1
|
Yip D, Zalcberg J, Blay JY, Eriksson M, Espinoza D, Price T, Marreaud S, Italiano A, Steeghs N, Boye K, Underhill C, Gebski V, Simes J, Gelderblom H, Joensuu H. Imatinib alternating with regorafenib compared to imatinib alone for the first-line treatment of advanced gastrointestinal stromal tumor: The AGITG ALT-GIST intergroup randomized phase II trial. Br J Cancer 2025; 132:897-904. [PMID: 40133509 DOI: 10.1038/s41416-025-02983-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/14/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND To determine if an alternating regimen of the tyrosine kinase inhibitors imatinib and regorafenib improved outcomes in patients with advanced gastrointestinal stromal tumors. METHODS ALTGIST (NCT02365441) was a randomized phase II study of standard treatment of imatinib (Arm A) compared with an experimental alternating regimen of imatinib and regorafenib (Arm B). Primary outcome was best objective tumor response (OTR) at nine months. RESULTS Seventy-six eligible patients (Arm A 36, Arm B 40) enrolled were evaluable. Median follow-up was 46.0 months (range 6.5-64.6). Best responses and OTR were similar at 9 months. Eighteen (50.0%) Arm A patients and twelve (30.0%) Arm B patients discontinued treatment due to progressive disease. No Arm A patients stopped protocol therapy due to unacceptable toxicity, with 12 (30.0%) stopping in Arm B. Twelve (33.2%) Arm A patients and 12 (30.0%) Arm B patients experienced at least one serious adverse event, mostly grade 3. Secondary endpoints of PFS at 1 and OS at 1 year were not statistically different. CONCLUSIONS Alternation of imatinib and regorafenib did not impact on 9 months objective response nor on the secondary objectives of PFS and OS. Patients in the alternating arm experienced more toxicity and protocol discontinuations. CLINICAL TRIAL REGISTRATION NCT02365441.
Collapse
Affiliation(s)
- Desmond Yip
- The Canberra Hospital and Australian National University, Canberra, ACT, Australia.
| | - John Zalcberg
- School of Public Health, Monash University, Melbourne, VIC, Australia
| | | | | | - David Espinoza
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
| | - Timothy Price
- Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia
| | - Sandrine Marreaud
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | | | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Craig Underhill
- Albury-Wodonga Regional Cancer Centre, Albury-Wodonga, NSW, Australia
| | - Val Gebski
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
| | - Hans Gelderblom
- Leiden University Medical Centre, Department of Medical Oncology, Leiden, The Netherlands
| | - Heikki Joensuu
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
2
|
Zhao Z, Qiu S, Zhang X, Liu S, Wang L, Guan H, He J, Hu Y, Li X, Luo S, Chen Z, Mo T, Zhang Y, Zhao X, Pan Y, Ding H, Cao J, Pan J. Characterization of a novel cell line established from mice gastrointestinal stromal model by chemical induction. Transl Oncol 2025; 56:102388. [PMID: 40233502 PMCID: PMC12022689 DOI: 10.1016/j.tranon.2025.102388] [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: 09/23/2024] [Revised: 02/03/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are a type of tumor that originates from gastrointestinal mesenchymal tissue. Although several somatic or germline mutation GIST mice were established, however, there is still a lack of an authentic mice GIST cell lines for further experimental study. METHODS We developed a chemically induced C57BL/6 J GIST model using 3- methylcholanthrene. Tumor characteristics were confirmed through histology and IHC. Primary cells were isolated to establish the mGSTc01 cell line, and molecular profiling was conducted. Additionally, we established GIST model in immunocompetent mice to evaluate their sensitivity to imatinib. RESULTS Our study successfully developed a chemically induced murine GIST model, characterized by positive staining of c-kit and DOG-1. The mGSTc01 monoclonal cell line exhibited slender morphology and expressed the c-kit marker, Whole exome sequencing uncovered mutations of Lamb1, MMP9, and c-kit in GIST cells and provided a detailed picture of the entire genome's copy number variations. RNA sequencing indicated genes associated with cell adhesion and focal adhesion were enriched in mGSTc01 cells. The mGSTc01 cells demonstrated obvious malignant behaviors, notably elevated migration, adhesion, and proliferation. In immunocompetent mice, subcutaneous xenografts not only reserved the aggressive phenotype but also displayed a response to imatinib, underscoring the model's applicability for advancing therapeutic research. CONCLUSION We firstly established a mGSTc01 cell line derived from C57BL/6 J mice GIST tumor offers, which closely mimicking human disease characteristics. It is a potent platform for investigating tumor microenvironment of GIST in mice model, and provides a novel way for new therapeutic discoveries in GIST.
Collapse
Affiliation(s)
- Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shenghui Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China
| | - Xiangwei Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shijin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, 510632, PR China
| | - Hanyang Guan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Jiashuai He
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yangzhi Hu
- The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, PR China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Simin Luo
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zuyang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Tianmu Mo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Xiaoxu Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Hui Ding
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| | - Jie Cao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China.
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| |
Collapse
|
3
|
Campaña MB, Perkins MR, McCabe MC, Neumann A, Larson ED, Fantauzzo KA. PDGFRα/β heterodimer activation negatively affects downstream ERK1/2 signaling and cellular proliferation. Nat Commun 2025; 16:4754. [PMID: 40404618 PMCID: PMC12098797 DOI: 10.1038/s41467-025-59938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
The platelet-derived growth factor receptor (PDGFR) family of receptor tyrosine kinases consists of two receptors, PDGFRα and PDGFRβ, that homodimerize and heterodimerize upon ligand binding. Here, we tested the hypothesis that differential internalization and trafficking dynamics of the various PDGFR dimers underlie differences in downstream intracellular signaling and cellular behavior. Using a bimolecular fluorescence complementation approach, we demonstrated that PDGFRα/β heterodimers are rapidly internalized into early endosomes. We showed that PDGFRα/β heterodimer activation does not induce downstream phosphorylation of ERK1/2 and significantly inhibits cell proliferation. Further, we identified MYO1D as a protein that preferentially binds PDGFRα/β heterodimers and demonstrated that knockdown of MYO1D leads to retention of PDGFRα/β heterodimers at the plasma membrane, increased phosphorylation of ERK1/2 and increased cell proliferation. Collectively, our findings impart valuable insight into the molecular mechanisms by which specificity is introduced downstream of PDGFR activation to differentially propagate signaling and generate distinct cellular responses.
Collapse
Affiliation(s)
- Maria B Campaña
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Madison R Perkins
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Neumann
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric D Larson
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine A Fantauzzo
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
4
|
Shulman DS, Crompton BD. Emerging Role of Blood-based Biomarkers in Sarcomas. Hematol Oncol Clin North Am 2025:S0889-8588(25)00040-1. [PMID: 40410056 DOI: 10.1016/j.hoc.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
We assess the emerging role of liquid biopsies, particularly circulating tumor DNA (ctDNA), in sarcoma management. Preliminary studies suggest that ctDNA has multiple potential applications including, early detection in patients with cancer predisposition syndromes, diagnosis, prognostication, therapy selection, and monitoring treatment response. Among patients with gastrointestinal stromal tumors, studies have demonstrated the capacity for identification of clinically relevant resistance mutations. In other sarcoma subtypes such as Ewing sarcoma and osteosarcoma, early findings indicate that ctDNA levels might correlate with tumor burden and outcomes, potentially aiding in risk stratification. Clinical utility has not been established for these applications.
Collapse
Affiliation(s)
- David S Shulman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Brian D Crompton
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA; Boston Children's Hospital, Boston, MA 02115, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
5
|
Kanda T, Ishikawa M, Techigawara K, Saginoya T, Hamada K, Saito M, Uesugi N, Teranishi Y. Pimitespib therapy for a patient with PDGFRA D842V-mutant gastrointestinal stromal tumor. Clin J Gastroenterol 2025:10.1007/s12328-025-02144-9. [PMID: 40347384 DOI: 10.1007/s12328-025-02144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Gastrointestinal stromal tumors (GISTs) with platelet-derived growth factor receptor alpha (PDGFRA) mutations are resistant to tyrosine kinase inhibitors. Pimitespib, a novel heat shock protein 90 inhibitor, was recently approved as a fourth-line treatment for advanced GISTs; however, data on its efficacy against PDGFRA-mutant GISTs remain scarce. We report a case of a 67-year-old male with a gastric GIST harboring a PDGFRA exon 18 Asp842Val mutation. The patient presented with a large peritoneal metastasis at the hepatic hilum and underwent proton beam therapy, achieving 8 months of disease control. However, the tumor progressed thereafter. Regorafenib was introduced but failed immediately owing to tumor penetration. The treatment was switched to pimitespib (160 mg daily, 5 days on, 2 days off, per 21-day cycle), and the patient completed four cycles of the therapy. Post-treatment 18F-fluorodeoxyglucose (FDG)-positron emission tomography showed a significant reduction in FDG uptake by the metastatic lesion. Pimitespib therapy was eventually discontinued because of duodenal bleeding, with a time to treatment failure of 13 weeks. Although based on a single case, this report demonstrates a significant metabolic response to pimitespib in PDGFRA-mutant GIST. More cases are required to fully elucidate the efficacy of this therapy against such rare tumors.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Department of Gastroenterology, Southern TOHOKU General Hospital, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8563, Japan.
| | - Masafumi Ishikawa
- Department of Gastroenterology, Southern TOHOKU General Hospital, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8563, Japan
| | - Kae Techigawara
- Department of Gastroenterology, Southern TOHOKU General Hospital, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8563, Japan
| | - Toshiyuki Saginoya
- Department of Diagnostic Radiology, Southern TOHOKU General Hospital, Koriyama, Fukushima, 963-8563, Japan
| | - Koichi Hamada
- Department of Minimally Invasive Surgical and Medical Oncology, Fukushima Medical University, Koriyama, Fukushima, 963-8563, Japan
| | - Motonobu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Cancer Genome Medicine, Fukushima Medical University Hospital, Fukushima City, Fukushima, 960-1295, Japan
| | - Noriyuki Uesugi
- Department of Pathology, Southern TOHOKU General Hospital, Koriyama, Fukushima, 963-8563, Japan
| | - Yasushi Teranishi
- Department of Surgery, Southern TOHOKU General Hospital, Koriyama, Fukushima, 963-8563, Japan
| |
Collapse
|
6
|
Naito Y, Iwagami S, Doi T, Takahashi T, Kurokawa Y. Pimitespib in patients with advanced gastrointestinal stromal tumors in Japan: an expanded access program. Int J Clin Oncol 2025; 30:935-943. [PMID: 40019689 PMCID: PMC12014775 DOI: 10.1007/s10147-025-02726-0] [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: 10/15/2024] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Pimitespib, an oral heat shock protein 90 inhibitor, significantly prolonged progression-free survival in patients with advanced gastrointestinal stromal tumors (GIST) in CHAPTER-GIST-301 study. This expanded access program was conducted to evaluate the safety and efficacy of pimitespib in Japanese patients with advanced GIST. METHODS This multicenter, open-label, single-arm study was conducted in patients (≥ 20 years) with histologically confirmed GIST who had been previously treated with imatinib, sunitinib and regorafenib and had an Eastern Cooperative Oncology Group performance status of 0-1. Patients received pimitespib 160 mg/day for five days, followed by a 2-day rest, in 21-day cycles. RESULTS Between February and August 2022, 23 patients were enrolled (median age 59.0 years). Over a median pimitespib treatment duration of 81.0 days, adverse events occurred in 22 patients (95.7%). The most common adverse events were diarrhea (73.9%), nausea (39.1%) and increased blood creatinine (30.4%). Serious adverse events occurred in two patients (tumor hemorrhage and tumor pain); neither was related to pimitespib. One patient had grade 3 diarrhea that was considered treatment-related. Four patients (17.4%) had eye disorders, all of which were grade 1 and treatment-related. The median progression-free survival was 4.2 months (95% confidence interval [CI] 1.9-6.2), the overall response rate was 0% (95% CI 0-16.1) and the disease control rate was 66.7% (95% CI 43.0-85.4). CONCLUSIONS Pimitespib was well tolerated and effective in patients with advanced GIST in real-world practice in Japan. No new safety signals were identified. TRIAL REGISTRATION jRCT2031210526 registered 1 February 2022.
Collapse
Affiliation(s)
- Yoichi Naito
- Department of General Internal Medicine, Medical Oncology, Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba-ken, Kashiwa, 277-8577, Japan.
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Surgery, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Toshihiko Doi
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
7
|
Xiang W, Yuan W, Ren L, Huang W, Liang H, Huang J, Luan L, Xu C, Hou Y. A case of quadruple wild-type gastrointestinal stromal tumor with CDC42BPB::NTRK3 fusion and abundant lymphoid infiltration. Diagn Pathol 2025; 20:31. [PMID: 40133893 PMCID: PMC11934696 DOI: 10.1186/s13000-025-01630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. The most common mutations in GISTs are those in receptor tyrosine kinase (KIT) and platelet-derived growth factor receptor alpha (PDGFRA). GISTs without KIT or PDGFRA mutations are defined as wild-type (WT) GISTs. The molecular changes, prognosis, and treatments of WT GISTs remain uncertain. Among WT GISTs, neurotrophic tyrosine receptor kinase (NTRK) fusions have rarely been reported. We report a case of quadruple wild-type GIST harboring a novel CDC42BPB::NTRK3 fusion. In this study, we described a 66-year-old male patient with intrajejunal lesion. This case showed massive lymphocytic and plasma cell infiltration, which caused diagnostic difficulties in morphology. CDC42BPB::NTRK3 fusion was detected via next-generation sequencing (NGS), and this finding was confirmed by fluorescence in situ hybridization (FISH), which revealed NTRK3 breakage. However, the expression of the Trk protein in tumor tissue was not detected by immunohistochemistry (IHC). This finding expands the genetic spectrum of NTRK rearrangements in GISTs.
Collapse
Affiliation(s)
- Wentao Xiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Yuan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Ren
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huaiyu Liang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lijuan Luan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Liu Y, Li XF. Characteristics and therapeutic strategies for familial gastrointestinal stromal tumors. World J Gastrointest Oncol 2025; 17:100463. [PMID: 40092960 PMCID: PMC11866256 DOI: 10.4251/wjgo.v17.i3.100463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 02/14/2025] Open
Abstract
This editorial discusses Wang et al's article on familial gastrointestinal stromal tumors (GISTs). We read with great interest this article concerning the diagnosis, treatment, and post-treatment management of patients with familial GISTs. The actual incidence of GISTs may be underestimated due to diagnostic limitations and the long-term low-risk behavior of some GISTs. The molecular landscape of GISTs is primarily driven by mutations in the KIT and platelet-derived growth factor receptor alpha (PDGFRA) genes. A subset of GISTs without these mutations known as wild-type GISTs, may harbor other rare mutations, impacting their response to targeted therapies. Clinically, patients with GISTs present with non-specific symptoms, often leading to delayed diagnosis. Genetic predispositions in familial GISTs provide insights into the genetic architecture and extragastrointestinal manifestations of GISTs. Management has evolved from surgical interventions to molecular-based therapies using tyrosine kinase inhibitors. The management of GISTs, especially in familial cases, requires a multidisciplinary approach. Cases of different gene mutations were reported in the same family, suggesting that incorporating genetic testing into routine clinical practice is crucial for the early identification of high-risk individuals and the implementation of tailored surveillance programs.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Xiao-Feng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| |
Collapse
|
9
|
Yang C, Li K, Xiang B. Case Report: Duodenal gastrointestinal stromal tumor misdiagnosed as tumor located on the major duodenal papilla leading to fatal gastrointestinal bleeding in a child. Front Pediatr 2025; 13:1546914. [PMID: 40123671 PMCID: PMC11925956 DOI: 10.3389/fped.2025.1546914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Background Although gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract, they are rare in children, particularly those located on the duodenum. Here, we present an interesting pediatric case involving a 13-year-old boy who experienced gastrointestinal hemorrhage, he was misdiagnosed with a tumor located on the major duodenal papilla and was ultimately confirmed to be duodenal GISTs. Case presentation A 13-year-old boy presented to a local hospital with fatigue and melena. Gastroscopy suggested a tumor located at the major duodenal papilla, and the patient was referred to our hospital for surgical evaluation. Upon further investigation and surgical exploration, the diagnosis was revised to a duodenal GIST with surface ulceration and active bleeding. The ulcer's morphology and location mimicked the appearance of the major duodenal papilla, leading to the initial diagnostic error. Conclusions Duodenal GISTs in pediatric patients often present asymptomatically but can manifest with severe complications such as fatal gastrointestinal bleeding. The tumor's morphology and location can obscure the major papilla, complicating preoperative diagnosis and influencing surgical decision-making. Comprehensive preoperative evaluation and careful intraoperative exploration are critical for accurate diagnosis and optimal management.
Collapse
Affiliation(s)
| | - Kewei Li
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Shahid G, Qudsia M, Naima I, Ibrahim K. Gastrointestinal stromal tumors with an uncommon primary mutation responded well to imatinib. CANCER PATHOGENESIS AND THERAPY 2025; 3:176-178. [PMID: 40182118 PMCID: PMC11963161 DOI: 10.1016/j.cpt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 04/05/2025]
Affiliation(s)
- Gilani Shahid
- Department of Oncology, University Hospital of North Midlands, Staffordshire, ST4 6QG, United Kingdom
| | - Mujeeb Qudsia
- Department of Oncology, University Hospital of North Midlands, Staffordshire, ST4 6QG, United Kingdom
| | - Ikram Naima
- Department of Oncology, University Hospital of North Midlands, Staffordshire, ST4 6QG, United Kingdom
| | - Khir Ibrahim
- Department of Oncology, University Hospital of North Midlands, Staffordshire, ST4 6QG, United Kingdom
| |
Collapse
|
11
|
Strauss G, George S. Gastrointestinal Stromal Tumors. Curr Oncol Rep 2025; 27:312-321. [PMID: 39985704 DOI: 10.1007/s11912-025-01636-8] [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] [Accepted: 01/07/2025] [Indexed: 02/24/2025]
Abstract
PURPOSE OF REVIEW This review aims to outline the current understanding of the molecular drivers and treatment paradigms of gastrointestinal stromal tumors, with a focus on recent developments in treatment in the advanced disease setting. RECENT FINDINGS There have been recent advancements in our understanding of the molecular biology of gastrointestinal stromal tumors, including the identification of new genetic drivers and complex resistance mechanisms. We review the most recent findings in these areas, focusing on how new research insights are reshaping treatment strategies. Recent advancements in our understanding of the biology and treatment of GIST are paving the way for more personalized and effective therapeutic options. As knowledge of rare molecular subtypes, resistance mechanisms, and novel genomic techniques grows, new approaches are emerging in an effort to improve patient outcomes.
Collapse
Affiliation(s)
- Gal Strauss
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Suzanne George
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Mohammadi M, Roets E, Bleckman RF, Oosten AW, Grunhagen D, Desar IME, Bonenkamp H, Reyners AKL, van Etten B, Hartgrink H, Fiocco M, Schrage Y, Steeghs N, Gelderblom H. Impact of Mutation Profile on Outcomes of Neoadjuvant Therapy in GIST. Cancers (Basel) 2025; 17:634. [PMID: 40002229 PMCID: PMC11852491 DOI: 10.3390/cancers17040634] [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: 12/10/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Neoadjuvant imatinib therapy plays a crucial role in the management of gastrointestinal stromal tumors (GISTs), but its impact across various mutational profiles remains uncertain. OBJECTIVE The aim of this study is to describe the clinicopathological features and to assess the response and surgical outcomes of neoadjuvant imatinib in GIST patients exhibiting diverse mutational profiles. METHODS We conducted a retrospective study, extracting data from the Dutch GIST Registry, including patients treated with neoadjuvant imatinib. Response rate was the primary outcome, and secondary outcomes were the time on neoadjuvant treatment and resection margins (R0 vs. R1/R2), respectively. RESULTS Between 2009 and 2021, 326 patients were treated with neoadjuvant imatinib, of which 264 (80.9%) underwent resection. A total of 197 (74.6%) of them had a KIT-exon 11 mutation, 19 (7.3%) had other KIT mutations, 10 (3.8%) had PDGFRA D842 mutations, 21 (6.8%) had other PDGFRA mutations, 2 (0.7%) had NTRK mutation, 1 (0.4%) had an SDH mutation, and 17 (6.4%) had WT GISTs. Patients with KIT-exon 11 mutations demonstrated a higher rate of partial response to imatinib (60.5% vs. 33.3%; p = 0.00). A positive resection margin (R1 or R2) was observed in 14 (21.2%) patients with a non-KIT exon 11 mutations and in 11 (5.5%) patients with a KIT-exon 11 mutation (p = 0.00). Moreover, non-KIT exon 11 mutation patients had a shorter median duration of neoadjuvant therapy (5.3 months, range 0.5-21.0) compared to patients with a KIT exon 11 mutation (8.8 months, range 0.2-31.3; p < 0.001). CONCLUSIONS Our study highlights the variability in treatment response associated with different GIST mutational profiles. Patients with a KIT-exon-11 mutation tended to respond more favorably to neoadjuvant imatinib in terms of partial response and surgical outcomes.
Collapse
Affiliation(s)
- Mahmoud Mohammadi
- Department of Medical Oncology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Evelyne Roets
- Department of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (E.R.)
| | - Roos F. Bleckman
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Astrid W. Oosten
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN Rotterdam, The Netherlands
| | - Dirk Grunhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 CN Rotterdam, The Netherlands
| | - Ingrid M. E. Desar
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Han Bonenkamp
- Department of Surgery, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Anna K. L. Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Boudewijn van Etten
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Henk Hartgrink
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Marta Fiocco
- Mathematical Institute, Leiden University, 2300 RA Leiden, The Netherlands
- Princess Máxima, Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Biomedical Data Science, Section Medical Statistics, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Yvonne Schrage
- Department of Surgical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (E.R.)
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
13
|
Makker S, Rammal R, Gu P, Dalbagni G, Al-Ahmadie H, Agaram NP, Iyer G, Kotecha RR. Case report: Clinical and molecular features of renal gastrointestinal tumor. Front Oncol 2025; 15:1508600. [PMID: 40018405 PMCID: PMC11865245 DOI: 10.3389/fonc.2025.1508600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/17/2025] [Indexed: 03/01/2025] Open
Abstract
While gastrointestinal stromal tumors (GISTs) often arise within the GI tract, it is well known that GISTs may also rarely emanate outside of the digestive system. Prior case reports have documented various primary sites in non-GI organs [extra-intestinal GIST (EGIST)], yet only one report has described a localized GIST of renal origin. Here, we describe a patient who presented with bilateral renal masses who was found to have a large unresectable renal GIST tumor treated with imatinib. We discuss treatment experience and response with systemic therapy and describe molecular data to contextualize this ultra-rare presentation within the landscape of EGIST tumors.
Collapse
Affiliation(s)
| | - Rayan Rammal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ping Gu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Guido Dalbagni
- Department of Surgery, Sloan Kettering Cancer Center, New York, NY, United States
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Narasimhan P. Agaram
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Gopa Iyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ritesh R. Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
14
|
Florou V, Jacobs MF, Casey R, Evans D, Owens B, Raygada M, Rothschild S, Greenberg SE. A Review of Genomic Testing and SDH- Deficiency in Gastrointestinal Stromal Tumors: Getting to the GIST. Cancer Med 2025; 14:e70669. [PMID: 39927693 PMCID: PMC11808740 DOI: 10.1002/cam4.70669] [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: 09/24/2024] [Revised: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Gastrointestinal Stromal Tumors (GISTs) have seen significant advancements in their diagnosis and management, driven by targeted therapeutic development and molecular testing. The identification of mutations in genes such as KIT and PDGFRA has transformed treatment approaches, particularly through targeted therapies like imatinib, which have improved patient outcomes. This review explores the critical role of genomic testing in GIST, highlighting its importance in accurate diagnosis, treatment planning, and long-term surveillance for KIT/PDGFRA negative, SDH-deficient GISTs. SDH-deficient GISTs arise from mutations or epigenetic changes affecting the succinate dehydrogenase complex. The complexity of SDH-deficient GISTs, including their association with hereditary syndromes such as Hereditary Paraganglioma-Pheochromocytoma and/or hypermethylation of the SDHC promoter, underscores the need for comprehensive germline testing. Despite the availability of guidelines, variability exists in genomic testing recommendations across different regions, necessitating a unified approach. This review proposes a simplified algorithm for the genomic workup of GIST, and suggests all individuals with SDH-deficient GIST, regardless of germline testing result, require monitoring for additional SDHx-related tumors, given the lack of widely available methylation and full gene SDHA analysis.
Collapse
Affiliation(s)
- Vaia Florou
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - Michelle F. Jacobs
- Division of Genetic Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Ruth Casey
- Department of Medical GeneticsCambridge UniversityCambridgeUK
| | | | | | - Margarita Raygada
- Pediatric Oncology and Neuro‐Oncology BranchNational Cancer Institute/National Institutes of HealthBethesdaMarylandUSA
| | | | | |
Collapse
|
15
|
Nigro MC, Marchetti A, Fumagalli ER, De Luca I, Bertuzzi AF, Grimaudo MS, Grignani G, D’Ambrosio L, Merlini A, Badalamenti G, Incorvaia L, Dimino A, Gasperoni S, Vincenzi B, Fanti S, Di Federico A, Campana D, Pantaleo MA, Nannini M. 18F-Fluorodeoxyglucose Uptake in PDGFRA-Mutant Gastrointestinal Stromal Tumors. JAMA Netw Open 2025; 8:e2456058. [PMID: 39853981 PMCID: PMC11762236 DOI: 10.1001/jamanetworkopen.2024.56058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/15/2024] [Indexed: 01/26/2025] Open
Abstract
Importance The D842V platelet-derived growth factor receptor α (PDGFRA) mutation identifies a molecular subgroup of gastrointestinal stromal tumors (GISTs), primarily resistant to standard tyrosine kinase inhibitors and with an overall more indolent behavior. Although functional imaging with 18F-fluorodeoxyglucose-labeled positron emission tomography ([18F]FDG-PET) plays a proven role in GISTs, especially in early assessment of tumor response, less is known about [18F]FDG uptake according to the GIST molecular subtypes. Objective To evaluate the degree of [18F]FDG uptake in PDGFRA-mutant GISTs and better define the role of functional imaging in this rare and peculiar subset of GISTs. Design, Setting, and Participants This multi-institutional retrospective cohort study involving 7 GIST reference centers in Italy included patients with PDGFRA-mutant GIST who underwent [18F]FDG-PET from January 1, 2000, to December 31, 2023. Data on the maximum standardized uptake value (SUVmax) of primary tumor or metastatic disease were collected. Exposure PDGFRA-mutant GIST and [18F]FDG-PET. Main Outcome and Measure The primary outcome was the degree of [18F]FDG uptake of PDGFRA-mutant GISTs, with a focus on the D842V-mutant subgroup. Secondary objectives were to assess the association between the degree of [18F]FDG uptake and main clinicopathologic features. Results A total of 71 patients with PDGFRA-mutant GISTs were included in the analysis: 37 (52.1%) in the D842V subgroup (group A) and 34 (47.9%) in the non-D842V subgroup (group B). Additionally, 70 patients with KIT exon 11-mutant GIST served as a control group (group C). For all 141 participants, the median age at diagnosis was 59 (range, 26-89) years, and 81 patients (57.4%) were male. Overall, the median SUVmax was 4.4 (IQR, 0-10.1), while the median SUVmax for group A was 0 (IQR, 0-3.2); for group B, 3.6 (IQR, 0-5.1); and for group C, 10.1 (IQR, 5.1-13.9). The median SUVmax of PDGFRA-mutant GISTs was significantly lower than the median value of KIT exon 11-mutant GISTs (0 [IQR, 0-4.3] vs 10.1 [IQR, 5.1-14.0]; P < .001). Median [18F]FDG uptake was significantly lower in the D842V subgroup compared with the non-D842V subgroup (0 [IQR, 0-3.2] vs 3.6 [IQR, 0-5.1]; P = .02). Moreover, the triad of gastric primary tumor, tumor size greater than 10 cm, and SUVmax of 5.75 or less was associated with identification of PDGFRA-mutant GISTs. Conclusions and Relevance In this cohort study of patients with PDGFRA-mutant GISTs, the D842V-mutant GISTs were associated with an overall lower [18F]FDG uptake compared with other GIST subgroups. Therefore, the role of functional imaging with [18F]FDG-PET in this subset of GISTs may be limited and should be further explored for its potential prognostic and predictive value.
Collapse
Affiliation(s)
- Maria Concetta Nigro
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Andrea Marchetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elena Rosa Fumagalli
- Medical Oncology Unit 2, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale dei Tumori, Milano, Italy
| | - Ida De Luca
- Medical Oncology Unit 2, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale dei Tumori, Milano, Italy
| | - Alexia Francesca Bertuzzi
- Department of Medical Oncology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milano, Italy
| | - Maria Susanna Grimaudo
- Department of Medical Oncology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milano, Italy
| | - Giovanni Grignani
- Department of Medical Oncology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza, Torino, Italy
- Candiolo Cancer Institute, Fondazione del Piemonte–IRCCS, Candiolo (Turin), Turin, Italy
| | - Lorenzo D’Ambrosio
- Department of Medical Oncology, University of Turin, Turin, Italy
- Azienda Ospedaliera Universitaria San Luigi Gonzaga University Hospital, Orbassano, Italy
| | - Alessandra Merlini
- Candiolo Cancer Institute, Fondazione del Piemonte–IRCCS, Candiolo (Turin), Turin, Italy
- Department of Medical Oncology, University of Turin, Turin, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandra Dimino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Silvia Gasperoni
- Department of Oncology, Clinical Oncology Unit, University Hospital Careggi, Firenze, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Biomedico University of Rome, Rome, Italy
| | - Stefano Fanti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero Universitaria Di Bologna, Bologna, Italy
| | | | - Davide Campana
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Medical Oncology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Medical Oncology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Margherita Nannini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Medical Oncology, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
16
|
Shah K, Gopal K, Kumar S, Saha S. Emerging AXL Inhibitors in Oncology: Chemical and Biological Advances in Targeted Cancer Therapy. Anticancer Agents Med Chem 2025; 25:460-467. [PMID: 39679460 DOI: 10.2174/0118715206351185241209053053] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
AXL, a receptor tyrosine kinase, has emerged as a critical player in tumorigenesis, metastasis, and resistance to conventional therapies. Its aberrant activation drives cell proliferation, survival, and angiogenesis, making it an attractive target for cancer treatment. In recent years, significant progress has been made in the development of AXL inhibitors. Chemical approaches have led to the discovery of small molecules that selectively bind to and inhibit AXL, disrupting its downstream signaling pathways. These inhibitors exhibit diverse structural features, including ATP-competitive and allosteric binding modes, offering potential advantages in terms of selectivity and potency. In addition to chemical approaches, biological strategies have also been explored to target AXL. These include the use of monoclonal antibodies, which can neutralize AXL ligands or induce receptor internalization and degradation. Furthermore, gene therapy techniques have been investigated to downregulate AXL expression or disrupt its signaling pathways. Despite these advancements, challenges remain in the development of AXL inhibitors. Selectivity is a critical concern, as AXL shares homology with other receptor tyrosine kinases. Drug resistance is another obstacle, as cancer cells can develop mechanisms to evade AXL inhibition. Furthermore, to address these challenges, combination therapies are being explored, such as combining AXL inhibitors with other targeted agents or conventional treatments. In conclusion, developing AXL inhibitors represents a promising avenue for improving cancer treatment outcomes. Continued research efforts are essential to overcome the existing challenges and translate these compounds into effective clinical therapies.
Collapse
Affiliation(s)
- Kamal Shah
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, 281406, India
| | - Krishan Gopal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, 281406, India
| | - Shivendra Kumar
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, 281406, India
| | - Sunam Saha
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, 281406, India
| |
Collapse
|
17
|
Ben Rejeb S, Aloui D, Ayari A, Chouchen A. Prognostic Significance of C-MYC and EGFR Overexpression in Gastrointestinal Stromal Tumors: An Immunohistochemical Study. Appl Immunohistochem Mol Morphol 2025; 33:43-48. [PMID: 39636317 DOI: 10.1097/pai.0000000000001235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 10/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In addition to mutations in KIT and PDGFRA, many other genetic alterations have been described in gastrointestinal stromal tumors (GISTs), including amplifications of C-MYC and EGFR, which are often associated with increased protein expression. The main of this study was to investigate the prognostic significance of C-MYC and EGFR expression in GISTs using immunohistochemistry (IHC). METHODS We collected all GIST cases over a 16-year period. These cases were tested using antibodies against C-MYC (Leica, clone EP121) and EGFR (Leica, clone 113). C-MYC staining was assessed using the H-score method for nuclear, cytoplasmic, and combined staining. For EGFR staining (either cytoplasmic or nuclear), the intensity was graded as follows: 0 (no staining), 1 (weak staining), 2 (moderate staining), and 3 (strong staining). The percentage of positive cells was evaluated using a semiquantitative approach. Statistical analysis was performed using SPSS24. RESULTS A total of 37 cases were included in our study. Nuclear expression of C-MYC was observed in 43% of the cases, with a high H-score in 43%. A statistically significant association was found between a high nuclear H-score for C-MYC and mitotic rate (P=0.046), as well as a high Ki-67 proliferation rate (P=0.046). However, no statistically significant associations were identified between the nuclear H-score of C-MYC and other clinical, pathologic, or survival data. Cytoplasmic expression of C-MYC was noted in 22% of cases, but no significant correlations were found with the clinicopathological data. EGFR staining was observed in 86% of cases, with a high score of 51%. EGFR expression was significantly associated with the mitotic index (P=0.012) and Ki-67 proliferation rate (P=0.046). CONCLUSIONS Our findings suggest that both C-MYC and EGFR may be overexpressed and/or amplified in GISTs, indicating their potential prognostic role. This could also pave the way for therapeutic strategies targeting these proteins.
Collapse
Affiliation(s)
| | | | - Asma Ayari
- Department of Pathology, Rabta's Hospital, Marsa, Tunisia
| | | |
Collapse
|
18
|
Hechtman JF, Baskovich B, Fussell A, Geiersbach KB, Iorgulescu JB, Sirohi D, Snow A, Sidiropoulos N. Charting the Genomic Frontier: 25 Years of Evolution and Future Prospects in Molecular Diagnostics for Solid Tumors. J Mol Diagn 2025; 27:6-11. [PMID: 39722285 DOI: 10.1016/j.jmoldx.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Jaclyn F Hechtman
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; Caris Life Sciences, Irving, Texas.
| | - Brett Baskovich
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; Mount Sinai Health System, New York, New York
| | - Amber Fussell
- The Association for Molecular Pathology, Rockville, Maryland
| | - Katherine B Geiersbach
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; Mayo Clinic, Rochester, Minnesota
| | - J Bryan Iorgulescu
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; Molecular Diagnostics Laboratory, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deepika Sirohi
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; University of California San Francisco, San Fransico, California
| | - Anthony Snow
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Nikoletta Sidiropoulos
- Solid Tumors Subdivision Leadership of the Association for Molecular Pathology, Rockville, Maryland; University of Vermont Medical Group, Burlington, Vermont
| |
Collapse
|
19
|
Liang SQ, Cui YT, Hu GB, Guo HY, Chen XR, Zuo J, Qi ZR, Wang XF. Development and validation of a machine-learning model for preoperative risk of gastric gastrointestinal stromal tumors. J Gastrointest Surg 2025; 29:101864. [PMID: 39448018 DOI: 10.1016/j.gassur.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) have malignant potential, and treatment varies according to risk. However, no specific protocols exist for preoperative assessment of the malignant potential of gastric GISTs (gGISTs). This study aimed to use machine learning (ML) to develop and validate clinically relevant preoperative models to predict the malignant potential of gGISTs. METHODS This study screened patients diagnosed with gGISTs at the Affiliated Hospital of North Sichuan Medical College. Moreover, this study employed the Least Absolute Shrinkage and Selection Operator (LASSO) and logistic regression to identify risk factors. Subsequently, an ensemble of ML models was used to determine the optimal classifier. In addition, this study used SHapley Additive exPlanations (SHAP) for tailored risk profiling. RESULTS This study included 318 patients with gGISTs. Using LASSO regression and multifactorial logistic regression, this study analyzed the training dataset, revealing that the presence of endoscopic ultrasound (EUS) high-risk features, tumor border clarity, tumor diameter, and monocyte-to-lymphocyte ratio (MLR) were significant predictors of high malignancy risk in gGIST. As determined by our ML approach, the logistic classification model demonstrated optimal performance, with area under the receiver operating characteristic curves of 0.919 for the training set and 0.925 for the test set. Furthermore, decision curve analysis confirmed the clinical relevance of the model. CONCLUSION High-risk EUS features, ill-defined tumor margins, larger tumor diameters, and elevated MLR independently predicted increased malignant potential in gGIST. This study developed logistic regression models based on these factors, which were further interpreted using the SHAP methodology. This analytical approach facilitated personalized therapeutic decision-making among diverse patient populations.
Collapse
Affiliation(s)
- Shi-Qi Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yu-Tong Cui
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guang-Bing Hu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hai-Yang Guo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xin-Rui Chen
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ji Zuo
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhi-Rui Qi
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xian-Fei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Digestive Endoscopy Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
20
|
Kanda T, Ichikawa H, Ishikawa T, Muneoka Y, Kano Y, Naito T, Suzuki S, Wakai T. Fourteen-year follow-up results of imatinib therapy in patients with unresectable and metastatic gastrointestinal stromal tumors. Int J Clin Oncol 2024; 29:1870-1877. [PMID: 39343814 DOI: 10.1007/s10147-024-02631-y] [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: 05/30/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Imatinib therapy is the gold standard for the treatment of unresectable and metastatic gastrointestinal stromal tumors (GISTs). However, few studies have reported the long-term outcomes of the treatment. METHODS Seventy patients who underwent imatinib therapy for unresectable and metastatic GISTs were enrolled between 2001 and 2009, and follow-up data were collected until October 2023. One hundred and sixty-eight months had passed since the final enrollment. The outcome measures were patient survival and the status of GIST and imatinib therapy. The cumulative incidence of disease progression (PD) and the chronological changes in PD hazard rate (HR) were also analyzed. RESULTS The 5-, 10-, 15-, and 20-year overall survival rates were 64.3%, 30.0%, 16.8%, and 12.2%, respectively. Sixty of the 70 enrolled patients died before the data cutoff date: 47 (78.3%) patients died of GIST progression while the remaining 13 (21.7%) died of diseases other than GISTs. The cumulative incidence of PD logarithmically increased, and PD continued even after 10 years of treatment. PD HR decreased over time to reach the lowest value at 9.6 years after the initiation of treatment (HR 0.00027; 95% CI 0.00007-0.00174) and after that formed a small peak at 13 years (HR 0.00144; 95% CI 0.00043-0.00436). CONCLUSIONS Imatinib therapy showed high clinical efficacy in terms of long-term survival in GIST patients. However, patients undergoing imatinib therapy were at continuous risk of PD even after the 10-year treatment. Long-term treatment and follow-up beyond 10 years are necessary for unresectable and metastatic GIST patients.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Department of Gastroenterology, Southern TOHOKU General Hospital, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8563, Japan.
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Takashi Ishikawa
- Department of Surgery, Saiseikai Niigata Kenoh Kikan Hospital, Sanjo, Niigata, Japan
| | - Yusuke Muneoka
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Yosuke Kano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| | - Tetsuya Naito
- Department of Surgery, Nagaoka Red Cross Hospital, Nagaoka, Niigata, Japan
| | - Satoshi Suzuki
- Department of Surgery, Tsuruoka Municipal Shonai Hospital, Tsuruoka, Yamagata, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Niigata, Japan
| |
Collapse
|
21
|
Xie F, Luo S, Liu D, Lu X, Wang M, Liu X, Jia F, Pang Y, Shen Y, Zeng C, Ma X, Tang D, Tu L, Yang L, Cheng Y, Luo Y, Xie F, Hou H, Huang T, Ni B, Zhuang C, Zhao W, Li K, Zheng X, Bi W, Jia X, He Y, Wang S, Cao H, Wu K, Wang Y. Genomic and transcriptomic landscape of human gastrointestinal stromal tumors. Nat Commun 2024; 15:9495. [PMID: 39489749 PMCID: PMC11532483 DOI: 10.1038/s41467-024-53821-1] [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: 12/23/2022] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Gastrointestinal stromal tumor (GISTs) are clinically heterogenous exhibiting varying degrees of disease aggressiveness in individual patients. We comprehensively describe the genomic and transcriptomic landscape of a cohort of 117 GISTs including 31 low-risk, 18 intermediate-risk, 29 high-risk, 34 metastatic and 5 neoadjuvant GISTs from 105 patients. GISTs have notably low tumor mutation burden but widespread copy number variations. Aggressive GISTs harbor remarkably more genomic aberrations than low-/intermediate-risk GISTs. Complex genomic alterations, chromothripsis and kataegis, occur selectively in aggressive GISTs. Despite the paucity of mutations, recurrent inactivating YLPM1 mutations are identified (10.3%, 7 of 68 patients), enriched in high-risk/metastatic GIST and functional study further demonstrates YLPM1 inactivation promotes GIST proliferation, growth and oxidative phosphorylation. Spatially and temporally separated GISTs from individual patients demonstrate complex tumor heterogeneity in metastatic GISTs. Finally, four prominent subtypes are proposed with different genomic features, expression profiles, immune characteristics, clinical characteristics and subtype-specific treatment strategies. This large-scale analysis depicts the landscape and provides further insights into GIST pathogenesis and precise treatment.
Collapse
Affiliation(s)
- Feifei Xie
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Shuzhen Luo
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Dongbing Liu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fujian Jia
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Yuzhi Pang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yanying Shen
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chunling Zeng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xinli Ma
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Daoqiang Tang
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Lin Tu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Linxi Yang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fanfan Xie
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Hao Hou
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Tao Huang
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, 200031, Shanghai, China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chun Zhuang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Wenyi Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Ke Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Wenbo Bi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yi He
- Department of Urology, No.1 Hospital of Jiaxing, 314000, Jiaxing, China
| | - Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| | - Kui Wu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China.
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
22
|
Ibrahim A, Montgomery EA. Gastrointestinal Stromal Tumors: Variants and Some Pitfalls That They Create. Adv Anat Pathol 2024; 31:354-363. [PMID: 39466697 DOI: 10.1097/pap.0000000000000463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The diagnosis of gastrointestinal stromal tumors (GISTs) is generally straightforward using a combination of histologic evaluation and pertinent immunohistochemical staining with CD117/kit and DOG-1 (discovered on GIST) antibodies. However, this tumor can be challenging in cases with an unusual morphology, in limited biopsies, for those in uncommon sites, post-treatment, and when other neoplasms express CD117/kit and DOG-1, thereby mimicking GIST. Finding epithelioid GISTs in the stomach in younger patients should prompt testing for succinate dehydrogenase (SHD)-deficiency using immunohistochemical staining for subunit B (SDHB). However, SDH-deficient GISTs can also arise in older patients, or as part of the Carney triad or Carney-Stratakis syndrome. GISTs with PDGFRA mutations can also prove difficult if they lack kit expression. It is also important to consider morphologic and immunophenotypic changes associated with treatment, including the potential absence of kit expression, particularly in GISTs that have metastasized. Therefore, obtaining clinical information regarding prior therapy with a tyrosine kinase inhibitor (TKI) is crucial.
Collapse
Affiliation(s)
- Ammoura Ibrahim
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL
| | | |
Collapse
|
23
|
He C, Wang Z, Yu J, Mao S, Xiang X. Current Drug Resistance Mechanisms and Treatment Options in Gastrointestinal Stromal Tumors: Summary and Update. Curr Treat Options Oncol 2024; 25:1390-1405. [PMID: 39441520 PMCID: PMC11541409 DOI: 10.1007/s11864-024-01272-7] [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] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
OPINION STATEMENT Gastrointestinal stromal tumor (GIST) is characterized by well-defined oncogenes. Despite the significant improvement in treatment outcomes with adjuvant imatinib therapy for patients, drug resistance remains a major challenge for GIST therapy. This review focuses on the mechanisms contributing to drug resistance phenotype in GIST, such as primary imatinib-resistant mutants, secondary mutations, non-covalent binding of TKI to its target, tumor heterogeneity, re-activation of pro-survival/proliferation pathways through non-KIT/PDGFRA kinases, and loss of therapeutic targets in wild-type GIST. Corresponding suggestions are proposed to overcome drug-resistance phenotype of GIST. This review also summarizes the suitability of currently approved TKIs on different KIT/PDGFRA mutations and updates related clinical trials. Recent potent drugs and emerging strategies against advanced GISTs in clinical trials are presented. Additionally, metabolic intervention offers a new avenue for clinical management in GIST. A landscape of metabolism in GIST and metabolic changes under imatinib treatment are summarized based on currently published data. The OXPHOS pathway is a promising therapeutic target in combination with TKI against sensitive KIT/PDGFRA mutants. Comprehensive understanding of the above resistance mechanisms, experimental drugs/strategies and metabolic changes is critical to implement the proper therapy strategy and improve the clinical therapy outcomes for GIST.
Collapse
Affiliation(s)
- Chunxiao He
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Zilong Wang
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiaying Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shuang Mao
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xi Xiang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
24
|
Mousavi S, Ono Y, VanderLaan PA, Guzmán-Arocho YD. Gastrointestinal stromal tumors in fine-needle aspiration biopsies. Diagn Cytopathol 2024; 52:575-581. [PMID: 38396207 DOI: 10.1002/dc.25285] [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: 01/15/2024] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024]
Abstract
Gastrointestinal stromal tumors (GISTs), although rare, are the most common mesenchymal neoplasms of the gastrointestinal tract. Their potential for malignancy underscores the significance of identifying them through cytomorphologic findings and pertinent immunohistochemical markers. GISTs can emerge anywhere along the gastrointestinal tract with a predilection for the stomach. The clinical manifestations vary from nonspecific abdominal symptoms to incidental discovery during diagnostic interventions for unrelated signs and symptoms. Cytologically, GIST aspirates contain spindle or epithelioid cells with immunoreactivity for CD117/c-KIT, DOG-1, and CD34. Molecularly, KIT or PDGFRA mutations are prevalent, guiding targeted therapy with tyrosine kinase inhibitors. Distinct subtypes like succinate dehydrogenase-deficient GISTs pose challenges, often affecting younger individuals and displaying unique features. Histologically, GISTs are graded by mitotic rates, aiding prognostication. Distinguishing GISTs from similar entities is pivotal, necessitating attention to their immunostaining patterns for making an accurate diagnosis and molecular alterations for effectively planning treatment. Common differential diagnoses include leiomyoma, schwannoma, and solitary fibrous tumor. This article presents a classic GIST case and showcases relatively simple diagnostic clues for identifying similar lesions that may occur in diverse locations.
Collapse
Affiliation(s)
- Seyedreza Mousavi
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuho Ono
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Paul A VanderLaan
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yaileen D Guzmán-Arocho
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Fujii H, Hirano H, Shiraishi K, Shoji H, Hirose T, Okita N, Takashima A, Koyama T, Kato K. Comprehensive Genomic Assessment of Advanced-Stage GI Stromal Tumors Using the Japanese National Center for Cancer Genomics and Advanced Therapeutics Database. JCO Precis Oncol 2024; 8:e2400284. [PMID: 39447098 PMCID: PMC11520344 DOI: 10.1200/po.24.00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Accepted: 09/06/2024] [Indexed: 10/26/2024] Open
Abstract
PURPOSE Clinical utility of comprehensive genomic profiling (CGP) for precision medicine has become evident. Although there are several reports on the genomic landscape of GI stromal tumors (GISTs), large-scale data specific to GIST are limited, especially in Asia. Additionally, the applicability of molecular-targeted agents identified using CGP has not been extensively examined. We investigated the status of genomic alterations in Japanese patients with advanced GISTs using the National Center for Cancer Genomics and Advanced Therapeutics (C-CAT) database to identify novel treatment strategies and drug development. MATERIALS AND METHODS We retrospectively reviewed the clinical and CGP data of patients with advanced-stage GIST registered in the C-CAT database to assess the genomic landscape and potential actionable alterations. RESULTS Data from 144 patients were reviewed. Oncogenic alterations were detected frequently in KIT (78%), CDKN2A (37%), CDKN2B (29%), RB1 (11%), STK11 (10%), TP53 (9%), PDGFRA (6%), and SDHB (6%). Loss of CDKN2A/CDKN2B was only observed in KIT/PDGFRA-mutated GISTs, while alterations in SDHA/SDHB were only detected in KIT/PDGFRA wild-type GISTs. Among 119 KIT/PDGFRA-mutated GISTs, 95 (80%) had oncogenic genomic alterations and 29 (24%) had actionable alterations, excluding KIT and PDGFRA. However, among 25 KIT/PDGFRA wild-type GISTs, 22 (88%) had oncogenic alterations and 11 (44%) had actionable alterations. Representative candidate drugs for genome-matched therapies in KIT/PDGFRA-mutated and wild-type GISTs were as follows: pembrolizumab for tumor mutation burden-high in one and two patients, respectively; poly-adenosine diphosphate ribose polymerase inhibitors for alterations related to homologous recombination deficiency in 12 and one patient, respectively; NTRK inhibitor for ETV6-NTRK3 fusion in one with KIT/PDGFRA wild-type GIST; and human epidermal growth factor receptor 2-antibody-drug conjugate in one with KIT/PDGFRA-mutated GIST. CONCLUSION This study highlights the genomic landscape of advanced GISTs and the important role of CGP in identifying rational molecular-targeted therapeutic options.
Collapse
Affiliation(s)
- Hiroyuki Fujii
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Hidekazu Hirano
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hirokazu Shoji
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Toshiharu Hirose
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Natsuko Okita
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
26
|
Yoshida M, Yuan J, Kihara T, Kimura N, Yamasaki T, Ohkouchi M, Hashikura Y, Isozaki K, Hagiyama M, Ito A, Hirota S. Anti-tumor effect of antibody-drug conjugate targeting cell adhesion molecule 1 on GIST cells representing small intestinal GIST. Exp Mol Pathol 2024; 139:104922. [PMID: 39096891 DOI: 10.1016/j.yexmp.2024.104922] [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: 03/27/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the alimentary tract. The prognosis depends on the primary site, and small intestinal GISTs have a worse prognosis than gastric GISTs. Molecularly targeted drugs to inhibit tyrosine kinase activity of KIT were used for unresectable or recurrent GISTs. However, secondary resistance to the drugs is often acquired, and treatments based on other mechanisms are needed. Previously, we reported that cell adhesion molecule 1 (CADM1) was highly expressed in most of small intestinal GISTs but not in most of gastric GISTs. In the present study, we examined whether the antibody-drug conjugate (ADC) with anti-CADM1 antibody and monomethyl auristatin E (anti-CAD-ADC) shows anti-tumor effect on CADM1-expressing human GIST cells. The ADC adhibited in this study was previously used for CADM1-expressing human mesothelioma cells and showed anti-tumor effect for them in vitro. GIST-T1 cell line of gastric origin which scarcely expresses CADM1 and GIST-T1 cells transfected with CADM1 cDNA (GIST-T1-CAD cells) which highly expresses CADM1 and represents small intestinal GIST were used. In vitro, anti-CAD-ADC showed remarkable cytotoxic activity on GIST-T1-CAD cells, but control ADC did not. Both anti-CAD-ADC and control ADC did not show anti-tumor effect on original GIST-T1 cells. When GIST-T1-CAD cells were subcutaneously injected to the nude mice, intravenous administration of anti-CAD-ADC showed inhibitory effect for tumor enlargement. Tumor of GIST-T1 cells grew even after anti-CAD-ADC injection. When GIST-T1-CAD cells were injected into peritoneal cavity of the SCID mice, intraperitoneal administration of anti-CAD-ADC showed reduction of the peritoneal tumor. On the other hand, peritoneal tumor grew after control ADC administration. Tissue and organ damage due to administration of anti-CAD-ADC was not apparent by macroscopic and histological examinations in mice. These results indicate that anti-CAD-ADC could have apparent anti-tumor effect on CADM1-expressing human GIST cells both in in vitro and in vivo mouse models.
Collapse
Affiliation(s)
- Makoto Yoshida
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Jiayin Yuan
- Department of Pathology, First People's Hospital of Foshan, Foshan City, Guangdong 528000, China
| | - Takako Kihara
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Neinei Kimura
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Takashi Yamasaki
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Mizuka Ohkouchi
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuka Hashikura
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Koji Isozaki
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan
| | - Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Ono-Higashi, Osaka-Sayama, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Ono-Higashi, Osaka-Sayama, Osaka, Japan.
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
27
|
Moura DS, López López D, di Lernia D, Martin-Ruiz M, Lopez-Alvarez M, Ramos R, Merino J, Dopazo J, Lopez-Guerrero J, Mondaza-Hernandez JL, Romero P, Hindi N, Garcia-Foncillas J, Martin-Broto J. Shared germline genomic variants in two patients with double primary gastrointestinal stromal tumours (GISTs). J Med Genet 2024; 61:927-934. [PMID: 39153853 DOI: 10.1136/jmg-2024-110109] [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: 05/07/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Gastrointestinal stromal tumours (GISTs) are prevalent mesenchymal tumours of the gastrointestinal tract, commonly exhibiting structural variations in KIT and PDGFRA genes. While the mutational profiling of somatic tumours is well described, the genes behind the susceptibility to develop GIST are not yet fully discovered. This study explores the genomic landscape of two primary GIST cases, aiming to identify shared germline pathogenic variants and shed light on potential key players in tumourigenesis. METHODS Two patients with distinct genotypically and phenotypically GISTs underwent germline whole genome sequencing. CNV and single nucleotide variant (SNV) analyses were performed. RESULTS Both patients harbouring low-risk GISTs with different mutations (PDGFRA and KIT) shared homozygous germline pathogenic deletions in both CFHR1 and CFHR3 genes. CNV analysis revealed additional shared pathogenic deletions in other genes such as SLC25A24. No particular pathogenic SNV shared by both patients was detected. CONCLUSION Our study provides new insights into germline variants that can be associated with the development of GISTs, namely, CFHR1 and CFHR3 deep deletions. Further functional validation is warranted to elucidate the precise contributions of identified germline mutations in GIST development.
Collapse
Affiliation(s)
- David S Moura
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Daniel López López
- Computational Medicine Platform, Fundación progreso y salud (FPS), Hospital Virgen del Rocío, Seville, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
| | - Davide di Lernia
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Marta Martin-Ruiz
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | | | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | - Jose Merino
- Pathology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Joaquin Dopazo
- Computational Medicine Platform, Fundación progreso y salud (FPS), Hospital Virgen del Rocío, Seville, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS; HUVR, CSIC, US), Sevilla, Spain
| | - Jose Lopez-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | - Jose L Mondaza-Hernandez
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Pablo Romero
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Nadia Hindi
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- General de Villalba University Hospital, Madrid, Spain
| | - Jesus Garcia-Foncillas
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- General de Villalba University Hospital, Madrid, Spain
| |
Collapse
|
28
|
Wang XK, Shen LF, Yang X, Su H, Wu T, Tao PX, Lv HY, Yao TH, Yi L, Gu YH. Two different mutational types of familial gastrointestinal stromal tumors: Two case reports. World J Gastrointest Oncol 2024; 16:4028-4036. [PMID: 39350996 PMCID: PMC11438775 DOI: 10.4251/wjgo.v16.i9.4028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal (GI) tract, and cases of GISTs tend to be of the disseminated type, with a global incidence of 10 to 15 cases/million each year. The rarer familial GISTs, which often represent a population, differ in screening, diagnosis, and treatment. Familial GISTs include primary familial GISTs with predominantly KIT/PDGFRA mutations and wild-type GISTs. However, whether the same genetic family has different phenotypes has not been reported. CASE SUMMARY We report two cases of rare GISTs in the same family: A male patient with the V561D mutation in exon 12 of the PDGFRA gene, who has been taking the targeted drug imatinib since undergoing surgery, and a female patient diagnosed with wild-type GIST, who has been taking imatinib for 3 years since undergoing surgery. The favorable prognosis of these patients during the 7-year follow-up period validates the accuracy of our treatment strategy, and we have refined the entire process of diagnosis and treatment of familial GISTs in order to better manage this rare familial disease. CONCLUSION Different mutation types of familial GISTs in the same family are very rare, thus it is very important to make the correct diagnosis and treatment strategies according to the results of molecular detection for the management of familial GISTs.
Collapse
Affiliation(s)
- Xiao-Ke Wang
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Lu-Fan Shen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Xin Yang
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - He Su
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Tao Wu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Peng-Xian Tao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Hong-Ying Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Tong-Han Yao
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yuan-Hui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
29
|
Boichuk S, Dunaev P, Galembikova A, Valeeva E. Fibroblast Growth Factor 2 (FGF2) Activates Vascular Endothelial Growth Factor (VEGF) Signaling in Gastrointestinal Stromal Tumors (GIST): An Autocrine Mechanism Contributing to Imatinib Mesylate (IM) Resistance. Cancers (Basel) 2024; 16:3103. [PMID: 39272961 PMCID: PMC11394061 DOI: 10.3390/cancers16173103] [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: 07/29/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
We showed previously that the autocrine activation of the FGFR-mediated pathway in GIST lacking secondary KIT mutations was a result of the inhibition of KIT signaling. We show here that the FGF2/FGFR pathway regulates VEGF-A/VEGFR signaling in IM-resistant GIST cells. Indeed, recombinant FGF2 increased the production of VEGF-A by IM-naive and resistant GIST cells. VEGF-A production was also increased in KIT-inhibited GIST, whereas the neutralization of FGF2 by anti-FGF2 mAb attenuated VEGFR signaling. Of note, BGJ 398, pan FGFR inhibitor, effectively and time-dependently inhibited VEGFR signaling in IM-resistant GIST T-1R cells, thereby revealing the regulatory role of the FGFR pathway in VEGFR signaling for this particular GIST cell line. This also resulted in significant synergy between BGJ 398 and VEGFR inhibitors (i.e., sunitinib and regorafenib) by enhancing their pro-apoptotic and anti-proliferative activities. The high potency of the combined use of VEGFR and FGFR inhibitors in IM-resistant GISTs was revealed by the impressive synergy scores observed for regorafenib or sunitinib and BGJ 398. Moreover, FGFR1/2 and VEGFR1/2 were co-localized in IM-resistant GIST T-1R cells, and the direct interaction between the aforementioned RTKs was confirmed by co-immunoprecipitation. In contrast, IM-resistant GIST 430 cells expressed lower basal levels of FGF2 and VEGF-A. Despite the increased expression VEGFR1 and FGFR1/2 in GIST 430 cells, these RTKs were not co-localized and co-immunoprecipitated. Moreover, no synergy between FGFR and VEGFR inhibitors was observed for the IM-resistant GIST 430 cell line. Collectively, the dual targeting of FGFR and VEGFR pathways in IM-resistant GISTs is not limited to the synergistic anti-angiogenic treatment effects. The dual inhibition of FGFR and VEGFR pathways in IM-resistant GISTs potentiates the proapoptotic and anti-proliferative activities of the corresponding RTKi. Mechanistically, the FGF2-induced activation of the FGFR pathway turns on VEGFR signaling via the overproduction of VEGF-A, induces the interaction between FGFR1/2 and VEGFR1, and thereby renders cancer cells highly sensitive to the dual inhibition of the aforementioned RTKs. Thus, our data uncovers the novel mechanism of the cross-talk between the aforementioned RTKs in IM-resistant GISTs lacking secondary KIT mutations and suggests that the dual blockade of FGFR and VEGFR signaling might be an effective treatment strategy for patients with GIST-acquired IM resistance via KIT-independent mechanisms.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
- Department of Radiotherapy and Radiology, Faculty of Surgery, Russian Medical Academy of Continuous Professional Education, Moscow 125993, Russia
- "Biomarker" Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
| | - Elena Valeeva
- Central Research Laboratory, Kazan State Medical University, Kazan 420012, Russia
| |
Collapse
|
30
|
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
|
31
|
Takaki EO, Kiyono K, Obuchi Y, Yamauchi T, Watanabe T, Matsumoto H, Karimine M, Kuniyoshi Y, Nishikori S, Yokoyama F, Nishimori H, Nabeshima H, Nakamura K. A PDE3A-SLFN12 Molecular Glue Exhibits Significant Antitumor Activity in TKI-Resistant Gastrointestinal Stromal Tumors. Clin Cancer Res 2024; 30:3603-3621. [PMID: 38864850 PMCID: PMC11325149 DOI: 10.1158/1078-0432.ccr-24-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/09/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE Gastrointestinal stromal tumor (GIST), the most common mesenchymal tumor with KIT or PDGFRA driver mutations, is typically treated with tyrosine kinase inhibitors (TKI). However, resistance to TKIs due to secondary mutations is a common challenge in advanced GISTs. In addition, there are currently no effective therapies for several other molecular subtypes, such as succinate dehydrogenase-deficient GISTs. Therefore, novel therapeutic strategies are needed. EXPERIMENTAL DESIGN To address this need, we tested the efficacy of a novel non-TKI compound, OPB-171775, using patient-derived xenograft models of GISTs. In parallel, we sought to elucidate the mechanism of action of the compound. RESULTS Our study revealed that OPB-171775 exhibited significant efficacy against GISTs regardless of their KIT mutation status by inducing complex formation between phosphodiesterase 3A (PDE3A) and Schlafen family member 12 (SLFN12), which are highly expressed in GISTs, leading to SLFN12 RNase-mediated cell death. Furthermore, we identified the activation of general control non-derepressible 2 and its downstream response as an effector pathway of SLFN12 in mediating anticancer activity and revealed potential pharmacodynamic markers. CONCLUSIONS These findings suggest that OPB-171775, with its significant efficacy, could potentially serve as a novel and effective treatment option for advanced GISTs, particularly those resistant to TKIs.
Collapse
Affiliation(s)
- Emiri O. Takaki
- Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Kunihiko Kiyono
- Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd., Otsu, Japan.
| | - Yutaka Obuchi
- Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Takeshi Yamauchi
- Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd., Otsu, Japan.
| | - Takashi Watanabe
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Hideki Matsumoto
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Miho Karimine
- Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Yuki Kuniyoshi
- Office of Bioinformatics, Department of Drug Discovery Strategy, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Shingo Nishikori
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Fumiharu Yokoyama
- Fujii Memorial Research Institute, Otsuka Pharmaceutical Co., Ltd., Otsu, Japan.
| | - Hikaru Nishimori
- Department of Drug Modality Development, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Hiroshi Nabeshima
- Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| | - Kazuhide Nakamura
- Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh, Japan.
| |
Collapse
|
32
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [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: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
33
|
Alvarez CS, Piazuelo MB, Fleitas-Kanonnikoff T, Ruhl J, Pérez-Fidalgo JA, Camargo MC. Incidence and Survival Outcomes of Gastrointestinal Stromal Tumors. JAMA Netw Open 2024; 7:e2428828. [PMID: 39158910 PMCID: PMC11333982 DOI: 10.1001/jamanetworkopen.2024.28828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/23/2024] [Indexed: 08/20/2024] Open
Abstract
Importance The incidence of gastrointestinal stromal tumors (GISTs) increased after the implementation of GIST-specific histology coding in 2001, but updated data on trends and survival are lacking. Objective To examine the evolving epidemiology of GISTs in major organ sites. Design, Setting, and Participants This descriptive, population-based cohort study used nationally representative data from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) Program, including the SEER-22 and SEER-17 registries. Data were from evaluated patients aged 20 years or older with GISTs diagnosed between January 1, 2000, and December 31, 2019. Analyses were last updated on October 29, 2023. Main Outcomes and Measures Organ site-specific trends in age-standardized incidence rates and annual percent changes (APCs) in rates were estimated by race and ethnicity and, when possible, by sex, age, and primary indicator. Multivariable Cox proportional hazards regression models were used to examine racial and ethnic differences in overall and GIST-specific survival by site. Results The SEER-22 and SEER-17 datasets contained 23 001 and 12 109 case patients with GISTs, respectively. Patients in the SEER-22 registry had a mean (SD) age of 64 (13) years and 51.3% were men. With regard to race and ethnicity, 9.7% of patients were Asian or Pacific Islander, 12.3% were Hispanic, 19.6% were non-Hispanic Black, and 57.7% were non-Hispanic White. Overall incidence rates of GISTs in the SEER-22 cohort increased substantially over time for all organ sites but the colon (APCs: esophagus, 7.3% [95% CI, 4.4% to 10.2%]; gastric, 5.1% [95% CI, 4.2% to 6.1%]; small intestine, 2.7% [95% CI, 1.8% to 3.7%]; colon, -0.2% [95% CI, -1.3% to 0.9%]; and rectum, 1.9% [95% CI, 0.1% to 3.8%]). There were similar increasing trends by age groups (<50 vs ≥50 years), sex, race and ethnicity, and primary indicator for gastric and small intestine GISTs. Increases were mainly restricted to localized stage disease. Patients in the SEER-17 cohort had a mean (SD) age of 64 (14) years and 51.9% were men. With regard to race and ethnicity, 13.3% of patients were Asian or Pacific Islander, 11.6% were Hispanic, 17.8% were non-Hispanic Black, and 56.6% were non-Hispanic White. Non-Hispanic Black individuals had higher overall mortality for esophageal (adjusted hazard ratio [HR], 6.4 [95% CI, 2.0 to 20.3]) and gastric (adjusted HR, 1.4 [95% CI, 1.2 to 1.5]) GISTs compared with non-Hispanic White individuals. Asian or Pacific Islander individuals also had higher overall mortality for esophageal GISTs (adjusted HR, 5.6 [95% CI, 1.5 to 20.2]). Results were similar for GIST-specific survival. Conclusions and Relevance In this cohort study using SEER data, the incidence of GISTs in major organ sites increased in the last 2 decades among several population groups. These findings suggest that additional studies are warranted to identify risk factors, because histologic reclassification and higher availability of endoscopy and imaging do not fully explain these unfavorable incidence trends. Prevention efforts are needed to reduce the substantial survival disparities among racial and ethnic minoritized populations.
Collapse
Affiliation(s)
- Christian S. Alvarez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tania Fleitas-Kanonnikoff
- Department of Medical Oncology, Instituto de Investigación Sanitaria (INCLIVA) Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Jennifer Ruhl
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland
| | - J. Alejandro Pérez-Fidalgo
- Medical Oncology Department, University Hospital of Valencia, INCLIVA Biomedical Research Institute, Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, Valencia, Spain
| | - M. Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
34
|
Hashimoto T, Nakamura Y, Komatsu Y, Yuki S, Takahashi N, Okano N, Hirano H, Ohtsubo K, Ohta T, Oki E, Nishina T, Yasui H, Kawakami H, Esaki T, Machida N, Doi A, Boku S, Kudo T, Yamamoto Y, Kanazawa A, Denda T, Goto M, Iida N, Ozaki H, Shibuki T, Imai M, Fujisawa T, Bando H, Naito Y, Yoshino T. Different efficacy of tyrosine kinase inhibitors by KIT and PGFRA mutations identified in circulating tumor DNA for the treatment of refractory gastrointestinal stromal tumors. BJC REPORTS 2024; 2:54. [PMID: 39516322 PMCID: PMC11523999 DOI: 10.1038/s44276-024-00073-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND While advanced gastrointestinal stromal tumors (GISTs) are primarily treated with tyrosine kinase inhibitors (TKIs), acquired resistance from specific mutations in KIT or PDGFRA frequently occurs. We aimed to assess the utility of circulating tumor DNA (ctDNA) as a modality of therapeutic decision-making in advanced GIST. METHODS We conducted a pooled analysis of SCRUM-Japan studies for advanced GIST patients. We compared patient characteristics analyzed with tissue and blood samples, assessed gene alteration profiles, and evaluated prognostic implications from ctDNA status. RESULTS In 133 patients, tissue and blood samples were analyzed for 89 and 44 patients, respectively. ctDNA was detected in 72.7% of cases; no prior treatment or progressive disease was significantly associated with ctDNA-positivity. ctDNA-positive patients had significantly shorter progression-free survival compared with ctDNA-negative patients (hazard ratio = 3.92; P = 0.007). ctDNA genotyping revealed a complex landscape of gene alterations, characterized by multi-exonic mutations in KIT, compared with tissue-based analysis. Patients who received TKIs matched to the identified KIT mutation in ctDNA demonstrated significantly longer PFS than those with unmatched treatment (median, 8.23 vs. 2.43 months; P < 0.001). CONCLUSIONS ctDNA-based analysis facilitates assessment of disease status and genomic profiles, thus potentially assisting in identifying optimal therapeutic strategies for advanced GIST patients.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan.
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Hidekazu Hirano
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tsukiji, Japan
| | - Koushiro Ohtsubo
- Department of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Hyogo, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Hospital, Osaka, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Nozomu Machida
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Ayako Doi
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University, Osaka, Japan
| | - Toshihiro Kudo
- Department of Medical Oncology, Osaka International Cancer Institute Osaka Prefectural Hospital Organization, Osaka, Japan
| | - Yoshiyuki Yamamoto
- Department of Gastroenterology, University of Tsukuba Hospital, Ibaraki, Japan
| | - Akiyoshi Kanazawa
- Department of Surgery Shimane Prefectural Central Hospital, Shimane, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Naoko Iida
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroshi Ozaki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
35
|
Yu G, Liu R, Li J, Zhao G, Wang Y. The immunotherapy in gastrointestinal stromal tumors. Heliyon 2024; 10:e33617. [PMID: 39040340 PMCID: PMC11260923 DOI: 10.1016/j.heliyon.2024.e33617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Using Tyrosine Kinase Inhibitors (TKIs) for gastrointestinal stromal tumors (GIST) has significantly reduced the risk of recurrence and prolonged survival. Immunotherapy has demonstrated efficacy in multiple solid tumors, but its effectiveness in GIST remains uncertain. Although early clinical studies indicate good tolerability of immunotherapy in patients, the efficacy is not as desired. Therefore, identifying the subset of GIST patients who benefit from immunotherapy and coordinating the relationship between immunotherapy and TKI treatment are crucial issues to be explored. In this review, we aims to provide a retrospective analysis of relevant literature and find that GIST patients exhibit a rich presence of tumor-infiltrating immune cells, which play critical roles in the immune surveillance and evasion processes of tumors. This review incorporates a selection of 48 articles published between 2002 and 2023, sourced from PubMed, EBSCO, and Google Scholar databases.
Collapse
Affiliation(s)
- Guilin Yu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Ruibin Liu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- Department of Clinical Integration of Traditional Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiayao Li
- Liaoning Normal University Haihua College,Liaoning, China
| | - Guohua Zhao
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yue Wang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
36
|
Yu Y, Yu M, Luo L, Zhang Z, Zeng H, Chen Y, Lin Z, Chen M, Wang W. Molecular characteristics and immune microenvironment of gastrointestinal stromal tumours: targets for therapeutic strategies. Front Oncol 2024; 14:1405727. [PMID: 39070147 PMCID: PMC11272528 DOI: 10.3389/fonc.2024.1405727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours, arising mainly from the interstitial cells of Cajal (ICCs) of the gastrointestinal tract. As radiotherapy and chemotherapy are generally ineffective for GISTs, the current primary treatment is surgical resection. However, surgical resection is not choice for most patients. Therefore, new therapeutic strategies are urgently needed. Targeted therapy, represented by tyrosine kinase inhibitors (TKIs), and immunotherapy, represented by immune checkpoint inhibitor therapies and chimeric antigen receptor T-cell immunotherapy (CAR-T), offer new therapeutic options in GISTs and have shown promising treatment responses. In this review, we summarize the molecular classification and immune microenvironment of GISTs and discuss the corresponding targeted therapy and immunotherapy options. This updated knowledge may provide more options for future therapeutic strategies and applications in GISTs.
Collapse
Affiliation(s)
- Yang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengdie Yu
- Guangzhou KingMed Diagnostics Group Co., Ltd., Guangzhou, Guangdong, China
| | - Lijie Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Zijing Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Haiping Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Zeyu Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengnan Chen
- Department of Thyroid and Breast Surgery, Baiyun Hospital, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Lu Q, Jiang Y. Diagnosis of Unresectable Primary Hepatic Gastrointestinal Stromal Tumor by Positron Emission Tomography/Computed Tomography Combined With Ultrasound-Guided Fine Needle Aspiration Biopsy. Gastroenterol Nurs 2024; 47:303-307. [PMID: 39087997 DOI: 10.1097/sga.0000000000000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/31/2023] [Indexed: 08/02/2024] Open
Affiliation(s)
- Qingqing Lu
- Qingqing Lu, MD, is from the Department of Gastroenterology, Hangzhou Red Cross Hospital/Integrated Traditional Chinese and Western Medicine Hospital of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- Yang Jiang, MD, is from the Department of Nuclear Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Jiang
- Qingqing Lu, MD, is from the Department of Gastroenterology, Hangzhou Red Cross Hospital/Integrated Traditional Chinese and Western Medicine Hospital of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- Yang Jiang, MD, is from the Department of Nuclear Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Pham AT, Pham AT, Truong CM, Nguyen TH, Trinh PH. Primary gastrointestinal stromal tumor of the liver: a case report. Ann Med Surg (Lond) 2024; 86:4284-4290. [PMID: 38989195 PMCID: PMC11230766 DOI: 10.1097/ms9.0000000000002228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction and importance Primary gastrointestinal stromal tumors of the liver are exceedingly rare entities, presenting diagnostic and therapeutic challenges. The authors present a case of a 64-year-old male with a primary gastrointestinal stromal tumor (GIST) of the liver, emphasizing the importance of comprehensive diagnostic evaluation and multidisciplinary management in such uncommon cases. Case presentation The patient presented with persistent hypochondriac pain, leading to the discovery of a hepatic mass. Diagnostic work-ups, including imaging studies and biopsy, confirmed the diagnosis of primary GIST in the liver. Following thorough multidisciplinary consultation, the patient underwent right anterior segmentectomy of the liver, performed by our experienced surgeon. Postoperative pathology confirmed the diagnosis of GIST, and the patient was advised to use adjuvant imatinib. Clinical discussion Primary GISTs of the liver pose diagnostic challenges due to their rarity and varied clinical presentations. Imaging modalities, immunohistochemistry, and molecular genotyping are crucial in accurate diagnosis and treatment planning. Surgical resection remains the cornerstone of treatment for localized GISTs, with adjuvant therapy considered based on recurrence risk factors and molecular characteristics. Conclusion This case highlights the need for multidisciplinary consultation in managing primary GISTs of the liver. Accurate diagnosis, surgical expertise, and personalized adjuvant therapy are crucial for better patient outcomes. Further research is necessary to enhance our understanding of prognostic factors and treatment strategies for these rare tumors.
Collapse
Affiliation(s)
| | - Anh The Pham
- Hepatobiliary and Pancreatic Surgery, Vietnam National Cancer Hospital, 30 Cau Buou, Tan Trieu, Hanoi, Vietnam
| | - Cuong Manh Truong
- Hepatobiliary and Pancreatic Surgery, Vietnam National Cancer Hospital, 30 Cau Buou, Tan Trieu, Hanoi, Vietnam
| | | | - Phuong Huy Trinh
- Hepatobiliary and Pancreatic Surgery, Vietnam National Cancer Hospital, 30 Cau Buou, Tan Trieu, Hanoi, Vietnam
| |
Collapse
|
39
|
Hirota S, Tateishi U, Nakamoto Y, Yamamoto H, Sakurai S, Kikuchi H, Kanda T, Kurokawa Y, Cho H, Nishida T, Sawaki A, Ozaka M, Komatsu Y, Naito Y, Honma Y, Takahashi F, Hashimoto H, Udo M, Araki M, Nishidate S. English version of Japanese Clinical Practice Guidelines 2022 for gastrointestinal stromal tumor (GIST) issued by the Japan Society of Clinical Oncology. Int J Clin Oncol 2024; 29:647-680. [PMID: 38609732 PMCID: PMC11130037 DOI: 10.1007/s10147-024-02488-1] [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: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 04/14/2024]
Abstract
The Japan Society of Clinical Oncology Clinical Practice Guidelines 2022 for gastrointestinal stromal tumor (GIST) have been published in accordance with the Minds Manual for Guideline Development 2014 and 2017. A specialized team independent of the working group for the revision performed a systematic review. Since GIST is a rare type of tumor, clinical evidence is not sufficient to answer several clinical and background questions. Thus, in these guidelines, we considered that consensus among the experts who manage GIST, the balance between benefits and harms, patients' wishes, medical economic perspective, etc. are important considerations in addition to the evidence. Although guidelines for the treatment of GIST have also been published by the National Comprehensive Cancer Network (NCCN) and the European Society for Medical Oncology (ESMO), there are some differences between the treatments proposed in those guidelines and the treatments in the present guidelines because of the differences in health insurance systems among countries.
Collapse
Affiliation(s)
- Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Japan.
| | - Ukihide Tateishi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetaka Yamamoto
- Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shinji Sakurai
- Department of Diagnostic Pathology, Japan Community Healthcare Organization Gunma Central Hospital, Maebashi, Japan
| | - Hirotoshi Kikuchi
- Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuo Kanda
- Department of Gastroenterology, Southern TOHOKU General Hospital, Koriyama, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Toshirou Nishida
- Department of Surgery, Japan Community Healthcare Organization Osaka Hospital, Osaka, Japan
| | - Akira Sawaki
- Department of Medical Oncology, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Fumiaki Takahashi
- Department of Information Science, Iwate Medical University, Morioka, Japan
| | | | - Midori Udo
- Nursing Department, Osaka Police Hospital, Osaka, Japan
| | - Minako Araki
- Association of Chubu GIST Patients and Their Families, Nagoya, Japan
| | | |
Collapse
|
40
|
Chai Y, Lin B, Zhong J, Wu X, Lin X, Ge X, Jiang J, Liang Z, Liu S, Gu C. Long-term outcomes of endoscopic therapy versus surgical resection for 2-5 cm gastric gastrointestinal stromal tumors: A population-based comparative study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108262. [PMID: 38531231 DOI: 10.1016/j.ejso.2024.108262] [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: 12/05/2023] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Endoscopic therapy (ET) of gastrointestinal stromal tumors (GIST) has become a viable treatment. We intended to compare long-term outcomes of ET versus surgical resection for 2-5 cm GIST using the Surveillance, Epidemiology, and End Results (SEER) database. METHODS A multicenter retrospective study was conducted to compare the long-term outcomes of patients treated with ET and surgical resection for GIST. The multivariate Cox proportional hazards models were used to identify predictors for patients survival. To balance the clinicopathologic characteristics, a 1:1 propensity score matching (PSM) was utilized. RESULTS A total of 749 patients with 2-5 cm GIST were enrolled, of whom 113 accepted ET and 636 underwent surgical resection. Before PSM, there was no significant difference in long-term outcomes between ET and surgical resection (5-year overall survival (OS): 93.5% vs. 91.6%, P=0.374; 5-year cancer-specific survival (CSS): 99.1% vs. 96.5%, P=0.546; 10-year OS: 71.1% vs. 78.2%, P=0.374; 10-year CSS: 93.6% vs. 92.7%, P=0.546). After adjusting for the relevant variables using the multivariable Cox proportional hazards models, we observed that the ET and surgical resection groups were similar in OS (HR 0.726, 95%CI 0.457-1.153, P=0.175) and CSS (HR 1.286, 95%CI 0.474-3.488, P=0.621). After PSM, the long-term OS and CSS of patients with 2-5 cm GIST after ET and surgical resection were comparable. CONCLUSIONS We found that the long-term survival of patients with 2-5 cm gastric GIST after ET and surgical resection were comparable. Further high-quality studies are needed to confirm the role of ET in 2-5 cm GIST.
Collapse
Affiliation(s)
- Yixia Chai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bitao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Zhong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaosheng Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyue Ge
- Department of General Practice, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayi Jiang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenye Liang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University · Zhao Qing Hospital, Zhaoqing 526000, China.
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Chuncai Gu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
41
|
Singh A, Chitti B, Aguiar C, Wernicke AG, Devoe CE, Rahman H, Sison C, Parashar B. Comparing gastrointestinal stromal tumor outcomes between geriatric and non-geriatric patients: A population-based analysis. World J Surg 2024; 48:1424-1432. [PMID: 38647223 DOI: 10.1002/wjs.12170] [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: 01/01/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Gastrointestinal Stromal Tumors (GISTs) are the most common mesenchymal tumors of the GI tract. SEER is an extensive cancer database which proves useful in analyzing population trends. This analysis investigated GIST outcomes between geriatric & non-geriatric patients. METHODS SEER*STAT 8.4.0.1 was used to extract relevant GIST data from 2000 to 2019. Geriatric age was defined as ≥70 years. Variables included age, sex, surgery, cancer-specific death, and overall survival. Statistical tests included univariate analysis using KM survival estimate (95% confidence interval) to calculate 5-year survival (5YS). Log-Rank tests determined statistical significance. Multivariable Cox's PH regression estimated the geriatric hazard death ratio adjusted for sex, stage, and surgery. RESULTS The number of patients included was 13,579, yielding overall 5YS of 68.6% (95% CI 67.7-69.5). Cancer-specific death was 39.11% in 2000 & 3.33% in 2019. Non-geriatric & geriatric patient data yielded 5YS of 77.4% (76.4%-78.3%) and 53.3% (51.7%-54.8%) respectively (p < 0.0001). For no surgery/surgery, younger patient data yielded 5YS of 48.7% (45.8%-51.4%) and 83.7% (82.7%-84.7%) respectively (p < 0.0001); geriatric data yielded 5YS of 29.3% (26.5%-32.1%) and 62.8% (60.8%-64.6%) respectively (p < 0.0001). Multivariable analysis yielded a geriatric hazard death of 2.56 (2.42-2.70) (p < 0.0001). CONCLUSIONS Cancer-specific death decreased since 2000, indicating an improvement in survival & treatment methods. Observed lower survival rates overall in the geriatric group. Surgery appeared to enhance survival rates in both groups, suggesting that surgery is an important factor in GIST survival regardless of age. Large prospective studies will help define clinical management for geriatric patients.
Collapse
Affiliation(s)
- Abhiram Singh
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California, USA
| | - Bhargava Chitti
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
| | | | | | - Craig E Devoe
- Department of Medical Oncology, Northwell, New Hyde Park, New York, USA
| | - Husneara Rahman
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, USA
| | - Cristina Sison
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, USA
| | - Bhupesh Parashar
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
| |
Collapse
|
42
|
Mühlenberg T, Falkenhorst J, Schulz T, Fletcher BS, Teuber A, Krzeciesa D, Klooster I, Lundberg M, Wilson L, Lategahn J, von Mehren M, Grunewald S, Tüns AI, Wardelmann E, Sicklick JK, Brahmi M, Serrano C, Schildhaus HU, Sievers S, Treckmann J, Heinrich MC, Raut CP, Ou WB, Marino-Enriquez A, George S, Rauh D, Fletcher JA, Bauer S. KIT ATP-Binding Pocket/Activation Loop Mutations in GI Stromal Tumor: Emerging Mechanisms of Kinase Inhibitor Escape. J Clin Oncol 2024; 42:1439-1449. [PMID: 38408285 PMCID: PMC11095889 DOI: 10.1200/jco.23.01197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 02/28/2024] Open
Abstract
PURPOSE Imatinib resistance in GI stromal tumors (GISTs) is primarily caused by secondary KIT mutations, and clonal heterogeneity of these secondary mutations represents a major treatment obstacle. KIT inhibitors used after imatinib have clinical activity, albeit with limited benefit. Ripretinib is a potent inhibitor of secondary KIT mutations in the activation loop (AL). However, clinical benefit in fourth line remains limited and the molecular mechanisms of ripretinib resistance are largely unknown. PATIENTS AND METHODS Progressing lesions of 25 patients with GISTs refractory to ripretinib were sequenced for KIT resistance mutations. Resistant genotypes were validated and characterized using novel cell line models and in silico modeling. RESULTS GISTs progressing on ripretinib were enriched for secondary mutations in the ATP-binding pocket (AP), which frequently occur in cis with preexisting AL mutations, resulting in highly resistant AP/AL genotypes. AP/AL mutations were rarely observed in a cohort of progressing GIST samples from the preripretinib era but represented 50% of secondary KIT mutations in patients with tumors resistant to ripretinib. In GIST cell lines harboring secondary KIT AL mutations, the sole genomic escape mechanisms during ripretinib drug selection were AP/AL mutations. Ripretinib and sunitinib synergize against mixed clones with secondary AP or AL mutants but do not suppress clones with AP/AL genotypes. CONCLUSION Our findings underscore that KIT remains the central oncogenic driver even in late lines of GIST therapy. KIT-inhibitor combinations may suppress resistance because of secondary KIT mutations. However, the emergence of KIT AP/AL mutations after ripretinib treatment calls for new strategies in the development of next-generation KIT inhibitors.
Collapse
Affiliation(s)
- Thomas Mühlenberg
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Johanna Falkenhorst
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Tom Schulz
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für Integrierte Wirkstoffforschung (ZIW), Dortmund, Germany
| | - Benjamin S. Fletcher
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alina Teuber
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für Integrierte Wirkstoffforschung (ZIW), Dortmund, Germany
| | - Dawid Krzeciesa
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Isabella Klooster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Meijun Lundberg
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lydia Wilson
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Jonas Lategahn
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für Integrierte Wirkstoffforschung (ZIW), Dortmund, Germany
| | - Margaret von Mehren
- Department of Hematology and Oncology, Fox Chase Cancer Center, Temple Health System, University, Philadelphia, PA
| | - Susanne Grunewald
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alicia Isabell Tüns
- Laboratory of Molecular Oncology, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Eva Wardelmann
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Jason K. Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, CA
- Department of Pharmacology, Moores Cancer Center, University of California San Diego, San Diego, CA
| | - Mehdi Brahmi
- Centre Leon Berard, Medical Oncology, Lyon, France
| | - César Serrano
- Sarcoma Translational Research Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Hans-Ulrich Schildhaus
- University Hospital Essen, Institute of Pathology, Essen, Germany
- Current affiliation: Discovery Life Sciences Biomarker Services & Institute of Pathology Nodhessen, Kassel, Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Jürgen Treckmann
- University of Duisburg-Essen, Medical School, Department of Visceral and Transplantation Surgery, Essen, Germany
| | - Michael C. Heinrich
- Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, OR
| | - Chandrajit P. Raut
- Department of Surgery, Brigham and Women's Hospital, Boston, MA
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Wen-Bin Ou
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Adrian Marino-Enriquez
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Suzanne George
- Dana-Farber Cancer Institute, Medical Oncology, Boston, MA
| | - Daniel Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für Integrierte Wirkstoffforschung (ZIW), Dortmund, Germany
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Sebastian Bauer
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
- DKTK partner site Essen, German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
43
|
Liu Z, Tao Q, Wu Y, Zeng C, Chen Y. Comparable long‑term survival outcomes of endoscopic treatment versus surgical treatment for gastrointestinal stromal tumors with a diameter of 5-10 cm. Sci Rep 2024; 14:8513. [PMID: 38609414 PMCID: PMC11014986 DOI: 10.1038/s41598-024-58802-4] [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: 01/15/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Currently, endoscopic treatment for small gastrointestinal stromal tumors (GIST) has been widely accepted. However, for tumors larger than 5 cm, endoscopic treatment has not been recognized by national guidelines as the standard therapy due to concerns about safety and adverse tumor outcomes. Therefore, this study compares the long-term survival outcomes of endoscopic treatment and surgical treatment for GIST in the range of 5-10 cm. We selected patients with GIST from the Surveillance, Epidemiology, and End Results (SEER) database between 2004 and 2015. Kaplan-Meier analysis and the log-rank test were employed to compare the long-term survival outcomes between endoscopic treatment and surgical treatment. A multivariate Cox proportional hazards model was used for analysis to identify risk factors influencing patient prognosis. To balance baseline data, we performed 1:1 propensity score matching (PSM). A total of 1223 GIST patients were included, with 144 patients (11.8%) received endoscopic treatment and 1079 patients (88.2%) received surgical treatment. Before PSM, there was no significant difference in the long-term survival rates between the two groups [5-year OS (86.5% vs. 83.5%, P = 0.42), 10-year OS (70.4% vs. 66.7%, P = 0.42)]. After adjusting for covariates, we found that the overall survival (HR = 1.26, 95% CI 0.89-1.77, P = 0.19) and cancer-specific survival (HR = 1.69, 95% CI 0.99-2.89, P = 0.053) risks were comparable between the endoscopic treatment group and the surgical treatment group. In the analysis after PSM, there was no significant difference between the endoscopic treatment group and the surgical treatment group. Our study found that for GIST patients with tumor sizes between 5 and 10 cm, the long-term OS and CSS outcomes were similar between the endoscopic treatment group and the surgical treatment group.
Collapse
Affiliation(s)
- Zide Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qing Tao
- Department of Gastroenterology, Digestive Disease Hospital, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yonghui Wu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chunyan Zeng
- Department of Gastroenterology, Digestive Disease Hospital, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China.
| | - Youxiang Chen
- Department of Gastroenterology, Digestive Disease Hospital, The First Affliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China.
| |
Collapse
|
44
|
Ewongwo A, Hui C, Moding EJ. Opportunity in Complexity: Harnessing Molecular Biomarkers and Liquid Biopsies for Personalized Sarcoma Care. Semin Radiat Oncol 2024; 34:195-206. [PMID: 38508784 DOI: 10.1016/j.semradonc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Due to their rarity and complexity, sarcomas represent a substantial therapeutic challenge. However, the incredible diversity within and across sarcoma subtypes presents an opportunity for personalized care to maximize efficacy and limit toxicity. A deeper understanding of the molecular alterations that drive sarcoma development and treatment response has paved the way for molecular biomarkers to shape sarcoma treatment. Genetic, transcriptomic, and protein biomarkers have become critical tools for diagnosis, prognostication, and treatment selection in patients with sarcomas. In the future, emerging biomarkers like circulating tumor DNA analysis offer the potential to improve early detection, monitoring response to treatment, and identifying mechanisms of resistance to personalize sarcoma treatment. Here, we review the current state of molecular biomarkers for sarcomas and highlight opportunities and challenges for the implementation of new technologies in the future.
Collapse
Affiliation(s)
- Agnes Ewongwo
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Caressa Hui
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA.; Stanford Cancer Institute, Stanford University, Stanford, CA..
| |
Collapse
|
45
|
Liu Z, Gao J, Zeng C, Chen Y. Development and validation of a preoperative risk nomogram prediction model for gastric gastrointestinal stromal tumors. Surg Endosc 2024; 38:1933-1943. [PMID: 38334780 DOI: 10.1007/s00464-024-10674-5] [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: 08/07/2023] [Accepted: 12/30/2023] [Indexed: 02/10/2024]
Abstract
BACKGROUND AND STUDY AIMS Gastrointestinal stromal tumors (GIST) carry a potential risk of malignancy, and the treatment of GIST varies for different risk levels. However, there is no systematic preoperative assessment protocol to predict the malignant potential of GIST. The aim of this study was to develop a reliable and clinically applicable preoperative nomogram prediction model to predict the malignant potential of gastric GIST. PATIENTS AND METHODS Patients with a pathological diagnosis of gastric GIST from January 2015 to December 2021 were screened retrospectively. Univariate and multivariate logistic analyses were used to identify independent risk factors for gastric GIST with high malignancy potential. Based on these independent risk factors, a nomogram model predicting the malignant potential of gastric GIST was developed and the model was validated in the validation group. RESULTS A total of 494 gastric GIST patients were included in this study and allocated to a development group (n = 345) and a validation group (n = 149). In the development group, multivariate logistic regression analysis revealed that tumor size, tumor ulceration, CT growth pattern and monocyte-to- lymphocyte ratio (MLR) were independent risk factors for gastric GIST with high malignancy potential. The AUC of the model were 0.932 (95% CI 0.890-0.974) and 0.922 (95% CI 0.868-0.977) in the development and validation groups, respectively. The best cutoff value for the development group was 0.184, and the sensitivity and specificity at this value were 0.895 and 0.875, respectively. The calibration curves indicated good agreement between predicted and actual observed outcomes, while the DCA indicated that the nomogram model had clinical application. CONCLUSIONS Tumor size, tumor ulceration, CT growth pattern and MLR are independent risk factors for high malignancy potential gastric GIST, and a nomogram model developed based on these factors has a high ability to predict the malignant potential of gastric GIST.
Collapse
Affiliation(s)
- Zide Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Jiaxin Gao
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Chunyan Zeng
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China.
| | - Youxiang Chen
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi, China.
| |
Collapse
|
46
|
Guglielmo A, Zengarini C, Agostinelli C, Motta G, Sabattini E, Pileri A. The Role of Cytokines in Cutaneous T Cell Lymphoma: A Focus on the State of the Art and Possible Therapeutic Targets. Cells 2024; 13:584. [PMID: 38607023 PMCID: PMC11012008 DOI: 10.3390/cells13070584] [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: 12/06/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Cutaneous T cell lymphomas (CTCLs), encompassing mycosis fungoides (MF) and Sézary syndrome (SS), present a complex landscape influenced by cytokines and cellular responses. In this work, the intricate relationship between these inflammatory proteins and disease pathogenesis is examined, focusing on what is known at the clinical and therapeutic levels regarding the most well-known inflammatory mediators. An in-depth look is given to their possible alterations caused by novel immunomodulatory drugs and how they may alter disease progression. From this narrative review of the actual scientific landscape, Interferon-gamma (IFN-γ) emerges as a central player, demonstrating a dual role in both promoting and inhibiting cancer immunity, but the work navigates through all the major interleukins known in inflammatory environments. Immunotherapeutic perspectives are elucidated, highlighting the crucial role of the cutaneous microenvironment in shaping dysfunctional cell trafficking, antitumor immunity, and angiogenesis in MF, showcasing advancements in understanding and targeting the immune phenotype in CTCL. In summary, this manuscript aims to comprehensively explore the multifaceted aspects of CTCL, from the immunopathogenesis and cytokine dynamics centred around TNF-α and IFN-γ to evolving therapeutic modalities. Including all the major known and studied cytokines in this analysis broadens our understanding of the intricate interplay influencing CTCL, paving the way for improved management of this complex lymphoma.
Collapse
Affiliation(s)
- Alba Guglielmo
- Institute of Dermatology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
| | - Corrado Zengarini
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Agostinelli
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giovanna Motta
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elena Sabattini
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Alessandro Pileri
- Dipartimento di Scienze Mediche e Chirurgiche, University of Bologna, 40138 Bologna, Italy
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
47
|
Doi T, Yamamoto N, Ohkubo S. Pimitespib for the treatment of advanced gastrointestinal stromal tumors and other tumors. Future Oncol 2024; 20:507-519. [PMID: 38050698 DOI: 10.2217/fon-2022-1172] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023] Open
Abstract
Pimitespib (TAS-116) is the first heat shock protein 90 (HSP90) inhibitor approved in Japan, and it is indicated for the treatment of gastrointestinal stromal tumors (GIST) that have progressed after treatment with imatinib, sunitinib and regorafenib. This review describes the preclinical and clinical research with pimitespib, including its mechanism of action, pharmacokinetics, clinical antitumour activity and safety. In a phase III study, pimitespib significantly prolonged progression-free survival compared with placebo (median 2.8 vs 1.4 months; hazard ratio 0.51; 95% CI 0.30-0.87; p = 0.006). Common treatment-related adverse events were diarrhoea, decreased appetite, increase in serum creatinine, malaise, nausea and eye disorders. The efficacy and safety of pimitespib are being investigated in other tumour types and in combination with other anticancer therapies.
Collapse
Affiliation(s)
- Toshihiko Doi
- Department of Experimental Therapeutics, National Cancer Centre Hospital East, Kashiwa, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Centre Hospital, Tokyo, Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Ibaraki, Japan
| |
Collapse
|
48
|
Nowak KM, Chetty R. Predictive and prognostic biomarkers in gastrointestinal tract tumours. Pathology 2024; 56:205-213. [PMID: 38238239 DOI: 10.1016/j.pathol.2023.12.412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 02/18/2024]
Abstract
Tumours of the gastrointestinal tract represent nearly a quarter of all newly diagnosed tumours diagnosed in 2019. Various treatment modalities for gastrointestinal cancers exist, some of which may be guided by biomarkers. Biomarkers act as gauges of either normal or pathogenic processes or responses to an exposure or intervention. They come in many forms. This review explores established and potential molecular/immunohistochemical (IHC) predictive and prognostic biomarkers of the gastrointestinal tract.
Collapse
Affiliation(s)
- Klaudia M Nowak
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| | | |
Collapse
|
49
|
Cui Z, Sun H, Gao Z, Li C, Xiao T, Bian Y, Liu Z, Gu T, Zhang J, Li T, Zhou Q, He Z, Li B, Li F, Xu Z, Xu H. TRIM21/USP15 balances ACSL4 stability and the imatinib resistance of gastrointestinal stromal tumors. Br J Cancer 2024; 130:526-541. [PMID: 38182686 PMCID: PMC10876985 DOI: 10.1038/s41416-023-02562-x] [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: 06/20/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Imatinib has become an exceptionally effective targeted drug for treating gastrointestinal stromal tumors (GISTs). Despite its efficacy, the resistance to imatinib is common in GIST patients, posing a significant challenge to the effective treatment. METHODS The expression profiling of TRIM21, USP15, and ACSL4 in GIST patients was evaluated using Western blot and immunohistochemistry. To silence gene expression, shRNA was utilized. Biological function of TRIM21, USP15, and ACSL4 was examined through various methods, including resistance index calculation, colony formation, shRNA interference, and xenograft mouse model. The molecular mechanism of TRIM21 and USP15 in GIST was determined by conducting Western blot, co-immunoprecipitation, and quantitative real-time PCR (qPCR) analyses. RESULTS Here we demonstrated that downregulation of ACSL4 is associated with imatinib (IM) resistance in GIST. Moreover, clinical data showed that higher levels of ACSL4 expression are positively correlated with favorable clinical outcomes. Mechanistic investigations further indicated that the reduced expression of ACSL4 in GIST is attributed to excessive protein degradation mediated by the E3 ligase TRIM21 and the deubiquitinase USP15. CONCLUSION These findings demonstrate that the TRIM21 and USP15 control ACSL4 stability to maintain the IM sensitive/resistant status of GIST.
Collapse
Affiliation(s)
- Zhiwei Cui
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Haoyu Sun
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Zhishuang Gao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chao Li
- Department of General Surgery, Zhongshan Hospital, Fudan University School of Medicine, #180 Fenglin Road, Shanghai, 200032, China
| | - Tingting Xiao
- Department of Cardiology, the Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Yibo Bian
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Rd, Xi'an, 710032, Shaanxi, China
| | - Zonghang Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Tianhao Gu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Jianan Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Tengyun Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Qianzheng Zhou
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Zhongyuan He
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Bowen Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Fengyuan Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Zekuan Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China
| | - Hao Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, 211166, China.
| |
Collapse
|
50
|
Martí-Obiol R, Martí-Fernández R, Fernández-Moreno MC, Barrios-Carvajal ME, López-Mozos F. Characteristics of gastrointestinal stromal tumors associated to other tumors: Características de los tumores del estroma gastrointestinal asociados a otras neoplasias. Cir Esp 2024; 102:135-141. [PMID: 38135151 DOI: 10.1016/j.cireng.2023.11.014] [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: 08/16/2023] [Accepted: 11/02/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Our aim is to analyze the differences between sporadic gastrointestinal stromal tumors and those associated with other tumors. METHODS Retrospective cohort study including patients with diagnosis of gastrointestinal stromal tumors operated at our center. Patients were divided into two groups, according to whether or not they had associated other tumors, both synchronously and metachronously. Disease free survival and overall survival were calculated for both groups. RESULTS 96 patients were included, 60 (62.5%) were male, with a median age of 66.8 (35-84). An association with other tumors was found in 33 cases (34.3%); 12 were synchronous (36.3%) and 21 metachronous (63.7%). The presence of mutations in associated tumors was 70% and in non-associated tumors 75%. Associated tumors were classified as low risk tumors based on Fletcher's stratification scale (p = 0.001) as they usually were smaller in size and had less than ≤5 mitosis per 50 HPF compared to non-associated tumors. When analyzing overall survival, there were statistically significant differences (p = 0,035) between both groups. CONCLUSION The relatively high proportion of gastrointestinal stromal tumors cases with associated tumors suggests the need to carry out a study to rule out presence of a second neoplasm and a long-term follow-up should be carried out in order to diagnose a possible second neoplasm. Gastrointestinal stromal tumors associated with other tumors have usually low risk of recurrence with a good long-term prognosis.
Collapse
Affiliation(s)
- Roberto Martí-Obiol
- Upper Gastrointestinal Surgery Unit, Hospital Clínico Universitario, Valencia, Spain
| | - Rosa Martí-Fernández
- Upper Gastrointestinal Surgery Unit, Hospital Clínico Universitario, Valencia, Spain.
| | | | | | - Fernando López-Mozos
- Upper Gastrointestinal Surgery Unit, Hospital Clínico Universitario, Valencia, Spain; Department of Surgery, Universidad de Valencia, Valencia, Spain
| |
Collapse
|