- Annalisa Astolfi
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Valentina Indio
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Margherita Nannini
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.,Medical Oncology Unit, S.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Maristella Saponara
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Angela Schipani
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Antonio De Leo
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Annalisa Altimari
- Laboratory of Oncologic Molecular Pathology, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - Danila Comandini
- Medical Oncology 1, Policlinico San Martino, University of Genova, Genova, Italy
| | - Giovanni Grignani
- Sarcoma Unit, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Milena Urbini
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Maria Abbondanza Pantaleo
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Madhala D, Sundaram S, Chinambedudandapani M, Balasubramanian A. Analysis of C-Kit Exon 9, Exon 11 and BRAFV600E Mutations Using Sangers Sequencing in Gastrointestinal Stromal Tumours. Cureus 2020; 12:e7369. [PMID: 32328381 PMCID: PMC7174865 DOI: 10.7759/cureus.7369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Gastrointestinal stromal tumors (GIST) are the most common mesenchymal neoplasms in the gastrointestinal (GI) tract. The mutation of C-KIT is considered to be the crucial step in the tumorigenesis. Targeted therapies are being developed focusing these mutations. Various exon mutations of GIST responded in varied patterns to this targeted therapy. This study was carried out to evaluate the C-KIT exon 11, exon 9 and BRAF V600E mutations among GIST specimens. Methods This retrospective study was carried out among 20 DNA extracted specimens from paraffin blocks of GIST received in our tertiary teaching institution for a period of three years. DNA sequencing was carried out for mutational analyses on C-KIT exon 9, C-KIT exon 11 and BRAF V600E genes using Sanger sequencing. Results Histologically, majority of the tumors had spindle cell morphology. About 19 cases were positive for CD117. The analysis of type of mutations showed that three cases carried Exon 11 and three cases carried Exon 9 mutations. BRAF V600E mutation was seen in one case. Conclusion It is essential to conduct molecular studies on GISTs in order to get a clear understanding of the pathogenesis and behavior pattern. This will also help in designing targeted therapies and assessing recurrence. With the advent of rapidly evolving personalized therapy, the evaluation of genetic mutations is essential for diagnosis and prognostic value.
Collapse
Affiliation(s)
- Divya Madhala
- Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Sandhya Sundaram
- Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | | | | |
Collapse
|
27
|
Coordinated targeting of CK2 and KIT in gastrointestinal stromal tumours. Br J Cancer 2019; 122:372-381. [PMID: 31776458 PMCID: PMC7000686 DOI: 10.1038/s41416-019-0657-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 09/26/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Background Most gastrointestinal stromal tumours (GIST) are driven by activating oncogenic mutations of KIT/PDGFRA, which provide a compelling therapeutic target. Our previous studies showed that CDC37, regulated by casein kinase 2 (CK2), is a crucial HSP90 cofactor for KIT oncogenic function and a promising and more selective therapeutic target in GIST. Methods Biologic mechanisms of CK2-mediated CDC37 regulation were assessed in GISTs by immunoblotting, immunoprecipitations, knockdown and inactivation assays. The effects of a combination of KIT and CK2 inhibition were assessed by immunoblotting, cell viability, colony growth, cell cycle analysis, apoptosis, migration and invasiveness. Results CK2 overexpression was demonstrated by immunoblotting in GIST cell lines and patient biopsies. Treatment with a specific CK2 inhibitor, CX4945, leads to CDC37 dephosphorylation and inhibits KIT signalling in imatinib-sensitive and in imatinib-resistant GIST cell lines. Immunoprecipitation demonstrated that CK2 inhibition blocks KIT:HSP90:CDC37 interaction in GIST cells. Coordinated inhibition of CK2 and KIT by CX4945 (or CK2 shRNA) and imatinib, respectively, leads to increased apoptosis, anti-proliferative effects and cell cycle arrest and decreased p-AKT and p-S6 expression, migration and invasiveness in all GIST cell lines compared with either intervention alone, indicating additive effects of inhibiting these two important regulators of GIST biology. Conclusion Our findings suggest that combinatorial inhibition of CK2 and KIT warrants evaluation as a novel therapeutic strategy in GIST, especially in imatinib-resistant GIST.
Collapse
|
28
|
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 Haematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University Medical Centre Freiburg, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| |
Collapse
|
29
|
Wu CE, Tzen CY, Wang SY, Yeh CN. Clinical Diagnosis of Gastrointestinal Stromal Tumor (GIST): From the Molecular Genetic Point of View. Cancers (Basel) 2019; 11:cancers11050679. [PMID: 31100836 PMCID: PMC6563074 DOI: 10.3390/cancers11050679] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) originating from the interstitial cells of Cajal are mesenchymal tumors of the gastrointestinal tract and have been found to harbor c-KIT mutations and KIT (CD117) expression since 1998. Later, PDGFRA mutations, SDH alterations, and other drive mutations were identified in GISTs. In addition, more and more protein markers such as DOG1, PKCθ were found to be expressed in GISTs which might help clinicians diagnose CD117-negative GISTs. Therefore, we plan to comprehensively review the molecular markers and genetics of GISTs and provide clinicians useful information in diagnostic and therapeutic strategies of GISTs. Twenty years after the discovery of KIT in GISTs, the diagnosis of GISTs became much more accurate by using immunohistochemical (IHC) panel (CD117/DOG1) and molecular analysis (KIT/PDGFRA), both of which constitute the gold standard of diagnosis in GISTs. The accurately molecular diagnosis of GISTs guides clinicians to precision medicine and provides optimal treatment for the patients with GISTs. Successful treatment in GISTs prolongs the survival of GIST patients and causes GISTs to become a chronic disease. In the future, the development of effective treatment for GISTs resistant to imatinib/sunitinib/regorafenib and KIT/PDGFRA-WT GISTs will be the challenge for GISTs.
Collapse
Affiliation(s)
- Chiao-En Wu
- GIST Team, Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, Chang Gung University, Taoyuan 333, Taiwan.
| | - Chin-Yuan Tzen
- Forlab Clinic, F2, No 14, Sec 2, Zhongxiao East Rd, Taipei 100, Taiwan.
| | - Shang-Yu Wang
- GIST Team, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
| | - Chun-Nan Yeh
- GIST Team, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
30
|
Chen T, Ye LY, Feng XY, Qiu HB, Zhang P, Luo YX, Yuan LY, Chen XH, Hu YF, Liu H, Li Y, Tao KX, Yu J, Li GX. Performance of risk stratification systems for gastrointestinal stromal tumors: A multicenter study. World J Gastroenterol 2019; 25:1238-1247. [PMID: 30886506 PMCID: PMC6421238 DOI: 10.3748/wjg.v25.i10.1238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumor type in the gastrointestinal system. Presently, various classification systems to prognosticate GISTs have been proposed. AIM To evaluate the application value of four different risk stratification systems for GISTs. METHODS Patients who were diagnosed with GISTs and underwent surgical resection at four hospitals from 1998 to 2015 were identified from a database. Risk of recurrence was stratified by the modified National Institute of Health (NIH) criteria, the Armed Forces Institute of Pathology (AFIP) criteria, the Memorial Sloan Kettering Cancer Center (MSKCC) prognostic nomogram, and the contour maps. Receiver operating characteristic (ROC) curves were established to compare the four abovementioned risk stratification systems based on the area under the curve (AUC). RESULTS A total of 1303 patients were included in the study. The mean age of the patients was 55.77 ± 13.70 yr; 52.3% of the patients were male. The mean follow-up period was 64.91 ± 35.79 mo. Approximately 67.0% the tumors were located in the stomach, and 59.5% were smaller than 5 cm; 67.3% of the patients had a mitotic count ≤ 5/50 high-power fields (HPFs). Thirty-four tumors ruptured before and during surgery. Univariate analysis demonstrated that tumor size > 5 cm (P < 0.05), mitotic count > 5/50 HPFs (P < 0.05), non-gastric location (P < 0.05), and tumor rupture (P < 0.05) were significantly associated with increased recurrence rates. According to the ROC curve, the AFIP criteria showed the largest AUC (0.754). CONCLUSION According to our data, the AFIP criteria were associated with a larger AUC than the NIH modified criteria, the MSKCC nomogram, and the contour maps, which might indicate that the AFIP criteria have better accuracy to support therapeutic decision-making for patients with GISTs.
Collapse
Affiliation(s)
- Tao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Liang-Ying Ye
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xing-Yu Feng
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong Province, China
| | - Hai-Bo Qiu
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Peng Zhang
- Department of General Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yi-Xin Luo
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Yi Yuan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xin-Hua Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yan-Feng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yong Li
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong Province, China
| | - Kai-Xiong Tao
- Department of General Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Guo-Xin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
31
|
Lu T, Chen C, Wang A, Jiang Z, Qi Z, Hu Z, Hu C, Liu F, Wang W, Wu H, Wang B, Wang L, Qi S, Wu J, Wang W, Tang J, Yan H, Bai M, Liu Q, Liu J. Repurposing cabozantinib to GISTs: Overcoming multiple imatinib-resistant cKIT mutations including gatekeeper and activation loop mutants in GISTs preclinical models. Cancer Lett 2019; 447:105-114. [PMID: 30684595 DOI: 10.1016/j.canlet.2019.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/05/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022]
Abstract
Despite of the great success of imatinib as the first-line treatment for GISTs, the majority of patients will develop drug-acquired resistance due to secondary mutations in the cKIT kinase. Sunitinib and regorafenib have been approved as the second and third line therapies to overcome some of these drug-resistance mutations; however, their limited clinical response, toxicity and resistance of the activation loop mutants still makes new therapies bearing different cKIT mutants activity spectrum profile highly demanded. Through a drug repositioning approach, we found that cabozantinib exhibited higher potency than imatinib against primary gain-of-function mutations of cKIT. Moreover, cabozantinib was able to overcome cKIT gatekeeper T670I mutation and the activation loop mutations that are resistant to imatinib or sunitinib. Cabozantinib demonstrated good efficacy in vitro and in vivo in the cKIT mutant-driven preclinical models of GISTs while displaying a long-lasting effect after treatment withdrawal. Furthermore, it also exhibited dose-dependent anti-proliferative efficacy in the GIST patient derived primary cells. Considering clinical safety and PK profile of cabozantinib, this report provides the basis for the future clinical applications of cabozantinib as an alternative anti-GISTs therapy in precision medicine.
Collapse
Affiliation(s)
- Tingting Lu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China; Anhui University of Chinese Medicine, Hefei, Anhui, 230012, PR China
| | - Cheng Chen
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Aoli Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Zongru Jiang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Ziping Qi
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Zhenquan Hu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Chen Hu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Feiyang Liu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Wenliang Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Hong Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Beilei Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Li Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Shuang Qi
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Jiaxin Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Wenchao Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Jun Tang
- The People's Liberation Army Joint Service Support Force No. 901 Hospital, Hefei, Anhui, 230031, PR China
| | - Hezhong Yan
- The People's Liberation Army Joint Service Support Force No. 901 Hospital, Hefei, Anhui, 230031, PR China
| | - Mingfeng Bai
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15219, United States
| | - Qingsong Liu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China; University of Science and Technology of China, Hefei, Anhui, 230026, PR China; Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, PR China.
| | - Jing Liu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.
| |
Collapse
|
32
|
Iwatsuki M, Harada K, Iwagami S, Eto K, Ishimoto T, Baba Y, Yoshida N, Ajani JA, Baba H. Neoadjuvant and adjuvant therapy for gastrointestinal stromal tumors. Ann Gastroenterol Surg 2019; 3:43-49. [PMID: 30697609 PMCID: PMC6345649 DOI: 10.1002/ags3.12211] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/05/2018] [Accepted: 08/23/2018] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) are rare and mesenchymal in origin with a yearly incidence of 10-15 cases per million people. If it is technically resectable, surgical resection is the mainstay of therapy regardless of tumor location,. Although complete (R0) resection can be achieved in up to 85% of patients with primary disease, approximately 50% of patients experience recurrence or metastases within 5 years of primary resection. Moreover, prior to 2000, the prognosis of patients with advanced, inoperable GIST was poor because the molecular mechanism had not sufficiently been elucidated, thus effective therapy was lacking. The tyrosine kinase inhibitor imatinib, which selectively inhibits tyrosine kinase KIT, has shown substantial clinical benefit for patients with GIST. In clinical trials, imatinib treatment resulted in response rates of 40%-55% and longer progression-free survival for patients with a KIT-positive unresectable or metastatic GIST. Furthermore, recent clinical trials have shown that giving imatinib after curative resection for high-risk cases prolonged recurrence-free survival and overall survival in an adjuvant setting. Several clinical trials of imatinib treatment in a neoadjuvant setting are ongoing; however, in clinical settings, there are problems to resolve, such as optimal agents, duration of administration, and postoperative management. In this review, we discuss the application of surgical options, combined with adjuvant/neoadjuvant or perioperative imatinib treatment and their potential impact on survival for patients with primary, recurrent, or metastatic GIST.
Collapse
Affiliation(s)
- Masaaki Iwatsuki
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Gastrointestinal Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonUSA
| | - Kazuto Harada
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Gastrointestinal Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonUSA
| | - Shiro Iwagami
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kojiro Eto
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Takatsugu Ishimoto
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshifumi Baba
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Naoya Yoshida
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonUSA
| | - Hideo Baba
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
33
|
Chen T, Xu L, Ye L, Qiu H, Hu Y, Liu H, Zhou Z, Li G, Yu J. A new nomogram for recurrence-free survival prediction of gastrointestinal stromal tumors: Comparison with current risk classification methods. Eur J Surg Oncol 2018; 45:1109-1114. [PMID: 30594406 DOI: 10.1016/j.ejso.2018.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/08/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study aimed to build a new risk stratification nomogram for gastrointestinal stromal tumors (GISTs) focused on a popular factor Ki-67 to enable individualized and precise predictions of the most suitable candidates for imatinib therapy. METHODS We retrospectively collected clinicopathologic data of the patients diagnosed with GISTs from January 1998 to December 2015 at Southern Medical University Nanfang Hospital as the experiment group. And patients with GISTs at the Sun Yat-sen University Cancer Center from January 2007 to December 2012 were included as the validation group. The nomogram was built using Kaplan-Meier method and the Cox proportional hazards regression model. The receiver operating characteristic (ROC) curves were established to compare the discriminative ability of the new nomogram with other risk stratification systems, including the modified National Institute of Health (modified NIH) criteria, Armed Forces Institute of Pathology (AFIP) criteria, Memorial Sloan Kettering Cancer Center (MSKCC) prognostic nomogram, and contour maps. RESULTS In univariate analysis, the tumor size, site, mitotic count, tumor rupture and Ki-67 labeling index were significant factors (all P < 0.05) and included in the Cox model to build our nomogram. According to the ROC curve, our new nomogram showed the largest AUC value (0.778) compared with that of the other classification methods (contour maps, AUC = 0.743; AFIP, AUC = 0.719; MSKCC, AUC = 0.712; and modified NIH, AUC = 0.719). CONCLUSION Our new nomogram exhibits an excellent performance and might become a potential risk stratification to support therapeutic decision-making for GISTs.
Collapse
Affiliation(s)
- Tao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China.
| | - Lili Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China
| | - Liangying Ye
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China
| | - Haibo Qiu
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong Province, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong Province, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
34
|
Tang J, Zhao R, Zheng X, Xu L, Wang Y, Feng L, Ren S, Wang P, Zhang M, Xu M. Using the recurrence risk score by Joensuu to assess patients with gastrointestinal stromal tumor treated with adjuvant imatinib: A retrospective cohort study. Medicine (Baltimore) 2018; 97:e11400. [PMID: 30024511 PMCID: PMC6086542 DOI: 10.1097/md.0000000000011400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In 2014, Joensuu and colleagues devised the first recurrence risk score (RRS) to identify the risk factors for gastrointestinal stromal tumor (GIST) recurrence. However, there are scarce data available on RRS effectiveness and efficiency. Therefore, we retrospectively analyzed clinical data to validate Joensuu's RRS in patients treated with adjuvant imatinib.In this retrospective cohort study, data were collected from patients with GIST who were treated with adjuvant imatinib between December 2005 and May 2017 in the West China Hospital. The study consisted of 137 patients, after application of inclusion and exclusion criteria. Recurrence-free survival (RFS) was the primary end point.The RRSs for 137 patients were divided into 3 groups: low (n = 46), medium (n = 48), and high (n = 43). The RFSs of the 3 groups were significantly different (P < .001). In patients who received adjuvant imatinib for <36 months, the RFS difference was also significant (P < .001), and the result was similar in patients treated with adjuvant imatinib for ≥36 months (P = .03). The area under the curve of the RRS was 0.84 ([95% confidence interval] 0.76-0.92, P < .001), suggesting that the RRS method could accurately assess recurrence risks for patients with GIST who were treated with adjuvant imatinib.It is appropriate to apply the RRS method to assess recurrence risks for patients with GIST who were treated with adjuvant imatinib. A longer adjuvant imatinib duration is recommended for high-risk patients with GIST. It is also important to identify a more effective treatment for patients who are resistant to imatinib.
Collapse
Affiliation(s)
| | - Rui Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | - Yong Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | | | | | |
Collapse
|
35
|
Thacoor A. Gastrointestinal stromal tumours: advances in surgical and pharmacological management options. J Gastrointest Oncol 2018; 9:573-578. [PMID: 29998023 DOI: 10.21037/jgo.2018.01.10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal neoplasm of the gastrointestinal tract. The stomach is most commonly affected. Advances in immunopathology have identified a mutation in the c-KIT proto-oncogene, leading to the development of the tyrosine-kinase inhibitor Imatinib as targeted therapy for advanced disease. Surgery, however, remains the only curative treatment of resectable primary GIST. This review article aims at reviewing the management of primary GISTs, investigating recurrences, exploring the role of surgery in metastatic disease and looking into combined surgery and molecular therapy for GISTs.
Collapse
|
36
|
Kondi-Pafiti A, Arkadopoulos N, Gennatas C, Michalaki V, Frangou-Plegmenou M, Chatzipantelis P. Expression of c-kit in Common Benign and Malignant Breast Lesions. TUMORI JOURNAL 2018. [DOI: 10.1177/548.6519] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Aims and background c-kit (CD117) is a transmembrane tyrosine kinase that acts as a type III receptor for mast cell growth factor. In recent years, the role of c-kit in the development of preinvasive and invasive breast carcinomas has been investigated. The aim of our study was to detect c-kit expression in the entire spectrum of common benign and malignant breast lesions in correlation with a well-studied myoepithelial or stem-cell like marker (p63). Methods and study design We evaluated 270 cases of benign and malignant breast lesions including fibrocystic disease, fibroadenoma, sclerosing adenosis, atypical ductal hyperplasia, ductal/lobular carcinoma in situ, and ductal/lobular/mixed type carcinoma. C-kit staining was evaluated in the cytoplasm/cell membrane in epithelial and myoepithelial cells and p63 in the nuclei of myoepithelial cells. Results c-kit was highly expressed (85.3%) in benign lesions (fibrocystic disease, sclerosing adenosis, fibroadenoma), and p63 expression was 95.5% in the aforementioned lesions. c-kit distribution in preinvasive and invasive lesions was as follows: ductal/lobular carcinoma in-situ, 43%/35%; ductal/lobular carcinoma, 36%/39%; and mixed type carcinoma, 20%. c-kit was highly expressed in myofibroblast/fibroblast cells only in grade III ductal/lobular carcinomas. c-kit was totally absent in stromal cells in benign lesions and in situ carcinomas whereas expression was weak in grade I and II carcinomas. Conclusions Combined overexpression of c-kit and p63 is indicative of benign breast lesions. In contrast, there is reduced expression of c-kit in in situ and invasive breast carcinomas, with simultaneous overexpression in the stromal cells. This suggests that c-kit may play a role in breast cancer progression. Free full text available at www.tumorionline.it
Collapse
Affiliation(s)
- Agatha Kondi-Pafiti
- Department of Pathology, University of Athens Medical School, Athens, Greece
| | | | - Constantinos Gennatas
- Oncology Clinic, Aretaieion Hospital, University of Athens Medical School, Athens, Greece
| | - Vassiliki Michalaki
- Oncology Clinic, Aretaieion Hospital, University of Athens Medical School, Athens, Greece
| | | | | |
Collapse
|
37
|
Niinuma T, Suzuki H, Sugai T. Molecular characterization and pathogenesis of gastrointestinal stromal tumor. Transl Gastroenterol Hepatol 2018; 3:2. [PMID: 29441367 DOI: 10.21037/tgh.2018.01.02] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/04/2018] [Indexed: 12/11/2022] Open
Abstract
Most gastrointestinal stromal tumors (GISTs) harbor activating mutations in the receptor tyrosine kinase gene KIT or platelet-derived growth factor receptor alpha (PDGFRA), and the resultant activation of downstream signals plays a pivotal role in the development of GISTs. The sites of the tyrosine kinase gene mutations are associated with the biological behavior of GISTs, including risk category, clinical outcome and drug response. Mutations in RAS signaling pathway genes, including KRAS and BRAF, have also been reported in KIT/PDGFRA wild-type GISTs, though they are rare. Neurofibromin 1 (NF1) is a tumor suppressor gene mutated in neurofibromatosis type 1. Patients with NF1 mutations are at high risk of developing GISTs. Recent findings suggest that altered expression or mutation of members of succinate dehydrogenase (SDH) heterotetramer are causally associated with GIST development through induction of aberrant DNA methylation. At present, GISTs with no alterations in KIT, PDGFRA, RAS signaling genes or SDH family genes are referred to as true wild-type GISTs. KIT and PDGFRA mutations are thought as the earliest events in GIST development, and subsequent accumulation of chromosomal aberrations and other molecular alterations are required for malignant progression. In addition, recent studies have shown that epigenetic alterations and noncoding RNAs also play key roles in the pathogenesis of GISTs.
Collapse
Affiliation(s)
- Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, Morioka, Japan
| |
Collapse
|
38
|
Mei L, Smith SC, Faber AC, Trent J, Grossman SR, Stratakis CA, Boikos SA. Gastrointestinal Stromal Tumors: The GIST of Precision Medicine. Trends Cancer 2017; 4:74-91. [PMID: 29413424 DOI: 10.1016/j.trecan.2017.11.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/06/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
Abstract
The discovery of activated KIT mutations in gastrointestinal (GI) stromal tumors (GISTs) in 1998 triggered a sea change in our understanding of these tumors and has ushered in a new paradigm for the use of molecular genetic diagnostics to guide targeted therapies. KIT and PDGFRA mutations account for 85-90% of GISTs; subsequent genetic studies have led to the identification of mutation/epimutation of additional genes, including the succinate dehydrogenase (SDH) subunit A, B, C, and D genes. This review focuses on integrating findings from clinicopathologic, genetic, and epigenetic studies, which classify GISTs into two distinct clusters: an SDH-competent group and an SDH-deficient group. This development is important since it revolutionizes our current management of affected patients and their relatives, fundamentally, based on the GIST genotype.
Collapse
Affiliation(s)
- Lin Mei
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Steven C Smith
- Departments of Pathology and Surgery, VCU School of Medicine, Richmond, VA, USA
| | - Anthony C Faber
- VCU Phillips Institute for Oral Health Research, School of Dentistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Steven R Grossman
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Constantine A Stratakis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Rockville, MD, USA
| | - Sosipatros A Boikos
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
39
|
Kasireddy V, von Mehren M. Emerging drugs for the treatment of gastrointestinal stromal tumour. Expert Opin Emerg Drugs 2017; 22:317-329. [DOI: 10.1080/14728214.2017.1411479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Vineela Kasireddy
- Fellow (PGY5), Department of Hematology Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Margaret von Mehren
- Director of Sarcoma Oncology, Associate Director for Clinical Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
40
|
Adjuvant imatinib for patients with high-risk gastrointestinal stromal tumors: a retrospective cohort study. Sci Rep 2017; 7:16834. [PMID: 29203825 PMCID: PMC5715066 DOI: 10.1038/s41598-017-17266-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/23/2017] [Indexed: 02/07/2023] Open
Abstract
The duration of adjuvant imatinib for high-risk patients with gastrointestinal stromal tumors (GISTs) is still controversial. Therefore, we retrospectively analyzed the data of high-risk patients with GISTs to investigate the appropriate duration. All 185 patients were divided into 4 groups: <1 year (Group A), 1–2 years (Group B), 2–3 years (Group C) and >3 years (Group D). The mean recurrence-free survival (RFS) in Groups A, B, and C were 44.3, 62.1, and 86.8 months, respectively (P < 0.001); the mean overall survival (OS) in Groups A, B and C was 75.2, 88.1, and 94.7 months, respectively (P = 0.009). The 5-year RFS in Groups A, B, C, and D was 15%, 26%, 83%, and 100%, respectively (P < 0.001); and the 5-year OS was 64%, 88%, 88%, and 100%, respectively (P < 0.001). The greatest impact on unfavorable outcomes was the tumor mitotic rate (HR, 2.01, 95% CI, 1.38–2.94; P < 0.001). Duration of adjuvant imatinib was the only favorable factor (HR, −0.95, 95% CI, 0.93–0.97; P < 0.001). For high-risk patients with high tumor size or mitotic rate, or non-gastric GISTs, we recommend that more than 3 years of adjuvant imatinib is feasible.
Collapse
|
41
|
Kim BJ, Kays JK, Koniaris LG, Valsangkar NP. Understanding the critical role for surgery in the management of wild-type gastrointestinal stromal tumor (GIST). Transl Gastroenterol Hepatol 2017; 2:91. [PMID: 29264429 DOI: 10.21037/tgh.2017.11.03] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/06/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Bradford J Kim
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua K Kays
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leonidas G Koniaris
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nakul P Valsangkar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
42
|
Bleloch JS, Ballim RD, Kimani S, Parkes J, Panieri E, Willmer T, Prince S. Managing sarcoma: where have we come from and where are we going? Ther Adv Med Oncol 2017; 9:637-659. [PMID: 28974986 PMCID: PMC5613860 DOI: 10.1177/1758834017728927] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are a heterogeneous group of neoplasms of mesenchymal origin. Approximately 80% arise from soft tissue and 20% originate from bone. To date more than 100 sarcoma subtypes have been identified and they vary in molecular characteristics, pathology, clinical presentation and response to treatment. While sarcomas represent <1% of adult cancers, they account for approximately 21% of paediatric malignancies and thus pose some of the greatest risks of mortality and morbidity in children and young adults. Metastases occur in one-third of all patients and approximately 10-20% of sarcomas recur locally. Surgery in combination with preoperative and postoperative therapies is the primary treatment for localized sarcoma tumours and is the most promising curative possibility. Metastasized sarcomas, on the other hand, are treated primarily with single-agent or combination chemotherapy, but this rarely leads to a complete and robust response and often becomes a palliative form of treatment. The heterogeneity of sarcomas results in variable responses to current generalized treatment strategies. In light of this and the lack of curative strategies for metastatic and unresectable sarcomas, there is a need for novel subtype-specific treatment strategies. With the more recent understanding of the molecular mechanisms underlying the pathogenesis of some of these tumours, the treatment of sarcoma subtypes with targeted therapies is a rapidly evolving field. This review discusses the current management of sarcomas as well as promising new therapies that are currently underway in clinical trials.
Collapse
Affiliation(s)
- Jenna S Bleloch
- Department of Human Biology, University of Cape Town, South Africa
| | - Reyna D Ballim
- Department of Human Biology, University of Cape Town, South Africa
| | - Serah Kimani
- Department of Human Biology, University of Cape Town, South Africa
| | - Jeannette Parkes
- Department of Radiation Oncology, University of Cape Town, South Africa
| | - Eugenio Panieri
- Department of Surgery, University of Cape Town, South Africa
| | - Tarryn Willmer
- Department of Human Biology, University of Cape Town, South Africa
| | - Sharon Prince
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| |
Collapse
|
43
|
Huang K, Chen Z, Zhang X. Two new KIT exon 13 mutations in one gastric gastrointestinal stromal tumor (GIST). INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8863-8867. [PMID: 31966753 PMCID: PMC6965401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/25/2017] [Indexed: 06/10/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequently mesenchymal tumors found in the gastrointestinal tract. These tumors are usually composed of spindle-shaped cells. More than 80% of GISTs harbor mutations of KIT gene which encodes for an important receptor tyrosine kinase (RTK) type III. Within KIT gene mutations, the mutation of exon 13 is very rare. Here we described a case of GIST in a 42-year-old male which carried two new KIT exon 13 mutations (R634W and N655T). This patient was diagnosed with upper digestive tract hemorrhage and later CT scan revealed a 2.2 cm × 4.0 cm soft-tissue mass on the posterior wall of the stomach. The patient went through a laparoscopic gastrectomy. Following pathological examination revealed this tumor to be a low-risk GIST. Gene sequencing analysis shown that the tumor had two mutations in KIT exon 13, which were not found in the literature. The postoperative course was uneventful and no recurrence was observed after 6 months. Furthermore, we also gave a short review of previously published papers describing KIT exon 13 mutations.
Collapse
Affiliation(s)
- Ke Huang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, Zhejiang Province, China
| | - Zhen Chen
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, Zhejiang Province, China
| | - Xiuming Zhang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, Zhejiang Province, China
| |
Collapse
|
44
|
Dual Targeting of Insulin Receptor and KIT in Imatinib-Resistant Gastrointestinal Stromal Tumors. Cancer Res 2017; 77:5107-5117. [DOI: 10.1158/0008-5472.can-17-0917] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 11/16/2022]
|
45
|
Zeichner SB, Goldstein DA, Kohn C, Flowers CR. Cost-effectiveness of precision medicine in gastrointestinal stromal tumor and gastric adenocarcinoma. J Gastrointest Oncol 2017; 8:513-523. [PMID: 28736638 DOI: 10.21037/jgo.2016.04.03] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Over the past 20 years, with the incorporation of genetic sequencing and improved understanding regarding the mechanisms of cancer growth/metastasis, novel targets and their associated treatments have emerged in oncology and are now regularly incorporated into the clinical care of patients in the US. Novel, more tumor-specific, non-chemotherapy agents, including agents that are commonly used in the treatment of patients with gastric adenocarcinoma (GA) and gastrointestinal stromal tumor (GIST), fall under a broader treatment strategy, termed "precision medicine". While diagnostic testing and associated treatments in metastatic GA (mGA) are costly and may produce marginal benefit, those associated with GIST, despite being costly, produce significant improvements in patient outcomes. Despite the significant difference in impact, the agents associated with these cancers have similar acquisition costs. In this paper, we will review the current literature regarding cost and cost-effectiveness associated with precision medicine diagnosis and treatment strategies for GA and GIST.
Collapse
Affiliation(s)
- Simon B Zeichner
- Winship Cancer Institute at Emory University, Division of Hematology & Oncology, Atlanta, GA 30322, USA
| | - Daniel A Goldstein
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva 4941492, Israel
| | - Christine Kohn
- University of Saint Joseph School of Pharmacy, Hartford Hospital Evidence-based Practice Center, Hartford, CT 06103, USA
| | - Christopher R Flowers
- Winship Cancer Institute at Emory University, Division of Hematology & Oncology, Atlanta, GA 30322, USA
| |
Collapse
|
46
|
Cho J, Kim SY, Kim YJ, Sim MH, Kim ST, Kim NKD, Kim K, Park W, Kim JH, Jang KT, Lee J. Emergence of CTNNB1 mutation at acquired resistance to KIT inhibitor in metastatic melanoma. Clin Transl Oncol 2017; 19:1247-1252. [PMID: 28421416 DOI: 10.1007/s12094-017-1662-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/10/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE The KIT inhibitor, imatinib, has shown promising efficacy in patients with KIT-mutated melanoma; however, acquisition of resistance to imatinib occurs rapidly in the majority of patients. The mechanisms of acquired resistance to imatinib in melanoma remain unclear. METHODS We analyzed biopsy samples from paired baseline and post-treatment tumor lesions in one patient with KIT-mutated melanoma who had had an initial objective tumor regression in response to imatinib treatment followed by disease progression 8 months later. RESULTS Targeted deep sequencing from post-treatment biopsy samples detected an additional mutation in CTNNB1 (S33C) with original KIT L576P mutation. We examined the functional role of the additional CTNNB1 S33C mutation in resistance to imatinib indirectly using the Ba/F3 cell model. Ba/F3 cell lines transfected with both the L576P KIT mutation and the CTNNB1 S33C mutation demonstrated no growth inhibition despite imatinib treatment, whereas growth inhibition was observed in the Ba/F3 cell line transfected with the L576 KIT mutation alone. CONCLUSIONS We report the first identification of the emergence of a CTNNB1 mutation that can confer acquired resistance to imatinib. Further investigation into the causes of acquired resistance to imatinib will be essential to improve the prognosis for patients with KIT-mutated melanoma.
Collapse
Affiliation(s)
- J Cho
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - S Y Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - Y J Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.,Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - M H Sim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - S T Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - N K D Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - K Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| | - W Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - J H Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - K-T Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - J Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea.
| |
Collapse
|
47
|
Bannon AE, Klug LR, Corless CL, Heinrich MC. Using molecular diagnostic testing to personalize the treatment of patients with gastrointestinal stromal tumors. Expert Rev Mol Diagn 2017; 17:445-457. [PMID: 28317407 DOI: 10.1080/14737159.2017.1308826] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION The diagnosis and treatment of gastrointestinal stromal tumor (GIST) has emerged as a paradigm for modern cancer treatment ('precision medicine'), as it highlights the importance of matching molecular defects with specific therapies. Over the past two decades, the molecular classification and diagnostic work up of GIST has been radically transformed, accompanied by the development of molecular therapies for specific subgroups of GIST. This review summarizes the developments in the field of molecular diagnosis of GIST, particularly as they relate to optimizing medical therapy. Areas covered: Based on an extensive literature search of the molecular and clinical aspects of GIST, the authors review the most important developments in this field with an emphasis on the differential diagnosis of GIST including mutation testing, therapeutic implications of each molecular subtype, and emerging technologies relevant to the field. Expert commentary: The use of molecular diagnostics to classify GIST has been shown to be successful in optimizing patient treatment, but these methods remain under-utilized. In order to facilitate efficient and comprehensive molecular testing, the authors have developed a decision tree to aid clinicians.
Collapse
Affiliation(s)
- Amber E Bannon
- a Portland VA Health Care System and OHSU Knight Cancer Institute , Portland , OR , USA
| | - Lillian R Klug
- a Portland VA Health Care System and OHSU Knight Cancer Institute , Portland , OR , USA
| | - Christopher L Corless
- a Portland VA Health Care System and OHSU Knight Cancer Institute , Portland , OR , USA
| | - Michael C Heinrich
- a Portland VA Health Care System and OHSU Knight Cancer Institute , Portland , OR , USA
| |
Collapse
|
48
|
Xue A, Gao X, Fang Y, Shu P, Ling J, Shen K, Sun Y, Qin J, Qin X, Hou Y. Incorporation of NLR into NIH stratification system increases predictive accuracy for surgically resected gastrointestinal stromal tumors. Acta Biochim Biophys Sin (Shanghai) 2017; 49:179-185. [PMID: 28039147 DOI: 10.1093/abbs/gmw123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/25/2016] [Indexed: 12/11/2022] Open
Abstract
Previous studies have reported that preoperative neutrophil-to-lymphocyte (NLR) and platelet-to-lymphocyte ratios were prognostic for various types of cancers. The aim of this study was to investigate the predictive utilities of preoperative peripheral blood counts in patients with gastrointestinal stromal tumors (GISTs). We enrolled 510 consecutive, previously untreated patients who underwent surgery for primary, localized GISTs. The relationship between recurrence-free survival and outcome variables was assessed by univariate and multivariate analyses, while the clinicopathologic relevance of NLR was determined using the Chi-square test. A preoperative NLR ≥2 was associated with poor prognosis in patients undergoing surgeries for primary, localized GISTs. It was an independent predictor only in patients classified as National Institutes of Health high risk but not in the entire population. Preoperative NLR is a feasible and reproducible peripheral biomarker that helps identify patients for intensive adjuvant therapy and frequent surveillance.
Collapse
Affiliation(s)
- Anwei Xue
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaodong Gao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Fang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ping Shu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiaqian Ling
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Kxuntang Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Qin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinyu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
49
|
Surgical management of patients with progressing metastatic gastrointestinal stromal tumors receiving sunitinib treatment: A prospective cohort study. Int J Surg 2017; 39:30-36. [PMID: 28110026 DOI: 10.1016/j.ijsu.2017.01.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND For selected patients with metastatic gastrointestinal stromal tumor (GIST) who have received first-line imatinib (IM) and are undergoing cytoreductive surgery, response to IM at time of surgery correlates with resection completeness as well as favorable progression-free survival (PFS) and overall survival (OS). However, surgical impact in GIST patients receiving second-line sunitinib (SU) is still not well clarified. MATERIALS AND METHODS Between 2001 and 2014, 86 of 311 metastatic GIST patients received SU. Among them, 69 patients eventually experienced progression. Twenty-six patients who experienced local progression (LP) and underwent surgeries were investigated. Each tumor was assessed for genetic alterations before and after surgery. RESULTS Twenty-six GIST patients receiving SU who experienced LP underwent surgery after a median of 6.2 months of SU use. Nineteen patients (73.1%) had undergone prior surgery on IM. The complication rate was 15.3%, and no additional operation was required for GIST patients receiving SU and experiencing LP who underwent surgery. The median PFS and OS times after surgery and start of SU were 5.2 and 18.9 months, respectively, and 16.4 and 26.0 months, respectively (median follow-up of 15.2 months). GIST patients receiving SU with LP (N = 26) may gain a significant PFS and OS benefit with surgery when compared with those not undergoing surgery (N = 43) (p = 0.003 and p = 0.02, respectively). Secondary exon 17 mutation occurred commonly and might explain SU resistance (8/23; 34.8%). CONCLUSION Surgery is feasible in highly selected patients with metastatic GIST who are receiving SU and experiencing LP. Those patients may also have significantly prolonged PFS and OS after surgery. Secondary exon 17 mutation might explain SU resistance.
Collapse
|
50
|
Tales of Personalized Cancer Treatment. Semin Nephrol 2016; 36:462-467. [PMID: 27987546 DOI: 10.1016/j.semnephrol.2016.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|