1
|
Mo YK, Chen XP, Hong LL, Hu YR, Lin DY, Xie LC, Dai ZZ. Gastric schwannoma: Computed tomography and perigastric lymph node characteristics. World J Gastrointest Oncol 2025; 17:102085. [DOI: 10.4251/wjgo.v17.i4.102085] [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: 10/08/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Gastric schwannoma (GS) is often misdiagnosed as gastrointestinal stromal tumors due to the high incidence of the latter. However, these two types differ significantly in pathology and biological behavior.
AIM To evaluate the computed tomography characteristics of GS and provide insights into its accurate diagnosis.
METHODS Twenty-three cases of GS confirmed between January 2011 and December 2023 were assessed clinically and radiologically. Imaging characteristics, including tumor location, size, contour, ulceration, growth pattern, enhancement degree and pattern, cystic change, calcification, and perigastric lymph nodes (PLNs), were reviewed by two experienced radiologists.
RESULTS Our sample included 18 females and 5 males, with a median age of 54.7 years. A total of 39.1% of cases were asymptomatic. GSs appeared as oval and well-defined submucosal tumors, with exophytic (43.5%) or mixed (endoluminal + exophytic; 43.5%) growth patterns. The tumors were primarily located in the gastric body (78.3%). Ulcerations were observed in 8 cases (34.5%), and PLNs were observed in 15 cases (65%). The average degree of enhancement was 48.3 Hounsfield units. Twenty cases (87%) showed peak enhancement in the delayed phase. Most GSs were homogeneous, while cystic change (13.0%) and calcification (17.4%) were rare.
CONCLUSION GS predominantly showed gradual homogenous enhancement with peak enhancement in the delayed phase. PLNs around GS are helpful in differentiating GS from other gastric submucosal tumors.
Collapse
Affiliation(s)
- Yong-Kang Mo
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515000, Guangdong Province, China
| | - Xia-Pu Chen
- Department of Radiology, Shantou Central Hospital, Shantou 515000, Guangdong Province, China
| | - Liang-Li Hong
- Department of Pathology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515000, Guangdong Province, China
| | - Yi-Ru Hu
- Clinical Research Center, Shantou Central Hospital, Shantou 515000, Guangdong Province, China
| | - Dai-Ying Lin
- Department of Radiology, Shantou Central Hospital, Shantou 515000, Guangdong Province, China
| | - Li-Chun Xie
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515000, Guangdong Province, China
| | - Zhuo-Zhi Dai
- Department of Radiology, Shantou Central Hospital, Shantou 515000, Guangdong Province, China
| |
Collapse
|
2
|
Tardy KJ, Kwak HV, Tieniber AD, Mangold AK, Perez JE, Do K, Zeng S, Rossi F, DeMatteo RP. Intratumoral Bacteria are Uncommon in Gastrointestinal Stromal Tumor. Ann Surg Oncol 2025; 32:1504-1510. [PMID: 39578323 PMCID: PMC11811456 DOI: 10.1245/s10434-024-16526-9] [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: 06/11/2024] [Accepted: 10/30/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Gastrointestinal stromal tumor (GIST) is the most common human sarcoma with over 5000 new patients diagnosed in the USA each year. The tumor originates from the interstitial cells of Cajal and forms an intramural lesion most commonly in the stomach or small intestine. The gut microbiome has been linked to other gastrointestinal cancers and a recent paper purported that GISTs contain substantial intratumoral bacteria. The purpose of this study is to further evaluate the presence of bacteria in GISTs. PATIENTS AND METHODS We collected 25 tumor samples of varying size and location from 24 patients under sterile conditions in the operating room immediately following surgical resection. 16S quantitative polymerase chain reaction (qPCR) and 16S ribosomal RNA (rRNA) gene amplicon sequencing were performed to evaluate the bacterial species present in each tumor. Retrospective chart review was performed to determine tumor characteristics, including tumor size, location, imatinib exposure, and mucosal involvement. RESULTS In 23 of the 25 tumor samples, there were fewer than 100 copy numbers of 16S rRNA per uL, indicating an absence of a significant bacterial load. 16S rRNA gene amplicon sequencing of the remaining two samples, one gastric tumor and one duodenal tumor, revealed the presence of normal intestinal bacteria. These two tumors, along with three others, had disruption of the mucosal lining. CONCLUSIONS GISTs generally lack substantial bacteria, except in some cases when the tumor disrupts the mucosa.
Collapse
Affiliation(s)
- Katherine J Tardy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hyunjee V Kwak
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D Tieniber
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alina K Mangold
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Juan E Perez
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Do
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shan Zeng
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ferdinando Rossi
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ronald P DeMatteo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Larrain C, Pu T, Cox P, Sprott K, Blakely AM, Bauer S, George S. INSIGHT: A Phase III Trial of Ripretinib Versus Sunitinib in Patients with Advanced GIST with KIT Exon 11 and Exon 17/18 Mutations Who Were Previously Treated with Imatinib. Ann Surg Oncol 2025:10.1245/s10434-024-16853-x. [PMID: 39907875 DOI: 10.1245/s10434-024-16853-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025]
Affiliation(s)
- Carolina Larrain
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tracey Pu
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paulina Cox
- Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Kam Sprott
- Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Andrew M Blakely
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| | | |
Collapse
|
4
|
Florou V, Jacobs MF, Casey R, Evans D, Owens B, Raygada M, Rothschild S, Greenberg SE. A Review of Genomic Testing and SDH- Deficiency in Gastrointestinal Stromal Tumors: Getting to the GIST. Cancer Med 2025; 14:e70669. [PMID: 39927693 PMCID: PMC11808740 DOI: 10.1002/cam4.70669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Gastrointestinal Stromal Tumors (GISTs) have seen significant advancements in their diagnosis and management, driven by targeted therapeutic development and molecular testing. The identification of mutations in genes such as KIT and PDGFRA has transformed treatment approaches, particularly through targeted therapies like imatinib, which have improved patient outcomes. This review explores the critical role of genomic testing in GIST, highlighting its importance in accurate diagnosis, treatment planning, and long-term surveillance for KIT/PDGFRA negative, SDH-deficient GISTs. SDH-deficient GISTs arise from mutations or epigenetic changes affecting the succinate dehydrogenase complex. The complexity of SDH-deficient GISTs, including their association with hereditary syndromes such as Hereditary Paraganglioma-Pheochromocytoma and/or hypermethylation of the SDHC promoter, underscores the need for comprehensive germline testing. Despite the availability of guidelines, variability exists in genomic testing recommendations across different regions, necessitating a unified approach. This review proposes a simplified algorithm for the genomic workup of GIST, and suggests all individuals with SDH-deficient GIST, regardless of germline testing result, require monitoring for additional SDHx-related tumors, given the lack of widely available methylation and full gene SDHA analysis.
Collapse
Affiliation(s)
- Vaia Florou
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer InstituteUniversity of UtahSalt Lake CityUtahUSA
| | - Michelle F. Jacobs
- Division of Genetic Medicine, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Ruth Casey
- Department of Medical GeneticsCambridge UniversityCambridgeUK
| | | | | | - Margarita Raygada
- Pediatric Oncology and Neuro‐Oncology BranchNational Cancer Institute/National Institutes of HealthBethesdaMarylandUSA
| | | | | |
Collapse
|
5
|
Gabellone S, Vanni S, Fausti V, Miserocchi G, Liverani C, Spadazzi C, Cocchi C, Calabrese C, Cavaliere D, Pacilio CA, Ercolani G, Pieri F, Gurrieri L, Riva N, Jones R, De Vita A. Exploring nanotechnology solutions for improved outcomes in gastrointestinal stromal tumors. Heliyon 2024; 10:e40596. [PMID: 39687122 PMCID: PMC11647801 DOI: 10.1016/j.heliyon.2024.e40596] [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: 11/21/2023] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Objectives Gastrointestinal stromal tumors, the most prevalent mesenchymal tumors (80 %) of the gastrointestinal tract, comprise less than 1 % of all gastrointestinal neoplasms and about 5 % of all sarcomas. Despite their rarity, Gastrointestinal stromal tumors present diverse clinical manifestations, anatomic locations, histological subtypes, and prognostic outcomes. Methods This scoping review comprehensively explores the epidemiology, clinical characteristics, diagnostic and prognostic modalities, as well as new therapeutic options for Gastrointestinal stromal tumors. Results A particular focus is placed on the promising role of bio-nanomaterials as multifunctional agents for drug delivery and 3D tumor microenvironment modeling. Bio-nanomaterials offer promising opportunities for targeted drug delivery, overcoming treatment resistance, and improving therapeutic efficacy. Conclusion Despite significant advancements, Gastrointestinal stromal tumors remain a complex clinical entity with ongoing challenges. The integration of nanotechnology into Gastrointestinal stromal tumors management offers the potential to enhance patient outcomes. Future studies should prioritize the development and evaluation of nanomaterial-based therapies in clinical trials to facilitate the translation of laboratory discoveries into real-world clinical applications.
Collapse
Affiliation(s)
- Sofia Gabellone
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Chiara Calabrese
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Davide Cavaliere
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121, Forlì, Italy
| | | | - Giorgio Ercolani
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121, Forlì, Italy
| | - Federica Pieri
- Pathology Unit, “Morgagni-Pierantoni” Hospital, 47121, Forlì, Italy
| | - Lorena Gurrieri
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Nada Riva
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| | - Robin Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, SW3 6JJ, London, UK
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014, Meldola, Italy
| |
Collapse
|
6
|
Grabill N, Louis M, Cawthon M, Conway J, Chambers J. Managing locally advanced GIST complicated by perforation: A case report and comprehensive review. Radiol Case Rep 2024; 19:4824-4831. [PMID: 39228941 PMCID: PMC11367461 DOI: 10.1016/j.radcr.2024.07.105] [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: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/05/2024] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract, frequently characterized by mutations in the KIT or PDGFRA genes. This case report details the complex clinical course of a 71-year-old female with a history of HIV and metastatic GIST presenting with acute abdominal symptoms indicative of perforated viscus. Initial imaging revealed a massive pneumoperitoneum and a large abdominal mass, necessitating immediate surgical intervention. The patient underwent multiple surgeries, including bowel resections and colostomy creation, to address the extensive tumor burden and complications. Postoperatively, she required intensive care management, including mechanical ventilation, vasopressor support, and hemodialysis for acute kidney injury. Pathological examination confirmed metastatic GIST with extensive mesenteric and omental involvement. Immunohistochemical staining was positive for CD117 (c-KIT) and DOG-1. Despite aggressive surgical and supportive measures, the patient's condition highlighted the significant challenges in managing advanced GIST with perforation. This case highlights the importance of a multidisciplinary approach, integrating surgical, medical, and intensive care to optimize outcomes. The prognosis of GIST varies widely, with localized tumors having favorable outcomes following resection, while metastatic cases often face a poorer prognosis despite advances in targeted therapies. This case exemplifies the critical need for personalized treatment plans and ongoing research to improve the management and prognosis of GIST patients.
Collapse
Affiliation(s)
- Nathaniel Grabill
- Northeast Georgia Medical Center, General Surgery Department. Gainesville, GA 30501, USA
| | - Mena Louis
- Northeast Georgia Medical Center, General Surgery Department. Gainesville, GA 30501, USA
| | - Mariah Cawthon
- Northeast Georgia Medical Center, General Surgery Department. Gainesville, GA 30501, USA
| | - Joseph Conway
- Northeast Georgia Medical Center, Pathology Department. Gainesville, GA 30501, USA
| | - James Chambers
- Northeast Georgia Medical Center, General Surgery Department, Braselton, GA 30517, USA
| |
Collapse
|
7
|
Li J, Zhang H, Chen XD. Effect and safety of ripretinib in the treatment of advanced gastrointestinal stromal tumor: A systematic review and meta-analysis. World J Clin Oncol 2024; 15:1092-1101. [PMID: 39193156 PMCID: PMC11346076 DOI: 10.5306/wjco.v15.i8.1092] [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: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Imatinib (IMA) has received approval as the primary treatment for gastrointestinal stromal tumors (GIST). Nonetheless, approximately half of the patients with advanced GIST show disease advancement following IMA treatment. Presently, the efficacy of secondary and tertiary medications in addressing various GIST secondary mutations is somewhat restricted. Consequently, there is a significant medical demand for the creation of kinase inhibitors that extensively block secondary drug-resistant mutations in advanced GIST. Ripretinib (RPT) is a new, switch-control tyrosine kinase inhibitors that can suppress different mutations of KIT and PDGFRA via a dual mechanism of action. AIM To investigate the literature on RPT to assess an effective, safe, and successful treatment strategy against advanced GIST. METHODS The present systematic review and meta-analysis was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. PubMed, Embase, Cochrane, Web of Science and ClinicalTrials.gov databases were screened from January 1, 2003 to May 1, 2024. RESULTS A total of 4 studies were included, with a total of 507 patients enrolled. The objective response rate (ORR) of the RPT-treated advanced GIST was 17% (95%CI: 0.11-0.27), while the disease control rate (DCR) was 66% (95%CI: 0.59-0.73). The overall occurrence of adverse events with varying degrees was 97% (95%CI: 0.93-1), whereas that of grade ≥ 3 adverse reactions was 42% (95%CI: 0.28-0.63). The sensitivity analysis revealed that omitting some studies did not yield statistically notable variances in the aggregate data regarding the ORR, DCR, and the occurrence of adverse events of grade 3 or higher. The publication bias was absent because no significant asymmetry was observed in Begg's funnel plot in all studies. CONCLUSION RPT has favorable efficacy profiles in GIST patients, but the adverse reactions are obvious, and patient management needs to be strengthened to achieve better safety and tolerability.
Collapse
Affiliation(s)
- Ji Li
- Department of General Surgery, Chongqing Western Hospital, Chongqing 400051, China
| | - Hao Zhang
- Department of General Surgery, Chongqing Western Hospital, Chongqing 400051, China
| | - Xiao-Dong Chen
- Department of General Surgery, Chongqing Western Hospital, Chongqing 400051, China
| |
Collapse
|
8
|
Gómez-Peregrina D, Cicala CM, Serrano C. Monitoring advanced gastrointestinal stromal tumor with circulating tumor DNA. Curr Opin Oncol 2024; 36:282-290. [PMID: 38726808 DOI: 10.1097/cco.0000000000001040] [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: 06/07/2024]
Abstract
PURPOSE OF REVIEW This review explores the role of circulating tumor (ct)DNA as a biomarker for clinical decision-making and monitoring purposes in metastatic gastrointestinal stromal tumor (GIST) patients. We discuss key insights from recent clinical trials and anticipate the future perspectives of ctDNA profiling within the clinical landscape of GIST. RECENT FINDINGS The identification and molecular characterization of KIT/platelet-derived growth factor receptor alpha (PDGFRA) mutations from ctDNA in metastatic GIST is feasible and reliable. Such identification through ctDNA serves as a predictor of clinical outcomes to tyrosine-kinase inhibitors (TKIs) in metastatic patients. Additionally, conjoined ctDNA analysis from clinical trials reveal the evolving mutational landscapes and increase in intratumoral heterogeneity across treatment lines. Together, this data positions ctDNA determination as a valuable tool for monitoring disease progression and guiding therapy in metastatic patients. These collective efforts culminated in the initiation of a ctDNA-based randomized clinical trial in GIST, marking a significant milestone in integrating ctDNA testing into the clinical care of GIST patients. SUMMARY The dynamic field of ctDNA technologies is rapidly evolving and holds significant promise for research. Several trials have successfully validated the clinical utility of ctDNA in metastatic GIST, laying the foundations for its prospective integration into the routine clinical management of GIST patients.
Collapse
Affiliation(s)
- David Gómez-Peregrina
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO)
| | - Carlo Maria Cicala
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO)
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - César Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO)
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
9
|
Liu AB, Liu J, Wang S, Ma L, Zhang JF. Biological role and expression of translationally controlled tumor protein (TCTP) in tumorigenesis and development and its potential for targeted tumor therapy. Cancer Cell Int 2024; 24:198. [PMID: 38835077 DOI: 10.1186/s12935-024-03355-9] [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: 12/07/2023] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Translationally controlled tumor protein (TCTP), also known as histamine-releasing factor (HRF) or fortilin, is a highly conserved protein found in various species. To date, multiple studies have demonstrated the crucial role of TCTP in a wide range of cellular pathophysiological processes, including cell proliferation and survival, cell cycle regulation, cell death, as well as cell migration and movement, all of which are major pathogenic mechanisms of tumorigenesis and development. This review aims to provide an in-depth analysis of the functional role of TCTP in tumor initiation and progression, with a particular focus on cell proliferation, cell death, and cell migration. It will highlight the expression and pathological implications of TCTP in various tumor types, summarizing the current prevailing therapeutic strategies that target TCTP.
Collapse
Affiliation(s)
- An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Jia Liu
- Medical Experimental Center, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Sheng Wang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| |
Collapse
|
10
|
Singh A, Chitti B, Aguiar C, Wernicke AG, Devoe CE, Rahman H, Sison C, Parashar B. Comparing gastrointestinal stromal tumor outcomes between geriatric and non-geriatric patients: A population-based analysis. World J Surg 2024; 48:1424-1432. [PMID: 38647223 DOI: 10.1002/wjs.12170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Gastrointestinal Stromal Tumors (GISTs) are the most common mesenchymal tumors of the GI tract. SEER is an extensive cancer database which proves useful in analyzing population trends. This analysis investigated GIST outcomes between geriatric & non-geriatric patients. METHODS SEER*STAT 8.4.0.1 was used to extract relevant GIST data from 2000 to 2019. Geriatric age was defined as ≥70 years. Variables included age, sex, surgery, cancer-specific death, and overall survival. Statistical tests included univariate analysis using KM survival estimate (95% confidence interval) to calculate 5-year survival (5YS). Log-Rank tests determined statistical significance. Multivariable Cox's PH regression estimated the geriatric hazard death ratio adjusted for sex, stage, and surgery. RESULTS The number of patients included was 13,579, yielding overall 5YS of 68.6% (95% CI 67.7-69.5). Cancer-specific death was 39.11% in 2000 & 3.33% in 2019. Non-geriatric & geriatric patient data yielded 5YS of 77.4% (76.4%-78.3%) and 53.3% (51.7%-54.8%) respectively (p < 0.0001). For no surgery/surgery, younger patient data yielded 5YS of 48.7% (45.8%-51.4%) and 83.7% (82.7%-84.7%) respectively (p < 0.0001); geriatric data yielded 5YS of 29.3% (26.5%-32.1%) and 62.8% (60.8%-64.6%) respectively (p < 0.0001). Multivariable analysis yielded a geriatric hazard death of 2.56 (2.42-2.70) (p < 0.0001). CONCLUSIONS Cancer-specific death decreased since 2000, indicating an improvement in survival & treatment methods. Observed lower survival rates overall in the geriatric group. Surgery appeared to enhance survival rates in both groups, suggesting that surgery is an important factor in GIST survival regardless of age. Large prospective studies will help define clinical management for geriatric patients.
Collapse
Affiliation(s)
- Abhiram Singh
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California, USA
| | - Bhargava Chitti
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
| | | | | | - Craig E Devoe
- Department of Medical Oncology, Northwell, New Hyde Park, New York, USA
| | - Husneara Rahman
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, USA
| | - Cristina Sison
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, New York, USA
| | - Bhupesh Parashar
- Department of Radiation Oncology, Northwell, New Hyde Park, New York, USA
| |
Collapse
|
11
|
Wang H, Zhao W, Wang D, Chen J. ANO6 (TMEM16F) inhibits gastrointestinal stromal tumor growth and induces ferroptosis. Open Med (Wars) 2024; 19:20240941. [PMID: 38756246 PMCID: PMC11097043 DOI: 10.1515/med-2024-0941] [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: 10/03/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Abstract
Herein, we elucidate the potential role of ANO6 (TMEM16F) in gastrointestinal stromal tumors (GISTs). ANO6 expression in GIST and adjacent normal tissues was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. Cell proliferation, apoptosis, and pyroptosis were examined utilizing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, terminal deoxynucleotidyl transferase dUTP Nick-End Labeling staining, and flow cytometry. In addition, the total iron and Fe2+ levels were assessed. IL-18 and IL-1β levels were also evaluated. Lipid reactive oxygen species (ROS), cystine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) levels were evaluated using appropriate kits. Ferroptotic markers, including Ptgs2, Chac1, SLC7A11, and SLC3A2, were analyzed by RT-qPCR, western blotting, and immunohistochemistry. ANO6 expression decreased in GIST tissues. ANO6-plasmid inhibits proliferation, induces apoptosis, and promotes pyroptosis in GIST-T1 and GIST-T1 IR cells. The ANO6-plasmid induced ferroptosis, as confirmed by enhanced lipid ROS levels, increased intracellular concentrations of total iron and Fe2+, promoted Ptgs2 and Chac1 expression, reduced Cys, GSH, and GPX4 levels, and downregulated SLC7A11 and SLC3A2 expression after in vitro and in vivo treatment with ANO6-plasmid. Moreover, the ANO6-plasmid inhibited GIST growth in vivo. Therefore, ANO6 may be a promising therapeutic target for blocking the development of GIST via the induction of apoptosis, pyroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Hao Wang
- School of Public Health, Nanjing Medical University, Nanjing211166, China
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Wei Zhao
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
| | - Daorong Wang
- Department of Gastrointestinal Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou225001, China
| | - Jin Chen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Jiangning District, Nanjing211166, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing211166, China
| |
Collapse
|
12
|
Cantú-Soriano GN, Padilla-Rodríguez ÁL. [Intestinal calcifying fibrous tumor: case report]. REVISTA ESPANOLA DE PATOLOGIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ANATOMIA PATOLOGICA Y DE LA SOCIEDAD ESPANOLA DE CITOLOGIA 2024; 57:137-140. [PMID: 38599735 DOI: 10.1016/j.patol.2023.11.003] [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: 08/28/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 04/12/2024]
Abstract
Calcifying fibrous tumor (CFT) is a rare benign lesion of mesenchymal origin that may present similar characteristics to other more common tumors. We present the case of a 36-year-old woman with a tumor in the proximal jejunum, initially suspected to be a gastrointestinal stromal tumor (GIST). Surgical resection was performed, revealing a well-demarcated nodule at the anti-mesenteric border with microscopic features typical of a calcifying fibrous tumor. The tumor cells were positive for CD34 and negative for other markers, differentiating it from other neoplasms. Calcifying fibrous tumors can be confused with more common tumors because of its appearance, but an accurate diagnosis supported by immunohistochemistry is essential. Complete surgical excision is usually curative.
Collapse
Affiliation(s)
- Gonzalo Nathaniel Cantú-Soriano
- DIGIPATH (Digital Pathology Laboratory), Escuela de Medicina Universidad Panamericana, Campus Ciudad de México, Ciudad de México, México
| | - Álvaro Lezid Padilla-Rodríguez
- DIGIPATH (Digital Pathology Laboratory), Escuela de Medicina Universidad Panamericana, Campus Ciudad de México, Escuela de Medicina Instituto Tecnológico y de Estudios Superiores de Monterrey, Campus Ciudad de México, Ciudad de México, México.
| |
Collapse
|
13
|
Cicala CM, Olivares-Rivas I, Aguirre-Carrillo JA, Serrano C. KIT/PDGFRA inhibitors for the treatment of gastrointestinal stromal tumors: getting to the gist of the problem. Expert Opin Investig Drugs 2024; 33:159-170. [PMID: 38344849 DOI: 10.1080/13543784.2024.2318317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
INTRODUCTION Approximately 90% of gastrointestinal stromal tumors (GISTs) are driven by activating mutations in receptor tyrosine-kinases KIT or PDGFRA. Despite the outstanding results of first-line imatinib in advanced GIST, resistance ultimately occurs mainly through secondary mutations in KIT/PDGFRA. Other tyrosine-kinase inhibitors (TKIs) with a broader spectrum of activity against these mutations are approved after imatinib failure. However, response rates and progression-free survival are drastically lower compared to imatinib. Notably, imatinib also triggers early tolerance adaptation mechanisms, which precede the occurrence of secondary mutations. AREAS COVERED In this review, we outline the current landscape of KIT inhibitors, discuss the novel agents, and present additional biological pathways that may be therapeutically exploitable. EXPERT OPINION The development of broad-spectrum and highly selective TKIs able to induce a sustained KIT/PDGFRA inhibition is the pillar of preclinical and clinical investigation in GIST. However, it is now recognized that the situation is more intricate, with various factors interacting with KIT and PDGFRA, playing a crucial role in the response and resistance to treatments. Future strategies in the management of advanced GIST should integrate driver inhibition with the blockade of other molecules to enhance cell death and establish enduring responses in patients.
Collapse
Affiliation(s)
- Carlo María Cicala
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Iván Olivares-Rivas
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - César Serrano
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
14
|
Abdullah ST, Abdullah SR, Hussen BM, Younis YM, Rasul MF, Taheri M. Role of circular RNAs and gut microbiome in gastrointestinal cancers and therapeutic targets. Noncoding RNA Res 2024; 9:236-252. [PMID: 38192436 PMCID: PMC10771991 DOI: 10.1016/j.ncrna.2023.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/10/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024] Open
Abstract
Gastrointestinal cancers are a huge worldwide health concern, which includes a wide variety of digestive tract cancers. Circular RNAs (circRNAs), a kind of non-coding RNA (ncRNAs), are a family of single-stranded, covalently closed RNAs that have become recognized as crucial gene expression regulators, having an impact on several cellular functions in cancer biology. The gut microbiome, which consists of several different bacteria, actively contributes to the regulation of host immunity, inflammation, and metabolism. CircRNAs and the gut microbiome interact significantly to greatly affect the growth of GI cancer. Several studies focus on the complex functions of circRNAs and the gut microbiota in GI cancers, including esophageal cancer, colorectal cancer, gastric cancer, hepatocellular cancer, and pancreatic cancer. It also emphasizes how changed circRNA expression profiles and gut microbiota affect pathways connected to malignancy as well as how circRNAs affect hallmarks of gastrointestinal cancers. Furthermore, circRNAs and gut microbiota have been recommended as biological markers for therapeutic targets as well as diagnostic and prognostic purposes. Targeting circRNAs and the gut microbiota for the treatment of gastrointestinal cancers is also being continued to study. Despite significant initiatives, the connection between circRNAs and the gut microbiota and the emergence of gastrointestinal cancers remains poorly understood. In this study, we will go over the most recent studies to emphasize the key roles of circRNAs and gut microbiota in gastrointestinal cancer progression and therapeutic options. In order to create effective therapies and plan for the future gastrointestinal therapy, it is important to comprehend the functions and mechanisms of circRNAs and the gut microbiota.
Collapse
Affiliation(s)
- Sara Tharwat Abdullah
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Yousif Mohammed Younis
- Department of Nursing, College of Nursing, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Heinrich MC, Jones RL, George S, Gelderblom H, Schöffski P, von Mehren M, Zalcberg JR, Kang YK, Razak AA, Trent J, Attia S, Le Cesne A, Siontis BL, Goldstein D, Boye K, Sanchez C, Steeghs N, Rutkowski P, Druta M, Serrano C, Somaiah N, Chi P, Reichmann W, Sprott K, Achour H, Sherman ML, Ruiz-Soto R, Blay JY, Bauer S. Ripretinib versus sunitinib in gastrointestinal stromal tumor: ctDNA biomarker analysis of the phase 3 INTRIGUE trial. Nat Med 2024; 30:498-506. [PMID: 38182785 PMCID: PMC10878977 DOI: 10.1038/s41591-023-02734-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/22/2023] [Indexed: 01/07/2024]
Abstract
INTRIGUE was an open-label, phase 3 study in adult patients with advanced gastrointestinal stromal tumor who had disease progression on or intolerance to imatinib and who were randomized to once-daily ripretinib 150 mg or sunitinib 50 mg. In the primary analysis, progression-free survival (PFS) with ripretinib was not superior to sunitinib. In clinical and nonclinical studies, ripretinib and sunitinib have demonstrated differential activity based on the exon location of KIT mutations. Therefore, we hypothesized that mutational analysis using circulating tumor DNA (ctDNA) might provide further insight. In this exploratory analysis (N = 362), baseline peripheral whole blood was analyzed by a 74-gene ctDNA next-generation sequencing-based assay. ctDNA was detected in 280/362 (77%) samples with KIT mutations in 213/362 patients (59%). Imatinib-resistant mutations were found in the KIT ATP-binding pocket (exons 13/14) and activation loop (exons 17/18). Mutational subgroup assessment showed 2 mutually exclusive populations with differential treatment effects. Patients with only KIT exon 11 + 13/14 mutations (ripretinib, n = 21; sunitinib, n = 20) had better PFS with sunitinib versus ripretinib (median, 15.0 versus 4.0 months). Patients with only KIT exon 11 + 17/18 mutations (ripretinib, n = 27; sunitinib, n = 25) had better PFS with ripretinib versus sunitinib (median, 14.2 versus 1.5 months). The results of this exploratory analysis suggest ctDNA sequencing may improve the prediction of the efficacy of single-drug therapies and support further evaluation of ripretinib in patients with KIT exon 11 + 17/18 mutations. ClinicalTrials.gov identifier: NCT03673501.
Collapse
Affiliation(s)
- Michael C Heinrich
- Division of Hematology/Oncology, Portland VA Health Care System, Portland, OR, USA
- Department of Medicine, OHSU Knight Cancer Institute, Portland, OR, USA
| | - Robin L Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Suzanne George
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Patrick Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Margaret von Mehren
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - John R Zalcberg
- Department of Medical Oncology, Monash University School of Public Health and Preventive Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, Korea
| | - Albiruni Abdul Razak
- Division of Medical Oncology, Toronto Sarcoma Program, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Jonathan Trent
- Department of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | - Steven Attia
- Department of Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, Villejuif, France
| | | | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Kjetil Boye
- Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Cesar Sanchez
- Department of Hematology-Oncology, Centro de Cáncer, Hospital Clínico Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Mihaela Druta
- Sarcoma Program, Moffitt Cancer Center, Tampa, FL, USA
| | - César Serrano
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Kam Sprott
- Biometrics, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
- Translational Medicine, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Haroun Achour
- Biometrics, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Matthew L Sherman
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Rodrigo Ruiz-Soto
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Sebastian Bauer
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| |
Collapse
|
16
|
Mirovic M, Stojanovic MD, Jovanovic M, Stankovic V, Milosev D, Zdravkovic N, Milosevic B, Cvetkovic A, Spasic M, Vekic B, Jovanovic I, Stojanovic BS, Petrovic M, Bogut A, Peulic M, Stojanovic B. Exploring Perforated Jejunal GIST: A Rare Case Report and Review of Molecular and Clinical Literature. Curr Issues Mol Biol 2024; 46:1192-1207. [PMID: 38392194 PMCID: PMC10887764 DOI: 10.3390/cimb46020076] [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: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
This case report details a rare instance of a perforated jejunal gastrointestinal stromal tumor (GIST) in a 76-year-old female patient. The patient presented with acute abdominal pain and distension without any changes in bowel habits or episodes of nausea and vomiting. Initial diagnostics, including abdominal plain radiography and ultrasonography, were inconclusive; however, a computed tomography (CT) scan revealed pneumoperitoneum and an irregular fluid collection suggestive of small intestine perforations. Surgical intervention uncovered a 35 mm jejunal GIST with a 10 mm perforation. Histopathological examination confirmed a mixed cell type GIST with high malignancy potential, further substantiated by immunohistochemistry markers CD117, DOG1, and vimentin. Molecular analysis illuminated the role of key oncogenes, primarily KIT and PDGFRA mutations, emphasizing the importance of molecular diagnostics in GIST management. Despite the severity of the presentation, the patient's postoperative recovery was favorable, highlighting the effectiveness of prompt surgical and multidisciplinary approaches in managing complex GIST cases.
Collapse
Affiliation(s)
- Milos Mirovic
- Department of General Surgery, Clinical Hospital Center Kotor, 85330 Kotor, Montenegro
| | - Milica Dimitrijevic Stojanovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Pathology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Marina Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Vesna Stankovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Pathology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Danijela Milosev
- Department of Pathology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Natasa Zdravkovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Bojan Milosevic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Aleksandar Cvetkovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Marko Spasic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Berislav Vekic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Bojana S Stojanovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Marko Petrovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ana Bogut
- City Medical Emergency Department, 11000 Belgrade, Serbia
| | - Miodrag Peulic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
17
|
Thomas M, Mandal A, Eisinger A, Doolittle T, Nat A. Rectal Hematoma Mimicking as a Gastrointestinal Stromal Tumor: An Atypical Endoscopic Ultrasound Finding. Cureus 2024; 16:e51898. [PMID: 38333511 PMCID: PMC10851035 DOI: 10.7759/cureus.51898] [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: 01/08/2024] [Indexed: 02/10/2024] Open
Abstract
Acute gastrointestinal bleeding (GIB) represents a frequently encountered condition that prompts individuals to seek urgent medical attention at the emergency department, often leading to subsequent hospitalization. GIB can range from self-limited bleeding to hemorrhagic shock. Multiple etiologies contribute to the occurrence of GIB. In this report, we present the case of an 84-year-old male with multiple medical comorbidities admitted with hemodynamically stable lower GIB. Colonoscopy demonstrated a submucosal mass without evidence of bleeding. He subsequently underwent an endoscopic ultrasound (EUS) with sonographic findings concerning for a gastrointestinal stromal tumor. However, pathological analysis from both colonoscopy and EUS indicated the presence of blood, but no evidence of malignancy. A follow-up EUS performed two months later showed a complete resolution of the previously observed submucosal mass, suggesting that the initial evaluation was likely a hematoma that has resolved completely.
Collapse
Affiliation(s)
- Mathew Thomas
- Internal Medicine, State University of New York Upstate Medical University, Syracuse, USA
| | - Amrendra Mandal
- Internal Medicine/Gastroenterology, State University of New York Upstate Medical University, Syracuse, USA
| | - Amanda Eisinger
- Internal Medicine, State University of New York Upstate Medical University, Syracuse, USA
| | - Tessa Doolittle
- Internal Medicine, State University of New York Upstate Medical University, Syracuse, USA
| | - Amitpal Nat
- Internal Medicine, State University of New York Upstate Medical University, Syracuse, USA
| |
Collapse
|
18
|
Serrano C, Rothschild S, Villacampa G, Heinrich MC, George S, Blay JY, Sicklick JK, Schwartz GK, Rastogi S, Jones RL, Rutkowski P, Somaiah N, Navarro V, Evans D, Trent JC. Rethinking placebos: embracing synthetic control arms in clinical trials for rare tumors. Nat Med 2023; 29:2689-2692. [PMID: 37828359 DOI: 10.1038/s41591-023-02578-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Affiliation(s)
- César Serrano
- Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain.
- Sarcoma Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | | | - Guillermo Villacampa
- Oncology Data Science, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- The Institute of Cancer Research, London, UK
| | - Michael C Heinrich
- Portland VA Health Care System and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Suzanne George
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- Université Claude Bernard, Lyon, France
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Gary K Schwartz
- Case Comprehensive Cancer Center in Cleveland, Cleveland, OH, USA
| | - Sameer Rastogi
- Department of Medical Oncology, All India Institute of Medical Science (AIIMS), New Delhi, India
| | - Robin L Jones
- Division of Clinical Studies, The Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, London, UK
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Víctor Navarro
- Oncology Data Science, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Jonathan C Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
19
|
Campanella NC, Gomes INF, Alves ALV, Leal LF, Evangelista AF, Rosa MN, Melendez ME, Silva VAO, Dias RLK, Abrahão-Machado LF, Santana I, Martinho O, Guimarães DP, Faça VM, Reis RM. Biological and therapeutic implications of RKIP in Gastrointestinal Stromal Tumor (GIST): an integrated transcriptomic and proteomic analysis. Cancer Cell Int 2023; 23:256. [PMID: 37907993 PMCID: PMC10619323 DOI: 10.1186/s12935-023-03102-6] [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: 12/22/2022] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GIST) represent a significant clinical challenge due to their metastatic potential and limited treatment options. Raf kinase inhibitor protein (RKIP), a suppressor of the MAPK signaling pathway, is downregulated in various cancers and acts as a metastasis suppressor. Our previous studies demonstrated low RKIP expression in GIST and its association with poor outcomes. This study aimed to expand on the previous findings and investigate the biological and therapeutic implications of RKIP loss on GIST. METHODS To validate the RKIP prognostic significance, its expression was evaluated by immunohistochemistry in 142 bona fide GIST cases. The functional role of RKIP was evaluated in vitro, using the GIST-T1 cell line, which was knocked out for RKIP. The biological and therapeutic implications of RKIP were evaluated by invasion, migration, apoptosis, and 2D / 3D viability assays. Additionally, the transcriptome and proteome of RKIP knockout cells were determined by NanoString and mass spectrometry, respectively. RESULTS Immunohistochemical analysis revealed the absence of RKIP in 25.3% of GIST cases, correlating with a tendency toward poor prognosis. Functional assays demonstrated that RKIP knockout increased GIST cells' invasion and migration potential by nearly 60%. Moreover, we found that RKIP knockout cells exhibited reduced responsiveness to Imatinib treatment and higher cellular viability in 2D and 3D in vitro models, as assessed by apoptosis-related protein expression. Through comprehensive genetic and proteomic profiling of RKIP knockout cells, we identified several putative RKIP-regulated proteins in GIST, such as COL3A1. CONCLUSIONS Using a multidimensional integrative analysis, we identified, for the first time in GIST, molecules and pathways modulated by RKIP that may potentially drive metastasis and, consequently, poor prognosis in this disease.
Collapse
Affiliation(s)
- Nathália Cristina Campanella
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Izabela Natalia Faria Gomes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Ana Laura Vieira Alves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- School of Health Sciences Dr. Paulo Prata (FACISB), Barretos, 14785-002, Brazil
| | - Adriane Feijó Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Marcela Nunes Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Matias Eliseo Melendez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Molecular Carcinogenesis Program, National Cancer Institute, Rio de Janeiro, 20231-050, Brazil
| | - Viviane Aline Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Department of Pathology, School of Medicine, Federal University of Bahia, Salvador, 40110-909, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, 40296-710, Brazil
| | - Richard Lucas Konichi Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- School of Health Sciences Dr. Paulo Prata (FACISB), Barretos, 14785-002, Brazil
| | | | - Iara Santana
- Department of Pathology, Barretos Cancer Hospital, Barretos, 14784-400, Brazil
| | - Olga Martinho
- ICVS/3B's - PT Government Associate Laboratory, Braga, 4806-909, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, 14784-400, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil.
- ICVS/3B's - PT Government Associate Laboratory, Braga, 4806-909, Portugal.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal.
| |
Collapse
|
20
|
Han S, Song M, Wang J, Huang Y, Li Z, Yang A, Sui C, Zhang Z, Qiao J, Yang J. Intelligent identification system of gastric stromal tumors based on blood biopsy indicators. BMC Med Inform Decis Mak 2023; 23:214. [PMID: 37833709 PMCID: PMC10576280 DOI: 10.1186/s12911-023-02324-y] [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: 07/20/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The most prevalent mesenchymal-derived gastrointestinal cancers are gastric stromal tumors (GSTs), which have the highest incidence (60-70%) of all gastrointestinal stromal tumors (GISTs). However, simple and effective diagnostic and screening methods for GST remain a great challenge at home and abroad. This study aimed to build a GST early warning system based on a combination of machine learning algorithms and routine blood, biochemical and tumour marker indicators. METHODS In total, 697 complete samples were collected from four hospitals in Gansu Province, including 42 blood indicators from 318 pretreatment GST patients, 180 samples of gastric polyps and 199 healthy individuals. In this study, three algorithms, gradient boosting machine (GBM), random forest (RF), and logistic regression (LR), were chosen to build GST prediction models for comparison. The performance and stability of the models were evaluated using two different validation techniques: 5-fold cross-validation and external validation. The DeLong test assesses significant differences in AUC values by comparing different ROC curves, the variance and covariance of the AUC value. RESULTS The AUC values of both the GBM and RF models were higher than those of the LR model, and this difference was statistically significant (P < 0.05). The GBM model was considered to be the optimal model, as a larger area was enclosed by the ROC curve, and the axes indicated robust model classification performance according to the accepted model discriminant. Finally, the integration of 8 top-ranked blood indices was proven to be able to distinguish GST from gastric polyps and healthy people with sensitivity, specificity and area under the curve of 0.941, 0.807 and 0.951 for the cross-validation set, respectively. CONCLUSION The GBM demonstrated powerful classification performance and was able to rapidly distinguish GST patients from gastric polyps and healthy individuals. This identification system not only provides an innovative strategy for the diagnosis of GST but also enables the exploration of hidden associations between blood parameters and GST for subsequent studies on the prevention and disease surveillance management of GST. The GST discrimination system is available online for free testing of doctors and high-risk groups at https://jzlyc.gsyy.cn/bear/mobile/index.html .
Collapse
Affiliation(s)
- Shangjun Han
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Meijuan Song
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jiarui Wang
- Department of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yalong Huang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Zuxi Li
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Aijia Yang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Changsheng Sui
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Zeping Zhang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jiling Qiao
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jing Yang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
21
|
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
|
22
|
Zhang X, Zhang P, Qiu H, Fang Y, Liu H, Zhou Y, Xu H, Yu J, Zhang J, Wang M, Shen L, Li J. Large-Scale, Multicenter, Prospective Registry Study of Ripretinib in Advanced GIST: A Real-World Study from China. Adv Ther 2023; 40:3817-3829. [PMID: 37356078 PMCID: PMC10427548 DOI: 10.1007/s12325-023-02576-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/02/2023] [Indexed: 06/27/2023]
Abstract
INTRODUCTION Tyrosine-kinase inhibitors (TKIs) have become the standard treatment for patients with advanced gastrointestinal stromal tumor (GIST); however, secondary mutations can still drive disease progression. Studies have shown that ripretinib, a novel switch-control TKI, inhibits various primary and secondary drug-resistant mutations. There is a paucity of data on the effectiveness and safety of ripretinib in a real-world setting. This prospective, large-scale, real-world registry study aimed to evaluate the effectiveness and safety of ripretinib as a fourth-line treatment in Chinese patients with advanced GIST. METHODS Patients ≥ 18 years of age having recurrent/metastatic GIST were enrolled. Key endpoints were median progression-free survival (mPFS), median overall survival (mOS), and adverse events (AEs) incidence. Univariate and multivariate analyses were conducted to identify various parameters associated with PFS. RESULTS A total of 240 patients were enrolled. After a median follow-up period of 6.5 months, the mPFS [95% confidence interval (CI)] was 7.70 (6.60, 8.60) months and the mOS was not reached. Multivariate analysis revealed association of Eastern Cooperative Oncology Group (ECOG) performance status score with PFS and superior benefits for non-gastric was observed as compared to gastric GISTs [hazard ratio (HR) 0.58, 95% CI (0.39-0.86)]. Disease control rate and tumor shrinkage (any magnitude) was 73% and 43%, respectively. Ripretinib was also effective in the subgroup of patients with different gene mutations. The toxicities were tolerable, and most reported AEs were alopecia (17.1%) and hand-foot syndrome (15.4%). CONCLUSION Ripretinib demonstrated effectiveness and a tolerable safety profile, making it a viable option as a fourth- or later-line treatment in Chinese patients with advanced GISTs, especially for non-gastric GISTs. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT05697107.
Collapse
Affiliation(s)
- Xinhua Zhang
- The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Zhang
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haibo Qiu
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yong Fang
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Heli Liu
- Xiangya Hospital, Central South University, Changsha, China
| | - Yongjian Zhou
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Hao Xu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - JiRen Yu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Wang
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Shen
- Peking University Cancer Hospital and Institute, 52 Fucheng Road, Beijing, China
| | - Jian Li
- Peking University Cancer Hospital and Institute, 52 Fucheng Road, Beijing, China.
| |
Collapse
|
23
|
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: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
24
|
Godesi S, Lee J, Nada H, Quan G, Elkamhawy A, Choi Y, Lee K. Small Molecule c-KIT Inhibitors for the Treatment of Gastrointestinal Stromal Tumors: A Review on Synthesis, Design Strategies, and Structure-Activity Relationship (SAR). Int J Mol Sci 2023; 24:ijms24119450. [PMID: 37298401 DOI: 10.3390/ijms24119450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The proto-oncogenic protein, c-KIT, plays a crucial role in regulating cellular transformation and differentiation processes, such as proliferation, survival, adhesion, and chemotaxis. The overexpression of, and mutations, in c-KIT can lead to its dysregulation and promote various human cancers, particularly gastrointestinal stromal tumors (GISTs); approximately 80-85% of cases are associated with oncogenic mutations in the KIT gene. Inhibition of c-KIT has emerged as a promising therapeutic target for GISTs. However, the currently approved drugs are associated with resistance and significant side effects, highlighting the urgent need to develop highly selective c-KIT inhibitors that are not affected by these mutations for GISTs. Herein, the recent research efforts in medicinal chemistry aimed at developing potent small-molecule c-KIT inhibitors with high kinase selectivity for GISTs are discussed from a structure-activity relationship perspective. Moreover, the synthetic pathways, pharmacokinetic properties, and binding patterns of the inhibitors are also discussed to facilitate future development of more potent and pharmacokinetically stable small-molecule c-KIT inhibitors.
Collapse
Affiliation(s)
- Sreenivasulu Godesi
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Joohan Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Hossam Nada
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Guofeng Quan
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Ahmed Elkamhawy
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Yongseok Choi
- College of Biosciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Kyeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| |
Collapse
|
25
|
Ruiz-Demoulin S, Trenquier E, Dekkar S, Deshayes S, Boisguérin P, Serrano C, de Santa Barbara P, Faure S. LIX1 Controls MAPK Signaling Reactivation and Contributes to GIST-T1 Cell Resistance to Imatinib. Int J Mol Sci 2023; 24:ijms24087138. [PMID: 37108337 PMCID: PMC10138740 DOI: 10.3390/ijms24087138] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST), the most common sarcoma, is mainly caused by an oncogenic mutation in the KIT receptor tyrosine kinase. Targeting KIT using tyrosine kinase inhibitors, such as imatinib and sunitinib, provides substantial benefit; however, in most patients, the disease will eventually progress due to KIT secondary mutations leading to treatment failure. Understanding how GIST cells initially adapt to KIT inhibition should guide the selection of appropriate therapies to overcome the emergence of resistance. Several mechanisms have been broadly implicated in the resistance to imatinib anti-tumoral effects, including the reactivation of MAPK signaling upon KIT/PDGFRA targeted inhibition. This study provides evidence that LImb eXpression 1 (LIX1), a protein we identified as a regulator of the Hippo transducers YAP1 and TAZ, is upregulated upon imatinib or sunitinib treatment. LIX1 silencing in GIST-T1 cells impaired imatinib-induced MAPK signaling reactivation and enhanced imatinib anti-tumor effect. Our findings identified LIX1 as a key regulator of the early adaptative response of GIST cells to targeted therapies.
Collapse
Affiliation(s)
- Salomé Ruiz-Demoulin
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Eva Trenquier
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Sanaa Dekkar
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Sébastien Deshayes
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Prisca Boisguérin
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - César Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Pascal de Santa Barbara
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Sandrine Faure
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| |
Collapse
|
26
|
Zhao Y, Weng Z, Zhou X, Xu Z, Cao B, Wang B, Li J. Mesenchymal stromal cells promote the drug resistance of gastrointestinal stromal tumors by activating the PI3K-AKT pathway via TGF-β2. J Transl Med 2023; 21:219. [PMID: 36966336 PMCID: PMC10040136 DOI: 10.1186/s12967-023-04063-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the prevailing sarcomas of the gastrointestinal tract. Tyrosine kinase inhibitors (TKIs) therapy, exemplified by Imatinib mesylate (IM), constitutes the established adjuvant therapy for GISTs. Nevertheless, post-treatment resistance poses a challenge that all patients must confront. The presence of tumor heterogeneity and secondary mutation mechanisms fail to account for some instances of acquired drug resistance. Certain investigations suggest a strong association between tumor drug resistance and mesenchymal stromal cells (MSC) in the tumor microenvironment, but the underlying mechanism remains obscure. Scarce research has explored the connection between GIST drug resistance and the tumor microenvironment, as well as the corresponding mechanism. METHODS Immunofluorescence and fluorescence-activated cell sorting (FACS) methodologies were employed to detect the presence of MSC in GIST samples. The investigation encompassed the examination of MSC migration towards tumor tissue and the impact of MSC on the survival of GIST cells under IM treatment. Through ELISA, western blotting, and flow cytometry analyses, it was confirmed that Transforming Growth Factor Beta 2 (TGF-β2) triggers the activation of the PI3K-AKT pathway by MSC, thereby facilitating drug resistance in GIST. RESULTS Our findings revealed a positive correlation between a high proportion of MSC and both GIST resistance and a poor prognosis. In vitro studies demonstrated the ability of MSC to migrate towards GIST. Additionally, MSC were observed to secrete TGF-β2, consequently activating the PI3K-AKT pathway and augmenting GIST resistance. CONCLUSIONS Our investigation has revealed that MSC within GISTs possess the capacity to augment drug resistance, thereby highlighting their novel mechanism and offering a promising target for intervention in GIST therapy.
Collapse
Affiliation(s)
- Yu Zhao
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Zuyi Weng
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Xuan Zhou
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Zhi Xu
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Bei Cao
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| | - Juan Li
- Phase I Clinical Trials Unit, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| |
Collapse
|
27
|
Neuropsychiatric Adverse Drug Reactions with Tyrosine Kinase Inhibitors in Gastrointestinal Stromal Tumors: An Analysis from the European Spontaneous Adverse Event Reporting System. Cancers (Basel) 2023; 15:cancers15061851. [PMID: 36980737 PMCID: PMC10046586 DOI: 10.3390/cancers15061851] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 03/22/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are widely used in gastrointestinal stromal tumors (GISTs). The aim of this study is to evaluate the reporting frequency of neuropsychiatric adverse drug reactions (ADRs) for TKIs through the analysis of European individual case safety reports (ICSRs). All ICSRs collected in EudraVigilance up to 31 December 2021 with one TKI having GISTs as an indication (imatinib (IM), sunitinib (SU), avapritinib (AVA), regorafenib (REG), and ripretinib (RIP)) were included. A disproportionality analysis was performed to assess the frequency of reporting for each TKI compared to all other TKIs. The number of analyzed ICSRs was 8512, of which 57.9% were related to IM. Neuropsychiatric ADRs were reported at least once in 1511 ICSRs (17.8%). A higher reporting probability of neuropsychiatric ADRs was shown for AVA. Most neuropsychiatric ADRs were known, except for a higher frequency of lumbar spinal cord and nerve root disorders (reporting odds ratio, ROR 4.46; confidence interval, CI 95% 1.58–12.54), olfactory nerve disorders (8.02; 2.44–26.33), and hallucinations (22.96; 8.45–62.36) for AVA. The analyses of European ICSRs largely confirmed the safety profiles of TKIs in GISTs, but some ADRs are worthy of discussion. Further studies are needed to increase the knowledge of the neuropsychiatric disorders of newly approved TKIs.
Collapse
|
28
|
Obi F, Anguiano-Albarran R, Cain D, Mudrovich S, Simien M. Peculiar Presentation of Gastrointestinal Stromal Tumor in a Patient With Early Satiety. Cureus 2023; 15:e36523. [PMID: 37090310 PMCID: PMC10120845 DOI: 10.7759/cureus.36523] [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: 03/22/2023] [Indexed: 04/25/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are one of the most common, potentially malignant, subepithelial lesions identified in the gastrointestinal tract. Hypothesized to derive from the interstitial cells of Cajal (ICC), GISTs commonly demonstrate gain of function mutations in proto-oncogenic receptor tyrosine kinase CD117 (KIT). Depending on mitotic activity and tumor size characteristics, GISTs may transform from benign to malignant neoplasms. Increasing evidence suggests that early identification of a GIST is paramount for optimal prognostic outcomes. We present a rare case of a GIST located in the uncinate pancreas identified via endoscopic ultrasound (EUS) and diagnosed with an EUS-guided fine needle aspiration (EUS-FNA) biopsy.
Collapse
Affiliation(s)
- Franklin Obi
- Internal Medicine, Baylor Scott & White All Saints Medical Center, Fort Worth, USA
| | | | - Daniel Cain
- Internal Medicine, Baylor Scott & White All Saints Medical Center, Fort Worth, USA
| | - Steven Mudrovich
- Medicine, Baylor Scott & White All Saints Medical Center, Fort Worth, USA
| | - Melvin Simien
- Medicine, Baylor Scott & White All Saints Medical Center, Fort Worth, USA
- Interventional Endoscopy, Baylor Scott & White Digestive Diseases, Fort Worth, USA
| |
Collapse
|
29
|
Yue L, Sun Y, Wang X, Hu W. Advances of endoscopic and surgical management in gastrointestinal stromal tumors. Front Surg 2023; 10:1092997. [PMID: 37123546 PMCID: PMC10130460 DOI: 10.3389/fsurg.2023.1092997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
As one of the most common mesenchymal malignancies in the digestive system, gastrointestinal stromal tumors (GISTs) occur throughout the alimentary tract with diversified oncological characteristics. With the advent of the tyrosine kinase inhibitor era, the treatment regimens of patients with GISTs have been revolutionized and GISTs have become the paradigm of multidisciplinary therapy. However, surgery resection remains recognized as the potentially curative management for the radical resection and provided with favorable oncological outcomes. The existing available surgery algorithms in clinical practice primarily incorporate open procedure, and endoscopic and laparoscopic surgery together with combined operation techniques. The performance of various surgery methods often refers to the consideration of risk evaluation of recurrence and metastases; the degree of disease progression; size, location, and growth pattern of tumor; general conditions of selected patients; and indications and safety profile of various techniques. In the present review, we summarize the fundamental principle of surgery of GISTs based on risk assessment as well as tumor size, location, and degree of progress with an emphasis on the indications, strengths, and limitations of current surgery techniques.
Collapse
Affiliation(s)
- Lei Yue
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Yingchao Sun
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Xinjie Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University (IGZJU), Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- Correspondence: Weiling Hu
| |
Collapse
|
30
|
Falkenhorst J, Grunewald S, Krzeciesa D, Herold T, Ketzer J, Christoff M, Hamacher R, Kostbade K, Treckmann J, Köster J, Farzaliyev F, Fletcher BS, Dieckmann N, Kaths M, Mühlenberg T, Schildhaus HU, Bauer S. Plasma Sequencing for Patients with GIST-Limitations and Opportunities in an Academic Setting. Cancers (Basel) 2022; 14:5496. [PMID: 36428589 PMCID: PMC9688348 DOI: 10.3390/cancers14225496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Circulating tumor DNA (ctDNA) from circulating free DNA (cfDNA) in GIST is of interest for the detection of heterogeneous resistance mutations and treatment monitoring. However, methodologies for use in a local setting are not standardized and are error-prone and difficult to interpret. We established a workflow to evaluate routine tumor tissue NGS (Illumina-based next generation sequencing) panels and pipelines for ctDNA sequencing in an academic setting. Regular blood collection (Sarstedt) EDTA tubes were sufficient for direct processing whereas specialized tubes (STRECK) were better for transportation. Mutation detection rate was higher in automatically extracted (AE) than manually extracted (ME) samples. Sensitivity and specificity for specific mutation detection was higher using digital droplet (dd)PCR compared to NGS. In a retrospective analysis of NGS and clinical data (133 samples from 38 patients), cfDNA concentration correlated with tumor load and mutation detection. A clinical routine pipeline and a novel research pipeline yielded different results, but known and resistance-mediating mutations were detected by both and correlated with the resistance spectrum of TKIs used. In conclusion, NGS routine panel analysis was not sensitive and specific enough to replace solid biopsies in GIST. However, more precise methods (hybridization capture NGS, ddPCR) may comprise important research tools to investigate resistance. Future clinical trials need to compare methodology and protocols.
Collapse
Affiliation(s)
- Johanna Falkenhorst
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Susanne Grunewald
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Dawid Krzeciesa
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Thomas Herold
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Institute of Pathology, University Medical Center Essen, 45147 Essen, Germany
| | - Julia Ketzer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Miriam Christoff
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Rainer Hamacher
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Karina Kostbade
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Jürgen Treckmann
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Department of Visceral Surgery, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Johannes Köster
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Farhad Farzaliyev
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Department of Visceral Surgery, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Benjamin Samulon Fletcher
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Nils Dieckmann
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Moritz Kaths
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Department of Visceral Surgery, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Thomas Mühlenberg
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Hans-Ulrich Schildhaus
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
- Institute of Pathology, University Medical Center Essen, 45147 Essen, Germany
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
31
|
Khan J, Ullah A, Waheed A, Karki NR, Adhikari N, Vemavarapu L, Belakhlef S, Bendjemil SM, Mehdizadeh Seraj S, Sidhwa F, Ghleilib I, Foroutan S, Blakely AM, Del Rivero J, Karim NA, Vail E, Heneidi S, Mesa H. Gastrointestinal Stromal Tumors (GIST): A Population-Based Study Using the SEER Database, including Management and Recent Advances in Targeted Therapy. Cancers (Basel) 2022; 14:3689. [PMID: 35954353 PMCID: PMC9367571 DOI: 10.3390/cancers14153689] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Introduction: Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasm of the gastrointestinal (GI) system. Most GISTs originate from the interstitial cells of Cajal (ICC), the pacemaker cell situated between the circular and longitudinal layers of the muscularis propria along the GI tract. In this population-based study using the SEER database, we sought to identify demographic, clinical, and pathologic factors that affect the prognosis and survival of patients with this neoplasm. Molecular genetic advances, current management guidelines, and advances in targeted therapy are discussed. Methods: Demographic and clinical data from GIST patients were retrieved from the SEER research plus database for the period 2000−2018. Statistical analysis was performed with IBM SPSS® v20.2 software using the Chi-square test, paired t-test, multivariate analysis, and Kaplan−Meier functions. Results: A total of 10,833 patients with GIST were identified. Most patients were between 60−74 years of age: 40%, Caucasian: 68%, and the male to female ratio was 1.1:1. The most common primary tumor sites were stomach: 63%, small intestine: 30%, rectum: 3%, and esophagus: 0.7%. When reported, the grade of differentiation was well: 38%, moderately: 32%, undifferentiated: 19%, poorly: 12%. The size of most tumors ranged between 6−10 cm: 36% and they were treated by surgical intervention: 82% and/or chemotherapy/targeted therapy: 39%. The stage was localized: 66%, advanced: 19%, and regional: 15%. The 5-year survival was 74% (95% confidence interval (95% CI) = 72.6−74.7), and the 5-year cause-specific survival 82% (95% CI = 80.7−82.6). The 5-year cause-specific survival by treatment included surgery at 86% (95% CI = 85.4−87.3), chemotherapy/targeted therapy with or without surgery at 77% (95% CI = 75.7−78.9), and radiation at 75% (95% CI = 74.5−80). On multivariable analysis tumor size > 5 cm, poorly and undifferentiated grade, age > 60, and distant metastases at presentation were associated with worse overall survival. Conclusion: GISTs comprise 1−2% of malignancies of the GI tract, usually affect male Caucasians between the ages of 60 and 74 years, most tumors occur in the stomach and small intestine, and are usually >5 cm, but still localized, at the time of diagnosis. Most tumors receive multimodality surgical and chemotherapy/targeted therapy treatment, with a 5-year overall survival of 74% and cause-specific survival of 82%. GIST patients would benefit from enrollment in large clinical trials to establish better therapy guidelines for unresectable, treatment-refractory, and recurrent tumors.
Collapse
Affiliation(s)
- Jaffar Khan
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Asad Ullah
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Abdul Waheed
- Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA; (A.W.); (S.M.B.); (S.M.S.); (F.S.); (S.F.)
| | - Nabin Raj Karki
- Georgia Cancer Center, Department of Hematology Oncology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Nawaraj Adhikari
- Department of Medicine, Bon Secours Memorial Regional Medical Center, Mechanicsville, VA 23116, USA;
| | - Lakshmi Vemavarapu
- Department of Pathology, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA;
| | - Sami Belakhlef
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (S.B.); (I.G.)
| | - Samy Malik Bendjemil
- Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA; (A.W.); (S.M.B.); (S.M.S.); (F.S.); (S.F.)
| | - Siamak Mehdizadeh Seraj
- Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA; (A.W.); (S.M.B.); (S.M.S.); (F.S.); (S.F.)
| | - Feroze Sidhwa
- Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA; (A.W.); (S.M.B.); (S.M.S.); (F.S.); (S.F.)
| | - Intisar Ghleilib
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (S.B.); (I.G.)
| | - Shahin Foroutan
- Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA; (A.W.); (S.M.B.); (S.M.S.); (F.S.); (S.F.)
| | - Andrew M. Blakely
- National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.M.B.); (J.D.R.)
| | - Jaydira Del Rivero
- National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.M.B.); (J.D.R.)
| | - Nagla Abdel Karim
- Inova Schar Cancer Institute, Department of Medicine, University of Virginia, Fairfax, VA 22031, USA;
| | - Eric Vail
- Molecular Pathology Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (E.V.); (S.H.)
| | - Saleh Heneidi
- Molecular Pathology Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (E.V.); (S.H.)
| | - Hector Mesa
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| |
Collapse
|