101
|
Annicchiarico A, Montali F, Baldinu M, Casali L, Virgilio E, Costi R. Leiomyosarcoma of the rectum: A systematic review of recent literature. J Surg Oncol 2024; 129:365-380. [PMID: 37814590 DOI: 10.1002/jso.27481] [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: 07/11/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Leiomyosarcomas (LMSs) are rare tumors originating from the muscular layer. We performed a literature review of cases of confirmed rectal leiomyosarcomas (rLMSs) to clarify the history of such an infrequent tumor arising at such an uncommon location. In this research local recurrence was related to poorly differentiated rLMS and no other association between recurrence and any criteria was found. Concerning overall survival (OS), rLMS patients developing recurrence presented shorter longevity compared with the group without.
Collapse
Affiliation(s)
- Alfredo Annicchiarico
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of General Surgery, Vaio Hospital, Fidenza, Italy
| | - Filippo Montali
- Department of General Surgery, Vaio Hospital, Fidenza, Italy
| | - Manuel Baldinu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Lorenzo Casali
- Department of General Surgery, Vaio Hospital, Fidenza, Italy
| | - Edoardo Virgilio
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of General Surgery, Vaio Hospital, Fidenza, Italy
| | - Renato Costi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of General Surgery, Vaio Hospital, Fidenza, Italy
| |
Collapse
|
102
|
Bleckman RF, Broekman KE, Roets E, Mohammadi M, Desar IME, Gelderblom H, Mathijssen RHJ, Steeghs N, de Graeff P, Reyners AKL. Individualized Dosing Patterns in the Treatment of Older Patients with Gastrointestinal Stromal Tumors: Results of a Registry-Based Observational National Cohort Study Including 871 Patients. Drugs Aging 2024; 41:165-176. [PMID: 38123766 DOI: 10.1007/s40266-023-01084-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND While the effectiveness of tyrosine kinase inhibitors (TKIs) seems similar in older patients with gastrointestinal stromal tumors (GIST) compared with younger patients, toxicities in older patients treated with TKIs more often lead to discontinuation of treatment. OBJECTIVE To better understand the age-related pharmacology and pharmacodynamic differences in patients with GIST treated with TKIs, the primary aim of this study was to evaluate TKI dosing patterns in older patients with GIST, while the secondary aims were to evaluate differences in imatinib trough plasma concentrations between age groups and to compare the overall survival (OS) in patients with and without dose reductions in all treatment lines in a palliative setting. METHODS Patients (18 years of age or older) with histologically proven GIST diagnosed between January 2009 and June 2021 and treated with one or more lines of TKIs were selected from the Dutch GIST Registry (DGR) database. Age groups were divided into younger patients (age <70 years) and older patients (age ≥70 years). All imatinib trough plasma concentrations of blood withdrawals taken from initiation of imatinib until a maximum of 1 year of treatment with imatinib were collected. Reasons for first adjustment of treatment were classified as adverse event, dose modification, progressive disease and other reasons. The next treatment steps after first adjustment of treatment were defined as dose escalation, dose reduction, dose interruption, or end of treatment. The association of dose reduction and OS was analyzed using the landmark approach. RESULTS Overall, 871 patients were included in this study, including 577 younger patients and 294 older patients. Older patients more often had an adverse event as the reason for first adjustment of treatment with both imatinib (45.6%; p < 0.001) and sunitinib (58.6%; p = 0.224) compared with younger patients (19.5% and 42.7%, respectively). Adjustment of imatinib and sunitinib after starting on a standard dose because of an adverse event most often resulted in dose reduction in both age groups. Median trough plasma concentrations of all samples taken within the first year after initiation of imatinib were higher in older patients (1228 ng/mL, interquartile range [IQR] 959-1687) compared with younger patients (1035 ng/mL [IQR 773-1377]; p < 0.001). No significant differences were seen between OS in patients with or without dose reduction in all treatment lines (imatinib: p = 0.270; sunitinib: p = 0.547; and regorafenib: p = 0.784). CONCLUSION Older patients showed higher imatinib trough plasma concentrations compared with younger patients and also had earlier and more often adverse events as the reason for first adjustment of treatment with imatinib followed by dose reduction. However, in a landmark analysis, patients with imatinib dose reductions had no poorer outcomes compared with patients not requiring a dose reduction.
Collapse
Affiliation(s)
- Roos F Bleckman
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - K Esther Broekman
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Evelyne Roets
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | - Neeltje Steeghs
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Pauline de Graeff
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Anna K L Reyners
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
103
|
Sasa K, Son R, Oguchi A, Ashizawa K, Hasegawa N, Kubota D, Suehara Y, Takagi T, Okubo T, Akaike K, Sugimoto K, Takahashi M, Sakamoto K, Hashimoto T, Mine S, Fukunaga T, Ishijima M, Hayashi T, Yao T, Murakawa Y, Saito T. NTRK2 expression in gastrointestinal stromal tumors with a special emphasis on the clinicopathological and prognostic impacts. Sci Rep 2024; 14:768. [PMID: 38191907 PMCID: PMC10774370 DOI: 10.1038/s41598-024-51211-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are typically characterized by activating mutations of the KIT proto-oncogene receptor tyrosine kinase (KIT) or platelet-derived growth factor receptor alpha (PDGFRA). Recently, the neurotrophic tyrosine receptor kinase (NTRK) fusion was reported in a small subset of wild-type GIST. We examined trk IHC and NTRK gene expressions in GIST. Pan-trk immunohistochemistry (IHC) was positive in 25 (all 16 duodenal and 9 out of 16 small intestinal GISTs) of 139 cases, and all pan-trk positive cases showed diffuse and strong expression of c-kit. Interestingly, all of these cases showed only trkB but not trkA/trkC expression. Cap analysis of gene expression (CAGE) analysis identified increased number of genes whose promoters were activated in pan-trk/trkB positive GISTs. Imbalanced expression of NTRK2, which suggests the presence of NTRK2 fusion, was not observed in any of trkB positive GISTs, despite higher mRNA expression. TrkB expression was found in duodenal GISTs and more than half of small intestinal GISTs, and this subset of cases showed poor prognosis. However, there was not clear difference in clinical outcomes according to the trkB expression status in small intestinal GISTs. These findings may provide a possible hypothesis for trkB overexpression contributing to the tumorigenesis and aggressive clinical outcome in GISTs of duodenal origin.
Collapse
Affiliation(s)
- Keita Sasa
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Raku Son
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Akiko Oguchi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Karin Ashizawa
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobuhiko Hasegawa
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Daisuke Kubota
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Tatsuya Takagi
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Taketo Okubo
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
| | - Keisuke Akaike
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
- Department of Orthopaedic Surgery, Yamanashi Central Hospital, Yamanashi, Japan
| | - Kiichi Sugimoto
- Department of Coloproctological Surgery, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
| | - Makoto Takahashi
- Department of Coloproctological Surgery, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
| | - Kazuhiro Sakamoto
- Department of Coloproctological Surgery, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
| | - Takashi Hashimoto
- Department of Upper Gastroenterological Surgery, Juntendo University Hospital, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Shinji Mine
- Department of Upper Gastroenterological Surgery, Juntendo University Hospital, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Tetsu Fukunaga
- Department of Upper Gastroenterological Surgery, Juntendo University Hospital, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University School of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Yao
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuhiro Murakawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan.
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, 113-8421, Japan.
| |
Collapse
|
104
|
Incorvaia L, De Biase D, Nannini M, Fumagalli E, Vincenzi B, De Luca I, Brando C, Perez A, Pantaleo MA, Gasperoni S, D’Ambrosio L, Grignani G, Maloberti T, Pedone E, Bazan Russo TD, Mazzocca A, Algeri L, Dimino A, Barraco N, Serino R, Gristina V, Galvano A, Bazan V, Russo A, Badalamenti G. KIT/PDGFRA Variant Allele Frequency as Prognostic Factor in Gastrointestinal Stromal Tumors (GISTs): Results From a Multi-Institutional Cohort Study. Oncologist 2024; 29:e141-e151. [PMID: 37463014 PMCID: PMC10769785 DOI: 10.1093/oncolo/oyad206] [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: 04/04/2023] [Accepted: 06/09/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND The patient selection for optimal adjuvant therapy in gastrointestinal stromal tumors (GISTs) is provided by nomogram based on tumor size, mitotic index, tumor location, and tumor rupture. Although mutational status is not currently used to risk assessment, tumor genotype showed a prognostic influence on natural history and tumor relapse. Innovative measures, such as KIT/PDGFRA-mutant-specific variant allele frequency (VAF) levels detection from next-generation sequencing (NGS), may act as a surrogate of tumor burden and correlate with prognosis and overall survival of patients with GIST, helping the choice for adjuvant treatment. PATIENTS AND METHODS This was a multicenter, hospital-based, retrospective/prospective cohort study to investigate the prognostic role of KIT or PDGFRA-VAF of GIST in patients with radically resected localized disease. In the current manuscript, we present the results from the retrospective phase of the study. RESULTS Two-hundred (200) patients with GIST between 2015 and 2022 afferent to 6 Italian Oncologic Centers in the EURACAN Network were included in the study. The receiver operating characteristic (ROC) curves analysis was used to classify "low" vs. "high" VAF values, further normalized on neoplastic cellularity (nVAF). When RFS between the low and high nVAF groups were compared, patients with GIST with KIT/PDGFRA nVAF > 50% showed less favorable RFS than patients in the group of nVAF ≤ 50% (2-year RFS, 72.6% vs. 93%, respectively; P = .003). The multivariable Cox regression model confirmed these results. In the homogeneous sub-population of intermediate-risk, patients with KIT-mutated GIST, the presence of nVAF >50% was statistically associated with higher disease recurrence. CONCLUSION In our study, we demonstrated that higher nVAF levels were independent predictors of GIST prognosis and survival in localized GIST patients with tumors harboring KIT or PDGFRA mutations. In the cohort of intermediate-risk patients, nVAF could be helpful to improve prognostication and the use of adjuvant imatinib.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Dario De Biase
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Margherita Nannini
- Department of Experimental, Diagnostic and Specialized Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Elena Fumagalli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Biomedico University of Rome, Rome, Italy
| | - Ida De Luca
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Maria A Pantaleo
- Department of Experimental, Diagnostic and Specialized Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Silvia Gasperoni
- Department of Oncology and Robotic Surgery, Translational Oncology Unit, University Hospital Careggi, Firenze, Italy
| | - Lorenzo D’Ambrosio
- Division of Medical Oncology, Candiolo Cancer Institute, FPO - IRCCS, Candiolo, TO, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Candiolo Cancer Institute, FPO - IRCCS, Candiolo, TO, Italy
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Erika Pedone
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandro Mazzocca
- Department of Medical Oncology, Campus Biomedico University of Rome, Rome, Italy
| | - Laura Algeri
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandra Dimino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Roberta Serino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
105
|
Teuber A, Schulz T, Fletcher BS, Gontla R, Mühlenberg T, Zischinsky ML, Niggenaber J, Weisner J, Kleinbölting SB, Lategahn J, Sievers S, Müller MP, Bauer S, Rauh D. Avapritinib-based SAR studies unveil a binding pocket in KIT and PDGFRA. Nat Commun 2024; 15:63. [PMID: 38167404 PMCID: PMC10761696 DOI: 10.1038/s41467-023-44376-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Avapritinib is the only potent and selective inhibitor approved for the treatment of D842V-mutant gastrointestinal stromal tumors (GIST), the most common primary mutation of the platelet-derived growth factor receptor α (PDGFRA). The approval was based on the NAVIGATOR trial, which revealed overall response rates of more than 90%. Despite this transformational activity, patients eventually progress, mostly due to acquired resistance mutations or following discontinuation due to neuro-cognitive side effects. These patients have no therapeutic alternative and face a dismal prognosis. Notable, little is known about this drug's binding mode and its medicinal chemistry development, which is instrumental for the development of the next generation of drugs. Against this background, we solve the crystal structures of avapritinib in complex with wild-type and mutant PDGFRA and stem cell factor receptor (KIT), which provide evidence and understanding of inhibitor binding and lead to the identification of a sub-pocket (Gα-pocket). We utilize this information to design, synthesize and characterize avapritinib derivatives for the determination of key pharmacophoric features to overcome drug resistance and limit potential blood-brain barrier penetration.
Collapse
Grants
- BA 5214/1-2 Deutsche Forschungsgemeinschaft (German Research Foundation)
- This work was co-funded by the German Research Foundation (DFG; BA 5214/1-2 (SB) | RA 1055/3-2 (DR)), the State of North Rhine-Westphalia (NRW), the European Union (European Regional Development Fund: Investing In Your Future) (EFRE-800400), DDHD (Drug Discovery Hub Dortmund, (DR)), the German Federal Ministry of Education and Research (InCa (01ZX2201B, (DR)), the Mercator Research Center Ruhr (MERCUR), IGNITE (Ex-2021-0033, (DR and SB)) and was supported by the "Netzwerke 2021" program, an initiative of the Ministry of Culture and Science of the State of North Rhine-Westphalia (CANcer TARgeting, NW21-062C, (DR and SB)). This work was supported by the Swiss Light Source of the Paul Scherrer Institute (SLS, Villingen, Switzerland) and The European Synchrotron Radiation Facility (ESRF, Grenoble, France, proposal MX-2391, DOI: 10.15151/ESRF-ES-744176088 and DOI: 10.15151/ESRF-ES-925653639, (DR and MPM)).
Collapse
Affiliation(s)
- A Teuber
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - T Schulz
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - B S Fletcher
- Department of Medical Oncology and Sarcoma Center and West German Cancer Center, DKTK partner site Essen, German Cancer Consortium (DKTK), University Duisburg-Essen, Medical School, Essen, Germany
| | - R Gontla
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - T Mühlenberg
- Department of Medical Oncology and Sarcoma Center and West German Cancer Center, DKTK partner site Essen, German Cancer Consortium (DKTK), University Duisburg-Essen, Medical School, Essen, Germany
| | - M-L Zischinsky
- Lead Discovery Center GmbH, Department for in vitro ADME and PK, Otto-Hahn-Strasse 15, 44227, Dortmund, Germany
| | - J Niggenaber
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - J Weisner
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - S B Kleinbölting
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - J Lategahn
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - S Sievers
- Compound Management and Screening Center, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - M P Müller
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany
| | - S Bauer
- Department of Medical Oncology and Sarcoma Center and West German Cancer Center, DKTK partner site Essen, German Cancer Consortium (DKTK), University Duisburg-Essen, Medical School, Essen, Germany
| | - D Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227, Dortmund, Germany.
| |
Collapse
|
106
|
Sharobim M, Matkovic E, Schwalbe M, Matkowskyj KA. Pathologic Features of Miscellaneous Foregut Malignancies. Cancer Treat Res 2024; 192:49-66. [PMID: 39212915 DOI: 10.1007/978-3-031-61238-1_3] [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: 09/04/2024]
Abstract
Neuroendocrine neoplasms are a heterogeneous group of tumors that can occur in almost any organ and share a common neuroendocrine phenotype.
Collapse
Affiliation(s)
- Mark Sharobim
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Eduard Matkovic
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | | | |
Collapse
|
107
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [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: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
108
|
Liu A, Zhang S, Wang M, Zhang L, Xu S, Nasimian A, Li S, Zhao S, Cao X, Tian J, Yu Y, Fan Z, Xiao K, Zhao H, Kazi JU, Ma L, Sun J. DDR1/2 enhance KIT activation and imatinib resistance of primary and secondary KIT mutants in gastrointestinal stromal tumors. Mol Carcinog 2024; 63:75-93. [PMID: 37737519 DOI: 10.1002/mc.23637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) are predominantly initiated by KIT mutations. In this study, we observed that discoidin domain receptors 1 and 2 (DDR1 and DDR2) exhibited high expression in GISTs, were associated with KIT, and enhanced the activation of both wild-type KIT and primary KIT mutants. Inhibition of DDR1/2 led to a reduction in the activation of KIT and its downstream signaling molecules, ultimately impairing GIST cell survival and proliferation in vitro. Consequently, treatment of mice carrying germline KIT/V558A mutation with DDR1/2 inhibitor significantly impeded tumor growth, and the combined use of DDR1/2 inhibitor and imatinib, the first-line targeted therapeutic agent for GISTs, markedly enhanced tumor growth suppression. In addition, DDR1/2 inhibition resulted in decreased KIT expression, while KIT inhibition led to upregulation of DDR1/2 expression in GISTs. The presence of DDR1/2 also decreased the sensitivity of wild-type KIT or primary KIT mutants to imatinib, indicating a possible role for DDR1/2 in promoting GIST survival during KIT-targeted therapy. The development of drug-resistant secondary KIT mutations is a primary factor contributing to GIST recurrence following targeted therapy. Similar to primary KIT mutants, DDR1/2 can associate with and enhance the activation of secondary KIT mutants, further diminishing their sensitivity to imatinib. In summary, our data demonstrate that DDR1/2 contribute to KIT activation in GISTs and strengthen resistance to imatinib for both primary and secondary KIT mutants, providing a rationale for further exploration of DDR1/2 targeting in GIST treatment.
Collapse
Affiliation(s)
- Anbu Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Shaoting Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ming Wang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Liangying Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Shidong Xu
- Department of Oncology, School of Medicine, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ahmad Nasimian
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Shujing Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Department of Pediatrics, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Sien Zhao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xu Cao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jinhai Tian
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Yu
- Department of Emergency, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhaoyang Fan
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Kun Xiao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Julhash U Kazi
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Lijun Ma
- Department of Oncology, School of Medicine, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianmin Sun
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
109
|
Tassinari E, Conci N, Battisti G, Porta F, Di Scioscio V, Pirini MG, de Biase D, Nigro MC, Iezza M, Castagnetti F, Lovato L, Fanti S, Pantaleo MA, Nannini M. Metabolic pseudoprogression in a patient with metastatic KIT exon 11 GIST after 1 month of first-line imatinib: a case report. Front Oncol 2023; 13:1310452. [PMID: 38188286 PMCID: PMC10769864 DOI: 10.3389/fonc.2023.1310452] [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: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Positron emission tomography (PET) with 18-fluorodeoxyglucose (18FDG) has proven to be highly sensitive in the early assessment of tumor response in gastrointestinal stromal tumors (GIST), especially in cases where there is doubt or when the early prediction of the response could be clinically useful for patient management. As widely known, kinase mutations have an undoubtful predictive value for sensitivity to imatinib, and the inclusion of KIT and PDGFRa mutational analysis in the diagnostic workup of all GIST is now considered standard practice. CASE PRESENTATION Herein, we described in detail a case of an exon 11 KIT mutated-metastatic GIST patient, who presented an unexpected metabolic progression at the early 18FDG-PET evaluation after 1 month of first-line imatinib, unconfirmed at the liver biopsy performed near after, which has conversely shown a complete pathological response. CONCLUSIONS This report aims to highlight the existence of this metabolic pseudoprogression in GIST at the beginning of imatinib therapy in order to avoid early treatment discontinuation. Therefore, an early metabolic progression during a molecular targeted therapy always deserves to be evaluated in the context of the disease molecular profiling, and in case of a discordant finding between functional imaging and molecular background, a short-term longitudinal control should be suggested.
Collapse
Affiliation(s)
- Elisa Tassinari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Nicole Conci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Giacomo Battisti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna-Policlinico di Sant’Orsola, Bologna, Italy
| | - Francesco Porta
- Department of Pediatric and Adult Cardio-Thoracovascular, Oncoematologic and Emergencies Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valerio Di Scioscio
- Department of Pediatric and Adult Cardio-Thoracovascular, Oncoematologic and Emergencies Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria Giulia Pirini
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna-Policlinico di Sant’Orsola, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Maria Concetta Nigro
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Miriam Iezza
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Hematology “Lorenzo E Ariosto Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Fausto Castagnetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Hematology “Lorenzo E Ariosto Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Luigi Lovato
- Department of Pediatric and Adult Cardio-Thoracovascular, Oncoematologic and Emergencies Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna-Policlinico di Sant’Orsola, Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Margherita Nannini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
110
|
Giger OT, Ten Hoopen R, Shorthouse D, Abdullahi S, Bulusu VR, Jadhav S, Maher ER, Casey RT. Preferential MGMT hypermethylation in SDH-deficient wild-type GIST. J Clin Pathol 2023; 77:34-39. [PMID: 36198483 PMCID: PMC10804026 DOI: 10.1136/jcp-2022-208462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022]
Abstract
AIMS Wild-type gastrointestinal stromal tumours (wtGIST) are frequently caused by inherited pathogenic variants, or somatic alterations in the succinate dehydrogenase subunit genes (SDHx). Succinate dehydrogenase is a key enzyme in the citric acid cycle. SDH deficiency caused by SDHx inactivation leads to an accumulation of succinate, which inhibits DNA and histone demethylase enzymes, resulting in global hypermethylation. Epigenetic silencing of the DNA repair gene MGMT has proven utility as a positive predictor of the therapeutic efficacy of the alklyating drug temozolomide (TMZ) in tumours such as glioblastoma multiforme. The aim of this study was to examine MGMT promoter methylation status in a large cohort of GIST. METHODS MGMT methylation analysis was performed on 65 tumour samples including 47 wtGIST (33 SDH-deficient wtGIST and 11 SDH preserved wtGIST) and 21 tyrosine kinase (TK) mutant GIST. RESULTS MGMT promoter methylation was detected in 8 cases of SDH-deficient (dSDH) GIST but in none of the 14 SDH preserved wild-type GIST or 21 TK mutant GIST samples analysed. Mean MGMT methylation was significantly higher (p 0.0449) and MGMT expression significantly lower (p<0.0001) in dSDH wtGIST compared with TK mutant or SDH preserved GIST. No correlation was identified between SDHx subunit gene mutations or SDHC epimutation status and mean MGMT methylation levels. CONCLUSION MGMT promoter hypermethylation occurs exclusively in a subset of dSDH wtGIST. Data from this study support testing of tumour MGMT promoter methylation in patients with dSDH wtGIST to identify those patients who may benefit from most from TMZ therapy.
Collapse
Affiliation(s)
| | | | - David Shorthouse
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | | | | | - Saili Jadhav
- Oncology, University of Cambridge, Cambridge, UK
| | - Eamonn R Maher
- Department of Medical Genetics and Cancer Research, University of Cambridge, Cambridge, UK
| | - Ruth T Casey
- Department of Medical Genetics and Cancer Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
111
|
Shi J, Sun K, Kong F, Shen D. Morphological, immunohistochemical, and genetic analyses of epithelioid gastrointestinal stromal tumors. Ann Diagn Pathol 2023; 67:152208. [PMID: 37696133 DOI: 10.1016/j.anndiagpath.2023.152208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
Epithelioid gastrointestinal stromal tumors (GISTs) are rare and may be confused with other tumors with epithelioid morphology. Therefore, herein, we collected 12 epithelioid GIST samples and summarized their morphological and immunohistochemical characteristics. Through genetic testing, we explored the correlation between morphology and gene mutations. The results showed that eight tumors showed focal or diffuse myxoid stromal changes with less cohesively arranged rhabdoid tumor cells; among these, five showed platelet-derived growth factor receptor alpha gene (PDGFRA) mutations. Signet ring cells with sclerosing stroma and receptor tyrosine kinase type III gene (KIT) mutations were present in two cases, which might be a KIT mutation-associated growth pattern in epithelioid GISTs. Succinate dehydrogenase gene (SDH) mutations were detected in three cases. Simultaneously, PDGFRA mutations were detected in two cases, and the Kirsten rat sarcoma viral oncogene homolog gene (KRAS) mutation was detected in another case. SDH-subunit B (SDHB) expression was partially weak and strongly diffuse in two cases with concurrent PDGFRA and SDHD mutations, respectively. The coexistence of PDGFRA and SDHD mutations may have affected SDHB expression. Altogether, we concluded that PDGFRA mutations may play an important role in co-mutant GIST pathogenesis.
Collapse
Affiliation(s)
- Jingli Shi
- Department of Pathology, Peking University People's Hospital, 100044, China
| | - Kunkun Sun
- Department of Pathology, Peking University People's Hospital, 100044, China
| | - Fangzhou Kong
- Department of Pathology, Peking University People's Hospital, 100044, China
| | - Danhua Shen
- Department of Pathology, Peking University People's Hospital, 100044, China.
| |
Collapse
|
112
|
Li C, Wang Q, Jiang KW, Ye YJ. Hallmarks and novel insights for gastrointestinal stromal tumors: A bibliometric analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107079. [PMID: 37826966 DOI: 10.1016/j.ejso.2023.107079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Due to the increasing recognition of gastrointestinal stromal tumor (GIST), novel insights have appeared in both preclinical and clinical research and begun to reshape the field. This study aims to map the research landscape through bibliometric analysis and provide a brief overview for the future of the GIST field. METHODS We searched the Web of Science Core Collection without publication data restrictions for GISTs and performed a bibliometric analysis with CiteSpace and VOSviewer software. RESULTS In sum, 5,911 of 13,776 records were included, and these studies were published in 948 journals and written by 24,965 authors from 4,633 institutions in 100 countries. Referring to published reviews and bibliometric analysis, we classified the future trends in four groups. In epidemiological study, precise incidence and clinicopathological features in different regions and races might become potential hotspots. Novel therapy, such as drugs, modified strategies, radioligand therapy, was persistent hotspots in GIST fields, and ctDNA-guided diagnosis, monitoring, and treatment might meet future clinical needs. The debate over serosa surgery vs. mucosa surgery will remain active for a long time in GIST surgery, and function reserve surgery, biology-based surgery will play an important role in future. Moreover, rare GIST type, like NF-1-associated GIST, Carney triads and SDH mutant GIST, need more studies in pathogenesis and genetic mutation to provide appropriate treatment for this orphan GIST patients. CONCLUSIONS Potential hotspots in future GIST trends might involve epidemiology, agents, resection therapy and rare type GIST, moreover, researchers could pay more attention in these four fields.
Collapse
Affiliation(s)
- Chen Li
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China
| | - Quan Wang
- Ambulatory Surgery Center, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, China
| | - Ke-Wei Jiang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China.
| | - Ying-Jiang Ye
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
113
|
Schaefer MA, Roy P, Chava S, Meyerson A, Duncan AL, Chee L, Hewitt KJ. Physiological and regenerative functions of sterile-α motif protein-14 in hematopoiesis. Exp Hematol 2023; 128:38-47. [PMID: 37722652 PMCID: PMC10947990 DOI: 10.1016/j.exphem.2023.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Sterile α-motif domain-14 (Samd14) protein expression increases the regenerative capacity of the erythroid system. Samd14 is transcriptionally upregulated and promotes cell signaling via the receptor tyrosine kinase Kit in a critical window of acute erythroid regeneration. We generated a hematopoietic-specific conditional Samd14 knockout mouse model (Samd14-CKO) to study the role of Samd14 in hematopoiesis. The Samd14-CKO mouse was viable and exhibited no steady-state hematopoietic phenotype. Samd14-CKO mice were hypersensitive to 5-fluorouracil, resulting in more severe anemia during recovery and impaired erythroid progenitor colony formation. Ex vivo, Samd14-CKO hematopoietic progenitors were defective in their ability to form mast cells. Samd14-CKO mast cells exhibited altered Kit/stem cell factor (SCF), IL-3/IL-3R signaling, and less granularity than Samd14-FL/FL cells. Our findings indicate that Samd14 promotes both erythroid and mast cell functions. The Samd14-CKO mouse phenotype exhibits striking similarities to the KitW/W-v mice, which carry Kit mutations resulting in reduced tyrosine kinase-dependent signaling, causing mast cell and erythroid abnormalities. The Samd14-CKO mouse model is a new tool for studying hematologic pathologies involving Kit signaling.
Collapse
Affiliation(s)
- Meg A Schaefer
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Pooja Roy
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Srinivas Chava
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Ainsley Meyerson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Andrew L Duncan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Linda Chee
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Kyle J Hewitt
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
114
|
Towery EA, Papke DJ. Emerging mesenchymal tumour types and biases in the era of ubiquitous sequencing. J Clin Pathol 2023; 76:802-812. [PMID: 37550012 DOI: 10.1136/jcp-2022-208684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023]
Abstract
New tumour types are being described at increasing frequency, and most new tumour types are now identified via retrospective review of next-generation sequencing data. This contrasts with the traditional, morphology-based method of identifying new tumour types, and while the sequencing-based approach has accelerated progress in the field, it has also introduced novel and under-recognised biases. Here, we discuss tumour types identified based on morphology, including superficial CD34-positive fibroblastic tumour, pseudoendocrine sarcoma and cutaneous clear cell tumour with melanocytic differentiation and ACTIN::MITF fusion. We also describe tumour types identified primarily by next-generation sequencing, including epithelioid and spindle cell rhabdomyosarcoma, round cell neoplasms with EWSR1::PATZ1 fusion, cutaneous melanocytic tumour with CRTC1::TRIM11 fusion, clear cell tumour with melanocytic differentiation and MITF::CREM fusion and GLI1-altered mesenchymal neoplasms, including nested glomoid neoplasm.
Collapse
|
115
|
Abstract
Total synthesis of (-)-enigmazole B was achieved for the first time. Highlights of the present synthesis include an olefin cross-metathesis and hemiacetalization/intramolecular oxa-Michael addition sequence for accessing an (E)-configured enol tosylate, a Sonogashira cross-coupling to assemble all the carbon atoms of the target natural product, a remarkably chemo- and regioselective Au-catalyzed intramolecular alkyne hydroalkoxylation for the construction of the dihydropyran ring, and a Yamaguchi macrolactonization to close the 18-membered macrolactone skeleton.
Collapse
Affiliation(s)
- Yoshihiro Goda
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Haruhiko Fuwa
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| |
Collapse
|
116
|
Fleming AM, Herb J, Stiles ZE, Burkbauer L, Dickson PV, Glazer ES, Shibata D, Murphy AJ, Davidoff AM, Gleeson E, Kim HJ, Meyers MO, Stitzenberg K, Ollila DW, Deneve JL. Lymph node metastases in young patients with gastrointestinal stromal tumor: A nationwide analysis. J Surg Oncol 2023; 128:1268-1277. [PMID: 37650827 DOI: 10.1002/jso.27431] [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: 05/31/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Children, adolescents, and young adults (CAYA) (age ≤39 years) with GIST have high rates of LNM, but their clinical relevance is undefined. This study analyzed the impact of LNM on overall survival (OS) for CAYA with GIST. METHODS The National Cancer Database was queried for patients with resected GIST and pathologic nodal staging data from 2004-2019. Factors associated with LNM were identified. Survival was assessed stratified by presence of LNM. RESULTS Of 4420 patients with GIST, 238 were CAYA (5.4%). When compared to older adults, CAYA more often had small intestine primaries (51.8% vs. 36.6%, p < 0.0001), T4 tumors (30.7% vs. 24.5%, p = 0.0275) and pN1 disease (11.3% vs. 4.7%, p < 0.0001). Within a multivariable Cox proportional hazards regression model adjusting for age, comorbid disease, mitotic rate, tumor size, and primary site, LNM were associated with increased hazard of death for older adults (hazard ratio [HR]: 1.83; confidence interval [CI]: 1.35-2.42; p < 0.0001), but not CAYA (HR: 3.38; CI: 0.50-14.08; p = 0.13). For CAYA, only high mitotic rate predicted mortality (HR: 4.68; CI: 1.41-18.37: p = 0.02). CONCLUSIONS LNM are more commonly identified among CAYA with resected GIST who undergo lymph node evaluations, but do not appear to impact OS as observed in older adults. High mitotic rate remains a predictor of poor outcomes for CAYA with GIST.
Collapse
Affiliation(s)
- Andrew M Fleming
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Joshua Herb
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Zachary E Stiles
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Laura Burkbauer
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Paxton V Dickson
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Evan S Glazer
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - David Shibata
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Andrew J Murphy
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Andrew M Davidoff
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Elizabeth Gleeson
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hong J Kim
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael O Meyers
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Karyn Stitzenberg
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - David W Ollila
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jeremiah L Deneve
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
117
|
Trotter N, White J. Gastrointestinal stromal tumour-induced hypercalcaemia. BMJ Case Rep 2023; 16:e243613. [PMID: 37967927 PMCID: PMC10660897 DOI: 10.1136/bcr-2021-243613] [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: 11/17/2023] Open
Abstract
Hypercalcaemia is recognised as the most common oncological metabolic emergency, with several proposed underlying mechanisms. Nevertheless, hypercalcaemia has been rarely reported as a complication in patients with gastrointestinal stromal tumours (GISTs). GISTs are uncommon mesenchymal tumours of the gastrointestinal tract. There are only nine previous cases of hypercalcaemia occurring in patients with GIST reported in the literature. We report a case of a man in his 70s with a background of metastatic GIST on fourth-line treatment. The patient presented with new hypercalcaemia and acute kidney injury. Despite medical management, his calcium remained elevated and he deteriorated secondary to significant disease progression.
Collapse
Affiliation(s)
- Nicola Trotter
- Internal Medicine, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Jeff White
- Medical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| |
Collapse
|
118
|
Boichuk S, Dunaev P, Skripova V, Galembikova A, Bikinieva F, Shagimardanova E, Gazizova G, Deviatiiarov R, Valeeva E, Mikheeva E, Vasilyeva M, Kopnin P, Strelnikov V, Kiyamova R. Unraveling the Mechanisms of Sensitivity to Anti-FGF Therapies in Imatinib-Resistant Gastrointestinal Stromal Tumors (GIST) Lacking Secondary KIT Mutations. Cancers (Basel) 2023; 15:5354. [PMID: 38001614 PMCID: PMC10670741 DOI: 10.3390/cancers15225354] [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: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
We showed previously that inhibition of KIT signaling in GISTs activates FGFR-signaling pathway rendering cancer cells resistant to receptor tyrosine kinase inhibitor (RTKi) imatinib mesylate (IM) (Gleevec) despite of absence of secondary KIT mutations and thereby illustrating a rationale for the combined (e.g., KIT- and FGFR-targeted) therapies. We show here that long-term culture of IM-resistant GISTs (GIST-R1) with IM substantially down-regulates KIT expression and induces activation of the FGFR-signaling cascade, evidenced by increased expression of total and phosphorylated forms of FGFR1 and 2, FGF-2, and FRS-2, the well-known adaptor protein of the FGF-signaling cascade. This resulted in activation of both AKT- and MAPK-signaling pathways shown on mRNA and protein levels, and rendered cancer cells highly sensitive to pan-FGFR-inhibitors (BGJ 398, AZD 4547, and TAS-120). Indeed, we observed a significant decrease of IC50 values for BGJ 398 in the GIST subclone (GIST-R2) derived from GIST-R1 cells continuously treated with IM for up to 12 months. An increased sensitivity of GIST-R2 cells to FGFR inhibition was also revealed on the xenograft models, illustrating a substantial (>70%) decrease in tumor size in BGJ 398-treated animals when treated with this pan-FGFR inhibitor. Similarly, an increased intra-tumoral apoptosis as detected by immunohistochemical (IHC)-staining for cleaved caspase-3 on day 5 of the treatment was found. As expected, both BGJ 398 and IM used alone lacked the pro-apoptotic and growth-inhibitory activities on GIST-R1 xenografts, thereby revealing their resistance to these TKis when used alone. Important, the knockdown of FGFR2, and, in much less content, FGF-2, abrogated BGJ 398's activity against GIST-R2 cells both in vitro and in vivo, thereby illustrating the FGF-2/FGFR2-signaling axis in IM-resistant GISTs as a primary molecular target for this RTKi. Collectively, our data illustrates that continuous inhibition of KIT signaling in IM-resistant GISTs lacking secondary KIT mutations induced clonal heterogeneity of GISTs and resulted in accumulation of cancer cells with overexpressed FGF-2 and FGFR1/2, thereby leading to activation of FGFR-signaling. This in turn rendered these cells extremely sensitive to the pan-FGFR inhibitors used in combination with IM, or even alone, and suggests a rationale to re-evaluate the effectiveness of FGFR-inhibitors in order to improve the second-line therapeutic strategies for selected subgroups of GIST patients (e.g., IM-resistant GISTs lacking secondary KIT mutations and exhibiting the activation of the FGFR-signaling pathway).
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (P.D.); (A.G.); (F.B.); (E.M.)
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, Moscow 127051, Russia
- Central Research Laboratory, Kazan State Medical University, Kazan 420012, Russia;
- Biomarker Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (V.S.); (R.K.)
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (P.D.); (A.G.); (F.B.); (E.M.)
| | - Vera Skripova
- Biomarker Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (V.S.); (R.K.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (P.D.); (A.G.); (F.B.); (E.M.)
| | - Firyuza Bikinieva
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (P.D.); (A.G.); (F.B.); (E.M.)
| | - Elena Shagimardanova
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (E.S.); (G.G.); (R.D.)
- LIFT—Life Improvement by Future Technologies Institute, Moscow 121205, Russia
- Loginov Moscow Clinical Scientific Center, Moscow 111123, Russia
| | - Guzel Gazizova
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (E.S.); (G.G.); (R.D.)
| | - Ruslan Deviatiiarov
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (E.S.); (G.G.); (R.D.)
- LIFT—Life Improvement by Future Technologies Institute, Moscow 121205, Russia
| | - Elena Valeeva
- Central Research Laboratory, Kazan State Medical University, Kazan 420012, Russia;
| | - Ekaterina Mikheeva
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia; (P.D.); (A.G.); (F.B.); (E.M.)
| | - Maria Vasilyeva
- Cytogenetics Laboratory, Carcinogenesis Institute, N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; (M.V.); (P.K.)
| | - Pavel Kopnin
- Cytogenetics Laboratory, Carcinogenesis Institute, N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia; (M.V.); (P.K.)
| | - Vladimir Strelnikov
- Epigenetics Laboratory, Research Centre for Medical Genetics, Moscow 115522, Russia;
| | - Ramziya Kiyamova
- Biomarker Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia; (V.S.); (R.K.)
| |
Collapse
|
119
|
Berndsen M, Renberg S, Hølmebakk T, Hancke E, Puls F, Karlsson F, Stoldt S, Bjerkehagen B, Haglund de Flon F, Muth A, Papakonstantinou A, Boye K, Lindskog S. Long-term outcome after surgical resection of non-high-risk gastrointestinal stromal tumours without adjuvant therapy. Br J Surg 2023; 110:1857-1862. [PMID: 37758514 PMCID: PMC10638541 DOI: 10.1093/bjs/znad309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/04/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Gastrointestinal stromal tumour (GIST) is the most common intra-abdominal sarcoma. Risk classification systems, commonly the modified National Institutes of Health consensus criteria, identify tumour properties relating to patient outcomes. However, owing to limited long-term evidence, most guidelines recommend up to 10-year follow-up for all risk groups except very low-risk GIST. METHODS This retrospective multicentre study included patients who had complete resection of primary, non-metastatic GIST from three Scandinavian sarcoma centres: Gothenburg (2004-2020), Stockholm (2000-2019), and Oslo (2000-2017). Medical records were reviewed for clinical details regarding diagnosis, treatment, and follow-up, and recurrence-free and disease-specific survival evaluated. RESULTS The total cohort consisted of 1213 patients with GIST. High-risk patients and those treated with tyrosine kinase inhibitors were excluded. The remaining 649 patients were included in the present analysis: 118 with very low-, 381 with low-, and 150 with intermediate-risk GISTs. Five-year recurrence-free survival rates were 100, 98.5, and 100 per cent for the intermediate-, low-, and very low-risk groups respectively (P = 0.246). Disease-specific survival rates 10 years after surgery were 100, 98.4, and 100 per cent for the intermediate-, low-, and very low-risk groups respectively (P = 0.262). CONCLUSION Patients with completely resected non-high-risk GISTs have an excellent long-term outcome, irrespective of risk group. Follow-up programmes to detect disease recurrences in these patients are probably not indicated.
Collapse
Affiliation(s)
- Marta Berndsen
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sara Renberg
- Department of Head, Neck, Lung and Skin Tumours, Karolinska University Hospital, Stockholm, Sweden
| | - Toto Hølmebakk
- Department of Abdominal and Paediatric Surgery, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Emma Hancke
- Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Florian Puls
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fredrik Karlsson
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Breast Cancer, Endocrine Tumours and Sarcoma, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Stephan Stoldt
- Department of Abdominal and Paediatric Surgery, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Bodil Bjerkehagen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Felix Haglund de Flon
- Department of Oncology–Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Andreas Muth
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andri Papakonstantinou
- Department of Breast Cancer, Endocrine Tumours and Sarcoma, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology–Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Stefan Lindskog
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Section of Endocrine and Sarcoma Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Halland Hospital, Varberg, Sweden
| |
Collapse
|
120
|
Soleimani M. Not all kidney cysts are created equal: a distinct renal cystogenic mechanism in tuberous sclerosis complex (TSC). Front Physiol 2023; 14:1289388. [PMID: 38028758 PMCID: PMC10663234 DOI: 10.3389/fphys.2023.1289388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is an autosomal dominant genetic disease caused by mutations in either TSC1 or TSC2 genes. Approximately, two million individuals suffer from this disorder worldwide. TSC1 and TSC2 code for the proteins harmartin and tuberin, respectively, which form a complex that regulates the mechanistic target of rapamycin complex 1 (mTORC1) and prevents uncontrollable cell growth. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomas) and cysts whose presence eventually causes kidney failure. The factors promoting cyst formation and tumor growth in TSC are poorly understood. Recent studies on kidney cysts in various mouse models of TSC, including mice with principal cell- or pericyte-specific inactivation of TSC1 or TSC2, have identified a unique cystogenic mechanism. These studies demonstrate the development of numerous cortical cysts that are predominantly comprised of hyperproliferating A-intercalated (A-IC) cells that express both TSC1 and TSC2. An analogous cellular phenotype in cystic epithelium is observed in both humans with TSC and in TSC2+/- mice, confirming a similar kidney cystogenesis mechanism in TSC. This cellular phenotype profoundly contrasts with kidney cysts found in Autosomal Dominant Polycystic Kidney Disease (ADPKD), which do not show any notable evidence of A-IC cells participating in the cyst lining or expansion. RNA sequencing (RNA-Seq) and confirmatory expression studies demonstrate robust expression of Forkhead Box I1 (FOXI1) transcription factor and its downstream targets, including apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in the cyst epithelia of Tsc1 (or Tsc2) knockout (KO) mice, but not in Polycystic Kidney Disease (Pkd1) mutant mice. Deletion of FOXI1, which is vital to H+-ATPase expression and intercalated (IC) cell viability, completely inhibited mTORC1 activation and abrogated the cyst burden in the kidneys of Tsc1 KO mice. These results unequivocally demonstrate the critical role that FOXI1 and A-IC cells, along with H+-ATPase, play in TSC kidney cystogenesis. This review article will discuss the latest research into the causes of kidney cystogenesis in TSC with a focus on possible therapeutic options for this devastating disease.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Department of Medicine, New Mexico Veterans Health Care Center, Albuquerque, NM, United States
- Department of Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
121
|
Das A, Chakraborty J, Luikham S, Banerjee S, Bhattacharya J, Dutta S. Targeting aloe active compounds to c-KIT promoter G-quadruplex and comparative study of their anti proliferative property. J Biomol Struct Dyn 2023; 41:9686-9694. [PMID: 36379679 DOI: 10.1080/07391102.2022.2145370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Small molecules targeting G-quadruplex of oncogene promoter is considered as a promising anticancer therapeutics approach. Natural aloe compounds aloe emodin, and its glycoside derivative aloe emodin-8-glucoside and aloin have anticancer activity and also have potential DNA binding ability. These three compounds have promising binding ability towards quadruplex structures particularly c-KIT G-quadruplex. Here, this study demonstrates complete biophysical study of these compounds to c-KIT quadruplex structure. Aloe emodin showed highest binding stabilization with c-KIT which has been proved by absorbance, fluorescence, dye displacement, ITC and SPR studies. Moreover, comparative study of these compounds with HCT 116 cells line also agreed to their anti proliferative property which may be helpful to establish these aloe compounds as potential anticancer drugs. This study comprises a complete biophysical study along with their anti proliferative property and demonstrates aloe emodin as a potent c-KIT binding molecule.
Collapse
Affiliation(s)
- Abhi Das
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Jeet Chakraborty
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Soching Luikham
- Department of Chemistry, National Institute of Technology Nagaland, Dimapur, India
| | - Sayanika Banerjee
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Jhimli Bhattacharya
- Department of Chemistry, National Institute of Technology Nagaland, Dimapur, India
| | - Sanjay Dutta
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
122
|
Tashi T, Deininger MW. Management of Advanced Systemic Mastocytosis and Associated Myeloid Neoplasms. Immunol Allergy Clin North Am 2023; 43:723-741. [PMID: 37758409 DOI: 10.1016/j.iac.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Advanced systemic mastocytosis (AdvSM) is a heterogeneous group of disorders characterized by neoplastic mast cell-related organ damage and frequently associated with a myeloid neoplasm. The 3 clinical entities that comprise AdvSM are aggressive SM (ASM), SM-associated hematologic neoplasm, and mast cell leukemia. A gain-of-function KIT D816 V mutation is the primary oncogenic driver found in about 90% of all patients with AdvSM. Midostaurin, an oral multikinase inhibitor with activity against KIT D816V, and avapritinib, an oral selective KIT D816V inhibitor are approved for AdvSM.
Collapse
Affiliation(s)
- Tsewang Tashi
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, 2000, Circle of Hope, Salt Lake City, UT 84112, USA.
| | - Michael W Deininger
- Division of Hematology and Oncology, Medical College of Wisconsin, Versiti Blood Research Institute, 8727 West Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
123
|
Chauhan S, Shinde RK, Jain Y. Giant Exophytic Gastrointestinal Stromal Tumor (GIST) Arising From the Antral Stomach: A Surgical Case Report. Cureus 2023; 15:e48773. [PMID: 38098933 PMCID: PMC10719664 DOI: 10.7759/cureus.48773] [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: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
This comprehensive case report meticulously outlines the clinical manifestation, diagnostic trajectory, surgical intervention, pathology, chemotherapy, and patient follow-up in a challenging instance of a giant exophytic gastrointestinal stromal tumor (GIST) emerging from the antral part of the stomach in a 56-year-old male patient. Over the course of a year, the patient presented with symptoms including abdominal distension, a progressively enlarging lump, constipation, and abdominal fullness. Our diagnostic strategy, anchored by ultrasonography and contrast-enhanced computed tomography (CECT), yielded crucial insights into the tumor's precise dimensions and anatomical location. The subsequent surgical planning and execution were pivotal, entailing a meticulous dissection of the cystic mass from adjacent structures to ensure negative margins. Histopathological scrutiny of frozen sections from the lymph nodes and excised antral margins conclusively confirmed the absence of malignancy, facilitating primary closure. This case report underscores the decisive role of accurate diagnostic imaging in shaping surgical strategies and emphasizes the need for a multidisciplinary approach to managing GISTs. Beyond the surgical realm, the case highlights the significance of tyrosine kinase inhibitors (TKIs), particularly imatinib, in the treatment paradigm. Additionally, the report sheds light on ongoing research endeavors to refine treatment modalities in GISTs.
Collapse
Affiliation(s)
- Simran Chauhan
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Raju K Shinde
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Yashraj Jain
- General Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
124
|
Namløs HM, Khelik K, Nakken S, Vodák D, Hovig E, Myklebost O, Boye K, Meza‐Zepeda LA. Chromosomal instability and a deregulated cell cycle are intrinsic features of high-risk gastrointestinal stromal tumours with a metastatic potential. Mol Oncol 2023; 17:2432-2450. [PMID: 37622176 PMCID: PMC10620130 DOI: 10.1002/1878-0261.13514] [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: 02/28/2023] [Revised: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 08/26/2023] Open
Abstract
Patients with localised, high-risk gastrointestinal stromal tumours (GIST) benefit from adjuvant imatinib treatment. Still, approximately 40% of patients relapse within 3 years after adjuvant therapy and the clinical and histopathological features currently used for risk classification cannot precisely predict poor outcomes after standard treatment. This study aimed to identify genomic and transcriptomic profiles that could be associated with disease relapse and thus a more aggressive phenotype. Using a multi-omics approach, we analysed a cohort of primary tumours from patients with untreated, resectable high-risk GISTs. We compared patients who developed metastatic disease within 3 years after finishing adjuvant imatinib treatment and patients without disease relapse after more than 5 years of follow-up. Combining genomics and transcriptomics data, we identified somatic mutations and deregulated mRNA and miRNA genes intrinsic to each group. Our study shows that increased chromosomal instability (CIN), including chromothripsis and deregulated kinetochore and cell cycle signalling, separates high-risk samples according to metastatic potential. The increased CIN seems to be an intrinsic feature for tumours that metastasise and should be further validated as a novel prognostic biomarker for high-risk GIST.
Collapse
Affiliation(s)
- Heidi Maria Namløs
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
| | - Ksenia Khelik
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
| | - Sigve Nakken
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
- Department of InformaticsUniversity of OsloOsloNorway
| | - Daniel Vodák
- Bioinformatics Core Facility, Department of Core Facilities, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
- Department of InformaticsUniversity of OsloOsloNorway
| | - Ola Myklebost
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
- Department for Clinical ScienceUniversity of BergenBergenNorway
| | - Kjetil Boye
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
- Department of OncologyOslo University HospitalOsloNorway
| | - Leonardo A. Meza‐Zepeda
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium HospitalOslo University HospitalOsloNorway
- Genomics Core Facility, Department of Core Facilities, Institute for Cancer ResearchOslo University HospitalOsloNorway
| |
Collapse
|
125
|
Zhang QW, Zhang RY, Yan ZB, Zhao YX, Wang XY, Jin JZ, Qiu QX, Chen JJ, Xie ZH, Lin J, Cao H, Zhou Y, Chen HM, Li XB. Personalized radiomics signature to screen for KIT-11 mutation genotypes among patients with gastrointestinal stromal tumors: a retrospective multicenter study. J Transl Med 2023; 21:726. [PMID: 37845765 PMCID: PMC10577986 DOI: 10.1186/s12967-023-04520-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVES Gastrointestinal stromal tumors (GISTs) carrying different KIT exon 11 (KIT-11) mutations exhibit varying prognoses and responses to Imatinib. Herein, we aimed to determine whether computed tomography (CT) radiomics can accurately stratify KIT-11 mutation genotypes to benefit Imatinib therapy and GISTs monitoring. METHODS Overall, 1143 GISTs from 3 independent centers were separated into a training cohort (TC) or validation cohort (VC). In addition, the KIT-11 mutation genotype was classified into 4 categories: no KIT-11 mutation (K11-NM), point mutations or duplications (K11-PM/D), KIT-11 557/558 deletions (K11-557/558D), and KIT-11 deletion without codons 557/558 involvement (K11-D). Subsequently, radiomic signatures (RS) were generated based on the arterial phase of contrast CT, which were then developed as KIT-11 mutation predictors using 1408 quantitative image features and LASSO regression analysis, with further evaluation of its predictive capability. RESULTS The TC AUCs for K11-NM, K11-PM/D, K11-557/558D, and K11-D ranged from 0.848 (95% CI 0.812-0.884), 0.759 (95% CI 0.722-0.797), 0.956 (95% CI 0.938-0.974), and 0.876 (95% CI 0.844-0.908), whereas the VC AUCs ranged from 0.723 (95% CI 0.660-0.786), 0.688 (95% CI 0.643-0.732), 0.870 (95% CI 0.824-0.918), and 0.830 (95% CI 0.780-0.878). Macro-weighted AUCs for the KIT-11 mutant genotype ranged from 0.838 (95% CI 0.820-0.855) in the TC to 0.758 (95% CI 0.758-0.784) in VC. TC had an overall accuracy of 0.694 (95%CI 0.660-0.729) for RS-based predictions of the KIT-11 mutant genotype, whereas VC had an accuracy of 0.637 (95%CI 0.595-0.679). CONCLUSIONS CT radiomics signature exhibited good predictive performance in estimating the KIT-11 mutation genotype, especially in prediction of K11-557/558D genotype. RS-based classification of K11-NM, K11-557/558D, and K11-D patients may be an indication for choice of Imatinib therapy.
Collapse
Affiliation(s)
- Qing-Wei Zhang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ran-Ying Zhang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi-Bo Yan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Xuan Zhao
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin-Yuan Wang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Zheng Jin
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi-Xuan Qiu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie-Jun Chen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen-Hui Xie
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Rd., Shanghai, 200127, China
| | - Jiang Lin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Road, Shanghai, 200127, China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Rd., Shanghai, 200127, China.
| | - Hui-Min Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiao-Bo Li
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
126
|
Dong RX, Wang C, Zhou H, Yin HQ, Liu Y, Liang HT, Pan YB, Wang JW, Cao YQ. Rare rectal gastrointestinal stromal tumor case: A case report and review of the literature. World J Clin Cases 2023; 11:6797-6805. [PMID: 37901015 PMCID: PMC10600839 DOI: 10.12998/wjcc.v11.i28.6797] [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: 07/22/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are rare tumors of the gastrointestinal tract accounting for less than 1% of all gut tumors. GISTs occurring in the rectum are extremely rare, and these usually present at an advanced stage compared with other sites. CASE SUMMARY A 60-year-old male who presented with features of sensations of rectal tenesmus was referred to our department with a mass in the lower rectum that was detected during a routine checkup. Colonoscopy, transrectal ultrasound, perianal magnetic resonance imaging and ultrasonic contrast were used to diagnose the rectum GIST, and then the patient underwent complete transanal resection using the ultrasonic scalpel. The patient was discharged ten days after the operation and was defined as low risk. Therefore, he had no need to receive subsequent adjuvant therapies, and he had not suffered any anal dysfunction or had any evidence of recurrence at follow up. CONCLUSION Surgical resection with histologically negative margins is the standard curative treatment for rectal GISTs. Appropriate surgical techniques based on the location, size, and resectability of the tumor should attract great attention from clinicians.
Collapse
Affiliation(s)
- Ruo-Xi Dong
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chen Wang
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hao Zhou
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hao-Qiang Yin
- Department of Ultrasound, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yang Liu
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hong-Tao Liang
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yi-Bin Pan
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jia-Wen Wang
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yong-Qing Cao
- Department of Anorectal Surgery, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
127
|
Nagano H, Ohyama S, Sato A, Igarashi J, Yamamoto T, Kadoya M, Kobayashi M. Jejunal gastrointestinal stromal tumor that developed in a patient with neurofibromatosis type 1: a case report. Diagn Pathol 2023; 18:110. [PMID: 37789344 PMCID: PMC10546696 DOI: 10.1186/s13000-023-01398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is known to be associated with the frequent occurrence of unique gastrointestinal stromal tumors (GISTs), preferably occurring in the small intestine, with no mutations in the c-kit proto-oncogene or platelet-derived growth factor receptor-alpha (PDGFRA), with a high tendency for multifocal development, indolent nature, with low proliferation activity and favorable prognosis. CASE PRESENTATION A woman in her forties visited her local doctor complaining of menstrual pain; a large mass was detected in her lower abdomen, and she was referred to our hospital. The patient had hundreds of skin warts and café au lait spots. The patient's mother had been diagnosed with type 1 neurofibromatosis. The patient met the diagnostic criteria for NF1 and was diagnosed with NF1. Ultrasonography showed a large heterogeneous cystic mass with various echo patterns, solid compartments and multiple septations. Magnetic resonance imaging showed a multilocular cystic mass with liquid content exhibiting various intensities, including that of blood. A small round solid mass was also observed close to the cystic tumor. Contrast-enhanced computed tomography showed that the round solid mass showed strong enhancement in the early phase, unlike the cystic tumor component. Open laparotomy revealed a multicystic exophytic tumor measuring 11.5 cm originating from the jejunal wall, 20 cm distal to the duodenojejunal flexure. A solid tumor measuring 2.1 cm was also found on the anal side of the large tumor. We resected the short segment of the jejunum, including the two lesions. Microscopic findings revealed that the cystic and solid tumors consisted of spindle-shaped tumor cells showing little atypia with a fascicular or bundle arrangement. Nuclear mitosis was scarce. Immunostaining of the tumor cells showed positive staining for KIT and DOG1 and negative staining for S100 and desmin. The NF1 patient was diagnosed with multiple GISTs accompanied by intratumoral hemorrhagic denaturation arising from the jejunum. The TNM staging was pT4N0M0, stage IIIA. CONCLUSION We report a case of GISTs associated with NF1 that showed a jejunal origin, multifocal development and few mitotic figures. The recurrence risk, survival prognosis and need for adjuvant chemotherapy, particularly in cases where the initial GIST exhibits a very indolent pathology in NF1-related GISTs, remain to be elucidated.
Collapse
Affiliation(s)
- Hideki Nagano
- Department of Surgery, Marunouchi Hospital, 1-7-45, Nagisa Matsumoto, Nagano, 390-0841, Japan.
| | - Shigekazu Ohyama
- Department of Surgery, Marunouchi Hospital, 1-7-45, Nagisa Matsumoto, Nagano, 390-0841, Japan
| | - Atsushi Sato
- Department of Surgery, Marunouchi Hospital, 1-7-45, Nagisa Matsumoto, Nagano, 390-0841, Japan
| | - Jun Igarashi
- Department of Surgery, Marunouchi Hospital, 1-7-45, Nagisa Matsumoto, Nagano, 390-0841, Japan
| | - Tomoko Yamamoto
- Department of Surgery, Marunouchi Hospital, 1-7-45, Nagisa Matsumoto, Nagano, 390-0841, Japan
| | - Masumi Kadoya
- Department of Radiology, Marunouchi Hospital, Matsumoto Nagano, Japan
| | - Mikiko Kobayashi
- Department of Pathology, Marunouchi Hospital, Matsumoto Nagano, Japan
| |
Collapse
|
128
|
Guo C, Zhou H, Chen X, Feng Z. Computed tomography texture-based models for predicting KIT exon 11 mutation of gastrointestinal stromal tumors. Heliyon 2023; 9:e20983. [PMID: 37876490 PMCID: PMC10590931 DOI: 10.1016/j.heliyon.2023.e20983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Background KIT exon 11 mutation in gastrointestinal stromal tumors (GISTs) is associated with treatment strategies. However, few studies have shown the role of imaging-based texture analysis in KIT exon 11 mutation in GISTs. In this study, we aimed to show the association between computed tomography (CT)-based texture features and KIT exon 11 mutation. Methods Ninety-five GISTs confirmed by surgery and identified with mutational genotype of KIT were included in this study. By amplifying the samples using over-sampling technique, a total of 183 region of interest (ROI) segments were extracted from 63 patients as training cohort. The 63 new ROI segments were extracted from the 63 patients as internal validation cohort. Thirty-two patients who underwent KIT exon 11 mutation test during 2021-2023 was selected as external validation cohort. The textural parameters were evaluated both in training cohort and validation cohort. Least absolute shrinkage and selection operator (LASSO) algorithms and logistic regression analysis were used to select the discriminant features. Results Three of textural features were obtained using LASSO analysis. Logistic regression analysis showed that patients' age, tumor location and radiomics features were significantly associated with KIT exon 11 mutation (p < 0.05). A nomogram was developed based on the associated factors. The area under the curve (AUC) of clinical features, radiomics features and their combination in training cohort was 0.687 (95 % CI: 0.604-0.771), 0.829 (95 % CI: 0.768-0.890) and 0.874 (95 % CI: 0.822-0.926), respectively. The AUC of radiomics features in internal validation cohort and external cohort was 0.880 (95 % CI: 0.796-0.964) and 0.827 (95%CI: 0.667-0.987), respectively. Conclusion The CT texture-based model can be used to predict KIT exon 11 mutation in GISTs.
Collapse
Affiliation(s)
- Chuangen Guo
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Hao Zhou
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, 155 hanzhong Road, Nanjing, 210029, China
| | - Zhan Feng
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| |
Collapse
|
129
|
Duarte Hospital C, Tête A, Debizet K, Imler J, Tomkiewicz-Raulet C, Blanc EB, Barouki R, Coumoul X, Bortoli S. SDHi fungicides: An example of mitotoxic pesticides targeting the succinate dehydrogenase complex. ENVIRONMENT INTERNATIONAL 2023; 180:108219. [PMID: 37778286 DOI: 10.1016/j.envint.2023.108219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Succinate dehydrogenase inhibitors (SDHi) are fungicides used to control the proliferation of pathogenic fungi in crops. Their mode of action is based on blocking the activity of succinate dehydrogenase (SDH), a universal enzyme expressed by all species harboring mitochondria. The SDH is involved in two interconnected metabolic processes for energy production: the transfer of electrons in the mitochondrial respiratory chain and the oxidation of succinate to fumarate in the Krebs cycle. In humans, inherited SDH deficiencies may cause major pathologies including encephalopathies and cancers. The cellular and molecular mechanisms related to such genetic inactivation have been well described in neuroendocrine tumors, in which it induces an oxidative stress, a pseudohypoxic phenotype, a metabolic, epigenetic and transcriptomic remodeling, and alterations in the migration and invasion capacities of cancer cells, in connection with the accumulation of succinate, an oncometabolite, substrate of the SDH. We will discuss recent studies reporting toxic effects of SDHi in non-target organisms and their implications for risk assessment of pesticides. Recent data show that the SDH structure is highly conserved during evolution and that SDHi can inhibit SDH activity in mitochondria of non-target species, including humans. These observations suggest that SDHi are not specific inhibitors of fungal SDH. We hypothesize that SDHi could have toxic effects in other species, including humans. Moreover, the analysis of regulatory assessment reports shows that most SDHi induce tumors in animals without evidence of genotoxicity. Thus, these substances could have a non-genotoxic mechanism of carcinogenicity that still needs to be fully characterized and that could be related to SDH inhibition. The use of pesticides targeting mitochondrial enzymes encoded by tumor suppressor genes raises questions on the risk assessment framework of mitotoxic pesticides. The issue of SDHi fungicides is therefore a textbook case that highlights the urgent need for changes in regulatory assessment.
Collapse
Affiliation(s)
| | - Arnaud Tête
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Kloé Debizet
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Jules Imler
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | | | - Etienne B Blanc
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Robert Barouki
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris
| | - Xavier Coumoul
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris.
| | - Sylvie Bortoli
- Université Paris Cité, INSERM UMR-S 1124, T3S, 45 rue des Saints-Pères, 75006 Paris.
| |
Collapse
|
130
|
Alrashed R, AlHarbi H, Ali B, Mubarah A, AlGhamdi F. Is Bleeding Gastrointestinal Stromal Tumor Associated With Higher Mortality and Morbidity? Cureus 2023; 15:e47398. [PMID: 38021903 PMCID: PMC10657152 DOI: 10.7759/cureus.47398] [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: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Gastrointestinal stromal tumor (GIST) is the most common primary mesenchymal tumor of the gastrointestinal tract. The clinical presentation of GIST varies widely and ranges from being asymptomatic to being a life-threatening emergency in the form of gastrointestinal bleeding or bowel obstruction. Multiple prognostic factors have been identified for GIST, including, most importantly, larger tumor size (>5 cm), higher mitotic activity (>5 per 50 high-power fields), rupture of the mass, site of the mass, and personal history of GIST. Objective In this paper, we aim to study bleeding in gastrointestinal tumors as a predictor for morbidity and mortality and investigate possible factors influencing bleeding in GIST. Method A retrospective study design was used. Electronic medical records of 39 patients diagnosed with GIST in Prince Sultan Military Medical City in Riyadh between January 2015 and December 2020 were retrieved. Normal variables were presented as mean and standard deviation, while non-normal variables were presented as median and interquartile range. Student t-test or Mann-Whitney test was used to compare quantitative data when appropriate. The distribution of recurrence and survival was plotted with the Kaplan-Meier curve and compared between the two groups (bleeding and non-bleeding groups) with the log-rank test. Factors affecting recurrence and mortality were assessed using univariable Cox regression. Result A total of 39 cases of GIST tumors were included in this study. Patients were categorized according to the presentation into two groups: patients with no bleeding (n= 28) and those presented with bleeding (n= 11). The gender distribution was equal. The mean age was 58.14± 12.46 in patients with non-bleeding GIST and 58.18± 16.01 in patients with bleeding GIST. The most common location of GIST was the stomach (22 cases, 56%). Neither group had any significant differences regarding age (P-value = 0.993), gender (P-value = 0.648), tumor location (P-value = 0.057), size (P-value = 0.250), cluster of differentiation (CD) markers, histological types (P-value = 0.692), and shape (P-value = 0.079). There was no significant difference in recurrence between both groups (log-rank P = 0.972). There was no significant difference in survival between both groups (Log-rank P = 0.506). Conclusion We concluded that bleeding GIST was not a significant predictor for recurrence rate or higher mortality. This can help in the debate of whether bleeding GIST should be added as an independent factor in risk stratification. Despite that, further studies are needed to identify more variables that can add more accuracy to the pre-existing risk stratification systems.
Collapse
Affiliation(s)
- Rema Alrashed
- Department of Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| | - Hussam AlHarbi
- Department of Surgery, AlKharj Armed Forces Hospital, Riyadh, SAU
| | - Bandar Ali
- Department of Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| | - Alanoud Mubarah
- Department of Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| | - Faisal AlGhamdi
- Department of Surgery, Prince Sultan Military Medical City, Riyadh, SAU
| |
Collapse
|
131
|
Abdellateif MS, Bayoumi AK, Mohammed MA. c-Kit Receptors as a Therapeutic Target in Cancer: Current Insights. Onco Targets Ther 2023; 16:785-799. [PMID: 37790582 PMCID: PMC10544070 DOI: 10.2147/ott.s404648] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
c-Kit is a type III receptor tyrosine kinase (RTK) that has an essential role in various biological functions including gametogenesis, melanogenesis, hematopoiesis, cell survival, and apoptosis. c-KIT aberrations, either overexpression or loss-of-function mutations, have been implicated in the pathogenesis and development of many cancers, including gastrointestinal stromal tumors, mastocytosis, acute myeloid leukemia, breast, thyroid, and colorectal cancer, making c-KIT an attractive molecular target for the treatment of cancers. Therefore, a lot of effort has been put into investigating the utility of tyrosine kinase inhibitors for the management of c-KIT mutated tumors. This review of the literature illustrates the role of c-KIT mutations in many cancers, aiming to provide insights into the role of TKIs as a therapeutic option for cancer patients with c-KIT aberrations. In conclusion, c-KIT is implicated in different types of cancer, and it could be a successful molecular target; however, proper detection of the underlying mutation type is required before starting the appropriate personalized therapy.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Ahmed K Bayoumi
- Paediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
- Children’s Cancer Hospital 57357, Cairo, 11617, Egypt
| | - Mohammed Aly Mohammed
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| |
Collapse
|
132
|
Obata Y, Kurokawa K, Tojima T, Natsume M, Shiina I, Takahashi T, Abe R, Nakano A, Nishida T. Golgi retention and oncogenic KIT signaling via PLCγ2-PKD2-PI4KIIIβ activation in gastrointestinal stromal tumor cells. Cell Rep 2023; 42:113035. [PMID: 37616163 DOI: 10.1016/j.celrep.2023.113035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/19/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023] Open
Abstract
Most gastrointestinal stromal tumors (GISTs) develop due to gain-of-function mutations in the tyrosine kinase gene, KIT. We recently showed that mutant KIT mislocalizes to the Golgi area and initiates uncontrolled signaling. However, the molecular mechanisms underlying its Golgi retention remain unknown. Here, we show that protein kinase D2 (PKD2) is activated by the mutant, which causes Golgi retention of KIT. In PKD2-inhibited cells, KIT migrates from the Golgi region to lysosomes and subsequently undergoes degradation. Importantly, delocalized KIT cannot trigger downstream activation. In the Golgi/trans-Golgi network (TGN), KIT activates the PKD2-phosphatidylinositol 4-kinase IIIβ (PKD2-PI4KIIIβ) pathway through phospholipase Cγ2 (PLCγ2) to generate a PI4P-rich membrane domain, where the AP1-GGA1 complex is aberrantly recruited. Disruption of any factors in this cascade results in the release of KIT from the Golgi/TGN. Our findings show the molecular mechanisms underlying KIT mislocalization and provide evidence for a strategy for inhibition of oncogenic signaling.
Collapse
Affiliation(s)
- Yuuki Obata
- Laboratory of Intracellular Traffic & Oncology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Kazuo Kurokawa
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Takuro Tojima
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Miyuki Natsume
- Laboratory of Intracellular Traffic & Oncology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Applied Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Isamu Shiina
- Department of Applied Chemistry, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryo Abe
- Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama 351-0198, Japan
| | - Toshirou Nishida
- National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
133
|
Rahimi-Ardabily A, Murdande S, Dong M, Gu KW, Zhang B, Miller K, Aploks K, Da Dong X. Liver resection for metastatic GIST tumor improves survival in the era of tyrosine kinase inhibitors: a systematic review and meta-analysis. Langenbecks Arch Surg 2023; 408:373. [PMID: 37740754 DOI: 10.1007/s00423-023-03052-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/08/2023] [Indexed: 09/25/2023]
Abstract
INTRODUCTION Survival for gastrointestinal stromal tumor (GIST) has been increasing over the years after the introduction of tyrosine kinase inhibitors. However, the role of metastasectomy for GIST is still controversial. Patients are currently treated with imatinib or sunitinib in case of imatinib failures as optimal medical therapy for metastatic GIST. METHODS The Pubmed, EMBASE, and Cochrane Library were systematically searched. Overall survival following liver resection ± tyrosine kinase inhibitor treatment for metastatic GIST was compared to treatment with tyrosine kinase inhibitors alone. RESULTS Eleven studies including both randomized control trials and retrospective cohort studies were included in the final analysis with a total of 988 patients. Seven studies encompassed data on 556 patients with isolated liver metastases (219 surgery ± drug groups and 337 drug-only groups) were included. Overall survival was significantly improved in patients undergoing liver resection ± drug therapy in comparison to drug therapy alone. [HR (95%CI) = 2.10 (1.58, 2.79); p<0.00001]. Subgroup analysis showed that patients also had improved progression free survival based on 4 studies. [HR (95%CI) = 1.92 (1.43, 2.56); p<0.00001]. In case of concurrent liver and peritoneal metastases, patients showed improved overall survival with aggressive surgical approaches based on 10 studies. [HR (95%CI) = 1.90 (1.56, 2.31); p<0.00001]. CONCLUSION This meta-analysis found that liver resection for patients with metastatic GIST regardless of peritoneal metastases improved progression free and overall survival in conjunction with tyrosine kinase inhibitors as compared with medical therapy alone. Furthermore, liver resections did not have any immediate detrimental impact on survival in the group of patients selected.
Collapse
Affiliation(s)
- Arash Rahimi-Ardabily
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Sanjana Murdande
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Michael Dong
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Katie W Gu
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Brianna Zhang
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Kendall Miller
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Krist Aploks
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA
| | - Xiang Da Dong
- Division of Surgical Oncology, Department of Surgery, Nuvance Health, Whittingham Cancer Center, 34 Maple Street, Norwalk, CT, 06856, USA.
| |
Collapse
|
134
|
Sposito M, Belluomini L, Pontolillo L, Tregnago D, Trestini I, Insolda J, Avancini A, Milella M, Bria E, Carbognin L, Pilotto S. Adjuvant Targeted Therapy in Solid Cancers: Pioneers and New Glories. J Pers Med 2023; 13:1427. [PMID: 37888038 PMCID: PMC10608226 DOI: 10.3390/jpm13101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Targeted therapy (TT) has revolutionized cancer treatment, successfully applied in various settings. Adjuvant TT in resected early-stage gastrointestinal stromal tumors (GIST), melanoma, non-small cell lung cancer (NSCLC), and breast cancer has led to practice-changing achievements. In particular, standard treatments include BRAF inhibitors for melanoma, osimertinib for NSCLC, hormone therapy or HER2 TT for breast cancer, and imatinib for GIST. Despite the undeniable benefit derived from adjuvant TT, the optimal duration of TT and the appropriate managing of the relapse remain open questions. Furthermore, neoadjuvant TT is emerging as valuable, particularly in breast cancer, and ongoing studies evaluate TT in the perioperative setting for early-stage NSCLC. In this review, we aim to collect and describe the large amount of data available in the literature about adjuvant TT across different histologies, focusing on epidemiology, major advances, and future directions.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Letizia Pontolillo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (L.P.); (E.B.)
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Daniela Tregnago
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Ilaria Trestini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Jessica Insolda
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Alice Avancini
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Michele Milella
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Roma, Italy; (L.P.); (E.B.)
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Luisa Carbognin
- Gynecology Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Roma, Italy;
| | - Sara Pilotto
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, 37134 Verona, Italy; (M.S.); (L.B.); (D.T.); (I.T.); (J.I.); (A.A.); (M.M.)
| |
Collapse
|
135
|
Gouda MA, Janku F, Somaiah N, Hunt KK, Yedururi S, Subbiah V. Multi-disciplinary management of recurrent gastrointestinal stromal tumor harboring KIT exon 11 mutation with the switch-control kinase inhibitor ripretinib and surgery. Oncoscience 2023; 10:38-43. [PMID: 37736254 PMCID: PMC10511119 DOI: 10.18632/oncoscience.586] [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: 05/28/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Ripretinib is a tyrosine kinase inhibitor that was approved by the United States FDA in 2020 for treatment of advanced gastrointestinal stromal tumor (GIST) in patients who received prior treatment with three or more tyrosine kinase inhibitors. In this case report, we show the durable clinical benefit achieved in a patient with GIST by using ripretinib and repeated timely surgical resection of limited disease progression. The total time on ripretinib was 43 months which is longer than the current reported data from ripretinib clinical trials. Such approach for using multi-disciplinary disease management can improve the durability of response to tyrosine kinase inhibitors, including ripretinib, and associated clinical outcomes.
Collapse
Affiliation(s)
- Mohamed A. Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kelly K. Hunt
- Departments of Breast Surgical Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sireesha Yedururi
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Sarah Cannon Research Institute, Nashville, TN 37203, USA
| |
Collapse
|
136
|
Hu X, Su P, Liu B, Guo J, Wang Z, He C, Wang Z, Kou Y. Characterization of a Human Gastrointestinal Stromal Tumor Cell Line Established by SV40LT-Mediated Immortalization. Int J Mol Sci 2023; 24:13640. [PMID: 37686448 PMCID: PMC10487453 DOI: 10.3390/ijms241713640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors in the digestive tract and originate from the interstitial cells of Cajal (ICC), which is the pacemaker for peristaltic movement in the gastrointestinal tract. Existing GIST cell lines are widely used as cell models for in vitro experimental studies because the mutation sites are known. However, the immortalization methods of these cell lines are unknown, and no Chinese patient-derived GIST cell lines have been documented. Here, we transfected simian virus 40 large T antigen (SV40LT) into primary GIST cells to establish an immortalized human GIST cell line (ImGIST) for the first time. The ImGIST cells had neuronal cell-like irregular radioactive growth and retained the fusion growth characteristics of GIST cells. They stably expressed signature proteins, maintained the biological and genomic characteristics of normal primary GIST cells, and responded well to imatinib, suggesting that ImGIST could be a potential in vitro model for research in GIST to explore the molecular pathogenesis, drug resistance mechanisms, and the development of new adjuvant therapeutic options.
Collapse
Affiliation(s)
- Xiangchen Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; (X.H.)
| | - Peng Su
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang 117005, China
| | - Bo Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; (X.H.)
| | - Jingwei Guo
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zitong Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Cai He
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Youwei Kou
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; (X.H.)
| |
Collapse
|
137
|
Rafael-Perez CI, Paz-López AJ, Castañeda-Anaya PS. A Rare Case of Gastrointestinal Stromal Tumor of the Abdominal Cavity: A Case Report. Cureus 2023; 15:e44926. [PMID: 37818497 PMCID: PMC10560609 DOI: 10.7759/cureus.44926] [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: 09/08/2023] [Indexed: 10/12/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are rare gastrointestinal neoplasms. We are presenting a 57-year-old female patient with a GIST of considerable dimensions that was deemed unresectable during the initial intervention. The patient had a previous surgical record of a peritoneal sarcomatosis tumor that covered approximately 50% of the abdominal cavity. The patient underwent surgical procedures, including exploratory laparotomy, lumpectomy, splenectomy, proximal gastrectomy, esophagogastric anastomosis, pyloroplasty, jejunostomy, and left diaphragm plasty. One of the observed results was the presence of a 50×40 cm exophytic multilobed cerebroid-like tumor in the abdomen region, specifically dependent on the gastric fundus. The primary treatment for patients without metastases is surgical removal of the tumor, with wedge resection being recommended for the preservation of organ function and quality of life postoperatively.
Collapse
Affiliation(s)
| | - Alexis Jared Paz-López
- Digestive and Endocrine Surgery Department, Unidad Médica de Alta Especialidad No. 25, Monterrey, MEX
| | | |
Collapse
|
138
|
Lopes David BB, Nazareth Aguiar Junior P, Costa e Silva M, Dienstmann R, Gil Ferreira C, Serrano C. Cost Evaluation Analysis of Genetic Testing and Tailored Adjuvant Imatinib in Patients With Resected High-Risk GI Stromal Tumors: The Brazilian Perspective. JCO Glob Oncol 2023; 9:e2300070. [PMID: 37856732 PMCID: PMC10664859 DOI: 10.1200/go.23.00070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/31/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
PURPOSE Mutations of the KIT gene are the molecular hallmark of most GI stromal tumors (GISTs). Imatinib has revolutionized GIST treatment. Adjuvant imatinib for 3 years is the standard of care for high-risk resected GIST. However, the GIST molecular biologic profile has found different responses to this approach. Despite this, genetic testing at diagnosis is not a routine and empirical adjuvant imatinib remains the rule. Barriers to genetic profiling include concerns about the cost and utility of testing. This analysis aims to determine whether targeted genetic testing reduces costs as an ancillary tool for a limited-resource scenario instead of adjuvant empirical imatinib in patients with resected high-risk GIST. METHODS The cost evaluation analysis of molecular testing for GIST was based on the Cost of Preventing an Event (COPE), considering the Number Needed to Treat and the costs of each test compared with the cost of 3-year empirical adjuvant imatinib and real treatment costs (median number of cycles) from the public and private Brazilian Healthcare System's perspective. The analysis compared the costs of the molecular tests (broad next-generation sequencing [NGS], GS Infinity DNA/RNA assay, and targeted NGS: GS Focus GIST and the Fleury GIST Tumor DNA sequencing panel), costs of drug acquisition, considering discounts (imatinib mesylate and Glivec), and the costs of supportive care. RESULTS In both scenarios, public and private, regardless of the use of imatinib or Glivec, tailoring adjuvant treatment reduced costs, irrespective of the number of cycles. The only exception was the combination of the broad NGS test and imatinib in the Public Healthcare System. CONCLUSION The molecularly tailored adjuvant imatinib reduced costs considering the COPE of available NGS tests for both the public and private Brazilian health care systems.
Collapse
Affiliation(s)
- Bruna Bianca Lopes David
- Grupo Oncoclinicas, São Paulo, Brazil
- Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
139
|
Wang C, Yantiss RK, Lieberman MD, Tubito-Massarano F, Qin L, Yemelyanova A, Solomon JP, Hissong E. A Rare PDGFRA Exon 15 Germline Mutation Identified in a Patient With Phenotypic Manifestations Concerning for GIST-Plus Syndrome: A Case Report and Review of Literature. Int J Surg Pathol 2023; 31:1139-1145. [PMID: 36802986 DOI: 10.1177/10668969231152588] [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: 02/22/2023]
Abstract
Molecular alterations in PDGFRA are well-described as drivers of sporadic gastrointestinal stromal tumors (GISTs) and inflammatory fibroid polyps (IFPs). However, a small number of families with germline PDGFRA mutations in exons 12, 14, and 18 have been reported, forming the basis of an autosomal dominant inherited disorder with incomplete penetrance and variable expressivity, now referred to as PDGFRA-mutant syndrome or GIST-plus syndrome. Phenotypic manifestations of this rare syndrome include multiple gastrointestinal GISTS, IFPs, fibrous tumors, and other variable features. Herein, we report the case of a 58-year-old female who presented with a gastric GIST and numerous small intestinal IFPs, found to harbor a previously undescribed germline PDGFRA exon 15 p.G680R mutation. Somatic tumor testing was performed on the GIST, a duodenal IFP, and an ileal IFP utilizing a targeted next-generation sequencing panel, revealing additional and distinct secondary PDGFRA exon 12 somatic mutations in each of the 3 tumors. Our findings raise important considerations regarding mechanisms of tumor development in patients with underlying germline PDGFRA alterations and highlight the potential utility of expanding currently available germline and somatic testing panels to include exons outside the typical hotspot regions.
Collapse
Affiliation(s)
- Chiyun Wang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Rhonda K Yantiss
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | - Lihui Qin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Anna Yemelyanova
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - James P Solomon
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
140
|
Kelly CH, Sipok A, Landry JP, Ramsey L, Joyce CJ, Gnerlich JL. Utilization of Neoadjuvant Therapy in Gastrointestinal Stromal Tumors of the Stomach: Analysis of the 2006-2018 National Cancer Database. J Gastrointest Surg 2023; 27:1794-1803. [PMID: 37316761 DOI: 10.1007/s11605-023-05742-5] [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: 02/13/2023] [Accepted: 06/03/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Neoadjuvant tyrosine kinase inhibitor (TKI) therapy has reduced tumor burden and improved survival in both primary and recurrent gastrointestinal stromal tumors (GISTs). However, no clear guidelines exist on optimal patient selection for neoadjuvant therapy (NAT). Our aim was to analyze factors and outcomes associated with the therapeutic sequence of TKI therapy before and/or after surgery for gastric GISTs. METHODS We performed a retrospective study of patients surgically treated for a gastric GIST utilizing the 2006-2018 National Cancer Database. We examined demographic, clinical, and pathological characteristics associated with NAT versus adjuvant therapy (AT) using logistic regression. RESULTS Of the 3732 patients, 20.4% received NAT and 79.6% had AT. Among patients receiving therapy, NAT significantly increased over our study period (12% to 30.7%). A majority of the AT group received a partial gastrectomy (77.9%) compared with the NAT group who received more near-total/total gastrectomy or gastrectomy with en bloc resection (p < 0.001). In a multivariable model, patients were more likely to receive NAT when insured (private, aOR: 2.37, 95% CI: 1.31-4.29), treated at an academic/research program (aOR: 1.83, 95% CI: 1.49-2.56), had tumors located in the proximal stomach (aOR: 1.40, 95% CI: 1.06-1.86), tumor size > 10 cm (aOR: 1.88, 95% CI: 1.41-2.51), and received near-total/total gastrectomy (aOR: 1.81, 95% CI: 1.42-2.29). There were no differences in outcomes. CONCLUSION NAT for gastric GIST has increased in utilization. NAT was used in patients with larger tumors and who underwent more extensive resection. Despite these factors, outcomes were similar to patients receiving only AT. More studies are required to determine the therapeutic sequence for gastric GISTs.
Collapse
Affiliation(s)
| | - Arkadii Sipok
- Department of Surgery, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Jace P Landry
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Lolita Ramsey
- Department of Surgery, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Cara J Joyce
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | | |
Collapse
|
141
|
Li S, Zhao S, Liang N, Zhang S, Zhang L, Zhou L, Liu A, Cao X, Tian J, Yu Y, Fan Z, Xiao K, Wang M, Zhao H, Bai R, Sun J. SPRY4 inhibits and sensitizes the primary KIT mutants in gastrointestinal stromal tumors (GISTs) to imatinib. Gastric Cancer 2023; 26:677-690. [PMID: 37222910 DOI: 10.1007/s10120-023-01402-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND KIT is frequently mutated in gastrointestinal stromal tumors (GISTs), and the treatment of GISTs largely relies on targeting KIT currently. In this study, we aimed to investigate the role of sprouty RTK signaling antagonist 4 (SPRY4) in GISTs and related mechanisms. METHODS Ba/F3 cells and GIST-T1 cell were used as cell models, and mice carrying germline KIT/V558A mutation were used as animal model. Gene expression was examined by qRT-PCR and western blot. Protein association was examined by immunoprecipitation. RESULTS Our study revealed that KIT increased the expression of SPRY4 in GISTs. SPRY4 was found to bind to both wild-type KIT and primary KIT mutants in GISTs, and inhibited KIT expression and activation, leading to decreased cell survival and proliferation mediated by KIT. We also observed that inhibition of SPRY4 expression in KITV558A/WT mice led to increased tumorigenesis of GISTs in vivo. Moreover, our results demonstrated that SPRY4 enhanced the inhibitory effect of imatinib on the activation of primary KIT mutants, as well as on cell proliferation and survival mediated by the primary KIT mutants. However, in contrast to this, SPRY4 did not affect the expression and activation of drug-resistant secondary KIT mutants, nor did it affect the sensitivity of secondary KIT mutants to imatinib. These findings suggested that secondary KIT mutants regulate a different downstream signaling cascade than primary KIT mutants. CONCLUSIONS Our results suggested that SPRY4 acts as negative feedback of primary KIT mutants in GISTs by inhibiting KIT expression and activation. It can increase the sensitivity of primary KIT mutants to imatinib. In contrast, secondary KIT mutants are resistant to the inhibition of SPRY4.
Collapse
Affiliation(s)
- Shujing Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Department of Pediatrics, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Sien Zhao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Nianhai Liang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Shaoting Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Liangying Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Liangji Zhou
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Anbu Liu
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xu Cao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jinhai Tian
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yuanyuan Yu
- Department of Emergency, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhaoyang Fan
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Kun Xiao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ming Wang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ru Bai
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jianmin Sun
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology Center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
142
|
Toulmonde M, Dinart D, Brahmi M, Verret B, Jean-Denis M, Ducimetière F, Desolneux G, Méeus P, Palussière J, Buy X, Bouhamama A, Gillon P, Dufresne A, Hénon C, Le Loarer F, Karanian M, Ngo C, Mathoulin-Pélissier S, Bellera C, Le Cesne A, Blay JY, Italiano A. Evolution of Patterns of Care and Outcomes in the Real-Life Setting for Patients with Metastatic GIST Treated in Three French Expert Centers over Three Decades. Cancers (Basel) 2023; 15:4306. [PMID: 37686582 PMCID: PMC10487208 DOI: 10.3390/cancers15174306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Gastrointestinal stromal tumors (GIST) are rare mesenchymal tumors characterized by KIT or PDGFRA mutations. Over three decades, significant changes in drug discovery and loco-regional (LR) procedures have impacted treatment strategies. We assessed the evolution of treatment strategies for metastatic GIST patients treated in the three national coordinating centers of NetSarc, the French network of sarcoma referral centers endorsed by the National Institute of Cancers, from 1990 to 2018. The primary objective was to describe the clinical and biological profiles as well as the treatment modalities of patients with metastatic GIST in a real-life setting, including access to clinical trials and LR procedures in the metastatic setting. Secondary objectives were to assess (1) patients' outcome in terms of time to next treatment (TNT) for each line of systemic treatment, (2) patients' overall survival (OS), (3) evolution of patients' treatment modalities and OS according to treatment access: <2002 (pre-imatinib approval), 2002-2006 (pre-sunitinib approval), 2006-2014 (pre-regorafenib approval), post 2014, and (4) the impact of clinical trials and LR procedures on TNT and OS in the metastatic setting. 1038 patients with a diagnosis of GIST made in one of the three participating centers between 1990 and 2018 were included in the national prospective database. Among them, 492 patients presented metastasis, either synchronous or metachronous. The median number of therapy lines in the metastatic setting was 3 (range 0-15). More than half of the patients (55%) participated in a clinical trial during the course of their metastatic disease and half (51%) underwent additional LR procedures on metastatic sites. The median OS in the metastatic setting was 83.4 months (95%CI [72.7; 97.9]). The median TNT was 26.7 months (95%CI [23.4; 32.3]) in first-line, 10.2 months (95%CI [8.6; 11.8]) in second line, 6.7 months (95%CI [5.3; 8.5]) in third line, and 5.5 months (95%CI [4.3; 6.7]) in fourth line, respectively. There was no statistical difference in OS in the metastatic setting between the four therapeutic periods (log rank, p = 0.18). In multivariate analysis, age, AFIP Miettinen classification, mutational status, surgery of the primary tumor, participation in a clinical trial in the first line and LR procedure to metastatic sites were associated with longer TNT in the first line, whereas age, mitotic index, mutational status, surgery of the primary tumor and LR procedure to metastatic sites were associated with longer OS. This real-life study advocates for early reference of metastatic GIST patients to expert centers to orchestrate the best access to future innovative clinical trials together with LR strategies and further improve GIST patients' survival.
Collapse
Affiliation(s)
- Maud Toulmonde
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France;
| | - Derek Dinart
- Department of Epidemiology and Clinical Research, Institut Bergonié, 33076 Bordeaux, France; (D.D.); (S.M.-P.); (C.B.)
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Leon Berard, 69373 Lyon, France; (M.B.); (A.D.); (J.Y.B.)
| | - Benjamin Verret
- Department of Medical Oncology, Gustave Roussy, 94800 Villejuif, France; (B.V.); (C.H.); (A.L.C.)
| | - Myriam Jean-Denis
- Department of Epidemiology and Clinical Research, Centre Leon Berard, 69373 Lyon, France; (M.J.-D.); (F.D.)
| | - Françoise Ducimetière
- Department of Epidemiology and Clinical Research, Centre Leon Berard, 69373 Lyon, France; (M.J.-D.); (F.D.)
| | - Gregoire Desolneux
- Department of Surgical Oncology, Institut Bergonié, 33076 Bordeaux, France;
| | - Pierre Méeus
- Department of Surgical Oncology, Centre Leon Berard, 69373 Lyon, France;
| | - Jean Palussière
- Department of Radiodiagnostic and Interventional Radiology, Institut Bergonié, 33076 Bordeaux, France; (J.P.); (X.B.)
| | - Xavier Buy
- Department of Radiodiagnostic and Interventional Radiology, Institut Bergonié, 33076 Bordeaux, France; (J.P.); (X.B.)
| | - Amine Bouhamama
- Department of Radiodiagnostic and Interventional Radiology, Centre Leon Berard, 69373 Lyon, France;
| | - Pauline Gillon
- Department of Radiation Oncology, Institut Bergonié, 33076 Bordeaux, France;
| | - Armelle Dufresne
- Department of Medical Oncology, Centre Leon Berard, 69373 Lyon, France; (M.B.); (A.D.); (J.Y.B.)
| | - Clémence Hénon
- Department of Medical Oncology, Gustave Roussy, 94800 Villejuif, France; (B.V.); (C.H.); (A.L.C.)
| | | | - Marie Karanian
- Department of Pathology, Centre Leon Berard, 69373 Lyon, France;
| | - Carine Ngo
- Department of Pathology, Gustave Roussy, 94800 Villejuif, France;
| | - Simone Mathoulin-Pélissier
- Department of Epidemiology and Clinical Research, Institut Bergonié, 33076 Bordeaux, France; (D.D.); (S.M.-P.); (C.B.)
| | - Carine Bellera
- Department of Epidemiology and Clinical Research, Institut Bergonié, 33076 Bordeaux, France; (D.D.); (S.M.-P.); (C.B.)
| | - Axel Le Cesne
- Department of Medical Oncology, Gustave Roussy, 94800 Villejuif, France; (B.V.); (C.H.); (A.L.C.)
| | - Jean Yves Blay
- Department of Medical Oncology, Centre Leon Berard, 69373 Lyon, France; (M.B.); (A.D.); (J.Y.B.)
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, 33076 Bordeaux, France;
| |
Collapse
|
143
|
Bangolo A, Fwelo P, Al-Qatish T, Bukasa-Kakamba J, Lee T, Cayago AG, Potiguara S, Nagesh VK, Kawall J, Ahmed R, Asjad Abbas M, Nursjamsi N, Lee SH, Meti S, Arana GV, Joseph CA, Mohamed A, Alencar A, Hassan HG, Aryal P, Javed A, Kalinin M, Lawal G, Khalaf IY, Mathew M, Karamthoti P, Gupta B, Weissman S. Outcomes of Patients with Gastrointestinal Stromal Tumors in the Past Decade. Med Sci (Basel) 2023; 11:54. [PMID: 37755158 PMCID: PMC10536810 DOI: 10.3390/medsci11030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms of the gastrointestinal tract (GIT) that represent approximately 1 to 2 percent of primary gastrointestinal (GI) cancers. Owing to their rarity, very little is known about their overall epidemiology, and the prognostic factors of their pathology. The current study aimed to evaluate the independent determinants of mortality in patients diagnosed with GISTs over the past decade. METHODS Our study comprised 2374 patients diagnosed with GISTs from 2000 to 2017 from the Surveillance, Epidemiology, and End Results (SEER) database. We analyzed the baseline characteristics, and overall mortality (OM), as well as the cancer-specific mortality (CSM) of GISTs. Variables with a p value < 0.01 in the univariate Cox regression were incorporated into the multivariate Cox model, to determine the independent prognostic factors. RESULTS Multivariate Cox proportional hazard regression analyses of factors affecting the all-cause mortality and GIST-related mortality among US patients between 2010 and 2017 revealed a higher overall mortality in non-Hispanic Black patients (HR = 1.516, 95% CI 1.172-1.961, p = 0.002), patients aged 80+ (HR = 9.783, 95% CI 4.185-22.868, p = 0), followed by those aged 60-79 (HR = 3.408, 95% CI 1.488-7.807, p = 0.004); male patients (HR = 1.795, 95% CI 1.461-2.206, p < 0.001); patients with advanced disease with distant metastasis (HR = 3.865, 95% CI 2.977-5.019, p < 0.001), followed by cases with regional involvement via both direct extension and lymph node involvement (HR = 3.853, 95% CI 1.551-9.57, p = 0.004); and widowed patients (HR = 1.975, 95% CI 1.494-2.61, p < 0.001), followed by single patients (HR = 1.53, 95% CI 1.154-2.028, p = 0.003). The highest CSM was observed in the same groups, except widowed patients and patients aged 60-79. The highest CSM was also observed among patients that underwent chemotherapy (HR = 1.687, 95% CI 1.19-2.392, p = 0.003). CONCLUSION In this updated study on the outcomes of patients with GISTs, we found that non-Hispanic Black patients, male patients, and patients older than 60 years have a higher mortality with GISTs. Furthermore, patients who have received chemotherapy have a higher GIST-specific mortality, and married patients have a lower mortality. However, we do not know to what extent these independent prognostic factors interact with each other to influence mortality. This study paves the way for future studies addressing these interactions. The results of this study may help treating clinicians to identify patient populations associated with a dismal prognosis, as those may require closer follow-up and more intensive therapy; furthermore, with married patients having a better survival rate, we hope to encourage clinicians to involve family members of the affected patients early in the disease course, as the social support might impact the prognosis.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Pierre Fwelo
- Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, TX 77204, USA
| | - Tha’er Al-Qatish
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - John Bukasa-Kakamba
- Division of Endocrinology, Department of Medicine, Kinshasa University Clinics, Kinshasa 7948, Democratic Republic of the Congo;
| | - Tiffany Lee
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Akira G. Cayago
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Sarah Potiguara
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Vignesh K. Nagesh
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Jessica Kawall
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Rashid Ahmed
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Muhammad Asjad Abbas
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Narissa Nursjamsi
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Stacy H. Lee
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Shagi Meti
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Georgemar V. Arana
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Chrishanti A. Joseph
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Abdifitah Mohamed
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Arthur Alencar
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Huzaifa G. Hassan
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Pramanu Aryal
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Aleena Javed
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Maksim Kalinin
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Gbenga Lawal
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Ibtihal Y. Khalaf
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Midhun Mathew
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Praveena Karamthoti
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| | - Bhavna Gupta
- Division of Hematology and Oncology, Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA
| | - Simcha Weissman
- Department of Medicine, Hackensack Meridian Health/Palisades Medical Center, North Bergen, NJ 07047, USA; (T.A.-Q.); (S.P.); (G.V.A.); (C.A.J.); (M.K.); (G.L.); (I.Y.K.)
| |
Collapse
|
144
|
Rönnberg H. Signal Transduction Inhibitors. THERAPEUTIC STRATEGIES IN VETERINARY ONCOLOGY 2023:89-110. [DOI: 10.1079/9781789245820.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
145
|
Serrano C, Martín-Broto J, Asencio-Pascual JM, López-Guerrero JA, Rubió-Casadevall J, Bagué S, García-del-Muro X, Fernández-Hernández JÁ, Herrero L, López-Pousa A, Poveda A, Martínez-Marín V. 2023 GEIS Guidelines for gastrointestinal stromal tumors. Ther Adv Med Oncol 2023; 15:17588359231192388. [PMID: 37655207 PMCID: PMC10467260 DOI: 10.1177/17588359231192388] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST) is the most common malignant neoplasm of mesenchymal origin. GIST spans a wide clinical spectrum that ranges from tumors with essentially no metastatic potential to malignant and life-threatening spread diseases. Gain-of-function mutations in KIT or PDGFRA receptor tyrosine kinases are the crucial drivers of most GISTs, responsible for tumor initiation and evolution throughout the entire course of the disease. The introduction of tyrosine kinase inhibitors targeting these receptors has substantially improved the outcomes in this formerly chemoresistant cancer. As of today, five agents hold regulatory approval for the treatment of GIST: imatinib, sunitinib, regorafenib, ripretinib, and avapritinib. This, in turn, represents a success for a rare neoplasm. During the past two decades, GIST has become a paradigmatic model in cancer for multidisciplinary work, given the disease-specific particularities regarding tumor biology and tumor evolution. Herein, we review currently available evidence for the management of GIST. This clinical practice guideline has been developed by a multidisciplinary expert panel (oncologist, pathologist, surgeon, molecular biologist, radiologist, and representative of patients' advocacy groups) from the Spanish Group for Sarcoma Research, and it is conceived to provide, from a critical perspective, the standard approach for diagnosis, treatment, and follow-up.
Collapse
Affiliation(s)
- César Serrano
- Sarcoma Translational Research Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Vall d’Hebron Barcelona Hospital Campus, Carrer de Natzaret, 115-117, Barcelona 08035, Spain
| | - Javier Martín-Broto
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain Instituto de investigación Sanitaria Fundación Jimenez Diaz (IIS/FJD; UAM), Madrid, Spain
| | - José Manuel Asencio-Pascual
- Department of General Surgery, Gregorio Marañón University Hospital, Madrid, Spain
- Department of Surgery, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Jordi Rubió-Casadevall
- Department of Medical Oncology, Catalan Institute of Oncology, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Silvia Bagué
- Department of Pathology, Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| | - Xavier García-del-Muro
- Department of Medical Oncology, Institut Català d’Oncologia, IDIBELL and University of Barcelona, Barcelona, Spain
| | | | - Luís Herrero
- GIST advocacy group – Colectivo GIST, Valladolid, Spain
| | - Antonio López-Pousa
- Department of Pathology, Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| | - Andrés Poveda
- Initia Oncologia, Hospital Quironsalud, Valencia, Spain
| | | |
Collapse
|
146
|
Pharaon N, Habbal W, Monem F. Bioinformatic analysis of KIT juxtamembrane domain mutations in Syrian GIST patients: jigsaw puzzle completed. J Egypt Natl Canc Inst 2023; 35:25. [PMID: 37574490 DOI: 10.1186/s43046-023-00185-0] [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: 07/14/2021] [Accepted: 07/27/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND The huge number of detected somatic KIT mutations highlights the necessity of in silico analyses that are almost absent in the relevant medical literature. The aim of this study is to report the mutation spectrum analysis of exon 11 encoding the juxtamembrane (JM) domain of the KIT gene in a group of Syrian GIST patients. METHODS Forty-eight formalin-fixed paraffin-embedded GIST tissue samples, collected between 2006 and 2016, were retrieved from the pathological archives and analyzed for KIT exon 11 mutations by DNA sequencing. Structural/functional impact of detected variants was predicted using several bioinformatic tools. RESULTS Twenty-one different variants have been detected in intron 10, exon 11, and intron 11 of the KIT gene, eight of which were novel changes. Mutations in exon 11 of the KIT gene were detected in 28 of 48 (58.3%) GIST patients and predicted to be pathogenic and cancer promoting. Specifically, age above 60 was very significantly associated with the negative selection of deletion mutations (p = .007), a phenomenon that points to deletion severity. CONCLUSIONS Six bioinformatic tools have proved efficient in predicting the impact of detected KIT variations in view of published structural, experimental, and clinical findings.
Collapse
Affiliation(s)
- Nour Pharaon
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - Wafa Habbal
- Clinical Laboratories Department, Al-Assad Hospital, Damascus University, PO Box 10769, Damascus, Syria.
| | - Fawza Monem
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
- Clinical Laboratories Department, Al-Assad Hospital, Damascus University, PO Box 10769, Damascus, Syria
| |
Collapse
|
147
|
Tzikos G, Menni AE, Krokou D, Vouchara A, Doutsini S, Karlafti E, Karakatsanis A, Ioannidis A, Panidis S, Papavramidis T, Michalopoulos A, Paramythiotis D. Gastrointestinal Stromal Tumors: Our Ten-Year Experience of a Single-Center Tertiary Hospital. J Pers Med 2023; 13:1254. [PMID: 37623504 PMCID: PMC10455766 DOI: 10.3390/jpm13081254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal neoplasms of the gastrointestinal tract. They have variable clinical presentation, prognosis, and molecular characteristics. Here, we present the results of our retrospective study including patients operated on for GIST during the last decade. METHODS All the patients who underwent GIST resection during the decade 2008-2018 were included in the study. The diagnosis was based on the pathology report. All the data were collected and analyzed statistically using the Statistical Package for Social Science v25.0. Finally, after having applied the proper search terms, a comprehensive review of articles published in the Medline database was held. RESULTS Thirty-two patients (sixteen women) were included in the study with a mean age of 69.6 years old (SD = 13.9). Twenty-one patients had a GIST in the stomach, eight in the small intestine, and three had an extra GIST. Of the 29 patients contacted, 21 were alive with a mean survival time of 74.3 months (SD = 49.6 months, min: 3.0 months, max: 161.0 months), whereas eight patients passed away. Finally, 13 patients were treated with tyrosine kinase inhibitors (TKIs) of whom only one died, while 9 patients passed away from those treated with surgery alone (p = 0.031). CONCLUSIONS Our results were in concordance with the existing data in the literature. GISTs require patient-based therapeutical management depending on the histology of the tumors. Gastric tumors present a better prognosis than those localized in the intestine, while the use of TKIs has led to an improvement in patient survival rate.
Collapse
Affiliation(s)
- Georgios Tzikos
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Alexandra-Eleftheria Menni
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Despoina Krokou
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Angeliki Vouchara
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Soultana Doutsini
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Eleni Karlafti
- Emergency Department, AHEPA University General Hospital of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Anestis Karakatsanis
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Aristeidis Ioannidis
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Stavros Panidis
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Theodosios Papavramidis
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Antonios Michalopoulos
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| | - Daniel Paramythiotis
- 1st Propedeutic Department of Surgery, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (A.-E.M.); (D.K.); (A.V.); (S.D.); (A.K.); (A.I.); (S.P.); (T.P.); (A.M.); (D.P.)
| |
Collapse
|
148
|
Zagami P, Comandone A, Fiore M, Baldi GG, Grignani G, Vincenzi B, Gronchi A, Antonarelli G, Boglione A, Pennacchioli E, Curigliano G, Conforti F, De Pas TM. The rare entity of gastrointestinal leiomyosarcomas: An Italian multicenter retrospective study in high-volume referral centers. Cancer Med 2023; 12:17047-17055. [PMID: 37455549 PMCID: PMC10501229 DOI: 10.1002/cam4.6340] [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: 04/26/2023] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND After a huge efficacy of imatinib in treating patients with gastrointestinal stromal tumors (GISTs) was proven, a maximum effort was made to make a differential diagnosis between GISTs and gastrointestinal leiomyosarcomas (GI-LMS), showing the latter to be an extremely rare tumor entity. Limited data on GI-LMS biology, clinical behavior and drug-sensibility are available, and the clinical decision-making in this subgroup of patients is usually challenging. METHODS We conducted a multicenter, retrospective observational study on patients with diagnosed GI-LMS from 2004 to 2020 within six high-volume referral centers in Italy. RESULTS Thirty-three patients had diagnosis of KIT-negative GI-LMS confirmed by sarcoma-expert pathologist. The most common site of origin was the intestine. Twenty-two patients had localized disease and underwent surgery: with a median follow-up of 72 months, median disease-free survival was 42 months. Overall survival (OS)-rate at 5 years was 73% and median OS was 193 months. Five out of 10 patients with local relapse received a salvage surgery, and 2/5 remained with no evidence of disease. Thirteen patients received neoadjuvant (6) or adjuvant (7) chemotherapy, and 2/13 patients remained free from relapse. The median OS for patients with metastatic LMS was 16.4 months. CONCLUSION GI-LMS is very rare and extremely aggressive subgroup of sarcomas with a high tendency to systemic spread. Localized GI-LMS at diagnosis may be cured if treated with adequate surgery with or without (neo) adjuvant chemotherapy, while de-novo metastatic disease appeared to have a poor prognosis. Clinical effort to understand GI-LMS biology and clinical behavior and to develop active treatment strategy, especially for metastatic-disease, is warranted.
Collapse
Affiliation(s)
- Paola Zagami
- Department of Oncology and HematologyUniversity of MilanMilanItaly
| | | | - Marco Fiore
- Fondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
| | | | - Giovanni Grignani
- Medical OncologyCandiolo Cancer Institute‐FPO, IRCCSCandioloItaly
- Department of Oncology AOU Citta della Salute e della Scienza di TorinoTorinoItaly
| | - Bruno Vincenzi
- Department of Medical OncologyUniversità Campus Bio‐Medico di RomaRomeItaly
| | | | | | | | | | - Giuseppe Curigliano
- Department of Oncology and HematologyUniversity of MilanMilanItaly
- Division of New Drugs and Early Drug Development for Innovative TherapiesEuropean Institute of Oncology, IRCCSMilanItaly
| | - Fabio Conforti
- Division of Medical Oncology of MelanomaSarcoma and Rare tumors, European Institute of Oncology, IRCCSMilanItaly
- Oncology DepartmentHumanitas GavazzeniBergamoItaly
| | - Tommaso Martino De Pas
- Division of Medical Oncology of MelanomaSarcoma and Rare tumors, European Institute of Oncology, IRCCSMilanItaly
- Oncology DepartmentHumanitas GavazzeniBergamoItaly
| |
Collapse
|
149
|
De Sutter L, Wozniak A, Verreet J, Vanleeuw U, De Cock L, Linde N, Drechsler C, Esdar C, Sciot R, Schöffski P. Antitumor Efficacy of the Novel KIT Inhibitor IDRX-42 (Formerly M4205) in Patient- and Cell Line-Derived Xenograft Models of Gastrointestinal Stromal Tumor (GIST). Clin Cancer Res 2023; 29:2859-2868. [PMID: 37223931 DOI: 10.1158/1078-0432.ccr-22-3822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/17/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
PURPOSE The majority of gastrointestinal stromal tumors (GIST) are driven by constitutively activated KIT/PDGFRA kinases and are susceptible to treatment with tyrosine kinase inhibitors. During treatment, most of these tumors will develop secondary mutations in KIT or PDGFRA inducing drug resistance, so there is an unmet need for novel therapies. We tested the efficacy of IDRX-42, a novel selective KIT inhibitor with high activity toward the most relevant KIT mutations, in 4 GIST xenograft models. EXPERIMENTAL DESIGN NMRI nu/nu mice were transplanted with patient-derived GIST xenograft models UZLX-GIST9 (KIT:p.P577del;W557LfsX5;D820G), UZLX-GIST2B (KIT:p.A502_Y503dup), UZLX-GIST25 (KIT:p.K642E), and the cell line-derived model GIST882 (KIT:p.K642E). Mice were treated daily with vehicle (control), imatinib (100 mg/kg), sunitinib (20 mg/kg), avapritinib (5 mg/kg), or IDRX-42 (10 mg/kg, 25 mg/kg). Efficacy was assessed by tumor volume evolution, histopathology, grading of histologic response, and IHC. The Kruskal-Wallis and Wilcoxon matched-pairs tests were used for statistical analysis, with P < 0.05 considered as significant. RESULTS IDRX-42 (25 mg/kg) caused tumor volume shrinkage in UZLX-GIST25, GIST882, and UZLX-GIST2B, with a relative decrease to 45.6%, 57.3%, and 35.1% on the last day as compared with baseline, and tumor growth delay (160.9%) compared with control in UZLX-GIST9. Compared with controls, IDRX-42 (25 mg/kg) induced a significant decrease in mitosis. In UZLX-GIST25 and GIST882 grade 2-4 histologic response with myxoid degeneration was observed in all IDRX-42 (25 mg/kg)-treated tumors. CONCLUSIONS IDRX-42 showed significant antitumor activity in patient- and cell line-derived GIST xenograft models. The novel kinase inhibitor induced volumetric responses, decreased mitotic activity, and had antiproliferative effects. In models with KIT exon 13 mutation IDRX-42 induced characteristic myxoid degeneration.
Collapse
Affiliation(s)
- Luna De Sutter
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Jasper Verreet
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Ulla Vanleeuw
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Lore De Cock
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Nina Linde
- The healthcare business of Merck KGaA, Darmstadt, Germany
| | | | | | - Raf Sciot
- Department of Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
150
|
Fu Y, Karanian M, Perret R, Camara A, Le Loarer F, Jean-Denis M, Hostein I, Michot A, Ducimetiere F, Giraud A, Courreges JB, Courtet K, Laizet Y, Bendjebbar E, Du Terrail JO, Schmauch B, Maussion C, Blay JY, Italiano A, Coindre JM. Deep learning predicts patients outcome and mutations from digitized histology slides in gastrointestinal stromal tumor. NPJ Precis Oncol 2023; 7:71. [PMID: 37488222 PMCID: PMC10366108 DOI: 10.1038/s41698-023-00421-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 06/29/2023] [Indexed: 07/26/2023] Open
Abstract
Risk assessment of gastrointestinal stromal tumor (GIST) according to the AFIP/Miettinen classification and mutational profiling are major tools for patient management. However, the AFIP/Miettinen classification depends heavily on mitotic counts, which is laborious and sometimes inconsistent between pathologists. It has also been shown to be imperfect in stratifying patients. Molecular testing is costly and time-consuming, therefore, not systematically performed in all countries. New methods to improve risk and molecular predictions are hence crucial to improve the tailoring of adjuvant therapy. We have built deep learning (DL) models on digitized HES-stained whole slide images (WSI) to predict patients' outcome and mutations. Models were trained with a cohort of 1233 GIST and validated on an independent cohort of 286 GIST. DL models yielded comparable results to the Miettinen classification for relapse-free-survival prediction in localized GIST without adjuvant Imatinib (C-index=0.83 in cross-validation and 0.72 for independent testing). DL splitted Miettinen intermediate risk GIST into high/low-risk groups (p value = 0.002 in the training set and p value = 0.29 in the testing set). DL models achieved an area under the receiver operating characteristic curve (AUC) of 0.81, 0.91, and 0.71 for predicting mutations in KIT, PDGFRA and wild type, respectively, in cross-validation and 0.76, 0.90, and 0.55 in independent testing. Notably, PDGFRA exon18 D842V mutation, which is resistant to Imatinib, was predicted with an AUC of 0.87 and 0.90 in cross-validation and independent testing, respectively. Additionally, novel histological criteria predictive of patients' outcome and mutations were identified by reviewing the tiles selected by the models. As a proof of concept, our study showed the possibility of implementing DL with digitized WSI and may represent a reproducible way to improve tailoring therapy and precision medicine for patients with GIST.
Collapse
Affiliation(s)
- Yu Fu
- Owkin, Inc., New York, NY, USA
| | - Marie Karanian
- Cancer Research Center of Lyon, Centre Léon Bérard, Lyon, France
| | - Raul Perret
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | | | - François Le Loarer
- Department of Biopathology, Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | | | | | - Audrey Michot
- Department of Surgical Oncology, Institut Bergonié, Bordeaux, France
| | | | - Antoine Giraud
- Clinical Research and Clinical Epidemiology Unit, Institut Bergonié, Bordeaux, France
| | | | - Kevin Courtet
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | - Yech'an Laizet
- Department of Biopathology, Institut Bergonié, Bordeaux, France
| | | | | | | | | | - Jean-Yves Blay
- Cancer Research Center of Lyon, Centre Léon Bérard, Lyon, France
| | - Antoine Italiano
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | - Jean-Michel Coindre
- Department of Biopathology, Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| |
Collapse
|