Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Endocr Relat Cancer ; 31(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38642579

RESUMEN

Neuropilin 2 (NRP2), a transmembrane non-tyrosine kinase receptor, has been described as a potential critical player in the tumourigenesis of several solid cancers and particularly in neuroendocrine neoplasms (NENs). A soluble form of NRP2 (sNRP2) has been previously described and corresponds to a truncated splice isoform. Its prognostic value has never been studied in NEN. NRP2 expression was studied by immunochemistry on tissue microarrays (n = 437) and on circulating tumour cells (CTCs, n = 5 patients with neuroendocrine carcinoma, NEC). We described the levels of sNRP2 in 229 patients with NEN using the ELISA method to identify the factors associated with sNRP2 levels and to evaluate its prognostic role; 90 blood donors represented the healthy control group. NRP2 was found in 97% of neuroendocrine tumours (396/410) and in 74% of NEC (20/27). NRP2 was also expressed in CTC of all the studied patients. The receiver operating characteristic (ROC) analysis showed that sNRP2 had a weak capacity to discriminate between NEN patients and healthy controls (area under curve (AUC) = 0.601, P = 0.053). Abnormal sNRP2 levels were associated with inflammatory syndrome, bone and peritoneal metastases, and abnormal chromogranin A levels. Patients with high sNRP2 levels (sNRP2Q3-Q4) had significantly poorer overall survival in multivariate analysis (HR 0.16, 95% CI (0.04-0.67), P = 0.015). In conclusion, the present study found that sNRP2 and NRP2 could represent a new prognostic biomarker and a therapeutic target, respectively, particularly in aggressive NEN.


Asunto(s)
Biomarcadores de Tumor , Tumores Neuroendocrinos , Neuropilina-2 , Humanos , Femenino , Neuropilina-2/metabolismo , Neuropilina-2/genética , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/sangre , Anciano , Adulto , Biomarcadores de Tumor/metabolismo , Pronóstico , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Anciano de 80 o más Años , Adulto Joven
2.
Neuroendocrinology ; 111(8): 786-793, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32998140

RESUMEN

INTRODUCTION: Small-intestinal neuroendocrine tumors (SI-NET) are situated preferentially within the ileum. The aim was to describe a potential difference in location between unifocal and multiple ileal-NET. PATIENTS AND METHODS: Between December 2010 and December 2019, all consecutive patients who underwent resection in our European Neuroendocrine Tumor Society Center of Excellence, of at least 1 non-duodenal SI-NET, were retrospectively included. The main objective was to prove that multiple ileal-NET were mostly located on the left side of the superior mesenteric artery (SMA) axis (defined as 40 cm from the ileocecal valve), and unifocal ones on the right side. RESULTS: Ninety-four patients were included, 6 with unifocal jejunal-NET located 35 cm (range, 10-60) from the duodenojejunal angle (DJA), 44 (47%) with unifocal ileal-NET and 44 (47%) with multiple ileal-NET. The median number of tumors in multiple ileal-NET was 7 (range, 2-95), within a median small bowel segment of 105 cm (10-240). The median length between the proximal tumor and the DJA was 428 cm (300-635) and 540 cm (350-725) for the distal one; 40 (91%) of them were located on the left side of the SMA axis. In contrast, unifocal ileal-NET were located at a median distance of 577 cm (305-820) from the DJA (p < 0.001, compared to multiple ileal-NET); 30 (68%) of them were on the right side of the SMA axis (p < 0.001). CONCLUSION: Multiple ileal-NET are mostly located on the left side of the SMA axis. Further studies are warranted to explore the embryological origin of unifocal versus multiple ileal-NET.


Asunto(s)
Neoplasias del Íleon/patología , Neoplasias de Células Germinales y Embrionarias/patología , Tumores Neuroendocrinos/patología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
3.
Metabolites ; 9(12)2019 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-31835679

RESUMEN

To assess the metabolomic fingerprint of small intestine neuroendocrine tumors (SI-NETs) and related hepatic metastases, and to investigate the influence of the hepatic environment on SI-NETs metabolome. Ninety-four tissue samples, including 46 SI-NETs, 18 hepatic NET metastases and 30 normal SI and liver samples, were analyzed using 1H-magic angle spinning (HRMAS) NMR nuclear magnetic resonance (NMR) spectroscopy. Twenty-seven metabolites were identified and quantified. Differences between primary NETs vs. normal SI and primary NETs vs. hepatic metastases, were assessed. Network analysis was performed according to several clinical and pathological features. Succinate, glutathion, taurine, myoinositol and glycerophosphocholine characterized NETs. Normal SI specimens showed higher levels of alanine, creatine, ethanolamine and aspartate. PLS-DA revealed a continuum-like distribution among normal SI, G1-SI-NETs and G2-SI-NETs. The G2-SI-NET distribution was closer and clearly separated from normal SI tissue. Lower concentration of glucose, serine and glycine, and increased levels of choline-containing compounds, taurine, lactate and alanine, were found in SI-NETs with more aggressive tumors. Higher abundance of acetate, succinate, choline, phosphocholine, taurine, lactate and aspartate discriminated liver metastases from normal hepatic parenchyma. Higher levels of alanine, ethanolamine, glycerophosphocholine and glucose was found in hepatic metastases than in primary SI-NETs. The present work gives for the first time a snapshot of the metabolomic characteristics of SI-NETs, suggesting the existence of complex metabolic reality, maybe characteristic of different tumor evolution.

4.
J Pathol ; 249(3): 343-355, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31257576

RESUMEN

The identification of novel regulators of tumor progression is a key challenge to gain knowledge on the biology of small intestinal neuroendocrine tumors (SI-NETs). We recently identified the loss of the axon guidance protein semaphorin 3F as a protumoral event in SI-NETs. Interestingly the expression of its receptor neuropilin-2 (NRP-2) was still maintained. This study aimed at deciphering the potential role of NRP-2 as a contributor to SI-NET progression. The role of NRP-2 in SI-NET progression was addressed using an approach integrating human tissue and serum samples, cell lines and in vivo models. Data obtained from human SI-NET tissues showed that membranous NRP-2 expression is present in a majority of tumors, and is correlated with invasion, metastatic abilities, and neovascularization. In addition, NRP-2 soluble isoform was found elevated in serum samples from metastatic patients. In preclinical mouse models of NET progression, NRP-2 silencing led to a sustained antitumor effect, partly driven by the downregulation of VEGFR2. In contrast, its ectopic expression conferred a gain of aggressiveness, driven by the activation of various oncogenic signaling pathways. Lastly, NRP-2 inhibition led to a decrease of tumor cell viability, and sensitized to therapeutic agents. Overall, our results point out NRP-2 as a potential therapeutic target for SI-NETs, and will foster the development of innovative strategies targeting this receptor. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Carcinoma Neuroendocrino/metabolismo , Neoplasias Intestinales/metabolismo , Intestino Delgado/metabolismo , Neuropilina-2/metabolismo , Anciano , Animales , Antineoplásicos/farmacología , Apoptosis , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/secundario , Línea Celular Tumoral , Movimiento Celular , Everolimus/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Intestino Delgado/patología , Masculino , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica , Neuropilina-2/sangre , Neuropilina-2/genética , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Mol Cancer Ther ; 17(1): 60-72, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051320

RESUMEN

Streptozotocin-based chemotherapy is the first-line chemotherapy recommended for advanced pancreatic neuroendocrine tumors (pNETs), whereas targeted therapies, including mTOR inhibitors, are available in second-line treatment. Unfortunately, objective response rates to both treatments are limited. Because mTOR pathway activation, commonly observed in pNETs, has been reported as one of the major mechanisms accounting for chemoresistance, we investigated the potential benefit of mTOR inhibition combined with streptozotocin treatment in a subset of pNETs, namely insulinomas. To evaluate the potential of mTOR inhibition in combination with streptozotocin, we selected four different inhibitors acting at various levels of the pathway (everolimus: inhibition of mTORC1; MK-2206: inhibition of AKT; BKM120: inhibition of PI3K, mTORC1, and mTORC2; and BEZ235: inhibition of mTORC1 and mTORC2). Effects on cell viability and apoptosis were assessed in insulinoma cell lines INS-1E (rat) and MIN6 (mouse) in vitro and were confirmed in vivo by using a mouse model of hepatic tumor dissemination after intrasplenic xenograft. In vitro, all four combinations display synergistic effects. These combinations lead to heterogeneous mTOR pathway inhibition, in agreement with their respective target, and increased apoptosis. In vivo, tumor growth in the liver was significantly inhibited by combining streptozotocin with everolimus (P = 0.0014), BKM120 (P = 0.0092), or BEZ235 (P = 0.008) as compared to each agent alone. These results suggest that targeting the mTOR pathway in combination with streptozotocin could be of potential benefit for insulinomas and pNET patients and thus support further clinical investigations. Mol Cancer Ther; 17(1); 60-72. ©2017 AACR.


Asunto(s)
Insulinoma/tratamiento farmacológico , Estreptozocina/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/uso terapéutico , Animales , Femenino , Humanos , Insulinoma/patología , Ratones , Ratones Desnudos , Estreptozocina/farmacología , Serina-Treonina Quinasas TOR/farmacología
6.
Oncotarget ; 8(13): 20974-20987, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28423496

RESUMEN

mTOR and Unfolded Protein Response (UPR) are two signaling pathways frequently activated in cancer cells. The mTOR pathway has been shown to be up-regulated in most gastroenteropancreatic neuroendocrine tumors. In contrast, little is known about the UPR status in neoplastic neuroendocrine cells. However, these hormone-producing cells are likely to present distinctive adaptations of this pathway, as other secretory cells. We therefore analyzed the status of the three axes of UPR and their relation to mTOR pathway in two gastrointestinal neuroendocrine tumors (GI-NET) cell lines STC-1 and GluTag. At baseline, pharmacological inducers activate the three arms of UPR: PERK, ATF6 and IRE1. Although hypoxia stimulates the PERK, ATF6 and IRE-1 pathways in both cell lines, glucose depletion activates UPR only in STC-1 cell line. Strikingly, P-p70S6K1 increases concomitantly to P-PERK and BiP in response to thapsigargin treatment, glucose depletion or hypoxia. We found that different mTOR inhibitors activate the PERK signaling pathway. To confirm that mTOR inhibition modulates PERK activation, we inhibited PERK and showed that it decreased cell viability when associated to mTOR inhibition, indicating that mTOR drives a PERK-dependent survival pathway. In conclusion, in GI-NET cell lines, UPR signaling is functional and PERK arm is induced by mTOR inhibition. These observations open up new perspectives for therapeutic strategies: the crosstalk between mTOR and UPR might contribute to the resistance to mTOR inhibitors and could be targeted by mTOR and PERK inhibitors in combination therapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias Gastrointestinales/patología , Proteínas de Choque Térmico/metabolismo , Tumores Neuroendocrinos/patología , Serina-Treonina Quinasas TOR/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Apoptosis , Biomarcadores de Tumor/metabolismo , Hipoxia de la Célula , Chaperón BiP del Retículo Endoplásmico , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/metabolismo , Glucosa , Proteínas de Choque Térmico/antagonistas & inhibidores , Humanos , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Células Tumorales Cultivadas , eIF-2 Quinasa/antagonistas & inhibidores
7.
Invest New Drugs ; 34(5): 654-62, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27230034

RESUMEN

Objectives Several targeted therapies are available for metastatic neuroendocrine tumours (NETs) but no predictive factor of response to these treatments has been identified yet. Our aim was to identify and evaluate clinical, biological, histological and functional markers of response to everolimus. Methods We retrospectively reviewed 53 patients with NETs treated with everolimus (68 % in clinical trials). Clinical, biological and histological data were analyzed. The functional marker p-p70S6K, a main effector of the mTOR pathway, was studied by immunohistochemistry in 43 cases. Prognostic factors of progression-free survival (PFS) were studied by Kaplan Meier analysis. Results All patients had metastatic and progressive disease before everolimus treatment. Objective response was 9 % and median PFS was 8.1 (4.7-11.5) months. Hypercholesterolemia (HR = 0.13, p < 0.0001) was associated with longer PFS, whereas presence of bone metastases (HR = 3.1, p < 0.001) and overexpression of p-p70S6K by tumor cells (HR = 2.5, p = 0.01) were associated with shorter PFS under everolimus at multivariate analysis. Conclusion Clinical markers are not useful to predict response to everolimus. However, occurrence of hypercholesterolemia under treatment may be an early marker of response. Prospective studies are required to confirm these results and to assess whether p-p70S6K immunostaining is a prognostic or predictive marker of no-response to everolimus.


Asunto(s)
Antineoplásicos/uso terapéutico , Everolimus/uso terapéutico , Hipercolesterolemia/inducido químicamente , Tumores Neuroendocrinos/tratamiento farmacológico , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Everolimus/efectos adversos , Everolimus/farmacología , Femenino , Humanos , Hipercolesterolemia/metabolismo , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/metabolismo , Pronóstico , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento
8.
Oncotarget ; 6(34): 36731-45, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26447612

RESUMEN

Gastro-intestinal neuroendocrine tumors (GI-NETs) are rare neoplasms, frequently metastatic, raising difficult clinical and therapeutic challenges due to a poor knowledge of their biology. As neuroendocrine cells express both epithelial and neural cell markers, we studied the possible involvement in GI-NETs of axon guidance molecules, which have been shown to decrease tumor cell proliferation and metastatic dissemination in several tumor types. We focused on the role of Semaphorin 3F (SEMA3F) in ileal NETs, one of the most frequent subtypes of GI-NETs.SEMA3F expression was detected in normal neuroendocrine cells but was lost in most of human primary tumors and all their metastases. SEMA3F loss of expression was associated with promoter gene methylation. After increasing endogenous SEMA3F levels through stable transfection, enteroendocrine cell lines STC-1 and GluTag showed a reduced proliferation rate in vitro. In two different xenograft mouse models, SEMA3F-overexpressing cells exhibited a reduced ability to form tumors and a hampered liver dissemination potential in vivo. This resulted, at least in part, from the inhibition of mTOR and MAPK signaling pathways.This study demonstrates an anti-tumoral role of SEMA3F in ileal NETs. We thus suggest that SEMA3F and/or its cellular signaling pathway could represent a target for ileal NET therapy.


Asunto(s)
Orientación del Axón/fisiología , Neoplasias del Íleon/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tumores Neuroendocrinos/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Neoplasias del Íleon/genética , Neoplasias del Íleon/patología , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/genética , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Transducción de Señal
9.
Mol Cell Endocrinol ; 399: 154-63, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25224486

RESUMEN

Small-intestinal neuroendocrine tumors (SI-NETs) are defined as locally invasive only after extension to the muscularis propria. To gain further insight into the molecular mechanisms, we applied a proteomic approach to an orthotopic xenograft model to identify candidate proteins evaluable in human SI-NETs. After grafting STC-1 neuroendocrine tumor cells on the caecum of nude mice, comparative proteomic studies were performed between the pre-invasive and the invasive stages, respectively 2 and 8 weeks after grafting. We identified 24 proteins displaying at least a 1.5-fold differential expression between 2 and 8 week-stages. Most were cytoskeleton-associated proteins, among which five showed decreasing expression levels (CRMP2, TCP1ε, TPM2, vimentin, desmin) and two increasing expression levels (14-3-3γ, CK8). Changes for CRMP2, TCP1ε, TPM2 and 14-3-3γ were confirmed in experimental tumors and in a series of 28 human SI-NETs. In conclusion, our results underline the relevance of proteomics to identify novel biomarkers of tissue invasion.


Asunto(s)
Proteínas del Citoesqueleto/biosíntesis , Células Enteroendocrinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Intestinales/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteómica , Animales , Línea Celular Tumoral , Células Enteroendocrinas/patología , Humanos , Neoplasias Intestinales/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias
10.
Neuroendocrinology ; 97(4): 331-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23343749

RESUMEN

BACKGROUND/AIMS: While the range of therapeutic options for well-differentiated gastroenteropancreatic neuroendocrine tumors has recently increased with the emergence of targeted therapies, such as mTOR inhibitors, there is no recent progress in the treatment of poorly differentiated neuroendocrine carcinomas (PDNECs). Since PDNECs have been shown to strongly express mTOR pathway components, the aim of the present study was to assess the antitumor effect of the mTOR inhibitor everolimus in preclinical models of PDNECs. METHODS: The expression of mTOR pathway components and their response to everolimus were assessed in two neuroendocrine cell lines: STC-1 and GluTag. A xenograft model of intrahepatic dissemination in the nude mouse, based on the intrasplenic injection of either STC-1 and GluTag tumor cells, was used. Animals were started on everolimus treatment 3 days after injection. The effects of treatment on tumor growth, proliferative capacities, apoptosis and in situ expression of mTOR pathway components were assessed. RESULTS: The expression of mTOR pathway components was comparable in STC-1 and GluTag cells and in human PDNECs and could be inhibited in vitro by everolimus. In vivo, the tumor volume of STC-1 and GluTag xenografts was significantly reduced in treated animals (6.05 ± 1.84% as compared to 21.76 ± 3.88% in controls). Everolimus treatment also induced a significant decrease in Ki67 index and in the phosphorylation levels of the two major effectors of mTOR, p70S6K and 4E-BP1. CONCLUSION: Our experimental data suggest that mTOR inhibition could be considered a therapeutic option for high-grade gastroenteropancreatic neuroendocrine tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Sirolimus/análogos & derivados , Neoplasias Gástricas/tratamiento farmacológico , Adulto , Anciano , Animales , Línea Celular Tumoral , Everolimus , Femenino , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/uso terapéutico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Neuroendocrinology ; 94(3): 200-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21677423

RESUMEN

BACKGROUND/AIMS: Experimental studies in vitro suggest that somatostatin and some of its analogues used in clinical practice, such as octreotide, may have potent antiangiogenic properties. However, the clinical transposition of these data is difficult. METHODS: To address this issue, we designed a comparative study of the effects of somatostatin and octreotide on the interactions between neoplastic endocrine cells and endothelial cells in several in vitro and in vivo experimental models, including primary cultures of human umbilical vein endothelial cells (HUVEC), indirect cocultures between HUVEC and the somatostatin-producing endocrine cell line STC-1, and an animal model of intrahepatic dissemination of STC-1 cells. RESULTS: 10(-8)M octreotide markedly inhibited both basal and VEGF-stimulated HUVEC proliferation, had no effect on endothelial cell migration, but inhibited endothelial tubule formation. HUVEC cocultured with the somatostatin- and VEGF-producing STC-1 cells presented a markedly decreased proliferation, a slightly increased motility and an increased capacity of tubule formation; in this system, the inhibition of endothelial cell proliferation was abolished by neutralizing anti-somatostatin but was restored in the presence of anti-VEGF antibodies. This suggests that somatostatin is able to antagonize the effects of VEGF on endothelial cell proliferation but not on endothelial cell sprouting. Finally, no significant effect of octreotide on tumor growth and intratumoral microvascular density was detected in an experimental model of intrahepatic dissemination of STC-1 cells. CONCLUSION: The in vitro antiangiogenic effects of somatostatin and its analogues are likely to be efficiently counterbalanced in the tumor microenvironment by the concomitant release of proangiogenic factors like VEGF.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Células Enteroendocrinas/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Octreótido/farmacología , Somatostatina/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma/patología , Comunicación Celular/fisiología , Células Cultivadas , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Femenino , Neoplasias Gastrointestinales/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Am J Pathol ; 178(1): 336-44, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224070

RESUMEN

The mammalian target of rapamycin (mTOR) inhibitors, such as rapalogues, are a promising new tool for the treatment of metastatic gastroenteropancreatic endocrine tumors. However, their mechanisms of action remain to be established. We used two murine intestinal endocrine tumoral cell lines, STC-1 and GLUTag, to evaluate the antitumor effects of rapamycin in vitro and in vivo in a preclinical model of liver endocrine metastases. In vitro, rapamycin inhibited the proliferation of cells in the basal state and after stimulation by insulin-like growth factor-1. Simultaneously, p70S6 kinase and 4EBP1 phosphorylation was inhibited. In vivo, rapamycin substantially inhibited the intrahepatic growth of STC-1 cells, irrespectively of the timing of its administration and even when the treatment was administered after cell intrahepatic engraftment. In addition, treated animals had significantly prolonged survival (mean survival time: 47.7 days in treated animals versus 31.8 days in controls) and better clinical status. Rapamycin treatment was associated with a significant decrease in mitotic index and in intratumoral vascular density within STC-1 tumors. Furthermore, the antitumoral effect obtained after treatment with a combination of rapamycin and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 was more significant than with rapamycin alone in both cell lines. Our results suggest that the antitumor efficacy of rapamycin in neuroendocrine tumors results from a combination of antiproliferative and antiangiogenic effects. Interestingly, a more potent antitumor efficiency could be obtained by simultaneously targeting several levels of the PI3K/mTOR pathway.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Carcinoma Neuroendocrino/tratamiento farmacológico , Cromonas/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Morfolinas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/patología , Ratones , Fosforilación/efectos de los fármacos
14.
J Cell Sci ; 123(Pt 19): 3256-65, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20826465

RESUMEN

The RNA-binding protein Musashi-1 (Msi1) has been proposed as a marker of intestinal epithelial stem cells. These cells are responsible for the continuous renewal of the intestinal epithelium. Although the function of Msi1 has been studied in several organs from different species and in mammalian cell lines, its function and molecular regulation in mouse intestinal epithelium progenitor cells are still undefined. We describe here that, in these cells, the expression of Msi1 is regulated by the canonical Wnt pathway, through a mechanism involving a functional Tcf/Lef binding site on its promoter. An in vitro study in intestinal epithelium primary cultures showed that Msi1 overexpression promotes progenitor proliferation and activates Wnt and Notch pathways. Moreover, Msi1-overexpressing cells exhibit tumorigenic properties in xenograft experiments. These data point to a positive feedback loop between Msi1 and Wnt in intestinal epithelial progenitors. They also suggest that Msi1 has oncogenic properties in these cells, probably through induction of both the Wnt and Notch pathways.


Asunto(s)
Biomarcadores/metabolismo , Mucosa Intestinal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Animales , Animales Recién Nacidos , Línea Celular Transformada , Proliferación Celular , Transformación Celular Neoplásica/genética , Mucosa Intestinal/patología , Mucosa Intestinal/trasplante , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Ratas , Receptores Notch/genética , Receptores Notch/metabolismo , Células Madre/patología , Activación Transcripcional/genética , Transgenes/genética , Trasplante Heterólogo , Proteínas Wnt/genética
15.
Neuroendocrinology ; 91(3): 268-78, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20389030

RESUMEN

Gastroenteropancreatic (GEP) endocrine tumors are hypervascular tumors able to synthesize and secrete high amounts of VEGF. We aimed to study the regulation of VEGF production in GEP endocrine tumors and to test whether some of the drugs currently used in their treatment, such as somatostatin analogues and mTOR inhibitors, may interfere with VEGF secretion. We therefore analyzed the effects of the somatostatin analogue octreotide, the mTOR inhibitor rapamycin, the PI3K inhibitor LY294002, the MEK1 inhibitor PD98059 and the p38 inhibitor SB203850 on VEGF secretion, assessed by ELISA and Western blotting, in three murine endocrine cell lines, STC-1, INS-r3 and INS-r9. Octreotide and rapamycin induced a significant decrease in VEGF production by all three cell lines; LY294002 significantly inhibited VEGF production by STC-1 and INS-r3 only. We detected no effect of PD98059 whereas SB203850 significantly inhibited VEGF secretion in INS-r3 and INS-r9 cells only. By Western blotting analysis, we observed decreased intracellular levels of VEGF and HIF-1alpha under octreotide, rapamycin and LY294002. For rapamycin and LY294002, this effect was likely mediated by the inhibition of the mTOR/HIF-1/VEGF pathway. In addition to its well-known anti-secretory effects, octreotide may also act through the inhibition of the PI3K/Akt pathway, as suggested by the decrease in Akt phosphorylation detected in all three cell lines. In conclusion, our study points out to the complex regulation of VEGF synthesis and secretion in neoplastic GEP endocrine cells and suggests that the inhibition of VEGF production by octreotide and rapamycin may contribute to their therapeutic effects.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/metabolismo , Octreótido/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Imidazoles/farmacología , Insulina/metabolismo , Secreción de Insulina , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Ratones , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Sirolimus/farmacología , Somatostatina/efectos de los fármacos , Somatostatina/metabolismo , Serina-Treonina Quinasas TOR , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
16.
Magn Reson Imaging ; 28(2): 264-72, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19695818

RESUMEN

Liver metastases in patients with gastroenteropancreatic (GEP) endocrine tumors represent the main factor of adverse prognosis in this tumor type and thus have a strong effect on the therapeutic strategies. Currently, magnetic resonance imaging (MRI) is considered the modality of choice for the noninvasive, in vivo detection of liver metastases. Dedicated MRI protocols suitable for following liver lesion evolution on an experimental model of endocrine tumors could be valuable. An experimental animal model mimicking the clinical situation of intrahepatic dissemination has been designed. The goal of this study was to characterize liver lesions in this athymic nude mouse model and assess the detection sensitivity of MRI using a physiological gating strategy optimized for high magnetic fields. The experiments were performed at 7 T using a dual cardiac-respiratory-triggered multiple spin-echo sequence. This protocol was used to carry out a longitudinal follow-up of hepatic lesions in a group of eight nude mice at different stages: Day 7 (D7), Day 12 (D12), Day 17 (D17) and Day 24 (D24). The hepatic lesion volume fraction (HLVF) was quantified using an adaptive segmentation procedure based on a dual-reference limit. Mean transverse relaxation time T(2) values were quantified from multiple spin-echo images. The first lesions were detected at stage D12 on images with 20-ms TE. From D12, the HLVF increased significantly with stage. The mean T(2) values also increased significantly at D17 and D24. In conclusion, the level of detection and characterization of liver lesions were performed using a devoted protocol with a dedicated high-field MRI synchronization strategy. In future studies, MRI could be used to monitor the effects of targeted therapies on liver endocrine metastases in preclinical animal models.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/secundario , Técnicas de Imagen Sincronizada Respiratorias/métodos , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
17.
J Surg Res ; 154(1): 68-77, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18708196

RESUMEN

BACKGROUND: Clinical observations suggest that in neuroendocrine digestive tumors a high intratumoral microvascular density is associated with good prognosis. We used an experimental orthotopic xenograft model to analyze the relations between angiogenic activity and tumor progression in this tumor subset. MATERIAL AND METHODS: We compared 2 endocrine cell lines: STC-1, a low vascular endothelial growth factor (VEGF)-producing cell line, and INS-r3, a high VEGF-producing cell line. Tumor cells were grafted in the adventitial layer of the caecal wall of nude mice, sacrificed after 8 wk. RESULTS: At 8 wk, "primary" tumors were present in all animals. STC-1 derived tumors were morphologically moderately differentiated, with high proliferative and apoptotic activities; in contrast, INS-r3 derived tumors were well differentiated, with low proliferative and apoptotic activities. VEGF was expressed in <50% grafted STC-1 cells but in >90% of grafted INS-r3 cells. Microvascular density was significantly higher in INS-r3 derived tumors than in STC-1 derived tumors. All STC-1 derived tumors (n = 8) have invaded the mucosa, in contrast to none of the INS-r3 derived tumors (n = 8); liver metastases were detected in 7/8 animals bearing STC-1 derived tumors and in 0/8 animals with INS-r3 derived tumors, despite the presence of lymph node metastases. CONCLUSIONS: Our experimental data concur with clinical findings to suggest that in well differentiated digestive neuroendocrine tumors angiogenesis is disconnected from tumor progression: the development of a highly vascular tumor microenvironment is correlated with VEGF secretion but is not associated with invasive and metastatic properties; it must therefore be regarded as an indirect marker of differentiation.


Asunto(s)
Carcinoma Neuroendocrino/patología , Neoplasias del Sistema Digestivo/patología , Neoplasias Intestinales/patología , Neovascularización Patológica/patología , Animales , Antígenos Virales de Tumores/genética , Carcinoma Neuroendocrino/irrigación sanguínea , Línea Celular Tumoral , Neoplasias del Sistema Digestivo/irrigación sanguínea , Progresión de la Enfermedad , Glucagón/genética , Insulina/genética , Neoplasias Intestinales/irrigación sanguínea , Metástasis Linfática/patología , Ratones , Ratones Desnudos , Microcirculación , Regiones Promotoras Genéticas , Ratas , Virus 40 de los Simios/inmunología , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética
18.
Gastroenterology ; 135(5): 1698-1709.e2, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18775714

RESUMEN

BACKGROUND & AIMS: Missense mutations account for 30% of mutations identified in patients with the multiple endocrine neoplasia type 1 (MEN1) syndrome. They raise several issues: the distinction between pathogenic mutations and polymorphisms is sometimes difficult and the functional effects of missense mutations are unclear. We aimed to evaluate the functional consequences of missense MEN1 mutations in an appropriate endocrine cellular context. METHODS: From the INS-1 insulinoma cell line, we established clones conditionally over expressing wild-type (WT) menin or its A160T, H317Y, and A541T variants. We compared the consequences of WT or variant menin over expression on apoptotic response after gamma-irradiation and analyzed the interactions of these proteins with p53. RESULTS: WT menin over expression sensitized INS-r3 cells to apoptosis through amplification of caspase-3 activation, increased p53 acetylation, and accelerated p21 activation; moreover, over expressed WT menin could be recovered in p53-containing complexes. For all 3 missense mutations tested, the functional effects observed with WT were impaired significantly and only low amounts of variant menin proteins were recovered in p53-containing complexes. CONCLUSIONS: Taking advantage of a new endocrine cellular model, we show a loss of function for 2 missense disease-related menin mutants and for a controversial variant as well. Furthermore, our results suggest the existence of functional interactions between p53 and menin for the control of apoptosis, which may cast new light on the mechanisms of endocrine tumorigenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , ADN de Neoplasias/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación Missense , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas/genética , Recuento de Células , Proliferación Celular , Técnica del Anticuerpo Fluorescente Indirecta , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Humanos , Immunoblotting , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/metabolismo , Células Tumorales Cultivadas
19.
Mol Cell Endocrinol ; 291(1-2): 109-15, 2008 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-18590796

RESUMEN

In a previous study, we demonstrated that the Men1 gene is mainly expressed in the proliferative crypt compartment of the small intestine and that a reduction of menin expression in the crypt-like IEC-17 cell line induces an increase in proliferation rate concomitant with an increase in cyclin D1 expression. The aim of the present study was to test the hypothesis that the NF-kappaB pathway may be involved in cyclin D1 overexpression. Transcriptional activity of the cyclin D1 gene promoter was increased upon reduction of menin expression. Blockade of the NF-kappaB pathway restored proliferation, cell cycle, cyclin D1 gene transcription and cyclin D1 expression levels to those observed in the presence of menin. These data support a correlation between cyclin D1 expression, NF-kappaB activity and menin expression in this epithelial cell line and are relevant to the physiological function of menin in regulating proliferation in the intestinal epithelium.


Asunto(s)
Células Epiteliales/fisiología , Mucosa Intestinal/citología , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Duodeno/citología , Duodeno/fisiología , Células Epiteliales/citología , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Ratas , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Transcripción Genética
20.
Nucl Med Biol ; 35(1): 123-30, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18158951

RESUMEN

PURPOSE: The aim of this study was to investigate (a) in vitro the relationship between [(18)F]fluorocholine ([(18)F]FCH) uptake and cell growth in endocrine cell lines and (b) in vivo the uptake of [(18)F]FCH by tumoral sites in an animal model of metastasized endocrine tumor. METHODS: In vitro studies were conducted on three endocrine and two nonendocrine digestive tumoral cell lines. The proliferative ratio was estimated using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. The uptake of [(18)F]FCH and that of [(18)F]fluorodeoxyglucose ([(18)F]FDG) were measured before and after cytotoxic therapy. [(18)F]FCH biodistribution was studied in nude mice and in an endocrine xenografted mice model. RESULTS: The [(18)F]FCH uptake in tumoral cell lines was related to their proliferative capacities as measured by the MTT assay in basal conditions. After cytotoxic therapy, the IC(50) values calculated with the [(18)F]FCH incorporation test were very close to those determined with the MTT assay. Biodistribution studies showed that [(18)F]FCH was predominantly concentrated in the liver and kidney of nude mice. In the STC-1 xenografted animal model, the uptake of [(18)F]FCH in the primary tumor was only 1.1%. On autoradiography and micro-positron emission tomography, there was no uptake of [(18)F]FCH in liver metastases but there was a significant uptake of [(18)F]FDG. CONCLUSIONS: In vitro studies suggested that the incorporation of [(18)F]FCH in endocrine tumor cell lines was related to their growth capacities; however, in vivo studies conducted in an endocrine xenografted animal model showed an uptake of [(18)F]FCH in hepatic metastases lower than that in normal liver cells. An influence of the microenvironment or a competition phenomenon for [(18)F]FCH uptake between normal liver and endocrine tumor cells cannot be excluded.


Asunto(s)
Colina/análogos & derivados , Neoplasias del Sistema Digestivo/diagnóstico por imagen , Neoplasias de las Glándulas Endocrinas/diagnóstico por imagen , Radioisótopos de Flúor , Animales , Línea Celular Tumoral , Proliferación Celular , Colina/metabolismo , Neoplasias del Sistema Digestivo/metabolismo , Neoplasias del Sistema Digestivo/patología , Neoplasias de las Glándulas Endocrinas/metabolismo , Neoplasias de las Glándulas Endocrinas/patología , Femenino , Fluorodesoxiglucosa F18 , Ratones , Metástasis de la Neoplasia , Cintigrafía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...