Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Clin Cancer Res ; 30(13): 2835-2845, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38630553

RESUMEN

PURPOSE: Multiple endocrine neoplasia type 1 (MEN1) is thought to increase the risk of meningioma and ependymoma. Thus, we aimed to describe the frequency, incidence, and specific clinical and histological features of central nervous system (CNS) tumors in the MEN1 population (except pituitary tumors). EXPERIMENTAL DESIGN: The study population included patients harboring CNS tumors diagnosed with MEN1 syndrome after 1990 and followed up in the French MEN1 national cohort. The standardized incidence ratio (SIR) was calculated based on the French Gironde CNS Tumor Registry. Genomic analyses were performed on somatic DNA from seven CNS tumors, including meningiomas and ependymomas from patients with MEN1, and then on 50 sporadic meningiomas and ependymomas. RESULTS: A total of 29 CNS tumors were found among the 1,498 symptomatic patients (2%; incidence = 47.4/100,000 person-years; SIR = 4.5), including 12 meningiomas (0.8%; incidence = 16.2/100,000; SIR = 2.5), 8 ependymomas (0.5%; incidence = 10.8/100,000; SIR = 17.6), 5 astrocytomas (0.3%; incidence = 6.7/100,000; SIR = 5.8), and 4 schwannomas (0.3%; incidence = 5.4/100,000; SIR = 12.7). Meningiomas in patients with MEN1 were benign, mostly meningothelial, with 11 years earlier onset compared with the sporadic population and an F/M ratio of 1/1. Spinal and cranial ependymomas were mostly classified as World Health Organization grade 2. A biallelic MEN1 inactivation was observed in 4/5 ependymomas and 1/2 meningiomas from patients with MEN1, whereas MEN1 deletion in one allele was present in 3/41 and 0/9 sporadic meningiomas and ependymomas, respectively. CONCLUSIONS: The incidence of each CNS tumor was higher in the MEN1 population than in the French general population. Meningiomas and ependymomas should be considered part of the MEN1 syndrome, but somatic molecular data are missing to conclude for astrocytomas and schwannomas.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Neoplasia Endocrina Múltiple Tipo 1 , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/epidemiología , Adolescente , Niño , Neoplasias del Sistema Nervioso Central/epidemiología , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/patología , Incidencia , Adulto Joven , Estudios de Cohortes , Preescolar , Anciano , Meningioma/genética , Meningioma/epidemiología , Meningioma/patología , Francia/epidemiología , Lactante , Ependimoma/genética , Ependimoma/epidemiología , Ependimoma/patología , Mutación , Sistema de Registros
2.
Cancers (Basel) ; 14(18)2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36139608

RESUMEN

Recurrent or high-grade meningiomas are an unmet medical need. Recently, we demonstrated that targeting mTOR by everolimus was relevant both in vitro and in humans. However, everolimus induces an AKT activation that may impact the anti-proliferative effect of the drug. Moreover, the MAP kinase pathway was shown to be involved in meningioma tumorigenesis. We therefore targeted both the Pi3k-AKT-mTOR and MAP kinase pathways by using combinations of the Pi3k inhibitor alpelisib and the MEK inhibitor trametinib. Our study was performed in vitro on the human meningioma cell lines and on a large series of primary cultures providing from 63 freshly operated meningiomas including 35 WHO grade 1, 23 grade 2, and five grade 3, half of which presented a NF2 genomic alteration. Alpelisib induced a higher inhibitory effect on cell viability and proliferation than everolimus in all cell lines and 32 randomly selected tumors no matter the genomic status, the histological subtype or grade. Trametinib also strongly inhibited cell proliferation and induced AKT activation. Combined treatment with alpelisib plus trametinib reversed the AKT activation induced by trametinib and induced an additive inhibitory effect irrespective of the cell lines or tumor features. Co-targeting pathways seems promising and may be considered particularly for aggressive meningioma.

3.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299200

RESUMEN

Forty percent of somatotroph tumors harbor recurrent activating GNAS mutations, historically called the gsp oncogene. In gsp-negative somatotroph tumors, GNAS expression itself is highly variable; those with GNAS overexpression most resemble phenotypically those carrying the gsp oncogene. GNAS is monoallelically expressed in the normal pituitary due to methylation-based imprinting. We hypothesize that changes in GNAS imprinting of gsp-negative tumors affect GNAS expression levels and tumorigenesis. We characterized the GNAS locus in two independent somatotroph tumor cohorts: one of 23 tumors previously published (PMID: 31883967) and classified by pan-genomic analysis, and a second with 82 tumors. Multi-omics analysis of the first cohort identified a significant difference between gsp-negative and gsp-positive tumors in the methylation index at the known differentially methylated region (DMR) of the GNAS A/B transcript promoter, which was confirmed in the larger series of 82 tumors. GNAS allelic expression was analyzed using a polymorphic Fok1 cleavage site in 32 heterozygous gsp-negative tumors. GNAS expression was significantly reduced in the 14 tumors with relaxed GNAS imprinting and biallelic expression, compared to 18 tumors with monoallelic expression. Tumors with relaxed GNAS imprinting showed significantly lower SSTR2 and AIP expression levels. Altered A/B DMR methylation was found exclusively in gsp-negative somatotroph tumors. 43% of gsp-negative tumors showed GNAS imprinting relaxation, which correlated with lower GNAS, SSTR2 and AIP expression, indicating lower sensitivity to somatostatin analogues and potentially aggressive behavior.


Asunto(s)
Cromograninas/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Neoplasias Hipofisarias/genética , Somatotrofos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Cromograninas/metabolismo , Metilación de ADN , Epigénesis Genética , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación Neoplásica de la Expresión Génica , Impresión Genómica , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Somatotrofos/patología , Adulto Joven
4.
Endocr Relat Cancer ; 28(1): 39-51, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112824

RESUMEN

Although there is evidence of a significant rise of neuroendocrine neoplasms (NENs) incidence, current treatments are largely insufficient due to somewhat poor knowledge of these tumours. Despite showing differentiated features, NENs exhibit therapeutic resistance to most common treatments, similar to other cancers in many instances. Molecular mechanisms responsible for this resistance phenomenon are badly understood. We aimed at identifying signalling partners responsible of acquired resistance to treatments in order to develop novel therapeutic strategies. We engineered QGP-1 cells resistant to current leading treatments, the chemotherapeutic agent oxaliplatin and the mTor inhibitor everolimus. Cells were chronically exposed to the drugs and assessed for acquired resistance by viability assay. We used microarray-based kinomics to obtain highthroughput kinase activity profiles from drug sensitive vs resistant cells and identified 'hit' kinases hyperactivated in drug-resistant cells, including kinases from FGFR family, cyclin-dependant kinases and PKCs in oxaliplatin-resistant (R-Ox) QGP-1 cells. We then validated these 'hit' kinases and observed that ERK signalling is specifically enhanced in QGP-1 R-Ox cells. Finally, we assessed drug-resistant cells sensitivity to pharmacological inhibition of 'hit' kinases or their signalling partners. We found that FGFR inhibition markedly decreased ERK signalling and cell viability in QGP-1 R-Ox cells. These results suggest that the FGFR/ERK axis is hyperactivated in response to oxaliplatin-based chemotherapeutic strategy. Thus, this sensitive approach, based on the study of kinome activity, allows identifying potential candidates involved in drug resistance in NENs and may be used to broadly investigate markers of NENs therapeutic response.


Asunto(s)
Antineoplásicos/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Proteómica/métodos , Anciano , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Persona de Mediana Edad
5.
Eur J Med Genet ; 63(12): 104072, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32987185

RESUMEN

SHANK2 code a scaffolding protein located at the postsynaptic membrane of glutamatergic neurons. To date, only nine patients were reported with a SHANK2-variation or microdeletion. Molecular anomalies were identified through screening of large cohorts of patients, but only poor patient clinical descriptions were available. However, this information is crucial for patient care. Here, we describe two additional unrelated patients carrying a SHANK2 de novo variant, improving the delineation of the condition. Phenotypic analysis of these 11 patients identified as major features of the condition: mild to moderate intellectual disability, speech delay, poor language skills, and autism spectrum disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Fenotipo , Trastorno del Espectro Autista/patología , Niño , Discapacidades del Desarrollo/patología , Femenino , Humanos , Discapacidad Intelectual/patología , Desarrollo del Lenguaje , Masculino , Mutación
7.
Clin Cancer Res ; 26(3): 552-557, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969329

RESUMEN

PURPOSE: Aggressive meningiomas that progress after surgery/radiotherapy represent an unmet medical need. Strong and constant expression of SSTR2A receptors and activation of the Pi3K/Akt/mTOR pathway have been demonstrated in meningiomas. The combination of everolimus, an mTOR inhibitor, and octreotide, a somatostatin agonist, has shown additive antitumor effect in vitro. The phase II CEVOREM trial investigated the efficacy of this combination on recurrent meningiomas. PATIENTS AND METHODS: Patients with documented recurrent tumor progression ineligible for further surgery/radiotherapy were eligible to receive octreotide (30 mg/d, day 1) and everolimus (10 mg/d, days 1-28). The primary endpoint was the 6-month progression-free survival rate (PFS6). The secondary endpoints were overall survival, response rate, tumor growth rate according to central review, and safety. RESULTS: A total of 20 patients were enrolled, including 2 with World Health Organization (WHO) grade I tumors, 10 with WHO grade II tumors, and 8 with WHO grade III tumors; furthermore, 4 patients harbored NF2 germline mutation. The overall PFS6 was 55% [95% confidence interval (CI), 31.3%-73.5%], and overall 6- and 12-month survival rates were 90% (95% CI, 65.6%-97.4%) and 75% (95% CI, 50.0%-88.7%), respectively. A major decrease (>50%) was observed in the growth rate at 3 months in 78% of tumors. The median tumor growth rate decreased from 16.6%/3 months before inclusion to 0.02%/3 months at 3 months (P < 0.0002) and 0.48%/3 months at 6 months after treatment (P < 0.0003). CONCLUSIONS: The combination of everolimus and octreotide was associated with clinical and radiological activity in aggressive meningiomas and warrants further studies. Decrease in the tumor volume growth rate should be considered a complementary and sensitive endpoint to select potentially effective drugs for recurrent meningiomas.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Meníngeas/tratamiento farmacológico , Meningioma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores de Somatostatina/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Carga Tumoral/efectos de los fármacos , Adulto , Anciano , Everolimus/administración & dosificación , Femenino , Humanos , Masculino , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Meningioma/metabolismo , Meningioma/patología , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Octreótido/administración & dosificación , Seguridad del Paciente , Estudios Prospectivos , Tasa de Supervivencia , Resultado del Tratamiento
8.
Pharmacogenomics ; 20(13): 931-938, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31486738

RESUMEN

Dihydropryimidine dehydrogenase (DPD) deficiency is a pharmacogenetic syndrome associated with severe or lethal toxicities with oral capecitabine. Usually, patients with history of 5-FU-based therapy with no signs for life-threatening toxicities are considered as not DPD-deficient individuals who can be safely treated next with capecitabine if required. Here we describe the case of a woman originally treated with standard FEC100 protocol for metastatic breast cancer with little severe toxicities but grade-3 mucosities that were quickly resolved by symptomatic treatment. When switched to capecitabine + vinorelbine combo, extremely severe toxicities with fatal outcome were unexpectedly observed. Pharmacogenetic investigations were performed on cytidine deaminase and DPYD, and showed that this patient was heterozygous for the 2846A>T mutation on the DPYD gene. DPD phenotyping (i.e., uracil plasma levels >250 ng/ml, dihydrouracil/uracil ratio <0.5) confirmed that this patient was profoundly DPD deficient. Differences in fluoropyrimidine dosing between FEC100 (i.e., 500 mg/m2 5-FU) and capecitabine (i.e., 2250 mg daily) could explain why initial 5-FU-based protocol did not lead to life-threatening toxicities, whereas capecitabine rapidly triggered toxic death. Overall, this case report suggests that any toxicity, even when not life threatening, should be considered as a warning signal for possible underlying profound DPD deficiency syndrome, especially with low-dose protocols.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Capecitabina/efectos adversos , Deficiencia de Dihidropirimidina Deshidrogenasa/inducido químicamente , Fluorouracilo/uso terapéutico , Antimetabolitos Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Capecitabina/administración & dosificación , Deficiencia de Dihidropirimidina Deshidrogenasa/metabolismo , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Femenino , Humanos , Persona de Mediana Edad
10.
Blood Adv ; 2(5): 462-469, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29490977

RESUMEN

Cytarabine (Ara-C) is the backbone of acute myeloid leukemia (AML) chemotherapy. Little is known about possible risk factors predictive for the frequent (ie, up to 16%) life-threatening or lethal toxicities caused by Ara-C. Ara-C is detoxified in the liver by a single enzyme, cytidine deaminase (CDA), coded by a gene known to be highly polymorphic. In this proof-of-concept study, we particularly investigated the role of the CDA poor metabolizer (PM) phenotype in Ara-C toxicities. CDA phenotyping (measurement of CDA residual activity in serum) and genotyping (search for the CDA*2 allelic variant) were performed in 58 adult patients with AML treated with the standard 7+3 (Ara-C + anthracyclines) protocol. Statistically significantly lower CDA activity was observed in patients experiencing severe/lethal toxicities as compared with patients who did not (1.5 ± 0.7 U/mg vs 3.95 ± 3.1 U/mg; Student t test P < .001). Subsequent receiver operating characteristic analysis identified a threshold in CDA activity (ie, 2 U/mg) associated with PM syndrome and increased risk of developing severe toxicities. Five percent of patients experienced lethal toxicities, all displaying CDA PM status (1.3 ± 0.5 U/mg). In terms of efficacy, a trend toward higher response rates and longer progression-free survival and overall survival were observed in patients with low CDA activity. Taken together, the results of this study strongly suggest that CDA is a predictive marker of life-threatening toxicities in patients with AML receiving induction therapy with standard Ara-C.


Asunto(s)
Citarabina/toxicidad , Citidina Desaminasa/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/uso terapéutico , Antimetabolitos Antineoplásicos/toxicidad , Biomarcadores/análisis , Citarabina/uso terapéutico , Regulación hacia Abajo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Adulto Joven
11.
Oncotarget ; 8(25): 41044-41063, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28454119

RESUMEN

Therapeutic management of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) is challenging. The mammalian target of rapamycin (mTOR) inhibitor everolimus recently obtained approval from the Food and Drug Administration for the treatment of patients with advanced pancreatic neuroendocrine tumors (pNETs). Despite its promising antitumor efficacy observed in cell lines, clinical benefit for patients is unsatisfactory. The limited therapeutic potential of everolimus in cancer cells has been attributed to Akt activation due to feedback loops relief following mTOR inhibition. Combined inhibition of Akt might then improve everolimus antitumoral effect. In this regard, the somatostatin analog (SSA) octreotide has been shown to repress the PI3K/Akt pathway in some tumor cell lines. Moreover, SSAs are well tolerated and routinely used to reduce symptoms caused by peptide release in patients carrying functional GEP-NETs. We have recently established and characterized primary cultures of human pNETs and demonstrated the anti-proliferative effects of both octreotide and pasireotide. In this study, we aim at determining the antitumor efficacy of everolimus alone or in combination with the SSAs octreotide and pasireotide in primary cultures of pNETs. Everolimus reduced both Chromogranin A secretion and cell viability and upregulated Akt activity in single treatment. Its anti-proliferative and anti-secretory efficacy was not improved combined with the SSAs. Both SSAs did not overcome everolimus-induced Akt upregulation. Furthermore, caspase-dependent apoptosis induced by SSAs was lost in combined treatments. These molecular events provide the first evidence supporting the lack of marked benefit in patients co-treated with everolimus and SSA.


Asunto(s)
Cromogranina A/metabolismo , Everolimus/farmacología , Octreótido/farmacología , Somatostatina/análogos & derivados , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
12.
Endocr Relat Cancer ; 23(7): 509-19, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27267119

RESUMEN

Pegvisomant (PEG), an antagonist of growth hormone (GH)-receptor (GHR), normalizes insulin-like growth factor 1 (IGF1) oversecretion in most acromegalic patients unresponsive to somatostatin analogs (SSAs) and/or uncontrolled by transsphenoidal surgery. The residual GH-secreting tumor is therefore exposed to the action of circulating PEG. However, the biological effect of PEG at the pituitary level remains unknown. To assess the impact of PEG in vitro on the hormonal secretion (GH and prolactin (PRL)), proliferation and cellular viability of eight human GH-secreting tumors in primary cultures and of the rat somatolactotroph cell line GH4C1. We found that the mRNA expression levels of GHR were characterized in 31 human GH-secreting adenomas (0.086 copy/copy ß-Gus) and the GHR was identified by immunocytochemistry staining. In 5/8 adenomas, a dose-dependent inhibition of GH secretion was observed under PEG with a maximum of 38.2±17% at 1µg/mL (P<0.0001 vs control). A dose-dependent inhibition of PRL secretion occurred in three mixed GH/PRL adenomas under PEG with a maximum of 52.8±11.5% at 10µg/mL (P<0.0001 vs control). No impact on proliferation of either human primary tumors or GH4C1 cell line was observed. We conclude that PEG inhibits the secretion of GH and PRL in primary cultures of human GH(/PRL)-secreting pituitary adenomas without effect on cell viability or cell proliferation.


Asunto(s)
Adenoma/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/análogos & derivados , Prolactina/metabolismo , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Hormona de Crecimiento Humana/farmacología , Humanos , Janus Quinasa 2/metabolismo , Masculino , Persona de Mediana Edad , Ratas , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/metabolismo , Células Tumorales Cultivadas , Adulto Joven
13.
PLoS One ; 10(3): e0120010, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25822178

RESUMEN

To test the role of wtPIT-1 (PITWT) or PIT-1 (R271W) (PIT271) in somatolactotroph cells, we established, using inducible lentiviral vectors, sublines of GH4C1 somatotroph cells that allow the blockade of the expression of endogenous PIT-1 and/or the expression of PITWT or PIT271, a dominant negative mutant of PIT-1 responsible for Combined Pituitary Hormone Deficiency in patients. Blocking expression of endogenous PIT-1 induced a marked decrease of cell proliferation. Overexpressing PITWT twofold led also to a dose-dependent decrease of cell proliferation that was accompanied by cell death. Expression of PIT271 induced a strong dose-dependent decrease of cell proliferation accompanied by a very pronounced cell death. These actions of PIT271 are independent of its interaction/competition with endogenous PIT-1, as they were unchanged when expression of endogenous PIT-1 was blocked. All these actions are specific for somatolactotroph cells, and could not be observed in heterologous cells. Cell death induced by PITWT or by PIT271 was accompanied by DNA fragmentation, but was not inhibited by inhibitors of caspases, autophagy or necrosis, suggesting that this cell death is a caspase-independent apoptosis. Altogether, our results indicate that under normal conditions PIT-1 is important for the maintenance of cell proliferation, while when expressed at supra-normal levels it induces cell death. Through this dual action, PIT-1 may play a role in the expansion/regression cycles of pituitary lactotroph population during and after lactation. Our results also demonstrate that the so-called "dominant-negative" action of PIT271 is independent of its competition with PIT-1 or a blockade of the actions of the latter, and are actions specific to this mutant variant of PIT-1.


Asunto(s)
Somatotrofos/fisiología , Factor de Transcripción Pit-1/fisiología , Adenoma/genética , Adenoma/patología , Adenoma/fisiopatología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Apoptosis/fisiología , Secuencia de Bases , Línea Celular , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Chlorocebus aethiops , Humanos , Lactotrofos/citología , Lactotrofos/fisiología , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Somatotrofos/citología , Factor de Transcripción Pit-1/antagonistas & inhibidores , Factor de Transcripción Pit-1/genética , Células Tumorales Cultivadas
14.
Gastroenterology ; 148(7): 1452-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683115

RESUMEN

BACKGROUND & AIMS: The KRAS gene is mutated in most pancreatic ductal adenocarcinomas (PDAC). Expression of this KRAS oncoprotein in mice is sufficient to initiate carcinogenesis but not progression to cancer. Activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) is required for KRAS for induction and maintenance of PDAC in mice. The somatostatin receptor subtype 2 (sst2) inhibits PI3K, but sst2 expression is lost during the development of human PDAC. We investigated the effects of sst2 loss during KRAS-induced PDAC development in mice. METHODS: We analyzed tumor growth in mice that expressed the oncogenic form of KRAS (KRAS(G12D)) in pancreatic precursor cells, as well as sst2+/- and sst2-/-, and in crossed KRAS(G12D);sst2+/- and KRAS(G12D);sst2-/- mice. Pancreatic tissues and acini were collected and assessed by histologic, immunoblot, immunohistochemical, and reverse-transcription polymerase chain reaction analyses. We also compared protein levels in paraffin-embedded PDAC samples from patients vs heathy pancreatic tissues from individuals without pancreatic cancer. RESULTS: In sst2+/- mice, PI3K was activated and signaled via AKT (PKB; protein kinase B); when these mice were crossed with KRAS(G12D) mice, premalignant lesions, tumors, and lymph node metastases developed more rapidly than in KRAS(G12D) mice. In crossed KRAS(G12D);sst2+/- mice, activation of PI3K signaling via AKT resulted in activation of nuclear factor-κB (NF-κB), which increased KRAS activity and its downstream pathways, promoting initiation and progression of neoplastic lesions. We found this activation loop to be mediated by PI3K-induced production of the chemokine CXCL16. Administration of a CXCL16-neutralizing antibody to KRAS(G12D) mice reduced activation of PI3K signaling to AKT and NF-κB, blocking carcinogenesis. Levels of CXCL16 and its receptor CXCR6 were significantly higher in PDAC tissues and surrounding acini than in healthy pancreatic tissues from mice or human beings. In addition, expression of sst2 was progressively lost, involving increased PI3K activity, in mouse lesions that expressed KRAS(G12D) and progressed to PDAC. CONCLUSIONS: Based on analyses of mice, loss of sst2 from pancreatic tissues activates PI3K signaling via AKT, leading to activation of NF-κB, amplification of oncogenic KRAS signaling, increased expression of CXCL16, and pancreatic tumor formation. CXCL16 might be a therapeutic target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Proliferación Celular , Quimiocina CXCL6/metabolismo , Mutación , Neoplasias Pancreáticas/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Somatostatina/deficiencia , Transducción de Señal , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/secundario , Estudios de Casos y Controles , Línea Celular Tumoral , Quimiocina CXCL16 , Quimiocinas CXC/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Metástasis Linfática , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Depuradores/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba
15.
Mol Cell Endocrinol ; 355(1): 106-13, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22348806

RESUMEN

OBJECTIVE: As prolactinomas fail to respond to dopamine agonist (DA) in 10-20% of cases, we hypothesized that somatostatin subtype 2 receptor (sst2) overexpression in DA-resistant prolactinomas may enhance suppression of prolactine (PRL) using chimeric agonist (dopastatin) that simultaneously binds sst2 and the dopamine subtype 2 receptor (D2DR). DESIGN AND METHODS: PRL suppression by octreotide, sst5 agonist, sst2-D2DR agonist (BIM-23A760 dopastatin) and cabergoline was assessed in primary cultures of seven DA-resistant prolactinomas overexpressing sst2. RESULTS: sst2 was effectively overexpressed via adenoviral expression in prolactinomas (38.1±7.4 vs. 0.1±0.1 copy/copy ß-Gus) and induced octreotide sst2-mediated PRL suppression that remained lower than that induced by DA. BIM-23A760 inhibited PRL similarly to cabergoline both in the control and sst2-expressing cells. Antagonist experiments confirmed predominant dopaminergic effect in dopastatin activity. CONCLUSION: sst2 was successfully overexpressed in prolactinomas. However BIM-23A760 was unable to enhance PRL suppression underlining a predominant dopaminergic contribution in its action.


Asunto(s)
Agonistas de Dopamina/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactina/antagonistas & inhibidores , Prolactinoma/tratamiento farmacológico , Receptores de Somatostatina/metabolismo , Adenoviridae , Adolescente , Adulto , Cabergolina , Dopamina/análogos & derivados , Dopamina/metabolismo , Dopamina/farmacología , Ergolinas/farmacología , Femenino , Vectores Genéticos , Humanos , Masculino , Octreótido/farmacología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Cultivo Primario de Células , Prolactina/genética , Prolactinoma/metabolismo , Prolactinoma/patología , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/genética , Somatostatina/análogos & derivados , Somatostatina/farmacología , Transfección
16.
Hum Gene Ther ; 23(1): 104-14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21942649

RESUMEN

The treatment of growth hormone (GH)- and prolactin (PRL)-secreting tumors resistant to current therapeutic molecules (somatostatin and dopamine analogues) remains challenging. To target these tumors specifically, we chose to inactivate a gene coding for a crucial factor in cell proliferation and hormonal regulation, specifically expressed in pituitary, by using a dominant-negative form of this gene involved in human pituitary deficiencies: transcription factor Pit-1 (POU1F1) mutated on arginine 271 to tryptophan (R271W). After lentiviral transfer, the effect of R271W was studied in vitro on human tumoral somatotroph and lactotroph cells and on the murine mammosomatotroph cell line GH4C1 and in vivo on GH4C1 subcutaneous xenografts in nude mice. R271W induced a decrease in GH and PRL hypersecretion by controlling the transcription of the corresponding hormones. This mutant decreased cell viability by an apoptotic mechanism and in vivo blocked the tumoral growth and GH secretion of xenografts obtained after transplantation of GH4C1 expressing mutant R271W. The strategy of using a dominant-negative form of a main factor controlling cell proliferation and hormonal secretion, and exclusively expressed in pituitary, seems promising for the gene therapy of human pituitary tumors and may be translated to other types of tumors maintaining some differentiation features.


Asunto(s)
Terapia Genética/métodos , Prolactina/metabolismo , Factor de Transcripción Pit-1/metabolismo , Activación Transcripcional , Adenoma/genética , Adenoma/metabolismo , Adenoma/terapia , Animales , Arginina/genética , Arginina/metabolismo , Western Blotting , Muerte Celular , Proliferación Celular , Supervivencia Celular , Trasplante de Células/métodos , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Vectores Genéticos/uso terapéutico , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/terapia , Hormona de Crecimiento Humana/análisis , Hormona de Crecimiento Humana/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Desnudos , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/terapia , Prolactina/análisis , Prolactina/sangre , Prolactinoma/genética , Prolactinoma/terapia , Factor de Transcripción Pit-1/genética , Transgenes , Triptófano/genética , Triptófano/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Endocrinology ; 152(10): 3884-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21810944

RESUMEN

Nonfunctioning pituitary adenomas (NFPA; gonadotroph derived), even not inducing hormonal hypersecretion, cause significant morbidity by compression neighboring structures. No effective and specific medical methods are available so far for treating these tumors. The pituitary homeobox 2 (PITX2) gene is a member of the bicoid-like homeobox transcription factor family, which is involved in the Wnt/Dvl/ß-catenin pathway. PITX2 is overexpressed in NFPA. PITX2 mutations are known to be responsible for Axenfield Rieger syndrome, a genetic disorder in which pituitary abnormalities have been detected. The R91P mutant identified in Axenfeld Rieger syndrome is a dominant-negative factor, which is able to block the expression of several pituitary genes activated by PITX2. To better understand the role of Pitx2 on gonadotroph tumorigenesis and to explore new approach for inhibiting tumoral growth, the R91P mutant was transferred via a lentiviral vector in tumoral gonadotroph cells of two kinds: the αT3-1 cell line and human adenoma cells. R91P mutant and small interfering RNA directed against Pitx2 both decreased the viability of αT3-1 cells via an apoptotic mechanism involving the activation of executioner caspase. Similar effects of the R91P mutant were observed on human gonadotroph cells in primary culture. Therefore, Pitx2 overexpression may play an antiapoptotic role during NFPA tumorigenesis.


Asunto(s)
Adenoma/etiología , Gonadotrofos/patología , Proteínas de Homeodominio/fisiología , Neoplasias Hipofisarias/etiología , Factores de Transcripción/fisiología , Adenoma/patología , Animales , Apoptosis , Supervivencia Celular , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Lentivirus/genética , Ratones , Neoplasias Hipofisarias/patología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteína del Homeodomínio PITX2
18.
J Cancer Res Ther ; 5(3): 198-202, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19841562

RESUMEN

BACKGROUND: Germline RET gene mutations are well known to be the genetic causes of multiple endocrine neoplasia type 2 (MEN2) and may be identified by genetic screening. AIM: The purpose of the present study was to screen nine MTC patients for RET sequence changes. MATERIALS AND METHODS: In this study, our sample was composed of 30 individuals: 9 index patients with medullary thyroid carcinoma (MTC) corresponding either to 3 subjects with clinical evidence of MEN2, 6 with apparently sporadic MTC (sMTC), and 21 relatives have been investigated for RET mutations. After DNA extraction from peripheral blood leukocytes, RET exons 8, 10, 11, 13-16 and exon/intron boundaries were analyzed by direct PCR sequencing. RESULTS: Three different known RET germline mutations in exon 11 (codon 634), p.Cys634Arg (c1900 T-->C) (de novo case), p.Cys634Phe (c1901 G-->T), p.Cys634Trp (c1902 C-->G), were detected in three individuals with MEN2 phenotype. Of the 21 relatives, 2 cases presented mutation. Among the six probands with sMTC, none was found to carry mutation. There was no difference between RET polymorphisms detected among both MEN2 and sMTC patients. CONCLUSIONS: These preliminary data suggest that the RET mutation spectra observed in Moroccan patients with MEN2 are similar to those previously reported in other countries.


Asunto(s)
Carcinoma Medular/diagnóstico , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/diagnóstico , Adulto , Carcinoma Medular/genética , Exones , Femenino , Tamización de Portadores Genéticos , Pruebas Genéticas , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Marruecos , Neoplasia Endocrina Múltiple Tipo 2a/diagnóstico , Neoplasia Endocrina Múltiple Tipo 2a/genética , Polimorfismo Genético , Análisis de Secuencia de ADN , Neoplasias de la Tiroides/genética
19.
Nephrol Ther ; 5(1): 41-51, 2009 Feb.
Artículo en Francés | MEDLINE | ID: mdl-18815088

RESUMEN

AIM AND BACKGROUND: To show results of the first year of an infection surveillance network for haemodialysis patients (Dialin). In order to improve the security and quality of care, six haemodialysis centers have organized an infection watching network. The purpose of the network is to compare of the watching results between centers. This comparison includes vascular access infection (VAI), bacteraemia and C viral hepatitis. The heterogeneous pattern has been also taken into account. SURVEY TYPE: Multicenter prospective permanent survey. POPULATION: Six hundred and sixty-four haemodialyzed chronic patients, followed during one year (2005), in six voluntary haemodialysis centers. This survey has based on 71,688 treatment sessions corresponding to 6257.5 months of haemodialysis (HM). METHODS: As with the heterogeneity among centers, the acquired infection standardized ratios (observed/expected) (AISR) and 95% confidence interval are computed with Cox model which includes confounding factors found in literature or in the preliminary stage of the survey. RESULTS: VAI crude rate was 0.47 per 100HM, 0.10 per 1000 native fistulae utilisation days, 0.45 per 1000 days of prosthetic graft utilisation and 0.44 per 1000 days of catheter utilisation. Bacteraemia crude incidence rate was 0.69 per 100HM, 0.02 per 1000 days of native fistulae utilisation, 0.00 per 1000 days of prosthetic graft utilisation and 0.39 per 1000 days of catheter utilisation. No new case of C viral hepatitis was found. Prevalence rate at the beginning of the survey was 5.3% (35 over 664). Two centers had a significantly high AISR for VAI and two centers had a significantly low AISR for VAI. One center had a significantly high AISR for bacteraemia and one center had a significantly low AISR for bacteraemia. CONCLUSIONS: The first year of Dialin running demonstrates the importance of standardised surveillance method in VAI and bacteraemia surveillance but not for viral hepatitis.


Asunto(s)
Infecciones Relacionadas con Catéteres/epidemiología , Vigilancia de la Población , Diálisis Renal/efectos adversos , Anciano , Femenino , Francia , Humanos , Masculino , Estudios Prospectivos , Factores de Riesgo
20.
Cancer Res ; 68(24): 10163-70, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19074883

RESUMEN

In human somatotroph adenomas, growth hormone (GH) hypersecretion can be inhibited by somatostatin analogues such as octreotide. Unfortunately, serum GH levels reach normal values in only 60% of treated patients. The decreased sensitivity to octreotide is strongly related to a lower expression of somatostatin receptor sst2. In this present study, the sst2 gene was transferred by an adenoviral vector (Ad-sst2) in human somatotroph (n = 7) and lactotroph (n = 2) adenomas in vitro. Sst2 mRNA levels and sst2 immunostaining dramatically increased after infection. Ten days after infection at 20 multiplicity of infection (MOI), sst2 gene transfer decreased cell viability from 19% to 90% by caspase-dependent apoptosis. At low viral doses (5 MOI), Ad-sst2 decreased GH or prolactin (PRL) basal secretion and mRNA expression. Somatotroph tumors were classified in three groups according to their octreotide sensitivity. Four days after infection by 5 MOI Ad-sst2, the maximal GH suppression by octreotide increased from 31% to 57% in the octreotide partially resistant group and from 0% to 27% in the resistant ones. In the octreotide-sensitive group, EC(50) values significantly decreased from 1.3 x 10(-11) to 6.6 x 10(-13) mol/L without improving maximal GH suppression. Finally, lactotroph tumors, nonresponding to octreotide in basal conditions, became octreotide sensitive with a maximal PRL suppression of 43% at 10(-8) mol/L. Therefore, sst2 reexpression is able to improve octreotide sensitivity. Sst2 gene transfer may open new therapeutic strategies in treatment combined with somatostatin analogues.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Hormona de Crecimiento Humana/metabolismo , Octreótido/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactina/metabolismo , Prolactinoma/tratamiento farmacológico , Adenoviridae/genética , Supervivencia Celular/fisiología , Resistencia a Antineoplásicos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/biosíntesis , Hormona de Crecimiento Humana/genética , Humanos , Inmunohistoquímica , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactina/biosíntesis , Prolactina/genética , Prolactinoma/genética , Prolactinoma/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Somatostatina/biosíntesis , Receptores de Somatostatina/genética , Transducción Genética , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA