Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299200

ABSTRACT

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.


Subject(s)
Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Pituitary Neoplasms/genetics , Somatotrophs/metabolism , Adult , Aged , Aged, 80 and over , Alleles , Chromogranins/metabolism , DNA Methylation , Epigenesis, Genetic , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation, Neoplastic , Genomic Imprinting , Humans , Male , Middle Aged , Mutation , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Somatotrophs/pathology , Young Adult
2.
J Pediatr ; 205: 281-285.e4, 2019 02.
Article in English | MEDLINE | ID: mdl-30442414

ABSTRACT

The GNAS postzygotic mosaic activating mutations involved in fibrous dysplasia and McCune-Albright syndrome (MAS) are not detectable in leukocytes by Sanger sequencing. Digital droplet polymerase chain reaction detects GNAS mutations in 7 of 12 patients (58.3%) suspected to have fibrous dysplasia/MAS from whole blood DNA, and in 4 of 5 patients (80%) from circulating cell-free DNA.


Subject(s)
Cell-Free Nucleic Acids/genetics , Chromogranins/genetics , DNA/genetics , Fibrous Dysplasia, Polyostotic/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Polymerase Chain Reaction/methods , Adolescent , Adult , Aged , Cell-Free Nucleic Acids/blood , Child , Child, Preschool , Chromogranins/metabolism , DNA/blood , DNA Mutational Analysis/methods , Female , Fibrous Dysplasia, Polyostotic/blood , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Infant , Male , Middle Aged , Young Adult
4.
Gastroenterology ; 148(7): 1452-65, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25683115

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Proliferation , Chemokine CXCL6/metabolism , Mutation , Pancreatic Neoplasms/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Somatostatin/deficiency , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/secondary , Case-Control Studies , Cell Line, Tumor , Chemokine CXCL16 , Chemokines, CXC/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Lymphatic Metastasis , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Scavenger/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Time Factors , Transfection , Tumor Burden , Up-Regulation
5.
Clin Cancer Res ; 30(13): 2835-2845, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38630553

ABSTRACT

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.


Subject(s)
Central Nervous System Neoplasms , Multiple Endocrine Neoplasia Type 1 , Humans , Male , Female , Adult , Middle Aged , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/epidemiology , Adolescent , Child , Central Nervous System Neoplasms/epidemiology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Incidence , Young Adult , Cohort Studies , Child, Preschool , Aged , Meningioma/genetics , Meningioma/epidemiology , Meningioma/pathology , France/epidemiology , Infant , Ependymoma/genetics , Ependymoma/epidemiology , Ependymoma/pathology , Mutation , Registries
6.
Cancers (Basel) ; 14(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36139608

ABSTRACT

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.

7.
Endocr Relat Cancer ; 28(1): 39-51, 2021 01.
Article in English | MEDLINE | ID: mdl-33112824

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Proteomics/methods , Aged , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged
8.
Eur J Med Genet ; 63(12): 104072, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32987185

ABSTRACT

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.


Subject(s)
Autism Spectrum Disorder/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Phenotype , Autism Spectrum Disorder/pathology , Child , Developmental Disabilities/pathology , Female , Humans , Intellectual Disability/pathology , Language Development , Male , Mutation
9.
Clin Cancer Res ; 26(3): 552-557, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31969329

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Receptors, Somatostatin/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Burden/drug effects , Adult , Aged , Everolimus/administration & dosage , Female , Humans , Male , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningioma/metabolism , Meningioma/pathology , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Octreotide/administration & dosage , Patient Safety , Prospective Studies , Survival Rate , Treatment Outcome
10.
Nephrol Ther ; 5(1): 41-51, 2009 Feb.
Article in French | MEDLINE | ID: mdl-18815088

ABSTRACT

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.


Subject(s)
Catheter-Related Infections/epidemiology , Population Surveillance , Renal Dialysis/adverse effects , Aged , Female , France , Humans , Male , Prospective Studies , Risk Factors
11.
Pharmacogenomics ; 20(13): 931-938, 2019 08.
Article in English | MEDLINE | ID: mdl-31486738

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Capecitabine/adverse effects , Dihydropyrimidine Dehydrogenase Deficiency/chemically induced , Fluorouracil/therapeutic use , Antimetabolites, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Capecitabine/administration & dosage , Dihydropyrimidine Dehydrogenase Deficiency/metabolism , Dihydrouracil Dehydrogenase (NADP)/metabolism , Female , Humans , Middle Aged
12.
Blood Adv ; 2(5): 462-469, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29490977

ABSTRACT

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.


Subject(s)
Cytarabine/toxicity , Cytidine Deaminase/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/toxicity , Biomarkers/analysis , Cytarabine/therapeutic use , Down-Regulation , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Young Adult
13.
Oncotarget ; 8(25): 41044-41063, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28454119

ABSTRACT

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.


Subject(s)
Chromogranin A/metabolism , Everolimus/pharmacology , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Somatostatin/pharmacology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
14.
Haematologica ; 91(2): 162-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16461299

ABSTRACT

BACKGROUND AND OBJECTIVES: Imatinib mesylate induces a complete cytogenetic response (CCR) in many patients with chronic myeloid leukemia (CML). However, the ultimate goal of therapy for CML is complete elimination of Philadelphia chromosome positive cells or BCR-ABL rearrangements. We studied molecular responses in CML patients in CCR after imatinib treatment. DESIGN AND METHODS: Real-time quantitative reverse transcriptase polymerase chain reaction analysis were used to monitor BCR-ABL levels in 59 CCR patients. Negative results were confirmed by two different techniques performed in two different laboratories. Patients were considered in complete molecular remission if they had four undetectable analyses from two separate samples taken three months apart. RESULTS: The median follow-up was 41 months (17-53). The median BCR-ABL/ABL ratio at the time of CCR was 0.3 % (0-9.88). Patients were split into two groups: group A (n=43) comprised patients with a detectable BCR-ABL/ABL ratio throughout the follow-up and group B (n=16) included those with an undetectable level of BCR-ABL/ABL (< 10(-5)) i.e. in complete molecular remission. No relapses were observed in group B, while 13 group A patients lost their CCR. The probability of losing CCR in this group was 33.2 % >+/-18.0. By Cox regression analysis the best factor for predicting the probability of achieving molecular remission was having a CCR at 6 months (p=0.038) or at 3 months (p=0.024). INTERPRETATION AND CONCLUSIONS: Molecular remission after imatinib treatment, i.e. BCR-ABL/ABL< 10-5 in peripheral blood, is not a rare event, particularly in patients achieving CCR at 6 months.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Adult , Aged , Aged, 80 and over , Benzamides , Cytogenetic Analysis , Female , Fusion Proteins, bcr-abl/analysis , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Male , Middle Aged , Polymerase Chain Reaction , Remission Induction/methods
15.
Endocr Relat Cancer ; 23(7): 509-19, 2016 07.
Article in English | MEDLINE | ID: mdl-27267119

ABSTRACT

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.


Subject(s)
Adenoma/metabolism , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Growth Hormone/metabolism , Human Growth Hormone/analogs & derivatives , Prolactin/metabolism , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Human Growth Hormone/pharmacology , Humans , Janus Kinase 2/metabolism , Male , Middle Aged , Rats , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/metabolism , Tumor Cells, Cultured , Young Adult
16.
PLoS One ; 10(3): e0120010, 2015.
Article in English | MEDLINE | ID: mdl-25822178

ABSTRACT

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.


Subject(s)
Somatotrophs/physiology , Transcription Factor Pit-1/physiology , Adenoma/genetics , Adenoma/pathology , Adenoma/physiopathology , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis/physiology , Base Sequence , Cell Line , Cell Proliferation/physiology , Cell Survival/genetics , Cell Survival/physiology , Chlorocebus aethiops , Humans , Lactotrophs/cytology , Lactotrophs/physiology , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/physiology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Somatotrophs/cytology , Transcription Factor Pit-1/antagonists & inhibitors , Transcription Factor Pit-1/genetics , Tumor Cells, Cultured
18.
J Endocrinol ; 183(1): 217-33, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15525589

ABSTRACT

Despite important advances in human therapeutics, no specific treatment for both non-functioning gonadotroph and resistant somatotroph adenomas is available. Gene transfer by viral vectors can be considered as a promising way to achieve a specific and efficient treatment. Here we show the possibility of efficient gene transfer in human pituitary adenoma cells in vitro using a human immunodeficiency virus (HIV)-type 1-derived vector. Using enhanced green fluorescent protein (eGFP) gene as a marker placed under the phosphoglycerate kinase (PGK) promoter, gonadotroph and somatotroph adenomas were transduced even with moderate viral loads. The expression started at day 2, reached a peak at day 5, and it was still present at day 90. For targeting somatotroph and gonadotroph adenomas, human growth hormone (GH) promoter (GH -481, +54 bp) and two fragments of the human glycoprotein hormone alpha-subunit promoter (alpha-subunit 1 -520, +33 bp, and alpha-subunit 2 -907, +33 bp) were tested. In gonadotroph adenomas, the percentage of identified fluorescent cells and the fluorescence intensity analyzed by fluorescence-activated cell sorting indicated that the strength of the alpha-subunit 1 and alpha-subunit 2 promoters were comparable to that of the PGK promoter. Primary cultures of rat pituitary cells showed that alpha-subunit 1 is more selective to thyreotroph and gonadotroph phenotypes than alpha-subunit 2. GH promoter activity appeared weak in somatotroph adenomas. The human GH enhancer did not increase the GH promoter activity at all but the human prolactin promoter (-250 bp) allowed 4-fold more fluorescent cells to be obtained than the GH promoter. Several cell lines appeared too permissive to test cell-specificity of pituitary promoters. However, on human non-pituitary cell cultures, the tested pituitary promoters seemed clearly selective to target endocrine pituitary phenotypes. This study gives a starting point for a gene-therapy program using lentiviral vectors to transfer therapeutic genes in human pituitary adenomas.


Subject(s)
Adenoma/therapy , Genetic Therapy/methods , Glycoprotein Hormones, alpha Subunit/genetics , HIV-1/genetics , Pituitary Neoplasms/therapy , Promoter Regions, Genetic , Adenoma/metabolism , Adenoma/virology , Adult , Aged , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression , Genetic Engineering , Genetic Vectors/therapeutic use , Gonadotropins, Pituitary/genetics , Gonadotropins, Pituitary/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Hormone/genetics , Growth Hormone/metabolism , Humans , Male , Microscopy, Fluorescence , Middle Aged , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/virology , Prolactin/genetics , Prolactin/metabolism , Transduction, Genetic/methods
19.
Mol Cell Endocrinol ; 355(1): 106-13, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22348806

ABSTRACT

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.


Subject(s)
Dopamine Agonists/pharmacology , Pituitary Neoplasms/drug therapy , Prolactin/antagonists & inhibitors , Prolactinoma/drug therapy , Receptors, Somatostatin/metabolism , Adenoviridae , Adolescent , Adult , Cabergoline , Dopamine/analogs & derivatives , Dopamine/metabolism , Dopamine/pharmacology , Ergolines/pharmacology , Female , Genetic Vectors , Humans , Male , Octreotide/pharmacology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Primary Cell Culture , Prolactin/genetics , Prolactinoma/metabolism , Prolactinoma/pathology , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Transfection
20.
Hum Gene Ther ; 23(1): 104-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21942649

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Prolactin/metabolism , Transcription Factor Pit-1/metabolism , Transcriptional Activation , Adenoma/genetics , Adenoma/metabolism , Adenoma/therapy , Animals , Arginine/genetics , Arginine/metabolism , Blotting, Western , Cell Death , Cell Proliferation , Cell Survival , Cell Transplantation/methods , Female , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genetic Vectors/therapeutic use , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Hormone-Secreting Pituitary Adenoma/genetics , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Growth Hormone-Secreting Pituitary Adenoma/therapy , Human Growth Hormone/analysis , Human Growth Hormone/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Nude , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/therapy , Prolactin/analysis , Prolactin/blood , Prolactinoma/genetics , Prolactinoma/therapy , Transcription Factor Pit-1/genetics , Transgenes , Tryptophan/genetics , Tryptophan/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL