1
|
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 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] [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
|
2
|
Rhodes NG, Olinger K, Galgano SJ, Pietryga JA. Imaging of Iatrogenic Injuries to the Bowel. Radiol Clin North Am 2025; 63:387-403. [PMID: 40221182 DOI: 10.1016/j.rcl.2024.11.003] [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] [Indexed: 04/14/2025]
Abstract
This review discusses the ways in which the health care provider (or more broadly, the provision of medical care) can cause harm to the bowel and highlights the appearance of such untoward events on imaging. The etiologies are myriad, and as such, the radiologist must remain vigilant in identifying the presence of suspected and unsuspected injuries. Specific attention is given to complications from medication and external radiation therapy, as these interventions and their timing are often not known by the radiologist.
Collapse
Affiliation(s)
- Nicholas G Rhodes
- Musculoskeletal and Hospital Imaging, Department of Radiology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| | - Kristen Olinger
- Department of Radiology, University of North Carolina, 2000 Old Clinic, 101 Manning Drive, Campus Box #7510, Chapel Hill, NC 27599, USA
| | - Samuel J Galgano
- Abdominal Imaging, Department of Radiology, University of Alabama at Birmingham, JTN 444, 619 19th Street South, Birmingham, AL 35294, USA
| | - Jason A Pietryga
- Department of Radiology, University of North Carolina, 2000 Old Clinic, 101 Manning Drive, Campus Box #7510, Chapel Hill, NC 27599, USA
| |
Collapse
|
3
|
Liang H, Huang S, Xu X, Yin Z, Hussain M, Song X, Yi J, He Y, Guo J, Tu Z, Zhang Z, Zhou Y, Lu X. Designing Macrocyclic Kinase Inhibitors Using Macrocycle Scaffold Hopping with Reinforced Learning (Macro-Hop). J Med Chem 2025; 68:6698-6717. [PMID: 40101196 DOI: 10.1021/acs.jmedchem.5c00087] [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: 03/20/2025]
Abstract
Macrocycles have gained significant attention in drug discovery, with over 70 macrocyclic compounds currently in clinical use. Despite this progress, the effective methods for designing macrocycles remain elusive. In this study, we present Macro-Hop, a reinforced learning framework designed to rapidly and comprehensively explore the macrocycle chemical space. Macro-Hop efficiently generates novel macrocyclic scaffolds that not only align with predefined physicochemical properties but also exhibit 3D structural similarities to a specified reference compound. As a proof of concept, we applied Macro-Hop to design a new series of macrocycle inhibitors targeting PDGFRαD842 V kinase. The representative compound L7 exhibited high potency against PDGFRαD842 V in both biochemical and cellular assays with IC50 values of 23.8 and 2.1 nM, respectively. L7 effectively inhibited clinically relevant secondary mutants PDGFRαD842 V/G680R (IC50 = 64.1 nM) and PDGFRαD842 V/T674I (IC50 = 27.6 nM), highlighting the rapid effectiveness of wet-leb validation with Macro-Hop.
Collapse
Affiliation(s)
- Hong Liang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Shengjie Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xinxin Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhao Yin
- Department of Hematology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province 510317, China
| | - Muzammal Hussain
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Xiaojuan Song
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jianqiao Yi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yingqi He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jing Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhengchao Tu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yang Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaoyun Lu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
- Department of Hematology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province 510317, China
| |
Collapse
|
4
|
Stavrou N, Memos N, Filippatos C, Sergentanis TN, Zagouri F, Gavriatopoulou M, Ntanasis-Stathopoulos I. Neoadjuvant Imatinib in Recurrent/Metastatic Gastrointestinal Stromal Tumors: A Systematic Review and Meta-analysis of Proportions. J Gastrointest Cancer 2025; 56:88. [PMID: 40140195 PMCID: PMC11947046 DOI: 10.1007/s12029-025-01210-2] [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: 03/15/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Metastatic and recurrent gastrointestinal stromal tumors (GISTs) present challenging clinical management. Imatinib is the standard first-line therapy, improving survival and reducing tumor burden in the neoadjuvant use, facilitating surgical intervention. This systematic review and meta-analysis assessed the efficacy of neoadjuvant imatinib in metastatic/recurrent GISTs, highlighting its potential to enhance surgical outcomes and overall patient management. METHODS A systematic search was conducted in PubMed, Embase and Scopus (end-of-search: February 13, 2025) for records on neoadjuvant imatinib therapy in recurrent/metastatic GISTs. Pooled proportions and 95% confidence intervals were calculated with common-effect and random-effects models. Subgroup and meta-regression analysis were performed, addressing heterogeneity and examining any potential association between the factors that varied and the outcomes reported. The present meta-analysis was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. RESULTS The search identified 957 articles, and 14 were analyzed. The meta-analysis of proportions indicated that 2-year and 5-year PFS were 76% (95% CI 58-88%) and 43% (95% CI 17-74%), respectively, while 2-year and 5-year OS were 84% (95% CI 78-89%) and 60% (95% CI 51-68%), respectively. The pooled R0 resection rate was 82% (95% CI 64-92%), associated positively with that of radiological partial response (PR) (β = 3.92, p < 0.001). Further meta-regression analysis yielded no significant association with preoperative imatinib duration. CONCLUSION The present meta-analysis of trials and studies on metastatic or recurrent GISTs highlights key insights into post-surgery patient outcomes following neoadjuvant treatment with imatinib. Pooled effect estimates revealed promising 2-year and 5-year PFS rates of 76% and 43%, respectively, and 2-year and 5-year OS rates of 84% and 60%, respectively. Furthermore, the high pooled R0 resection rate of 82% emphasizes a substantial surgical efficacy in this population, while it was significantly correlated with successful R0 resections in patients with favorable outcomes.
Collapse
Affiliation(s)
- Niki Stavrou
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Nikolaos Memos
- Surgical Department, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Filippatos
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Flora Zagouri
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
5
|
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 2025:10.1038/s41416-025-02983-w. [PMID: 40133509 DOI: 10.1038/s41416-025-02983-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|
6
|
Li HT, Du YY, Huang Z, Li JJ, Zhang J. Significance of monitoring imatinib plasma concentration in second-line treatment decisions for c-kit 11 gene-mutated gastrointestinal stromal tumors. World J Gastrointest Oncol 2025; 17:98746. [PMID: 40092944 PMCID: PMC11866230 DOI: 10.4251/wjgo.v17.i3.98746] [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: 07/05/2024] [Revised: 11/02/2024] [Accepted: 12/16/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND For patients with advanced gastrointestinal stromal tumors (GISTs) carrying the c-kit exon 11 mutation, imatinib (IM) at a standard dosage of 400 mg per day is the preferred first-line treatment. In cases where treatment with IM fails, there is an urgent need for a more precise assessment method to determine whether to switch therapies or escalate the IM dosage. This approach will enhance clinical decision-making and optimize patient outcomes. AIM To investigate IM plasma concentration's role in second-line treatment decisions for c-kit 11-mutated advanced GISTs post-IM failure. METHODS Patients with advanced GIST harboring c-kit 11 mutation who experienced failure with IM 400 mg per day as first-line treatment at our hospital were retrospectively analyzed. Patients were categorized into a low plasma (LP) concentration group (LP group, < 1100 ng/mL) and high plasma (HP) concentration group (HP group, ≥ 1100 ng/mL). Each group was further subdivided into Group A (dose-escalation group) and Group B (drug-switch group). Baseline characteristics were compared and Kaplan-Meier curves were used to analyze the survival of patients. RESULTS Seventy-five patients were included in the analysis. For the LP group (n = 28), Group A (n = 14) had longer overall survival (OS) than Group B (n = 14) (P = 0.02). No differences were observed between the two subgroups in disease control rate (DCR), objective response rate, and progression-free survival (PFS) (P > 0.05). For the HP group (n = 47), Group B (n = 18) had a higher DCR and longer PFS than Group A (n = 29) (P = 0.008 and P = 0.03, respectively). No difference in OS was observed between the two subgroups (P > 0.05). CONCLUSION Increasing IM dosage for c-kit 11-mutated advanced GISTs post-IM failure may prolong OS if plasma concentration is < 1100 ng/mL. Switching tyrosine kinase inhibitors may improve DCR and PFS if ≥ 1100 ng/mL.
Collapse
Affiliation(s)
- Hai-Tao Li
- Department of Gastrointestinal Surgery, Nanchuan Hospital of Chongqing Medical University, Chongqing 408400, China
| | - Yun-Yun Du
- Department of Oncology, Nanchuan Hospital of Chongqing Medical University, Chongqing 408400, China
| | - Zhen Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jin-Jin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jun Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
7
|
Hoe JTM, Wong EYT, Tay TKY, Yang VS. Systemic Therapy for Advanced Gastrointestinal Stromal Tumors in 2025: Current Standard of Care and Emerging Therapeutic Strategies. J Gastroenterol Hepatol 2025. [PMID: 40084405 DOI: 10.1111/jgh.16932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Joshua Tian Ming Hoe
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Evelyn Yi Ting Wong
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Data and Computational Science Core, National Cancer Centre Singapore, Singapore, Singapore
| | - Timothy Kwang Yong Tay
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Valerie Shiwen Yang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
8
|
Schneider MA, Vetter D, Gutschow CA. Management of subepithelial esophageal tumors. Innov Surg Sci 2025; 10:21-30. [PMID: 40144787 PMCID: PMC11934943 DOI: 10.1515/iss-2023-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/16/2024] [Indexed: 03/28/2025] Open
Abstract
Subepithelial esophageal tumors (SET) are normally benign intramural esophageal lesions of mesenchymal origin. Although rare, the incidence of SET has increased in recent decades due to the more widespread use of endoscopy and diagnostic imaging. The current review aims to provide an overview of the histopathologic spectrum and the most frequent entities including leiomyoma and gastrointestinal stromal tumor (GIST), diagnostic workup, and multidisciplinary treatment options. Staging for SET should include endoscopy, endoscopic ultrasonography (EUS), and tissue sampling. Current consensus guidelines recommend that SET suggestive of gastrointestinal stromal tumor (GIST) larger than 20 mm or lesions with high-risk stigmata should undergo tissue sampling. Most SET have an excellent long-term outcome, but malignancy may be present in certain subtypes. Asymptomatic SET without high-risk stigmata discovered incidentally usually do not require specific treatment. However, depending on the size and location of the lesion symptoms may occur. Therapeutic interventions range from endoscopic interventional resections to major surgical procedures. Enucleation via minimally invasive or robotic-assisted access remains the standard of care for most SET sub-entities.
Collapse
Affiliation(s)
- Marcel A. Schneider
- Department of Visceral and Transplant Surgery, University Hospital Zürich, Zurich, Switzerland
| | - Diana Vetter
- Department of Visceral and Transplant Surgery, University Hospital Zürich, Zurich, Switzerland
| | - Christian A. Gutschow
- Department of Visceral and Transplant Surgery, University Hospital Zürich, Zurich, Switzerland
| |
Collapse
|
9
|
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
|
10
|
Mauri G, Patelli G, Roazzi L, Valtorta E, Amatu A, Marrapese G, Bonazzina E, Tosi F, Bencardino K, Ciarlo G, Mariella E, Marsoni S, Bardelli A, Bonoldi E, Sartore-Bianchi A, Siena S. Clinicopathological characterisation of MTAP alterations in gastrointestinal cancers. J Clin Pathol 2025; 78:195-201. [PMID: 38350716 PMCID: PMC11874331 DOI: 10.1136/jcp-2023-209341] [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: 12/09/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Methylthioadenosine phosphorylase (MTAP) is an essential metabolic enzyme in the purine and methionine salvage pathway. In cancer, MTAP gene copy number loss (MTAP loss) confers a selective dependency on the related protein arginine methyltransferase 5. The impact of MTAP alterations in gastrointestinal (GI) cancers remains unknown although hypothetically druggable. Here, we aim to investigate the prevalence, clinicopathological features and prognosis of MTAP loss GI cancers. METHODS Cases with MTAP alterations were retrieved from The Cancer Genome Atlas (TCGA) and a real-world cohort of GI cancers profiled by next-generation sequencing. If MTAP alterations other than loss were found, immunohistochemistry was performed. Finally, we set a case-control study to assess MTAP loss prognostic impact. RESULTS Findings across the TCGA dataset (N=1363 patients) and our cohort (N=508) were consistent. Gene loss was the most common MTAP alteration (9.4%), mostly co-occurring with CDKN2A/B loss (97.7%). Biliopancreatic and gastro-oesophageal cancers had the highest prevalence of MTAP loss (20.5% and 12.7%, respectively), being mostly microsatellite stable (99.2%). In colorectal cancer, MTAP loss was rare (1.1%), while most MTAP alterations were mutations (5/7, 71.4%); among the latter, only MTAP-CDKN2B truncation led to protein loss, thus potentially actionable. MTAP loss did not confer worse prognosis. CONCLUSIONS MTAP alterations are found in 5%-10% of GI cancers, most frequently biliopancreatic and gastro-oesophageal. MTAP loss is the most common alteration, identified almost exclusively in MSS, CDKN2A/B loss, upper-GI cancers. Other MTAP alterations were found in colorectal cancer, but unlikely to cause protein loss and drug susceptibility.
Collapse
Affiliation(s)
- Gianluca Mauri
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Roazzi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Emanuele Valtorta
- Department of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giovanna Marrapese
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Gabriele Ciarlo
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elisa Mariella
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Silvia Marsoni
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Alberto Bardelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Emanuela Bonoldi
- Department of Pathology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Division of Research and Innovation, Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
11
|
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
|
12
|
Yalikong A, Song B, He D, Xu E, Qi Z, Zhong Y. Large proximal gastric GIST tumours: downsizing by imatinib and subsequent endoresection. Gut 2025; 74:346-349. [PMID: 39532477 DOI: 10.1136/gutjnl-2024-332993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Ayimukedisi Yalikong
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Baohui Song
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Dongli He
- Shanghai Xuhui Central Hospital, Shanghai, China
| | - Enpan Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Zhipeng Qi
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai, China
- Zhongshan Hospital Fudan University, Shanghai, China
| | - Yunshi Zhong
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai, China
- Zhongshan Hospital Fudan University, Shanghai, China
- Shanghai Geriatric Medical Center, Shanghai, China
| |
Collapse
|
13
|
Ecker BL, Maki RG, Cavnar MJ, DeMatteo RP. Surgical Management of Sarcoma Metastatic to Liver. Hematol Oncol Clin North Am 2025; 39:55-65. [PMID: 39510677 DOI: 10.1016/j.hoc.2024.08.002] [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] [Indexed: 11/15/2024]
Abstract
Sarcomas are rare mesenchymal tumors with a propensity for hematogenous metastasis. Gastrointestinal stromal tumor (GIST) is the most common histologic subtype and the most common source of hepatic metastases. In the case of metastatic GIST, neoadjuvant imatinib can be used as a selection tool for the judicious application of surgery, where treatment-responsive patients who undergo resection to prevent the development of treatment-resistant clones have associated 10-year actuarial survival of 40%. Further advances for many of the non-GIST sarcoma subtypes will depend on the development of improved systemic therapies and evaluation of their activity in subtype or molecularly defined trials.
Collapse
Affiliation(s)
- Brett L Ecker
- Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Cooperman Barnabas Medical Center 94 Old Short Hills Road, Suite 1172, Livingston, NJ 07039.
| | - Robert G Maki
- Department of Medicine, University of Pennsylvania, 3400 Spruce st, Philadelphia, PA 19104, USA
| | - Michael J Cavnar
- Department of Surgery, University of Kentucky, 800 Rose St First Floor, Lexington, KY 40536, USA
| | - Ronald P DeMatteo
- Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Cooperman Barnabas Medical Center 94 Old Short Hills Road, Suite 1172, Livingston, NJ 07039
| |
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
|
Suto H, Kawamura M, Morita M, Sakai H, Onoe T, Ikeuchi K, Kajimoto K, Matsumoto K. Low-dose Imatinib Efficacy in a Gastrointestinal Stromal Tumor Patient With KIT Exon 11 W557_K558 Deletion. In Vivo 2025; 39:532-538. [PMID: 39740915 PMCID: PMC11705098 DOI: 10.21873/invivo.13857] [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: 10/10/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM Gastrointestinal stromal tumors (GISTs) are rare cancers originating from Cajal's stromal cells in the gastrointestinal tract. The most common driver mutation in these cancers is the KIT mutation. This report presents a case of response to low-dose imatinib in a patient with GIST harboring KIT exon 11 W557_K558 deletion. CASE REPORT In June 2023, an 82-year-old male developed perineal pain. Computed tomography (CT) imaging revealed a mass measuring >9 cm, extending from the rectum to the prostate. Submucosal tumor biopsy revealed a tumor with CD117-positive and DOG1-positive spindle-shaped cells leading to a diagnosis of GIST. c-Kit gene mutation analysis detected a W557_K558 deletion in exon 11. Treatment with imatinib (400 mg/day) was initiated in late October 2023 to preserve organ function; the dose was reduced to 300 mg/day after 5 days of treatment and further reduced to 200 mg/day after 10 days, with continued treatment at this dose. The CT performed in January, April, and June 2024 showed that the rectal GIST had shrunk. The blood imatinib concentration remained at approximately 650 ng/ml from January to March 2024 and decreased to 391 ng/ml in May 2024. CONCLUSION The response rate of GISTs to imatinib is influenced by genetic mutations. GIST with KIT exon 11 W557_K558 deletion is associated with a high risk of recurrence. In vitro data showed that low-dose imatinib was effective in such cases. Low-dose imatinib is a treatment option for patients with GIST harboring KIT exon 11 W557_K558 deletion who are intolerant to high-dose imatinib.
Collapse
Affiliation(s)
- Hirotaka Suto
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan;
| | - Miyuki Kawamura
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Mitsunori Morita
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Hideki Sakai
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Takuma Onoe
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Kyoko Ikeuchi
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| | | | - Koji Matsumoto
- Department of Medical Oncology, Hyogo Cancer Center, Akashi, Japan
| |
Collapse
|
16
|
Verkerk K, Zeverijn LJ, van de Haar J, Roepman P, Geurts BS, Spiekman AC, van der Noort V, van Berge Henegouwen JM, Hoes LR, van der Wijngaart H, Jansen AML, de Leng WWJ, Gelderblom AJ, Verheul HMW, Voest EE. The pathway alteration load is a pan-cancer determinant of outcome of targeted therapies: results from the Drug Rediscovery Protocol (DRUP). ESMO Open 2025; 10:104112. [PMID: 39778224 PMCID: PMC11760820 DOI: 10.1016/j.esmoop.2024.104112] [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: 10/14/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Many patients with cancer exhibit primary or rapid secondary resistance to targeted therapy (TT). We hypothesized that a higher number of altered oncogenic signaling pathways [pathway alteration load (PAL)] would reduce the benefit of TT which only intervenes in one pathway. This hypothesis was tested in the Drug Rediscovery Protocol (DRUP). PATIENTS AND METHODS DRUP is a prospective, pan-cancer, non-randomized clinical trial (NCT02925234) that treats patients with therapy-refractory metastatic cancer and an actionable molecular profile using matched off-label targeted and immunotherapies. All patients treated with TT with available clinical outcomes and whole genome sequencing were included. PAL was determined based on driver gene alterations and correlated with clinical benefit rate (CBR), progression-free survival (PFS) and overall survival (OS). Outcomes were validated in the independent Hartwig Medical database of metastatic cancers. RESULTS In 154 patients treated with TT, the median PAL was 3. Patients with a PAL below median (n = 60) demonstrated a higher CBR (41.7% versus 25.5%, odds ratio 0.48, P = 0.051), longer PFS [median 4.7 versus 2.9 months, adjusted hazard ratio (aHR) 1.70, P = 0.020] and OS (median 13.7 versus 5.6 months, aHR 3.80, P < 0.001) compared with those with PAL ≥3. Two hundred and fifty-eight patients in the Hartwig database showed similar results for CBR (54.2% versus 36.7%, odds ratio 2.04, P = 0.009) and PFS (7.0 versus 4.2 months, aHR 1.55, P = 0.009). CONCLUSIONS In our population, PAL emerged as a pan-cancer determinant of outcome to TT. Our findings support refined patient selection for TT and highlight the rationale for combinatorial treatment strategies in patients with multiple affected pathways.
Collapse
Affiliation(s)
- K Verkerk
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - L J Zeverijn
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - J van de Haar
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - P Roepman
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - B S Geurts
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - A C Spiekman
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - V van der Noort
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J M van Berge Henegouwen
- Oncode Institute, Utrecht, The Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - L R Hoes
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - H van der Wijngaart
- Department of Medical Oncology, GROW, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - A M L Jansen
- Department of Pathology, University Medical Cancer Center Utrecht, Utrecht, The Netherlands
| | - W W J de Leng
- Department of Pathology, University Medical Cancer Center Utrecht, Utrecht, The Netherlands
| | - A J Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - H M W Verheul
- Department of Medical Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - E E Voest
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Cicala CM, Matito J, Quindos M, Gómez-Peregrina D, Romero-Lozano P, Fernández-Suárez P, Valverde C, González M, Landolfi S, Pérez-Albert P, Gros L, Vivancos A, Serrano C. Targeted Next-Generation Sequencing in Succinate Dehydrogenase-Deficient GI Stromal Tumor Identifies Actionable Alterations in the PI3K/mTOR Pathway. JCO Precis Oncol 2025; 9:e2400497. [PMID: 39787462 DOI: 10.1200/po-24-00497] [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: 07/17/2024] [Revised: 10/29/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE Less than 5% of GI stromal tumors (GISTs) are driven by the loss of the succinate dehydrogenase (SDH) complex, resulting in a pervasive DNA hypermethylation pattern that leads to unique clinical features. Advanced SDH-deficient GISTs are usually treated with the same therapies targeting KIT and PDGFRA receptors as those used in metastatic GIST. However, these treatments display less activity in the absence of alternative therapeutic options. Therefore, it is critical to identify novel actionable alterations in SDH-deficient GIST. PATIENTS AND METHODS We performed a single-center, retrospective analysis of patients with SDH-deficient GIST together with next-generation sequencing (NGS) analysis from their respective tumor samples to identify mutations and copy number alterations and chromosomal alterations. NGS-tailored treatment was implemented whenever possible. RESULTS Seventeen tumor samples from 14 patients with SDH-deficient GIST underwent NGS. Mutational load was low, although three patients (21%) displayed molecular events in relapse samples leading to PI3K/mTOR pathway hyperactivation. mTOR inhibition with everolimus obtained a sustained tumor response in a heavily pretreated patient. Other alterations, largely present in late-stage patients, uncovered genes involved in cell cycle regulation, telomere maintenance, and DNA damage repair. Chromosomal arm-level alterations differed from the canonical cytogenetic progression in KIT/PDGFRA-mutant GIST. CONCLUSION This molecular landscape of SDH-deficient GIST uncovers novel molecular alterations, mostly in relapse and/or previously pretreated patients. The identification of genetic events leading to PI3K/mTOR dysregulation together with the remarkable activity of everolimus in one patient showcases the clinical relevance of this pathway, validates the utility of NGS in this population, and poses everolimus as a novel therapeutic alternative. Several other alterations were found at the genetic and genomic levels, underscoring novel biological processes likely involved during tumor evolution.
Collapse
Affiliation(s)
- Carlo María Cicala
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Judit Matito
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - María Quindos
- Medical Oncology Department, Complexo Hospitalario Universitario de A Coruña. Biomedical Research Institute (INIBIC), A Coruña, Spain
| | - David Gómez-Peregrina
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paula Romero-Lozano
- Vall d'Hebron Institute of Research (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Paula Fernández-Suárez
- Abdominal Imaging, Radiodiagnostic Department, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Claudia Valverde
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Macarena González
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Stefania Landolfi
- Pathology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Paula Pérez-Albert
- Paediatric Oncology and Hematology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Luis Gros
- Paediatric Oncology and Hematology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - César Serrano
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
18
|
Yu JZ, Kiss Z, Ma W, Liang R, Li T. Preclinical Models for Functional Precision Lung Cancer Research. Cancers (Basel) 2024; 17:22. [PMID: 39796653 PMCID: PMC11718887 DOI: 10.3390/cancers17010022] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Patient-centered precision oncology strives to deliver individualized cancer care. In lung cancer, preclinical models and technological innovations have become critical in advancing this approach. Preclinical models enable deeper insights into tumor biology and enhance the selection of appropriate systemic therapies across chemotherapy, targeted therapies, immunotherapies, antibody-drug conjugates, and emerging investigational treatments. While traditional human lung cancer cell lines offer a basic framework for cancer research, they often lack the tumor heterogeneity and intricate tumor-stromal interactions necessary to accurately predict patient-specific clinical outcomes. Patient-derived xenografts (PDXs), however, retain the original tumor's histopathology and genetic features, providing a more reliable model for predicting responses to systemic therapeutics, especially molecularly targeted therapies. For studying immunotherapies and antibody-drug conjugates, humanized PDX mouse models, syngeneic mouse models, and genetically engineered mouse models (GEMMs) are increasingly utilized. Despite their value, these in vivo models are costly, labor-intensive, and time-consuming. Recently, patient-derived lung cancer organoids (LCOs) have emerged as a promising in vitro tool for functional precision oncology studies. These LCOs demonstrate high success rates in growth and maintenance, accurately represent the histology and genomics of the original tumors and exhibit strong correlations with clinical treatment responses. Further supported by advancements in imaging, spatial and single-cell transcriptomics, proteomics, and artificial intelligence, these preclinical models are reshaping the landscape of drug development and functional precision lung cancer research. This integrated approach holds the potential to deliver increasingly accurate, personalized treatment strategies, ultimately enhancing patient outcomes in lung cancer.
Collapse
Affiliation(s)
- Jie-Zeng Yu
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (J.-Z.Y.); (Z.K.); (W.M.); (R.L.)
| | - Zsofia Kiss
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (J.-Z.Y.); (Z.K.); (W.M.); (R.L.)
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (J.-Z.Y.); (Z.K.); (W.M.); (R.L.)
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Ruqiang Liang
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (J.-Z.Y.); (Z.K.); (W.M.); (R.L.)
| | - Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (J.-Z.Y.); (Z.K.); (W.M.); (R.L.)
- Medical Service, Hematology/Oncology, Veterans Affairs Northern California Health Care System, Mather, CA 10535, USA
| |
Collapse
|
19
|
Delgado-de la Mora J, Al Assaad M, Quitian S, Levine MF, Deshpande A, Sigouros M, Manohar J, Medina-Martínez JS, Sboner A, Elemento O, Jessurun J, Hissong E, Mosquera JM. Novel structural variants that impact cell cycle genes are elucidated in metastatic gastrointestinal stromal tumors. Pathol Res Pract 2024; 266:155782. [PMID: 39708519 DOI: 10.1016/j.prp.2024.155782] [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: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasm of the digestive tract. Despite multiple therapeutic advances, patients with advanced disease frequently develop resistance to tyrosine kinase inhibitors (TKIs), and therefore represent a therapeutic challenge. We employed whole genome sequencing (WGS) on three metastatic GISTs refractory to various TKIs and explored a publicly available cohort of 499 GISTs. This study sheds light on the clinical importance of alterations in cell cycle genes such as cyclin-dependent kinase 2 A (CDKN2A), and cyclin-dependent kinase 2B (CDKN2B), their frequent alteration in metastatic GISTs and their potential role in tumor progression of this neoplasm. Likewise, new structural variations were identified in cyclin-dependent kinase 12 (CDK12). Whole genome profiling of metastatic GIST provides new insights to advance precision care of the disease, focusing on new therapeutic possibilities, especially for emerging targets such as CDK12.
Collapse
Affiliation(s)
- Jesús Delgado-de la Mora
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Majd Al Assaad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Stephanie Quitian
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA
| | - Max F Levine
- Isabl Inc., 175 Greenwich Street, Fl 38, New York, NY 10007, USA
| | - Aditya Deshpande
- Isabl Inc., 175 Greenwich Street, Fl 38, New York, NY 10007, USA
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | | | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - José Jessurun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA
| | - Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA.
| |
Collapse
|
20
|
Meyer M, Ota H, Messiou C, Benson C, Henzler T, Mattonen SA, Marin D, Bartsch A, Schoenberg SO, Riedel RF, Hohenberger P. Prospective evaluation of quantitative response parameter in patients with Gastrointestinal Stroma Tumor undergoing tyrosine kinase inhibitor therapy-Impact on clinical outcome. Int J Cancer 2024; 155:2047-2057. [PMID: 39023303 DOI: 10.1002/ijc.35094] [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: 01/10/2024] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 07/20/2024]
Abstract
The purpose of this study was to determine if dual-energy CT (DECT) vital iodine tumor burden (ViTB), a direct assessment of tumor vascularity, allows reliable response assessment in patients with GIST compared to established CT criteria such as RECIST1.1 and modified Choi (mChoi). From 03/2014 to 12/2019, 138 patients (64 years [32-94 years]) with biopsy proven GIST were entered in this prospective, multi-center trial. All patients were treated with tyrosine kinase inhibitors (TKI) and underwent pre-treatment and follow-up DECT examinations for a minimum of 24 months. Response assessment was performed according to RECIST1.1, mChoi, vascular tumor burden (VTB) and DECT ViTB. A change in therapy management could be because of imaging (RECIST1.1 or mChoi) and/or clinical progression. The DECT ViTB criteria had the highest discrimination ability for progression-free survival (PFS) of all criteria in both first line and second line and thereafter treatment, and was significantly superior to RECIST1.1 and mChoi (p < .034). Both, the mChoi and DECT ViTB criteria demonstrated a significantly early median time-to-progression (both delta 2.5 months; both p < .036). Multivariable analysis revealed 6 variables associated with shorter overall survival: secondary mutation (HR = 4.62), polymetastatic disease (HR = 3.02), metastatic second line and thereafter treatment (HR = 2.33), shorter PFS determined by the DECT ViTB criteria (HR = 1.72), multiple organ metastases (HR = 1.51) and lower age (HR = 1.04). DECT ViTB is a reliable response criteria and provides additional value for assessing TKI treatment in GIST patients. A significant superior response discrimination ability for median PFS was observed, including non-responders at first follow-up and patients developing resistance while on therapy.
Collapse
Affiliation(s)
- Mathias Meyer
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim-Heidelberg University, Mannheim, Germany
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Hideki Ota
- Department of Diagnostic Radiology, Tohoku University Hospital, Miyagi, Japan
| | - Christina Messiou
- Department of Radiology, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | | | - Thomas Henzler
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim-Heidelberg University, Mannheim, Germany
| | - Sarah A Mattonen
- Department of Medical Biophysics, Western University, London, Canada
| | - Daniele Marin
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Anna Bartsch
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim-Heidelberg University, Mannheim, Germany
- Department of Orthopedics and Traumatology, University Hospital Basel, Basel, Switzerland
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim-Heidelberg University, Mannheim, Germany
| | - Richard F Riedel
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| |
Collapse
|
21
|
García-Trejo MA, Castañeda G, Ríos Á. Analytical control of imatinib in bioanalytical samples using graphene quantum dots sensing. Anal Bioanal Chem 2024; 416:7267-7276. [PMID: 38795213 DOI: 10.1007/s00216-024-05346-1] [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: 03/31/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/27/2024]
Abstract
An analytical method for the determination of imatinib (IMA, the primary treatment for chronic myeloid leukemia), based on the fluorescence properties of graphene quantum dots (GQDs), is reported in this work. The method is addressed to the analytical control of IMA in biological and pharmaceutical samples, due to the present interest in the control of the doses of this anticancer drug, as well as the therapeutic monitoring. The whole method involves the use of a solid-phase extraction (SPE) procedure, followed by an evaporation step, for the treatment of biological samples. For that, tC18 sorbent cartridges were used. After the sample treatment, the solution containing the analyte was mixed with an aqueous solution of GQDs at pH 7.2, and the fluorescent quenching of GQDs was measured. IMA was determined in the 10-250 µg L-1 range, with a limit of detection of 21 µg L-1 and a precision of 1.5% as relative standard deviation, measured in terms of reproducibility. The recovery for biological samples was in the 84-113% range.
Collapse
Affiliation(s)
- María A García-Trejo
- Department of Analytical Chemistry and Food Technology, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, Campus Universitario, 13071, Ciudad Real, Spain
- Regional Institute for Applied Scientific Research, IRICA, Camilo José Cela Avenue, E-13005, Ciudad Real, Spain
| | - Gregorio Castañeda
- Department of Analytical Chemistry and Food Technology, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, Campus Universitario, 13071, Ciudad Real, Spain
- Regional Institute for Applied Scientific Research, IRICA, Camilo José Cela Avenue, E-13005, Ciudad Real, Spain
| | - Ángel Ríos
- Department of Analytical Chemistry and Food Technology, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, Campus Universitario, 13071, Ciudad Real, Spain.
- Regional Institute for Applied Scientific Research, IRICA, Camilo José Cela Avenue, E-13005, Ciudad Real, Spain.
| |
Collapse
|
22
|
Hu B, Ye Y, Gao Z. The benefit of surgery during systematic therapy for gastrointestinal stromal tumor liver metastasis: a SEER-based retrospective study. Gastroenterol Rep (Oxf) 2024; 12:goae095. [PMID: 39588533 PMCID: PMC11587995 DOI: 10.1093/gastro/goae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 11/27/2024] Open
Abstract
Background The liver is the most common site of gastrointestinal stromal tumor (GIST) metastasis. Most patients who develop metastases gradually develop multiline drug resistance during long-term systematic treatment. We aimed to evaluate the benefit of surgery during the systematic treatment of GIST liver metastases. Methods Data on GISTs with liver metastasis were retrieved from the Surveillance, Epidemiology, and End Results database. This study included 607 patients, of whom 380 patients were treated with chemotherapy alone (Chemo group) and 227 patients underwent surgery in addition to chemotherapy (Chemo&Surg group). The primary outcomes were cancer-specific survival (CSS) and overall survival (OS). Propensity score matching (PSM) was performed to balance the baseline factors. Results According to the multivariate analysis, surgery benefitted both CSS and OS (P < 0.001). After PSM, surgical resection still showed significant benefits in terms of both CSS and OS (P < 0.001). Surgery combined with chemotherapy increased the median CSS by at least 63 months and the median OS by at least 76 months. Subgroup analysis of the Chemo&Surg group revealed that the timing of surgery was not an independent influencing factor for either CSS or OS. Conclusions We found that performing additional surgery, in addition to systematic therapy, for GIST liver metastasis resulted in improved CSS and OS. These benefits were not affected by the timing of surgery during systemic treatment.
Collapse
Affiliation(s)
- Bozhi Hu
- Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing, P. R. China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing, P. R. China
| | - Zhidong Gao
- Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing, P. R. China
| |
Collapse
|
23
|
Wu T, Sun Y, Wang D, Isaji T, Fukuda T, Suzuki C, Hanamatsu H, Nishikaze T, Tsumoto H, Miura Y, Furukawa JI, Gu J. The acetylglucosaminyltransferase GnT-Ⅲ regulates erythroid differentiation through ERK/MAPK signaling. J Biol Chem 2024; 300:108010. [PMID: 39571652 PMCID: PMC11699732 DOI: 10.1016/j.jbc.2024.108010] [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: 07/17/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Differentiation therapy is an alternative strategy used in treating chronic myelogenous leukemia to induce the differentiation of immature or cancerous cells toward mature cells and inhibit tumor cell proliferation. We aimed to explore N-glycans' roles in erythroid differentiation using the sodium butyrate (NaBu)-induced model of K562 cells (WT/NaBu cells). Here, using lectin blot, flow cytometry, real-time PCR, and mass spectrometry analyses, we demonstrated that the mRNA levels of N-acetylglucosaminyltransferase Ⅲ ((encoded by the MGAT3 gene) and its product (bisected N-glycans) were significantly increased during erythroid differentiation. To address the importance of GnTN-acetylglucosaminyltransferase-Ⅲ in this progress, we established a stable MGAT3 KO K562 cell line using the CRISPR/Cas9 technology. Compared to WT/NaBu cells, MGAT3 KO significantly impeded the progression of erythroid differentiation, as shown in decreased cell color and levels of erythroid markers, glycophorin A (CD235a), and β-globin. Consistently, MGAT3 KO mitigated the inhibitory impact of NaBu on cell proliferation. During induction, MGAT3 KO suppressed the cellular phosphorylated tyrosine and phospho-extracellular signal-regulated kinase (ERK)1/2 levels. Inhibition of the ERK/mitogen-activated protein kinase signaling pathway using U0126 blocked erythroid differentiation while concurrently suppressing the expression levels of MGAT3 and bisected N-glycans. Furthermore, the lack of bisecting GlcNAc modification on c-Kit and transferrin receptor 1 (CD71) suppressed cellular signaling and accelerated the degradation of the CD71 protein, respectively. Our study highlights the critical role of MGAT3 in regulating erythroid differentiation associated with the ERK/mitogen-activated protein kinase signaling pathway, which may shed light on identifying new differentiation therapy in chronic myelogenous leukemia.
Collapse
Affiliation(s)
- Tiangui Wu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Dan Wang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Chiharu Suzuki
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Hisatoshi Hanamatsu
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Takashi Nishikaze
- Solutions COE, Analytical & Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Hiroki Tsumoto
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Jun-Ichi Furukawa
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan; Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
24
|
Shi J, Sun D, Wu K, Jiang Z, Kong X, Wang W, Wu H, Zheng Y. Positional encoding-guided transformer-based multiple instance learning for histopathology whole slide images classification. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 258:108491. [PMID: 39549395 DOI: 10.1016/j.cmpb.2024.108491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Whole slide image (WSI) classification is of great clinical significance in computer-aided pathological diagnosis. Due to the high cost of manual annotation, weakly supervised WSI classification methods have gained more attention. As the most representative, multiple instance learning (MIL) generally aggregates the predictions or features of the patches within a WSI to achieve the slide-level classification under the weak supervision of WSI labels. However, most existing MIL methods ignore spatial position relationships of the patches, which is likely to strengthen the discriminative ability of WSI-level features. METHODS In this paper, we propose a novel positional encoding-guided transformer-based multiple instance learning (PEGTB-MIL) method for histopathology WSI classification. It aims to encode the spatial positional property of the patch into its corresponding semantic features and explore the potential correlation among the patches for improving the WSI classification performance. Concretely, the deep features of the patches in WSI are first extracted and simultaneously a position encoder is used to encode the spatial 2D positional information of the patches into the spatial-aware features. After incorporating the semantic features and spatial embeddings, multi-head self-attention (MHSA) is applied to explore the contextual and spatial dependencies of the fused features. Particularly, we introduce an auxiliary reconstruction task to enhance the spatial-semantic consistency and generalization ability of features. RESULTS The proposed method is evaluated on two public benchmark TCGA datasets (TCGA-LUNG and TCGA-BRCA) and two in-house clinical datasets (USTC-EGFR and USTC-GIST). Experimental results validate it is effective in the tasks of cancer subtyping and gene mutation status prediction. In the test stage, the proposed PEGTB-MIL outperforms the other state-of-the-art methods and respectively achieves 97.13±0.34%, 86.74±2.64%, 83.25±1.65%, and 72.52±1.63% of the area under the receiver operating characteristic (ROC) curve (AUC). CONCLUSION PEGTB-MIL utilizes positional encoding to effectively guide and reinforce MIL, leading to enhanced performance on downstream WSI classification tasks. Specifically, the introduced auxiliary reconstruction module adeptly preserves the spatial-semantic consistency of patch features. More significantly, this study investigates the relationship between position information and disease diagnosis and presents a promising avenue for further research.
Collapse
Affiliation(s)
- Jun Shi
- School of Software, Hefei University of Technology, Hefei, 230601, Anhui Province, China
| | - Dongdong Sun
- School of Computer Science and Information Engineering, Hefei University of Technology, Hefei, 230601, Anhui Province, China
| | - Kun Wu
- Image Processing Center, School of Astronautics, Beihang University, Beijing, 102206, China
| | - Zhiguo Jiang
- Image Processing Center, School of Astronautics, Beihang University, Beijing, 102206, China
| | - Xue Kong
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China
| | - Wei Wang
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China
| | - Haibo Wu
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui Province, China
| | - Yushan Zheng
- School of Engineering Medicine, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
25
|
Haller V, Reiff C, Hamacher R, Kostbade K, Kaths M, Treckmann J, Bertram S, Zaun Y, Bauer S, Falkenhorst J. Overall survival of patients with KIT-mutant metastatic GIST in the era of multiple kinase inhibitor availability. J Cancer Res Clin Oncol 2024; 150:489. [PMID: 39516299 PMCID: PMC11549121 DOI: 10.1007/s00432-024-05965-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE The prognosis of patients with metastatic GIST and imatinib-sensitive primary mutations has significantly improved. However, limited data are available to inform patients about outcomes across different lines of treatment. This retrospective analysis aims to evaluate patient outcomes at a large German GIST referral center over the past 15 years. PATIENTS AND METHODS Overall survival (OS) and progression-free survival (PFS) were analyzed in patients with metastatic GIST, with diagnosis of metastases between 2008 and 2021, when at least three lines of treatment were available in Germany (n = 174). RESULTS The median overall survival far exceeds historical data for patients with primary exon 11 and exon 9 mutations (median OS in palliative treatment with imatinib: 7.1 years; median OS in second-line palliative treatment with sunitinib: 2.9 years; median OS in third-line palliative treatment with regorafenib: 1.9 years). Among those patients who received palliative imatinib treatment, no significant difference in median OS survival was observed between those who had received perioperative imatinib for localized disease and those who did not. Furthermore, the location of metastases significantly impacted survival, whereas the time between the initial diagnosis and the diagnosis of metastases had no significant effect on survival. CONCLUSION In conclusion, this study provides a novel, real-world reference for survival outcomes in patients with metastatic GIST.
Collapse
Affiliation(s)
- Valerie Haller
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Carina Reiff
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Karina Kostbade
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Moritz Kaths
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Department of Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Juergen Treckmann
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Department of Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Stefanie Bertram
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Department of Pathology, University Hospital Essen, Essen, Germany
| | - Yasmin Zaun
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Sebastian Bauer
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| | - Johanna Falkenhorst
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
26
|
Tripathi M, Purwar R, Sinha R, Singh P, Pandey M. Gastrointestinal Stromal Tumor of the Rectum: Report of a Case With Long-Term Imatinib Treatment. Cureus 2024; 16:e74269. [PMID: 39717333 PMCID: PMC11663722 DOI: 10.7759/cureus.74269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Gastrointestinal stromal tumors (GIST) are rare in the rectum. These usually present with symptoms produced by compression of pelvic organs or bleeding. Surgery is the treatment of choice, however, at times the surgery can be mutilating and organ preservation may not be possible. In such cases imatinib can be used. We report a case of rectal GIST that presented with urinary obstruction and obstipation treated with long-term imatinib with very good response and relief of symptoms, which prompted the patient to refuse surgery and resulted in organ preservation.
Collapse
Affiliation(s)
- Madhumita Tripathi
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Roli Purwar
- Obstetrics and Gynaecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Richie Sinha
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Pooja Singh
- Oncological Research, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Manoj Pandey
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| |
Collapse
|
27
|
Scuoppo C, Cai B, Ofori K, Scholze H, Kumar R, D’Alessandro A, Basso K, Pasqualucci L, Dalla-Favera R. Repurposing NAMPT Inhibitors for Germinal Center B Cell-Like Diffuse Large B-Cell Lymphoma. Blood Cancer Discov 2024; 5:417-427. [PMID: 39105568 PMCID: PMC11528193 DOI: 10.1158/2643-3230.bcd-24-0020] [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: 01/23/2024] [Revised: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) includes the activated B cell-like (ABC) and germinal center B cell-like (GCB) subtypes, which differ in cell of origin, genetics, and clinical response. By screening the subtype-specific activity of 211 drugs approved or in active clinical development for other diseases, we identified inhibitors of nicotinamide phosphoribosyl transferase (NAMPTi) as active in a subset of GCB-DLBCL in vitro and in vivo. We validated three chemically distinct NAMPTis for their on-target activity based on biochemical and genetic rescue approaches and found the ratio between NAMPT and PARP1 RNA levels was predictive of NAMPTi sensitivity across DLBCL subtypes. Notably, the NAMPT:PARP1 transcript ratio predicts higher antitumor activity in BCL2-translocated GCB-DLBCL. Accordingly, pharmacologic and genetic inhibition of BCL2 was potently synergistic with NAMPT blockade. These data support the inhibition of NAMPT as a therapeutically relevant strategy for BCL2-translocated DLBCLs. Significance: Targeted therapies have emerged for the ABC subtype of DLBCL, but not for the GCB subtype, despite the evidence of a significant subset of high-risk cases. We identify a drug that specifically targets a subset of GCB-DLBCL and provide preclinical evidence for BCL2 translocations as biomarkers for their identification.
Collapse
Affiliation(s)
- Claudio Scuoppo
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Bowen Cai
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kenneth Ofori
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Hanna Scholze
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Rahul Kumar
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Katia Basso
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Riccardo Dalla-Favera
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
28
|
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
|
29
|
Huang X, Chen L, Liu L, Chen H, Gong Z, Lyu J, Li Y, Jiang Q, Zeng X, Zhang P, Zhou H. Untargeted metabolomics analysis reveals the potential mechanism of imatinib-induced skin rash in patients with gastrointestinal stromal tumor. Int Immunopharmacol 2024; 140:112728. [PMID: 39098227 DOI: 10.1016/j.intimp.2024.112728] [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: 05/10/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024]
Abstract
Imatinib-induced skin rash poses a significant challenge for patients with gastrointestinal stromal tumor, often resulting in treatment interruption or discontinuation and subsequent treatment failure. However, the underlying mechanism of imatinib-induced skin rashes in gastrointestinal stromal tumor patients remains unclear. A total of 51 patients (27 with rash and 24 without rash) were enrolled in our study. Blood samples were collected concomitantly with the onset of clinical manifestations of rashes, and simultaneously collecting clinical relevant information. The imatinib concentration and untargeted metabolomics were performed by ultra-high-performance liquid chromatography-tandem mass spectrometry. There were no significant differences in age, gender, imatinib concentration and white blood cells count between the rash group and the control group. However, the rash group exhibited a higher eosinophil count (P<0.05) and lower lymphocyte count (P<0.05) compared to the control group. Untargeted metabolomics analysis found that 105 metabolites were significantly differentially abundant. The univariate analysis highlighted erucamide, linoleoylcarnitine, and valine betaine as potential predictive markers (AUC≥0.80). Further enriched pathway analysis revealed primary metabolic pathways, including sphingolipid signaling pathway, sphingolipid metabolism, cysteine and methionine metabolism, biosynthesis of unsaturated fatty acids, arginine and proline metabolism, and biosynthesis of amino acids. These findings suggest that the selected differential metabolites could serve as a foundation for the prediction and management of imatinib-induced skin rash in gastrointestinal stromal tumor patients.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Linhua Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Li Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Hefen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhujun Gong
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Jianbo Lyu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yao Li
- Department of Gastrointestinal Surgery, Department of Nursing, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangyu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Hong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China.
| |
Collapse
|
30
|
Renne SL, Cammelli M, Santori I, Tassan-Mangina M, Samà L, Ruspi L, Sicoli F, Colombo P, Terracciano LM, Quagliuolo V, Cananzi FCM. True Mitotic Count Prediction in Gastrointestinal Stromal Tumors: Bayesian Network Model and PROMETheus (Preoperative Mitosis Estimator Tool) Application Development. J Med Internet Res 2024; 26:e50023. [PMID: 39437385 PMCID: PMC11538881 DOI: 10.2196/50023] [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: 06/16/2023] [Revised: 05/14/2024] [Accepted: 07/21/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) present a complex clinical landscape, where precise preoperative risk assessment plays a pivotal role in guiding therapeutic decisions. Conventional methods for evaluating mitotic count, such as biopsy-based assessments, encounter challenges stemming from tumor heterogeneity and sampling biases, thereby underscoring the urgent need for innovative approaches to enhance prognostic accuracy. OBJECTIVE The primary objective of this study was to develop a robust and reliable computational tool, PROMETheus (Preoperative Mitosis Estimator Tool), aimed at refining patient stratification through the precise estimation of mitotic count in GISTs. METHODS Using advanced Bayesian network methodologies, we constructed a directed acyclic graph (DAG) integrating pertinent clinicopathological variables essential for accurate mitotic count prediction on the surgical specimen. Key parameters identified and incorporated into the model encompassed tumor size, location, mitotic count from biopsy specimens, surface area evaluated during biopsy, and tumor response to therapy, when applicable. Rigorous testing procedures, including prior predictive simulations, validation utilizing synthetic data sets were employed. Finally, the model was trained on a comprehensive cohort of real-world GIST cases (n=80), drawn from the repository of the Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, with a total of 160 cases analyzed. RESULTS Our computational model exhibited excellent diagnostic performance on synthetic data. Different model architecture were selected based on lower deviance and robust out-of-sample predictive capabilities. Posterior predictive checks (retrodiction) further corroborated the model's accuracy. Subsequently, PROMETheus was developed. This is an intuitive tool that dynamically computes predicted mitotic count and risk assessment on surgical specimens based on tumor-specific attributes, including size, location, surface area, and biopsy-derived mitotic count, using posterior probabilities derived from the model. CONCLUSIONS The deployment of PROMETheus represents a potential advancement in preoperative risk stratification for GISTs, offering clinicians a precise and reliable means to anticipate mitotic counts on surgical specimens and a solid base to stratify patients for clinical studies. By facilitating tailored therapeutic strategies, this innovative tool is poised to revolutionize clinical decision-making paradigms, ultimately translating into improved patient outcomes and enhanced prognostic precision in the management of GISTs.
Collapse
Affiliation(s)
- Salvatore Lorenzo Renne
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Pathology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Manuela Cammelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Ilaria Santori
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Marta Tassan-Mangina
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Laura Samà
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Laura Ruspi
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federico Sicoli
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Piergiuseppe Colombo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Pathology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luigi Maria Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Pathology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Vittorio Quagliuolo
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ferdinando Carlo Maria Cananzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
31
|
Faouzi A, Arnaud A, Hallé F, Roussel J, Aymard M, Denavit V, Do CV, Mularoni A, Salah M, ElHady A, Pham TN, Bancet A, Le Borgne M, Terreux R, Barret R, Engel M, Lomberget T. Design, synthesis, and structure-activity relationship studies of 6 H-benzo[ b]indeno[1,2- d]thiophen-6-one derivatives as DYRK1A/CLK1/CLK4/haspin inhibitors. RSC Med Chem 2024; 16:d4md00537f. [PMID: 39430953 PMCID: PMC11487425 DOI: 10.1039/d4md00537f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
A series of sulfur-containing tetracycles was designed and evaluated for their ability to inhibit protein kinase DYRK1A, a target known to have several potential therapeutic applications including cancers, Down syndrome or Alzheimer's disease. Our medicinal chemistry strategy relied on the design of new compounds using ring contraction/isosteric replacement and constrained analogy of known DYRK1A inhibitors, thus resulting in their DYRK1A inhibitory activity enhancement. Whereas a good inhibitory effect of targeted DYRK1A protein was observed for 5-hydroxy compounds 4i-k (IC50 = 35-116 nM) and the 5-methoxy derivative 4e (IC50 = 52 nM), a fairly good selectivity towards its known DYRK1B off-target was observed for 4k. In addition, the most active compound 4k, having an ATP-competitive mechanism of action, proved to be also a potent inhibitor of CLK1/CLK4 (IC50 = 20 and 26 nM) and, to a lesser extent, of haspin (IC50 = 76 nM) kinases. In silico docking studies within the DYRK1A, CLK1/CLK4 and haspin ATP binding sites were carried out to understand the interactions of our tetracyclic derivatives 4 with these targets. Antiproliferative activities on U87/U373 glioblastoma cell lines of the most potent compound 4k showed a moderate effect (IC50 values between 33 and 46 μM). Microsomal stabilities of the designed compounds 4a-m were also investigated, showing great disparities, depending on benzo[b]thiophene ring 5-substitution.
Collapse
Affiliation(s)
- Abdelfattah Faouzi
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Alexandre Arnaud
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - François Hallé
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Jean Roussel
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Mandy Aymard
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Vincent Denavit
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Cong Viet Do
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
- University of Science and Technology of HanoÏ USTH 18 Hoang Quoc Viet Hanoi 100000 Vietnam
| | - Angélique Mularoni
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Mohamed Salah
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
- Department of Pharmaceutical Chemistry, School of Pharmacy, Newgiza University (NGU) Newgiza, km 22 Cairo-Alexandria Desert Road 12577 Cairo Egypt
| | - Ahmed ElHady
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Cairo 11865 Egypt
| | - Thanh-Nhat Pham
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Alexandre Bancet
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Marc Le Borgne
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Raphaël Terreux
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, LBTI, ECMO Team, Institut de Biologie et Chimie des Protéines 7 Passage du Vercors 69367 Lyon Cedex 07 France
| | - Roland Barret
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
| | - Thierry Lomberget
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| |
Collapse
|
32
|
Eschalier C, Lafont T, Marsili S, Yakoubi M, Brice A, Thomas F, White-Koning M, Allal B, Chatelut E. Therapeutic Drug Monitoring of Imatinib: Is There a Rationale for Also Quantifying Its Active Metabolite? Chemotherapy 2024; 70:26-36. [PMID: 39396500 DOI: 10.1159/000541936] [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: 07/30/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Therapeutic drug monitoring of imatinib is widely performed to individualize imatinib dosage. While N-desmethyl imatinib is an active metabolite of imatinib, its concentrations are not routinely determined. METHODS Imatinib and N-desmethyl imatinib trough plasma concentrations at steady-state were obtained from 295 patients with either chronic myeloid leukemia or gastrointestinal stromal tumor to see whether N-desmethyl imatinib provided additional information. Pharmacokinetic data were analyzed using a nonlinear mixed effect approach. Correlations between several pharmacokinetic metrics of drug exposure were evaluated. RESULTS The mean value of the N-desmethyl imatinib/imatinib ratio of trough concentrations was 0.31 with half of the values between 0.23 and 0.37. N-desmethyl imatinib and total (i.e., N-desmethyl imatinib plus imatinib) trough plasma concentrations or area under the curve values were closely correlated with imatinib values. The distribution of imatinib or total concentrations between patients requiring imatinib dosage adjustment, or not, was similar. CONCLUSION These results do not clearly support routine N-desmethyl monitoring since it does not provide additional information to imatinib data.
Collapse
Affiliation(s)
- Clara Eschalier
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France,
| | - Thierry Lafont
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Sabrina Marsili
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Malika Yakoubi
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Aurélie Brice
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Fabienne Thomas
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Mélanie White-Koning
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Ben Allal
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| | - Etienne Chatelut
- Oncopole Claudius-Regaud and Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
- Cancer Research Center of Toulouse, Inserm U1037, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
33
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [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: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
34
|
Zahra MA, Pessin J, Rastogi D. A clinician's guide to effects of obesity on childhood asthma and into adulthood. Expert Rev Respir Med 2024; 18:759-775. [PMID: 39257361 PMCID: PMC11473229 DOI: 10.1080/17476348.2024.2403500] [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: 05/29/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Obesity, one of the most common chronic conditions affecting the human race globally, affects several organ systems, including the respiratory system, where it contributes to onset and high burden of asthma. Childhood onset of obesity-related asthma is associated with high persistent morbidity into adulthood. AREAS COVERED In this review, we discuss the disease burden in children and adults to highlight the overlap between symptoms and pulmonary function deficits associated with obesity-related asthma in both age ranges, and then discuss the potential role of three distinct mechanisms, that of mechanical fat load, immune perturbations, and of metabolic perturbations on the disease burden. We also discuss interventions, including medical interventions for weight loss such as diet modification, that of antibiotics and anti-inflammatory therapies, as well as that of surgical intervention on amelioration of burden of obesity-related asthma. EXPERT OPINION With increase in obesity-related asthma due to increasing burden of obesity, it is evident that it is a disease entity distinct from asthma among lean individuals. The time is ripe to investigate the underlying mechanisms, focusing on identifying novel therapeutic targets as well as consideration to repurpose medications effective for other obesity-mediated complications, such as insulin resistance, dyslipidemia and systemic inflammation.
Collapse
Affiliation(s)
- Mahmoud Abu Zahra
- Division of Respiratory and Sleep Medicine, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jeffrey Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Deepa Rastogi
- Division of Respiratory and Sleep Medicine, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, United States
- Norman Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
35
|
Li Y, Zhang R, Fu C, Jiang Q, Zhang P, Zhang Y, Chen J, Tao K, Chen WH, Zeng X. Intratumoral microbiome promotes liver metastasis and dampens adjuvant imatinib treatment in gastrointestinal stromal tumor. Cancer Lett 2024; 601:217149. [PMID: 39117066 DOI: 10.1016/j.canlet.2024.217149] [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: 04/09/2024] [Revised: 06/06/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Understanding the determinants of long-term liver metastasis (LM) outcomes in gastrointestinal stromal tumor (GIST) patients is crucial. We established the feature selection model of intratumoral microbiome at the surgery, achieving robust predictive accuracies of 0.953 and 0.897 AUCs in discovery (n = 74) and validation (n = 34) cohorts, respectively. Notably, despite the significant reduction in LM occurrence with adjuvant imatinib (AI) treatment, intratumoral microbiome exerted independently stronger effects on post-operative LM. Employing both 16S and full-length rRNA sequencing, we pinpoint intracellular Shewanella algae as a foremost LM risk factor in both AI- and non-AI-treated patients. Experimental validation confirmed S. algae's intratumoral presence in GIST, along with migration/invasion-promoting effects on GIST cells. Furthermore, S. algae promoted LM and impeded AI treatment in metastatic mouse models. Our findings advocate for incorporating intratumoral microbiome evaluation at surgery, and propose S. algae as a therapeutic target for LM suppression in GIST.
Collapse
Affiliation(s)
- Yanze Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; Department of Computer Science, School of Science, Aalto University, Helsinki, Finland
| | - Ruizhi Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengbo Fu
- Department of Computer Science, School of Science, Aalto University, Helsinki, Finland
| | - Qi Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingchao Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China; Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, 264003, China.
| | - Xiangyu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Burda P, Hlavackova A, Polivkova V, Curik N, Laznicka A, Krizkova J, Suttnar J, Klener P, Polakova KM. Imatinib therapy of chronic myeloid leukemia significantly reduces carnitine cell intake, resulting in adverse events. Mol Metab 2024; 88:102016. [PMID: 39182842 PMCID: PMC11403060 DOI: 10.1016/j.molmet.2024.102016] [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: 06/18/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE A prominent, safe and efficient therapy for patients with chronic myeloid leukemia (CML) is inhibiting oncogenic protein BCR::ABL1 in a targeted manner with imatinib, a tyrosine kinase inhibitor. A substantial part of patients treated with imatinib report skeletomuscular adverse events affecting their quality of life. OCTN2 membrane transporter is involved in imatinib transportation into the cells. At the same time, the crucial physiological role of OCTN2 is cellular uptake of carnitine which is an essential co-factor for the mitochondrial β-oxidation pathway. This work investigates the impact of imatinib treatment on carnitine intake and energy metabolism of muscle cells. METHODS HTB-153 (human rhabdomyosarcoma) cell line and KCL-22 (CML cell line) were used to study the impact of imatinib treatment on intracellular levels of carnitine and vice versa. The energy metabolism changes in cells treated by imatinib were quantified and compared to changes in cells exposed to highly specific OCTN2 inhibitor vinorelbine. Mouse models were used to test whether in vitro observations are also achieved in vivo in thigh muscle tissue. The analytes of interest were quantified using a Prominence HPLC system coupled with a tandem mass spectrometer. RESULTS This work showed that through the carnitine-specific transporter OCTN2, imatinib and carnitine intake competed unequally and intracellular carnitine concentrations were significantly reduced. In contrast, carnitine preincubation did not influence imatinib cell intake or interfere with leukemia cell targeting. Blocking the intracellular supply of carnitine with imatinib significantly reduced the production of most Krebs cycle metabolites and ATP. However, subsequent carnitine supplementation rescued mitochondrial energy production. Due to specific inhibition of OCTN2 activity, the influx of carnitine was blocked and mitochondrial energy metabolism was impaired in muscle cells in vitro and in thigh muscle tissue in a mouse model. CONCLUSIONS This preclinical experimental study revealed detrimental effect of imatinib on carnitine-mediated energy metabolism of muscle cells providing a possible molecular background of the frequently occurred side effects during imatinib therapy such as fatigue, muscle pain and cramps.
Collapse
Affiliation(s)
- Pavel Burda
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Vendula Polivkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Nikola Curik
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Adam Laznicka
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jitka Krizkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jiri Suttnar
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic; First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Katerina Machova Polakova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
37
|
He B, Dymond L, Wood KH, Bastow ER, Satiaputra J, Li J, Johansson-Percival A, Hamzah J, Kumarasinghe MP, Ballal M, Foo J, Johansson M, Ee HC, White SW, Winteringham L, Ganss R. Immune priming and induction of tertiary lymphoid structures in a cord-blood humanized mouse model of gastrointestinal stromal tumor. Oncoimmunology 2024; 13:2406576. [PMID: 39314905 PMCID: PMC11418220 DOI: 10.1080/2162402x.2024.2406576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024] Open
Abstract
Gastrointestinal stromal tumors (GISTs) harbor diverse immune cell populations but so far immunotherapy in patients has been disappointing. Here, we established cord blood humanized mouse models of localized and disseminated GIST to explore the remodeling of the tumor environment for improved immunotherapy. Specifically, we assessed the ability of a cancer vascular targeting peptide (VTP) to bind to mouse and patient GIST angiogenic blood vessels and deliver the TNF superfamily member LIGHT (TNFS14) into tumors. LIGHT-VTP treatment of GIST in humanized mice improved vascular function and tumor oxygenation, which correlated with an overall increase in intratumoral human effector T cells. Concomitant with LIGHT-mediated vascular remodeling, we observed intratumoral high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), which resemble spontaneous TLS found in GIST patients. Thus, by overcoming the limitations of immunodeficient xenograft models, we demonstrate the therapeutic feasibility of vascular targeting and immune priming in human GIST. Since TLS positively correlate with patient prognosis and improved response to immune checkpoint inhibition, vascular LIGHT targeting in GIST is a highly translatable approach to improve immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Larissa Dymond
- Translational Cancer Research Program, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Kira H. Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Edward R. Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Jiulia Satiaputra
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Anna Johansson-Percival
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Juliana Hamzah
- Imaging & Therapy Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | | | - Mohammed Ballal
- Department of General Surgery, Fiona Stanley Hospital, WesternAustralia, Australia
- Division of Surgery, School of Medicine, University of Western Australia, WesternAustralia, Australia
| | - Jonathan Foo
- Division of Surgery, School of Medicine, University of Western Australia, WesternAustralia, Australia
- Sir Charles Gairdner Hospital, QEII Medical Centre, Perth, WesternAustralia, Australia
| | - Mikael Johansson
- Sir Charles Gairdner Hospital, QEII Medical Centre, Perth, WesternAustralia, Australia
| | - Hooi C. Ee
- Sir Charles Gairdner Hospital, QEII Medical Centre, Perth, WesternAustralia, Australia
- Division of Internal Medicine, School of Medicine, University of Western Australia, WesternAustralia, Australia
| | - Scott W. White
- Division of Obstetrics and Gynaecology, Faculty of Medicine, Dentistry, and Health Sciences, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Louise Winteringham
- Translational Cancer Research Program, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
- Translational Cancer Research Program, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WesternAustralia, Australia
| |
Collapse
|
38
|
Stecko H, Iyer S, Tsilimigras D, Pawlik TM. Demographic Trends, Co-Alterations, and Imatinib Resistance across Genomic Variants in Gastrointestinal Stromal Tumors: An AACR Project GENIE Analysis. Oncology 2024; 103:327-340. [PMID: 39307135 DOI: 10.1159/000541454] [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: 06/21/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasm of the gastrointestinal tract, the treatment of which represents a significant breakthrough in targeted cancer therapy. Given its overall rare nature, genomic differences and clinical implications between demographic groups have not been previously investigated. METHODS Anonymized demographic, clinical, and genomic data from 1,559 GIST patients in the American Association for Cancer Research Project GENIE database were analyzed using cBioPortal and custom Python scripts. Data on patient demographics, genomic alterations, and co-occurrence genetic alerations were collected and classified according to clinical implications using the OncoKB database. χ2 tests for differences in genomic alterations were used across various demographic factors and mutual exclusivity analysis was employed to identify co-mutation patterns. RESULTS Male patients demonstrated higher incidence of PDGFRA mutation (14.56% vs. 8.05%; p < 0.001), while female patients had higher likelihood of NF1 mutations (7.46% vs. 3.23%; p = 0.001). Asian patients had higher alteration rates at KIT (85.59%; p = 0.002). Co-occurrence analysis revealed KIT alterations frequently co-occurred with CDKN2A (q < 0.001), MTAP (q = 0.045), and PTEN (q = 0.056), while there was mutual exclusivity with PDGFRA (q < 0.001), NF1 (q < 0.001), and BRAF (q = 0.015). CDKN2A alterations co-occurred with MTAP (q < 0.001) and PIK3CA (q = 0.015), while being mutually exclusive with TP53 (q = 0.002) and NF1 (q = 0.007). Trends were similar among patients who had received no prior medical treatment. Imatinib-resistant mutations were more common among male patients (25.6% vs. 18.9%; p = 0.0056) and individuals under 55 (27.3% vs. 20.9%; p = 0.0228). Among patients with imatinib-resistant mutations, 77.78% had sunitinib resistance, while 70.25% maintained sensitivity to ripretinib. CONCLUSION Sex and race/ethnic differences in genomic alterations, as well as co-mutations, were prevalent among patients with GIST. Variations in mutational profiles highlight the importance of distinct genetic drivers that may be targeted to treat different patient populations.
Collapse
Affiliation(s)
- Hunter Stecko
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sidharth Iyer
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Diamantis Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
39
|
Sun X, Lin X, Zhang Q, Li C, Shu P, Gao X, Shen K. Safety, effectiveness and the optimal duration of preoperative imatinib in locally advanced gastric gastrointestinal stromal tumors: A retrospective cohort study. Cancer Med 2024; 13:e70237. [PMID: 39300931 PMCID: PMC11413410 DOI: 10.1002/cam4.70237] [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: 01/04/2024] [Revised: 07/14/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The optimal duration of preoperative imatinib (IM) remains controversial. This study aimed to evaluate the safety, therapeutic effectiveness, and optimal duration of preoperative IM in patients with locally advanced gastric gastrointestinal stromal tumors (GIST). METHODS The clinicopathologic data of 41 patients with locally advanced gastric GIST who received preoperative IM and underwent surgical resection from January 2014 and December 2021 were retrospectively analyzed. RESULTS After a median of 7.0 (IQR: 4.5-10) months of preoperative IM treatment, 30 patients experienced adverse events (AEs), 80% of which were grade 1/2 AEs. The mean tumor size decreased from 12.71 ± 5.34 cm to 8.26 ± 4.00 cm, with a reduction rate of 35%. Setting 8 months as the cut-off value according to the results of ROC analysis. The proportion of laparoscopic surgery was higher in patients with short-term (≤8 months) versus long-term (>8 months) preoperative IM. Compared with the subtotal/total gastrectomy group, patients in the local gastrectomy group exhibited less intraoperative blood loss, shorter length of postoperative hospital stay, and fewer postoperative complications. The 3-year recurrence-free survival (RFS) and overall survival (OS) rates were 82.9% and 97.6%, and the expected 5-year RFS and OS rates were 75.6% and 90.2% respectively. RFS was better in the short-term than in the long-term preoperative IM treatment group, and it was also better in pre- plus postoperative IM treatment group than that in the preoperative IM alone group. Both univariate and multivariate COX analysis showed that a higher mitotic index and long-term preoperative IM treatment were associated with worse RFS, while postoperative IM treatment could significantly improve RFS. CONCLUSIONS The study suggests that in patients with locally advanced gastric GIST, preoperative short-term (≤8 months) use of IM is associated with higher RFS than long-term use.
Collapse
Affiliation(s)
- Xiangfei Sun
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| | - Xiaohan Lin
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| | - Qiang Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Chao Li
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| | - Ping Shu
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| | - Xiaodong Gao
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| | - Kuntang Shen
- Department of General Surgery, Zhongshan HospitalFudan University School of MedicineShanghaiChina
| |
Collapse
|
40
|
Xu Y, Lyu J, Xu Z, Chen X. Research trends on the gastrointestinal stromal tumor: A bibliometric analysis. Asian J Surg 2024; 47:4083-4084. [PMID: 38749831 DOI: 10.1016/j.asjsur.2024.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/02/2024] [Indexed: 09/05/2024] Open
Affiliation(s)
- Yang Xu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, People's Republic of China.
| | - JianBo Lyu
- Department of Radiology, The Second Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, People's Republic of China.
| | - Zhaohui Xu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, People's Republic of China.
| | - Xin Chen
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, People's Republic of China.
| |
Collapse
|
41
|
Denu RA, Somaiah N. Imatinib in advanced GIST: if it's working, don't stop a good thing. Lancet Oncol 2024; 25:1105-1107. [PMID: 39127065 DOI: 10.1016/s1470-2045(24)00403-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Affiliation(s)
- Ryan A Denu
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Ninomiya K, Ennishi D, Okamoto K, Ando M, Nakamura S, Tomida S, Ayada Y, Makimoto G, Ichihara E, Okita N, Toyooka S, Maeda Y, Tabata M. Response to Imatinib in a Patient With Gastric Adenocarcinoma With KIT Q556_K558 In-Frame Deletion: A Case Report. JCO Precis Oncol 2024; 8:e2400228. [PMID: 39298692 DOI: 10.1200/po.24.00228] [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: 04/04/2024] [Revised: 06/28/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Imatinib may be a useful targeted agent for patients with advanced gastric adenocarcinoma who have KIT mutations.
Collapse
Affiliation(s)
- Kiichiro Ninomiya
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Kunio Okamoto
- Department of Medical Oncology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Midori Ando
- Department of Pathology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Satoko Nakamura
- Department of Pathology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshiyuki Ayada
- Department of Pathology, Okayama University Hospital, Okayama, Japan
| | - Go Makimoto
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Eiki Ichihara
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Natsuko Okita
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Shinichi Toyooka
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Tabata
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
43
|
Ma Z, Zhao J, Li S, Gao F, Zhang C, Wu L, Lin Y. Imatinib-induced ulcerative colitis. J Oncol Pharm Pract 2024; 30:1111-1117. [PMID: 38772691 DOI: 10.1177/10781552241255290] [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] [Indexed: 05/23/2024]
Abstract
INTRODUCTION Imatinib, a tyrosine kinase inhibitor, is the first-line therapy for patients with KIT mutation in gastrointestinal stromal tumor (GIST). Nausea, vomiting, diarrhea, dyspepsia and abdominal pain are common gastrointestinal adverse reactions of imatinib, but imatinib-induced ulcerative colitis (UC) is rarely reported. CASE REPORT We presented a case of UC induced by imatinib in a 56-year-old male patient who experienced this adverse event after 5 years of imatinib 400 mg/d treatment following GIST resection. MANAGEMENT AND OUTCOME The patient's diarrhea and bloody stools showed significant improvement following the discontinuation of imatinib therapy and administration of antidiarrheal medications. Then, imatinib was restarted at a daily dosage of 400 mg. DISCUSSION UC is a rare adverse event associated with imatinib. Physicians should consider the possibility of UC induced by imatinib when patients present with diarrhea and bloody stool after receiving imatinib treatment. This case offered objective evidence of UC induced by imatinib.
Collapse
Affiliation(s)
- Zengqing Ma
- Department of Pharmacy, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Jianguo Zhao
- Department of Pharmacy, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Susu Li
- Department of Pharmacy, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Fuping Gao
- Department of Pathology, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Chuanyang Zhang
- Department of Medical Imaging, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Lianping Wu
- Department of Pharmacy, Nanjing Gaochun People's Hospital, Nanjing, China
| | - Yu Lin
- Department of Respiratory, Nanjing Gaochun People's Hospital, Nanjing, China
| |
Collapse
|
44
|
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
|
45
|
Ganzon R, Chen W, Tinoco G. First Description of the Clinical Activity of Avapritinib in Sporadic Mesenteric Desmoid Tumor. Case Rep Oncol Med 2024; 2024:8684418. [PMID: 39135981 PMCID: PMC11319063 DOI: 10.1155/2024/8684418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/17/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024] Open
Abstract
Desmoid tumors (DTs) are rare and locally aggressive with a high rate of local recurrence even with optimal surgical resection. Systemic treatments are often utilized for desmoid cases with high risk of surgical morbidity or for local and symptomatic control of recurrent disease. However, the systemic treatment options for DTs are limited with limited responses. Avapritinib is a tyrosine kinase inhibitor (TKI) approved in 2020 for adults with unresectable or metastatic gastrointestinal (GI) stromal tumors (GISTs) harboring a platelet-derived growth factor receptor alpha (PDGFRA) Exon 18 mutation, including D842V mutations. In this case report, we describe a 55-year-old man with a history of D842V-mutant gastric GIST who presented several years after complete resection of the GIST with an enlarging soft tissue mass in the small intestine. After a nondiagnostic biopsy, the patient was started on avapritinib due to concerns for recurrent D842V-mutant GIST. The tumor had a partial response to treatment by RECIST 1.1 criteria, and the patient underwent surgical resection. The final pathology report revealed a sporadic DT. To our knowledge, this is the first known description of the activity of avapritinib in the treatment of a sporadic mesenteric DT, which is relevant given the limited treatment options for patients with this diagnosis. This clinical finding may be worth exploring in a dedicated clinical trial.
Collapse
Affiliation(s)
- Rebecca Ganzon
- Division of Medical OncologyThe Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA
| | - Wei Chen
- Department of PathologyThe Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA
| | - Gabriel Tinoco
- Division of Medical OncologyThe Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, Ohio, USA
| |
Collapse
|
46
|
Beecroft JR, Brar S, Feng X, Hamilton T, Han-Lee C, Henning JW, Josephy PD, Khalili K, Ko YJ, Lemieux C, Liu DM, MacDonald DB, Noujaim J, Pollett A, Salawu A, Saleh R, Smrke A, Warren BE, Zbuk K, Razak AA. Pan-Canadian consensus recommendations for GIST management in high- and low-throughput centres across Canada. Ther Adv Med Oncol 2024; 16:17588359241266179. [PMID: 39386314 PMCID: PMC11461906 DOI: 10.1177/17588359241266179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/18/2024] [Indexed: 10/12/2024] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are mesenchymal tumours that originate from the interstitial cells of Cajal. GISTs are mainly driven by gain-of-function mutations in receptor tyrosine kinase or platelet-derived growth factor receptor alpha. Surgical resection is the only curative treatment for localized tumours and all currently approved medical GIST treatments are based on orally available tyrosine kinase inhibitors. Recent discoveries in the molecular and clinical features of GISTs have greatly impacted GIST management. Due to the provincially rather than nationally administered Canadian healthcare system, there have been inconsistencies in the treatment of GISTs across the country. Therefore, guidance on the latest knowledge, clinical management and treatment of GIST is needed to standardize the approach to GIST management nationwide. To establish pan-Canadian guidance, provide up-to-date data and harmonize the clinical practice of GIST management in high- and low-throughput centres across Canada; a panel of 20 physicians with extensive clinical experience in GIST management reviewed relevant literature. This included radiologists, pathologists, interventional radiologists, surgeons and medical oncologists across Canada. The structured literature focused on seven key domains: molecular profiling, radiological techniques/reporting, targeted localized therapy, intricacies of systemic treatments, emerging tests, multidisciplinary care and patient advocacy. This literature review, along with clinical expertise and opinion, was used to develop this concise and clinically relevant consensus paper to harmonize the knowledge and clinical practice on GIST management across Canada. The content presented here will help guide healthcare providers, especially in Canada, in terms of approaching and managing GIST.
Collapse
Affiliation(s)
- J. Robert Beecroft
- Division of Interventional Radiology, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital, Toronto, ON, Canada
| | - Savtaj Brar
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Xiaolan Feng
- Division of Medical Oncology, Tom Baker Cancer Center, Calgary, AB, Canada
| | - Trevor Hamilton
- Department of Surgery, BC Cancer, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Cheng Han-Lee
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - Jan-Willem Henning
- Department of Oncology, Tom Baker Cancer Centre, Cuming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Korosh Khalili
- Department of Medical Imaging, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Yoo-Joung Ko
- Department of Medicine, St. Michael’s Hospital, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Christopher Lemieux
- Division of Hematology and Medical Oncology, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec City, QC, Canada
| | - David M. Liu
- Department of Radiology, University of British Columbia, School of Biomedical Engineering, Vancouver, BC, Canada
- Department of Interventional Radiology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - D. Blair MacDonald
- Department of Medical Radiology, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan Noujaim
- Division of Medical Oncology, Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, QC, Canada
| | - Aaron Pollett
- Pathology and Laboratory Medicine, Division of Diagnostic Medical Genetics, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Abdulazeez Salawu
- Division of Medical Oncology, Princess Margaret Cancer Centre, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Ramy Saleh
- Division of Medical Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alannah Smrke
- Division of Medical Oncology, BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Blair E. Warren
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Kevin Zbuk
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Albiruni Abdul Razak
- Division of Medical Oncology, Princess Margaret Cancer Centre, Mount Sinai Hospital, University of Toronto, 610 University Ave., Toronto, ON M2G 2M9, Canada
| |
Collapse
|
47
|
Fukuda M, Mukohara T, Kuwata T, Sunami K, Naito Y. Efficacy of Trametinib in Neurofibromatosis Type 1-Associated Gastrointestinal Stromal Tumors: A Case Report. JCO Precis Oncol 2024; 8:e2300649. [PMID: 39116355 PMCID: PMC11371073 DOI: 10.1200/po.23.00649] [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: 11/28/2023] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Trametinib, an MEK inhibitor, may offer a new therapeutic option for patients with NF1-related GIST.
Collapse
Affiliation(s)
- Misao Fukuda
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takeshi Kuwata
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Yoichi Naito
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
48
|
Weeda YA, Kalisvaart GM, Hartgrink HH, van der Molen AJ, Gelderblom H, Bovée JV, de Geus-Oei LF, Grootjans W, van der Hage JA. Monitoring neoadjuvant treatment-induced surgical benefit in GIST patients using CT-based radiological criteria. Surg Open Sci 2024; 20:169-177. [PMID: 39886063 PMCID: PMC11780385 DOI: 10.1016/j.sopen.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 01/05/2025] Open
Abstract
Objective This single-centre retrospective study aims to determine the incidence of therapy-induced surgical benefit in patients with non-metastatic gastrointestinal stromal tumour (GIST) treated with neoadjuvant tyrosine kinase inhibitors (TKI) and evaluate whether this can be predicted by radiological response criteria. Methods Thirty-nine non-metastatic GIST patients were treated with neoadjuvant TKI treatment, followed by curative-intended surgery, and monitored using contrast-enhanced computed tomography (CE-CT). Surgical benefit was independently assessed by two surgical oncologists and was defined by de-escalation of surgical strategy or reduced surgical complexity. Radiological response between baseline and the last preoperative scan was determined through RECIST 1.1, Choi and volumetric criteria. Results In this patient cohort, median neoadjuvant treatment interval was 8.3 (IQR, 3.9-10.6) months. Surgical benefit was gained in 22/39 patients. When comparing radiological criteria to findings on surgical benefit, accuracy, sensitivity, and specificity for RECIST 1.1 (90 %, 100.0 % and 82 %), Choi (64 %, 24 %, and 96 %) and volumetry (95 %, 100.0 %, and 91 %) were calculated. In 30/39 patients, temporal changes in tumour size over the course of treatment was assessed. Tumour volume reduced significantly in the surgical-benefit group compared to the non-benefit group (72 % vs. 25 %, p < 0.01) within three months. 14/19 surgical-benefit patients had an initial volume reduction above 66 %, after which volume reduced slightly with a median 3.1 % (IQR, 2.1-7.8 %) reduction. Conclusion Surgical benefit after neoadjuvant treatment was achieved in 56 % of patients and was most accurately reflected by size-based response criteria. In patients with therapy-induced surgical benefit, nearly all treatment-induced volume reductions were achieved within three months.
Collapse
Affiliation(s)
- Ylva A. Weeda
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Gijsbert M. Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Henk H. Hartgrink
- Department of Surgical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Aart J. van der Molen
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Judith V.M.G. Bovée
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, the Netherlands
- Department of Radiation Science & Technology, Delft University of Technology, 2629 JB Delft, the Netherlands
| | - Willem Grootjans
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jos A. van der Hage
- Department of Surgical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
49
|
Awad A, Pal K, Yevich S, Kuban JD, Tam A, Odisio BC, Gupta S, Habibollahi P, Bishop AJ, Conley AP, Somaiah N, Araujo DM, Zarzour MA, Ratan R, Roland CL, Keung EZ, Huang SY, Sheth RA. Safety and efficacy of percutaneous image-guided ablation for soft tissue sarcoma metastases to the liver. Cancer 2024; 130:2703-2712. [PMID: 38642369 DOI: 10.1002/cncr.35330] [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: 01/26/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024]
Abstract
PURPOSE To evaluate outcomes following percutaneous image-guided ablation of soft tissue sarcoma metastases to the liver. MATERIALS AND METHODS A single-institution retrospective analysis of patients with a diagnosis of metastatic soft tissue sarcoma who underwent percutaneous image-guided ablation of hepatic metastases between January 2011 and December 2021 was performed. Patients with less than 60 days of follow-up after ablation were excluded. The primary outcome was local tumor progression-free survival (LPFS). Secondary outcomes included overall survival, liver-specific progression-free survival. and chemotherapy-free survival. RESULTS Fifty-five patients who underwent percutaneous ablation for 84 metastatic liver lesions were included. The most common histopathological subtypes were leiomyosarcoma (23/55), followed by gastrointestinal stromal tumor (22/55). The median treated liver lesions was 2 (range, 1-8), whereas the median size of metastases were 1.8 cm (0.3-8.7 cm). Complete response at 2 months was achieved in 90.5% of the treated lesions. LPFS was 83% at 1 year and 80% at 2 years. Liver-specific progression-free survival was 66% at 1 year and 40% at 2 years. The overall survival at 1 and 2 years was 98% and 94%. The chemotherapy-free holiday from the start of ablation was 71.2% at 12 months. The complication rate was 3.6% (2/55); one of the complications was Common Terminology Criteria for Adverse Events grade 3 or higher. LPFS subgroup analysis for leiomyosarcoma versus gastrointestinal stromal tumor suggests histology-agnostic outcomes (2 years, 89% vs 82%, p = .35). CONCLUSION Percutaneous image-guided liver ablation of soft tissue sarcoma metastases is safe and efficacious.
Collapse
Affiliation(s)
- Ahmed Awad
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Koustav Pal
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Yevich
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Joshua D Kuban
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Alda Tam
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Bruno C Odisio
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanjay Gupta
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Peiman Habibollahi
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew J Bishop
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Anthony Paul Conley
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Dejka M Araujo
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ravin Ratan
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Christina L Roland
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Emily Z Keung
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven Y Huang
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Rahul A Sheth
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
50
|
Tsai HJ, Shan YS, Yang CY, Hsiao CF, Tsai CH, Wang CC, Lin MT, Ting CF, Chan DC, Chen TH, Yen CC, Chen YY, Lin HY, Yeh TS, Ho CL, Shieh TY, Bai LY, Hsu JT, Chen IS, Chen LT, Yeh CN. Survival of advanced/recurrent gastrointestinal stromal tumors treated with tyrosine kinase inhibitors in Taiwan: a nationwide registry study. BMC Cancer 2024; 24:828. [PMID: 38992597 PMCID: PMC11238460 DOI: 10.1186/s12885-024-12567-1] [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: 10/05/2023] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Most gastrointestinal stromal tumors (GISTs) harbor c-KIT or PDGFRA mutations. Administration of tyrosine kinase inhibitors (TKIs) has significantly improved the survival of patients with GISTs. We aimed to evaluate the clinical outcome of advanced or recurrent GIST patients in Taiwan. METHODS Patients diagnosed between 2010 and 2020 were enrolled. The collected data included baseline characteristics, treatment pattern, treatment outcome, genetic aberrations and survival status. Progression-free survival (PFS) and overall survival (OS) were analyzed and plotted with the Kaplan-Meier method. Cox regression analysis was used to analyze the prognostic factors of survival. RESULTS A total of 224 patients with advanced or recurrent GISTs treated with TKIs were enrolled. All patients received imatinib treatment. Ninety-three and 42 patients received sunitinib and regorafenib treatment, respectively. The 48-month PFS and OS rates for patients treated with imatinib were 50.5% and 79.5%, respectively. c-KIT exon 9 and PDGFRA mutations were prognostic factors for a poor PFS and PDGFRA mutation was a prognostic factor for a poor OS in patients treated with imatinib in multivariate Cox regression analysis. The median PFS of patients who received sunitinib treatment was 12.76 months (95% confidence interval (CI), 11.01-14.52). Patients with c-KIT exon 9 mutations had a longer PFS than those with other genetic aberrations. The median PFS of patients treated with regorafenib was 7.14 months (95% CI, 3.39-10.89). CONCLUSIONS We present real-world clinical outcomes for advanced GIST patients treated with TKIs and identify mutational status as an independent prognostic factor for patient survival.
Collapse
Affiliation(s)
- Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chin-Fu Hsiao
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chung-Hsin Tsai
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Chuan-Cheng Wang
- Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chun-Fu Ting
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - De-Chuan Chan
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-Hung Chen
- Department of Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chueh-Chuan Yen
- Division of Medical Oncology, Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Yang Chen
- Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsuan-Yu Lin
- Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Ta-Sen Yeh
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Liang Ho
- Division of Hematology and Oncology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Hematology and Oncology, Medical Department, Taipei Tzu Chi Hospital, Taipei, Taiwan
| | - Tze-Yu Shieh
- Division of Gastroenterology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Li-Yaun Bai
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jun-Te Hsu
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan, Taiwan
| | - I-Shu Chen
- Division of General Surgery, Department of Surgery, Kaohsiung Veterans General Hospital and National Yang Ming Chiao Tung University, Kaohsiung, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Chun-Nan Yeh
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan, Taiwan.
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan.
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|