Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 828
Filtrar
1.
Appl Immunohistochem Mol Morphol ; 32(5): 229-232, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38584487

RESUMEN

Gastrointestinal stromal tumors (GISTs) are mesenchymal neoplasms with variable behavior characterized by differentiation toward the interstitial cells of Cajal occurring anywhere in the gastrointestinal stromal tract. Frequently, GISTs have fibrous stroma within tumor cell proliferation areas, which is unlike other types of malignant tumors. If this desmoplasia is active, there is a possibility that some sort of transmitter exists between GIST cells and cells related to fibrosis in the tumor cell proliferation areas. Transforming growth factor (TGF)-ß isoforms, particularly TGF-ß1, are critical for fibrosis pathogenesis. TGF-ß1 regulation of myofibroblasts and fibroblasts during fibrosis is well described. The induced fibroblast activation resulting in myofibroblast differentiation has been reported as an important source of collagen, glycoproteins, proteoglycans, and matrix metallopeptidases in wound healing and fibrosis. However, there are a few reports on the relationship between TGF-ß1 and GISTs. This study aims to clarify TGF-ß1 expression in 30 gastric GISTs using immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR). For comparison, we also enrolled 30 samples of gastric tubular adenocarcinoma (GTAC). We confirmed TGF-ß1 expression (H-score ≥50 points) in 57% of GIST and 13% of GTAC samples, a significant difference between the 2 tumor types ( P =0.001). We examined the TGF-ß1 mRNA expression of 3 representative GIST samples, each having their respective immunostained areas detected by RT-PCR. Finding TGF-ß1 expression may indicate that this cytokine plays a part in the formation of desmoplasia within GIST cell proliferative areas.


Asunto(s)
Tumores del Estroma Gastrointestinal , Factor de Crecimiento Transformador beta1 , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Tumores del Estroma Gastrointestinal/genética , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Femenino , Masculino , Persona de Mediana Edad , Anciano , Inmunohistoquímica , Regulación Neoplásica de la Expresión Génica , Adulto , Fibrosis , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Anciano de 80 o más Años
3.
Cell Death Dis ; 14(12): 839, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110356

RESUMEN

Imatinib (IM) has significantly improved the prognosis of gastrointestinal stromal tumor (GIST) patients, but some patients still have primary resistance to IM, and approximately half of patients develop acquired drug resistance within 2 years of treatment, necessitating exploration of new treatment strategies. Targeting ferroptosis as a novel approach to tumor treatment has gained attention. Yet, there is limited research on ferroptosis in GIST, and the underlying mechanism remains unclear. In this study, we revealed that IM increased lipid reactive oxygen species and intracellular Fe2+ levels, and decreased glutathione levels in GIST. This effect could be partially inhibited by Ferrostatin-1. Additionally, knocking down STUB1 and overexpressing GPX4 reversed the IM-induced ferroptosis effect. Moreover, STUB1 was identified as a novel E3 ubiquitin ligase of GPX4, promoting the ubiquitination at site K191 of GPX4. The combination of the GPX4 inhibitor RSL3 and IM synergistically induces ferroptosis, inhibiting GIST proliferation both in vivo and in vitro. Furthermore, STUB1 and GPX4 expression serve as independent prognostic factors for GIST. In conclusion, This study is the first to demonstrate that IM induces ferroptosis by promoting STUB1-mediated GPX4 ubiquitination in GIST, and the combination of RSL3 and IM emerges as a promising therapeutic strategy for GIST.


Asunto(s)
Ferroptosis , Tumores del Estroma Gastrointestinal , Humanos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/metabolismo
4.
West Afr J Med ; 40(11 Suppl 1): S25, 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37978874

RESUMEN

Introduction: Gastrointestinal stromal tumours (GISTs) are neoplastic lesions that primarily affect the digestive tract and develop from interstitial cells of Cajal. Due to their malignant potential and personalized treatment, these lesions require histopathologic and immunohistochemical characterization. In this investigation, the sex, age, lesional sites of origin, histopathologic types, the prevalence of HER-2 expression, prognostic indices (based on tumour size and mitotic figures), expression of CD117 and DOG1, and characteristics of patients with GIST were all characterized. Methodology: This is a retrospective cross-sectional analysis of GIST cases seen at four tertiary healthcare centers in Nigeria over ten years (2008 to 2017) and investigated utilizing histopathological and immunohistochemical (CD117, DOG1, and HER-2) methods. Result: In this study, there were twenty GIST cases. Notably, the majority (40%) of the cases had tumours with sizes between 7.0 and 8.0, the stomach was the most frequent site (70%) and the spindle cell type of GIST was the most prevalent (80%) histopathological type. Additionally, the stomach was significantly associated with GIST as an origin site (with a P value of 0.001), and 100% and 50% of these tumours were immunoreactive with CD117 and DOG1 respectively. Finally, HER-2 immunoreactivity was negatively stained with GIST tumour. Conclusion: In our study, GISTs most frequently develop in the stomach, and CD117& DOG1 are essential for correctly diagnosing these tumours. However, HER-2 immunoreactivity is a predictive marker of survival for personalized care.


Asunto(s)
Tumores del Estroma Gastrointestinal , Humanos , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Estudios Retrospectivos , Nigeria/epidemiología , Estudios Transversales
5.
Cell Rep ; 42(9): 113035, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37616163

RESUMEN

Most gastrointestinal stromal tumors (GISTs) develop due to gain-of-function mutations in the tyrosine kinase gene, KIT. We recently showed that mutant KIT mislocalizes to the Golgi area and initiates uncontrolled signaling. However, the molecular mechanisms underlying its Golgi retention remain unknown. Here, we show that protein kinase D2 (PKD2) is activated by the mutant, which causes Golgi retention of KIT. In PKD2-inhibited cells, KIT migrates from the Golgi region to lysosomes and subsequently undergoes degradation. Importantly, delocalized KIT cannot trigger downstream activation. In the Golgi/trans-Golgi network (TGN), KIT activates the PKD2-phosphatidylinositol 4-kinase IIIß (PKD2-PI4KIIIß) pathway through phospholipase Cγ2 (PLCγ2) to generate a PI4P-rich membrane domain, where the AP1-GGA1 complex is aberrantly recruited. Disruption of any factors in this cascade results in the release of KIT from the Golgi/TGN. Our findings show the molecular mechanisms underlying KIT mislocalization and provide evidence for a strategy for inhibition of oncogenic signaling.


Asunto(s)
Tumores del Estroma Gastrointestinal , Humanos , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Proteína Quinasa D2 , Fosfolipasa C gamma/metabolismo , Aparato de Golgi/metabolismo , Red trans-Golgi/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo
6.
Cancer Res ; 83(21): 3624-3635, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37556508

RESUMEN

Imatinib mesylate (IM) has revolutionized the treatment of gastrointestinal stromal tumor (GIST). However, most patients inevitably acquire IM resistance. Second- and third-line treatments exhibit modest clinical benefits with a median time to disease progression of 4 to 6 months, highlighting the urgency for novel therapeutic approaches. Here, we report that the expression of BCL6, a known oncogenic driver and transcriptional repressor, was significantly induced in GIST cells following IM treatment. Elevated BCL6 levels suppressed apoptosis and contributed to IM resistance. Mechanistically, BCL6 recruited SIRT1 to the TP53 promoter to modulate histone acetylation and transcriptionally repress TP53 expression. The reduction in p53 subsequently attenuated cell apoptosis and promoted tolerance of GIST cells to IM. Concordantly, treatment of GIST cells showing high BCL6 expression with a BCL6 inhibitor, BI-3802, conferred IM sensitivity. Furthermore, BI-3802 showed striking synergy with IM in IM-responsive and IM-resistant GIST cells in vitro and in vivo. Thus, these findings reveal a role for BCL6 in IM resistance and suggest that a combination of BCL6 inhibitors and IM could be a potentially effective treatment for GIST. SIGNIFICANCE: BCL6 drives resistance to imatinib by inhibiting p53-mediated apoptosis and can be targeted in combination with imatinib to synergistically suppress tumor growth, providing a therapeutic strategy for treating gastrointestinal stromal tumor.


Asunto(s)
Antineoplásicos , Neoplasias Gastrointestinales , Tumores del Estroma Gastrointestinal , Humanos , Mesilato de Imatinib/farmacología , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Proteína p53 Supresora de Tumor/genética , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Apoptosis , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo
7.
Toxicol Appl Pharmacol ; 466: 116490, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36963523

RESUMEN

Ripretinib, a tyrosine kinase inhibitor (TKI), is the first FDA approved fourth-line therapy for adults with advanced gastrointestinal stromal tumor (GIST). Studies have shown that several TKIs for treating GIST were potent inhibitors of human UDP-glucosyltransferase (UGTs) enzymes. However, whether ripretinib affects the activity of UGTs remains unclear. The aim of this study was to investigate the effects of ripretinib on major UGT isoforms, as well as to evaluate its potential drug-drug interactions (DDIs) risk caused by the inhibition of UGTs activities. The inhibitory effects and inhibition modes of ripretinib on UGTs were systematically evaluated using high-performance liquid chromatography (HPLC) and enzyme kinetic studies, respectively. Our data showed that ripretinib exhibited potent inhibition against UGT1A1, UGT1A3, UGT1A4, UGT1A7 and UGT1A8. Enzyme kinetic studies indicated that ripretinib was not only a competitive inhibitor of UGT1A1, UGT1A4 and UGT1A7, but also a noncompetitive inhibitor of UGT1A3, as well as a mixed inhibitor of UGT1A8. The prediction results of in vitro-in vivo extrapolation (IVIVE) demonstrated that ripretinib might bring the potential risk of DDIs when combined with substrates of UGT1A1, UGT1A3, UGT1A4, UGT1A7 or UGT1A8. Therefore, special attention should be paid when ripretinib is used in conjunction with other drugs metabolized by UGTs to avoid risk of DDIs in clinic.


Asunto(s)
Tumores del Estroma Gastrointestinal , Microsomas Hepáticos , Humanos , Microsomas Hepáticos/metabolismo , Cinética , Tumores del Estroma Gastrointestinal/metabolismo , Glucuronosiltransferasa/metabolismo , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología
8.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36834926

RESUMEN

Activating mutations in KIT (CD117) have been associated with several diseases, including gastrointestinal stromal tumors and mastocytosis. Rapidly progressing pathologies or drug resistance highlight the need for alternative treatment strategies. Previously, we reported that the adaptor molecule SH3 binding protein 2 (SH3BP2 or 3BP2) regulates KIT expression at the transcriptional level and microphthalmia-associated transcription factor (MITF) expression at the post-transcriptional level in human mast cells and gastrointestinal stromal tumor (GIST) cell lines. Lately, we have found that the SH3BP2 pathway regulates MITF through miR-1246 and miR-5100 in GIST. In this study, miR-1246 and miR-5100 were validated by qPCR in the SH3BP2-silenced human mast cell leukemia cell line (HMC-1). MiRNA overexpression reduces MITF and MITF-dependent target expression in HMC-1. The same pattern was observed after MITF silencing. In addition, MITF inhibitor ML329 treatment reduces MITF expression and affects the viability and cell cycle progression in HMC-1. We also examine whether MITF downregulation affected IgE-dependent mast cell degranulation. MiRNA overexpression, MITF silencing, and ML329 treatment reduced IgE-dependent degranulation in LAD2- and CD34+-derived mast cells. These findings suggest MITF may be a potential therapeutic target for allergic reactions and deregulated KIT mast-cell-mediated disorders.


Asunto(s)
Leucemia de Mastocitos , MicroARNs , Factor de Transcripción Asociado a Microftalmía , Humanos , Muerte Celular/genética , Regulación hacia Abajo , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Inmunoglobulina E/metabolismo , Leucemia de Mastocitos/metabolismo , Mastocitos/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , MicroARNs/genética
9.
Cell Immunol ; 384: 104642, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36577281

RESUMEN

BACKGROUND: Tumor-associated macrophages (TAMs) are linked with the progression and poor prognosis of multifarious solid tumors, but the regulatory mechanisms involved in gastrointestinal stromal tumors (GIST) remain indistinct. This study intended to delve into the job of TAM-derived chemokines in promoting metastasis in GIST microenvironment. METHODS: Expression levels of M2-TAM markers and CXCL2 in primary and metastatic tissues of GIST were analyzed by bioinformatics methods, and we analyzed the correlation between CXCL2 and M2-TAM markers. Immunofluorescence was applied to assay CXCL2 and M2-TAM marker protein (CD68 and CD206) expression in tumor tissues. Serum CXCL2 concentration in metastatic and non-metastatic patients was assayed by ELISA. The differentiation of THP-1 cells was tested by flow cytometry. Cell function test was utilized to analyze the viability, invasion and migration of GIST cells. Western blot was used to examine the expression of epithelial-mesenchymal transition (EMT)-related proteins. The mouse liver metastasis model was established, and the effects of CXCL2 and EMT-related genes on metastasis were confirmed by hematoxylin-eosin staining and immunohistochemistry experiments. RESULTS: Bioinformatics analysis ascertained that M2-TAM marker proteins and chemokine CXCL2 were highly expressed in GIST metastatic tissues, and CXCL2 and TAM were co-located in tumor tissues. Results of in vitro cell function experiments displayed that CXCL2 secreted by M2-TAM promoted the invasion, migration and EMT of GIST tumor cells, and the anti-CXCL2 antibody could block the metastasis promoting effect of CXCL2. Additionally, the silencing of CXCR2 in GIST cells inhibited the metastasis promoting effect of CXCL2. Animal studies further confirmed that CXCL2 promoted liver metastasis of GIST in vivo. CONCLUSION: This study preliminarily revealed the mechanism of M2-TAM promoting tumor metastasis by secreting CXCL2 in GIST tumor microenvironment, and proffered theoretical reference for the development of immunotherapy strategies targeting M2-TAM.


Asunto(s)
Tumores del Estroma Gastrointestinal , Neoplasias Hepáticas , Animales , Ratones , Línea Celular Tumoral , Movimiento Celular , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Neoplasias Hepáticas/patología , Macrófagos , Microambiente Tumoral , Macrófagos Asociados a Tumores , Humanos
10.
Head Neck Pathol ; 17(2): 339-346, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36307634

RESUMEN

BACKGROUND: Pleomorphic Adenoma (PA) and Basal cell adenoma (BCA) are benign salivary gland tumors that may pose a diagnostic challenge if typical features are not present. Due to the increased relapse and malignant transformation rate of the former, a correct diagnosis carries relevant prognostic information. Even though immunohistochemistry (IHC) plays a limited role in the diagnosis of these tumors, the use of IHC panels could increase diagnostic accuracy. In the present work, we aimed to demonstrate that the use of an IHC panel consisting of Glial Fibrillary Acid Protein (GFAP), B-Catenin and Discovered On GIST 1 (DOG-1) can aid in the differential diagnosis between PA and BCA. METHODS: We analyzed 18 cases of benign salivary gland tumors (Pleomorphic adenomas and Basal cell adenomas) with overlapping histologic features. First, a head and neck pathologist diagnosed the cases relying on morphology alone. Afterwards, cases were re-evaluated considering the IHC panel results. Inter-observer IHC scoring concordance was evaluated with pre-defined marker cut-off points using Cohen's Kappa scores. RESULTS: Based on morphology alone, 9 cases were classified as PA while the remaining tumors were considered to be BCA. Five out of nine BCA cases showed GFAP staining and absent nuclear B-catenin and DOG-1 positivity. Conversely, 2 PA cases showed absent GFAP and positive nuclear B-catenin with concurrent DOG-1 expression. Therefore, after IHC evaluation, up to 40% of morphologic diagnoses were reconsidered. Overall, the inter-observer concordance for IHC evaluation was good (resulting Kappa Scores between 0.78 and 1). CONCLUSION: Our work supports the use of a concise IHC panel to improve the diagnostic accuracy of benign salivary gland tumors with overlapping histologic features.


Asunto(s)
Adenoma Pleomórfico , Adenoma , Tumores del Estroma Gastrointestinal , Neoplasias de las Glándulas Salivales , Humanos , Adenoma Pleomórfico/diagnóstico , Adenoma Pleomórfico/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Cateninas/metabolismo , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Biomarcadores de Tumor/metabolismo , Glándulas Salivales/metabolismo , Adenoma/diagnóstico , Adenoma/patología , Neoplasias de las Glándulas Salivales/patología
11.
Ann Afr Med ; 22(4): 501-507, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38358152

RESUMEN

Aims: Gastrointestinal stromal tumors (GISTs) are neoplastic lesions that primarily affect the digestive tract and develop from interstitial cells of Cajal. These lesions require histopathological and immunohistochemical characterization due to their malignant potential and personalized treatment. In this investigation, the sex, age, lesional sites of origin, histopathological types, the prevalence of human epidermal growth factor receptors (HER-2) expression, prognostic indices (based on tumor size and mitotic figures), expression of CD117 and DOG1, and characteristics of patients with GIST were all characterized. Materials and Methods: This was a retrospective cross-sectional analysis of GIST cases seen at four tertiary health-care centers in Nigeria over a 10-year period (2008-2017) and investigated utilizing histopathological and immunohistochemical (CD117, DOG1, and HER-2) methods. Results: In this investigation, there were twenty GIST cases. Notably, the majority (40%) of the cases had tumors with sizes between 7.0 and 8.0 cm; the stomach was the most frequent site (70%) and the spindle cell type of GIST was the most prevalent (80%) histopathological type. In addition, the stomach was significantly associated with GIST as an origin site (with a P = 0.001), and 100% and 50% of these tumors were immunoreactive with CD117 and DOG1, respectively. Conclusions: In our study, GISTs most frequently develop in the stomach, and CD117 and DOG1 are essential for correctly diagnosing these tumors. However, HER-2 immunoreactivity is a predictive marker of survival for personalized care.


Résumé Objectifs: Les tumeurs stromales gastro-intestinales (GIST) sont des lésions néoplasiques qui affectent principalement le tube digestif et se développent à partir des cellules interstitielles de Cajal. Ces lésions nécessitent une caractérisation histopathologique et immunohistochimique en raison de leur potentiel malin et d'un traitement personnalisé. Dans cette enquête, le sexe, l'âge, les sites d'origine des lésions, les types histopathologiques, la prévalence de l'expression des récepteurs du facteur de croissance épidermique humain (HER-2), les indices pronostiques (basés sur la taille de la tumeur et les chiffres mitotiques), l'expression de CD117 et DOG1, et les caractéristiques des patients atteints de GIST ont toutes été caractérisées. Matériels et méthodes: Il s'agissait d'une analyse transversale rétrospective de cas de GIST observés dans quatre centres de soins de santé tertiaires au Nigeria sur une période de 10 ans (2008-2017) et étudiée à l'aide d'analyses histopathologiques et immunohistochimiques (CD117, DOG1 et HER). 2) méthodes. Résultats: Dans cette enquête, il y a eu vingt cas de GIST. Notamment, la majorité (40 %) des cas présentaient des tumeurs mesurant entre 7,0 et 8,0 cm ; l'estomac était le site le plus fréquent (70 %) et le type de GIST à cellules fusiformes était le type histopathologique le plus répandu (80 %). De plus, l'estomac était significativement associé au GIST comme site d'origine (avec un P = 0,001) et 100 % et 50 % de ces tumeurs étaient immunoréactives avec CD117 et DOG1, respectivement. Conclusions: Dans notre étude, les GIST se développent le plus souvent dans l'estomac, et CD117 et DOG1 sont essentiels pour diagnostiquer correctement ces tumeurs. Cependant, l'immunoréactivité HER-2 est un marqueur prédictif de survie pour une prise en charge personnalisée. Mots-clés: Biomarqueurs, tumeurs stromales gastro-intestinales, histopathologie, immunohistochimie.


Asunto(s)
Biomarcadores de Tumor , Tumores del Estroma Gastrointestinal , Humanos , Biomarcadores de Tumor/análisis , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/epidemiología , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Estudios Retrospectivos , Nigeria/epidemiología , Estudios Transversales , Inmunohistoquímica
12.
Cell Commun Signal ; 20(1): 138, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36076237

RESUMEN

BACKGROUND: Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours of the gastrointestinal tract and are characterized by activating mutations of c-KIT or PDGFRa receptor tyrosine kinases (RTKs). Despite the clinical success of tyrosine kinase inhibitors (TKIs), more than half of GIST patients develop resistance due to a second mutation. Cyclin-dependent kinase 7 (CDK7) is the catalytic subunit of CDK-activating kinase (CAK), and it plays an important role in the regulation of cell cycle transitions and gene transcription. THZ1, a CDK7 inhibitor, exhibits a dose-dependent inhibitory effect in various cancers. METHODS: Data from the public GEO database and tissue microarray were used to analyse the gene expression levels of CDKs in GISTs. The impact of CDK7 knockdown and the CDK7 inhibitor THZ1 on GIST progression was investigated in vitro using CCK-8, colony formation, and flow cytometry assays and in vivo using a xenograft mouse model. RNA sequencing was performed to investigate the mechanism of GIST cell viability impairment mediated by THZ1 treatment. RESULTS: Our study demonstrated that CDK7 is relatively overexpressed in high-risk GISTs and predicts a poor outcome. A low concentration of THZ1 exhibited a pronounced antineoplastic effect in GIST cells in vivo and in vitro. Moreover, THZ1 exerted synergistic anticancer effects with imatinib. THZ1 treatment resulted in transcriptional modulation by inhibiting the phosphorylation of Ser2, Ser5, and Ser7 within RNA polymerase II (RNAPII). c-KIT, an oncogene driver of GIST, was transcriptionally repressed by THZ1 treatment or CDK7 knockdown. Transcriptome sequencing analysis showed that OSR1 acts as a downstream target of CDK7 and regulates c-KIT expression. Taken together, our results highlight elevated CDK7 expression as a predictor of poor outcome in GIST and present the combination of CDK7 and RTK inhibitors as a potent therapeutic strategy to improve the efficacy of GIST treatment. Video abstract.


Asunto(s)
Antineoplásicos , Tumores del Estroma Gastrointestinal , Fenilendiaminas/farmacología , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Humanos , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Quinasa Activadora de Quinasas Ciclina-Dependientes
13.
Drugs ; 82(13): 1413-1418, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35986838

RESUMEN

Pimitespib (Jeselhy®) is an oral small molecule inhibitor of the α and ß isoforms of heat shock protein 90 (HSP90). HSP90α and HSP90ß regulate the stability and activity of a number of proteins that are crucial for tumour development. Pimitespib is being developed by Taiho Pharmaceutical for the treatment of solid tumours, including gastrointestinal stromal tumour (GIST), and in June 2022 it received its first approval in Japan for GIST that has progressed after chemotherapy. Pimitespib is undergoing phase I development for the treatment of solid tumours in the EU and the USA. This article summarizes the milestones in the development of pimitespib leading to this first approval for GIST that has progressed after chemotherapy.


Asunto(s)
Antineoplásicos , Tumores del Estroma Gastrointestinal , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/uso terapéutico , Humanos , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/uso terapéutico
14.
Lipids Health Dis ; 21(1): 1, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34991597

RESUMEN

BACKGROUND: To investigate the characteristics and clinical significance of serum lipids in patients with gastrointestinal stromal tumors (GISTs). METHODS: The clinical and pathological data of 694 GIST patients in Liyuan hospital and Union hospital from 2012 to 2016 were retrospectively analyzed. Blood lipid levels in patients with varying degrees of risk were compared. RESULTS: The findings showed that LDL-C, HDL-C, and CHOL increased significantly in women, and CD34 positive. In patients with tumors size less than 5 cm in diameter, TG, HDL-C, and CHOL were significantly higher. TG levels were significantly higher in DOG-1 (a marker and has a high specificity and sensitivity in the diagnosis of GIST) positive patients than in DOG-1 negative patients (P < 0.05). S-100 positive patients had lower HDL-C levels than S-100 negative patients (P < 0.05). Lipids indexes were found to be correlated with GIST risk stratification and tumor site (P < 0.05). TG/HDL-C was were significantly different among patients with GIST in different locations (P < 0.05). CONCLUSION: The clinical and pathological characteristics of the patients with GIST are closely related to the level of blood lipids. To a certain extent, information about level of blood lipids can be helpful for distinguishing benign and malignant GIST.


Asunto(s)
Neoplasias Gastrointestinales/metabolismo , Tumores del Estroma Gastrointestinal/metabolismo , Metabolismo de los Lípidos , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Femenino , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/patología , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Triglicéridos/sangre
15.
J Clin Pathol ; 75(12): 825-831, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34155091

RESUMEN

AIMS: Novel prognostic markers are warranted for gastrointestinal stromal tumours. Caveolin-1 is a multifunctional protein that proved to be associated with outcome in multiple tumour types. Aim of this study was to investigate Caveolin-1 expression and prognostic efficacy in a series of gastrointestinal stromal tumours. METHODS: Caveolin-1 expression was assessed by immunohistochemistry in a retrospective series of 66 gastrointestinal stromal tumours representative of the different molecular subtypes. Correlations with clinical, histopathological and molecular features were investigated. Statistical analyses were performed as appropriate. RESULTS: Thirty-five cases out of 66 (53.0%) expressed Caveolin-1. Presence of Caveolin-1 expression correlated with favourable histopathologic and clinical traits, including a lower mitotic count (p=0.003) and lower relapse rate (p=0.005). Caveolin-1 expression also resulted associated with the presence of PDGFRA mutations (p=0.010). Outcome analyses showed a favourable prognostic significance of Caveolin-1 expression in terms of relapse-free survival (HR=0.14; 95% CI=0.03 to 0.63) and overall survival (HR=0.29; 95% CI=0.11 to 0.74), even after adjusting for the mutational subgroup (relapse-free survival: HR=0.14, 95% CI=0.04 to 0.44; overall survival: HR=0.29, 95% CI=0.11 to 0.51) and imatinib treatment (relapse-free survival: HR=0.14, 95% CI=0.02 to 0.81; overall survival: HR=0.29, 95% CI=0.17 to 0.48). CONCLUSION: Caveolin-1 represents a novel prognostic marker in gastrointestinal stromal tumours. Further studies are warranted to validate these results and to explore the mechanisms linking Caveolin-1 expression with the PDGFRA oncogenic pathway.


Asunto(s)
Tumores del Estroma Gastrointestinal , Humanos , Caveolina 1/genética , Caveolina 1/metabolismo , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Mutación , Recurrencia Local de Neoplasia/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estudios Retrospectivos
16.
Turk J Gastroenterol ; 32(12): 1038-1048, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34876394

RESUMEN

BACKGROUND: Considering the difficulty in predicting the biological behavior of gastrointestinal stromal tumors (GISTs) based on histological findings alone, genetic abnormalities have recently become an area of focus. Platelet-derived growth factor receptor (PDGFR), with 2 isoforms (α and ß) is one of the mutations that play a role in the development of GIST. There are very little data determining the relationship of GIST with PDGFRß which is associated with poor prognosis in other mesenchymal and epithelial tumors. In this study, we aimed to show the relationship between clinicopathological criteria and recurrence. We also wanted to evaluate the effect of PDGFRß expression on recurrence and clinicopathological findings. METHODS: We evaluated 40 GIST patients retrospectively for detailed clinicopathological findings, postoperative immunohistochemical tumor markers (CD117, Ki67), and also for tumor recurrence. Immunohistochemical examination for PDGFRß was performed for the all GIST cases. RESULTS: Tumor recurrence was related to male gender (P = .003), serosal localization (P = .004), surgical margins positivity (P = .001), risk group (P = .011), mitotic activity (P = .000), and Ki67 proliferation index (P = .000). PDGFRß was not significantly associated with tumor recurrence (P = .277). CONCLUSION: We can say that the most important parameters related with recurrence of GISTs are mitotic activity and the Ki67 proliferation index. The determination of the cut-off value of the Ki67 proliferation index as 13% instead of 10% would be much more specific and sensitive. Although PDGFRß may be used for the diagnosis of GIST as an alternative for PDGFRα in cases with cKIT negativity, it is not an indicator of tumor recurrence as in other tumors.


Asunto(s)
Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Antígeno Ki-67 , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Proteínas Proto-Oncogénicas c-kit/genética , Estudios Retrospectivos
17.
Oncogene ; 40(48): 6614-6626, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34621020

RESUMEN

KIT/PDGFRA oncogenic tyrosine kinase signaling is the central oncogenic event in most gastrointestinal stromal tumors (GIST), which are human malignant mesenchymal neoplasms that often feature myogenic differentiation. Although targeted inhibition of KIT/PDGFRA provides substantial clinical benefit, GIST cells adapt to KIT/PDGFRA driver suppression and eventually develop resistance. The specific molecular events leading to adaptive resistance in GIST remain unclear. By using clinically representative in vitro and in vivo GIST models and GIST patients' samples, we found that the E3 ubiquitin ligase Atrogin-1 (FBXO32)-the main effector of muscular atrophy in cachexia-resulted in the most critical gene derepressed in response to KIT inhibition, regardless the type of KIT primary or secondary mutation. Atrogin-1 in GISTs is transcriptionally controlled by the KIT-FOXO3a axis, thus indicating overlap with Atrogin-1 regulation mechanisms in nonneoplastic muscle cells. Further, Atrogin-1 overexpression was a GIST-cell-specific pro-survival mechanism that enabled the adaptation to KIT-targeted inhibition by apoptosis evasion through cell quiescence. Buttressed on these findings, we established in vitro and in vivo the preclinical proof-of-concept for co-targeting KIT and the ubiquitin pathway to maximize the therapeutic response to first-line imatinib treatment.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mesilato de Imatinib/farmacología , Proteínas Musculares/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Proteínas Ligasas SKP Cullina F-box/antagonistas & inhibidores , Sulfuros/farmacología , Sulfonamidas/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Quimioterapia Combinada , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Humanos , Ratones , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638938

RESUMEN

Metabolic adaptation to increased oxidative phosphorylation (OXPHOS) has been found in gastrointestinal stromal tumor (GIST) upon imatinib treatment. However, the underlying mechanism of imatinib-induced OXPHOS is unknown. Discovering molecules that mediate imatinib-induced OXPHOS may lead to the development of therapeutic strategies synergizing the efficacy of imatinib. In this study, we explored the role of microRNAs in regulating OXPHOS in GIST upon imatinib treatment. Using a microarray approach, we found that miR-483-3p was one of the most downregulated miRNAs in imatinib-treated tumors compared to untreated tumors. Using an extended series of GIST samples, we further validated the downregulation of miR-483-3p in imatinib-treated GIST samples by RT-qPCR. Using both gain- and loss-of-function experiments, we showed that miR-483-3p could regulate mitochondrial respiratory Complex II expression, suggesting its role in OXPHOS regulation. Functionally, miR-483-3p overexpression could rescue imatinib-induced cell death. These findings provide the molecular link for imatinib-induced OXPHOS expression and the biological role of miR-483-3p in regulating cell viability upon imatinib treatment.


Asunto(s)
Antineoplásicos/farmacología , Complejo II de Transporte de Electrones/metabolismo , Neoplasias Gastrointestinales/metabolismo , Tumores del Estroma Gastrointestinal/metabolismo , Mesilato de Imatinib/farmacología , MicroARNs/metabolismo , Mitocondrias/enzimología , Transducción de Señal/efectos de los fármacos , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib/uso terapéutico , MicroARNs/genética , Mitocondrias/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , ARN Mensajero/genética , Transducción de Señal/genética , Transfección
19.
Front Immunol ; 12: 715727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489967

RESUMEN

Gastrointestinal stromal tumors (GISTs) are a subtype of soft tissue sarcoma (STS), and have become a concept of oncogenic addiction and targeted therapies.The large majority of these tumors develop after a mutation in KIT or platelet derived growth factor receptor α (PDGFRα), resulting in uncontrolled proliferation. GISTs are highly sensitive to imatinib. GISTs are immune infiltrated tumors with a predominance of tumor-associated macrophages (TAMs) and T-cells, including many CD8+ T-cells, whose numbers are prognostic. The genomic expression profile is that of an inhibited Th1 response and the presence of tertiary lymphoid structures and B cell signatures, which are known as predictive to response to ICI. However, the microtumoral environment has immunosuppressive attributes, with immunosuppressive M2 macrophages, overexpression of indoleamine 2,3-dioxygenase (IDO) or PD-L1, and loss of major histocompatibility complex type 1. In addition to inhibiting the KIT oncogene, imatinib appears to act by promoting cytotoxic T-cell activity, interacting with natural killer cells, and inhibiting the expression of PD-L1. Paradoxically, imatinib also appears to induce M2 polarization of macrophages. There have been few immunotherapy trials with anti-CTLA-4 or anti-PD-L1drugs and available clinical data are not very promising. Based on this comprehensive analysis of TME, we believe three immunotherapeutic strategies must be underlined in GIST. First, patients included in clinical trials must be better selected, based on the identified driver mutation (such as PDGFRα D842V mutation), the presence of tertiary lymphoid structures (TLS) or PD-L1 expression. Moreover, innovative immunotherapeutic agents also provide great interest in GIST, and there is a strong rationale for exploring IDO targeting after disease progression during imatinib therapy. Finally and most importantly, there is a strong rationale to combine of c-kit inhibition with immune checkpoint inhibitors.


Asunto(s)
Biomarcadores de Tumor , Susceptibilidad a Enfermedades , Tumores del Estroma Gastrointestinal/etiología , Mutación , Oncogenes , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Tumores del Estroma Gastrointestinal/terapia , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Inmunoterapia , Mediadores de Inflamación/metabolismo , Especificidad de Órganos , Resultado del Tratamiento , Escape del Tumor , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
Ann Diagn Pathol ; 55: 151813, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34509898

RESUMEN

Malignant gastrointestinal neuroectodermal tumor (GNET) is a rare malignant primary gastrointestinal mesenchymal tumor which can be diagnosed via fine-needle aspiration (FNA) cytology. In the context of FNA, the diagnosis requires a cell block and the use of significant resources including immunohistochemical stains and molecular testing. The differential diagnosis of GNET includes clear cell sarcoma (CCS), gastrointestinal stromal tumor (GIST), gastric schwannoma, metastatic melanoma, malignant perivascular epithelioid cell tumor (PEComa) and granular cell tumor, among others. Here we describe a case which was initially diagnosed as malignant granular cell tumor by FNA which was later revised to GNET following the finding of an EWSR1-ATF1 fusion gene rearrangement.


Asunto(s)
Tracto Gastrointestinal/patología , Tumores Neuroectodérmicos , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Biopsia con Aguja Fina , Proteínas de Unión a Calmodulina/análisis , Proteínas de Unión a Calmodulina/metabolismo , Diagnóstico Diferencial , Femenino , Neoplasias Gastrointestinales/diagnóstico , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/patología , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Melanoma/diagnóstico , Melanoma/metabolismo , Melanoma/patología , Persona de Mediana Edad , Tumores Neuroectodérmicos/diagnóstico , Tumores Neuroectodérmicos/metabolismo , Tumores Neuroectodérmicos/patología , Sarcoma de Células Claras/diagnóstico , Sarcoma de Células Claras/metabolismo , Sarcoma de Células Claras/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...