Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Hum Mol Genet ; 26(18): 3495-3507, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28911199

ABSTRACT

Inactivating mutations in the Armadillo repeat-containing 5 (ARMC5) gene have recently been discovered in primary macronodular adrenal hyperplasia (PMAH), a cause of Cushing syndrome. Biallelic ARMC5 inactivation in PMAH suggested that ARMC5 may have tumor suppressor functions in the adrenal cortex. We generated and characterized a new mouse model of Armc5 deficiency. Almost all Armc5 knockout mice died during early embryonic development, around 6.5 and 8.5 days. Knockout embryos did not undergo gastrulation, as demonstrated by the absence of mesoderm development at E7.5. Armc5 heterozygote mice (Armc5+/-) developed normally but at the age of 1 year, their corticosterone levels decreased; this was associated with a decrease of protein kinase A (PKA) catalytic subunit α (Cα) expression both at the RNA and protein levels that were also seen in human patients with PMAH and ARMC5 defects. However, this was transient, as corticosterone levels normalized later, followed by the development of hypercorticosteronemia in one-third of the mice at 18 months of age, which was associated with increases in PKA and Cα expression. Adrenocortical tissue analysis from Armc5+/- mice at 18 months showed an abnormal activation of the Wnt/ß-catenin signaling pathway in a subset of zona fasciculata cells. These data confirm that Armc5 plays an important role in early mouse embryonic development. Our new mouse line can be used to study tissue-specific effects of Armc5. Finally, Armc5 haploinsufficiency leads to Cushing syndrome in mice, but only later in life, and this involves PKA, its catalytic subunit Cα, and the Wnt/ß-catenin pathway.


Subject(s)
Adrenocortical Hyperfunction/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Adrenal Cortex/pathology , Adrenal Glands/pathology , Adrenocortical Hyperfunction/metabolism , Adrenocortical Hyperfunction/pathology , Age Factors , Animals , Armadillo Domain Proteins , Corticosterone , Cushing Syndrome/metabolism , Disease Models, Animal , Female , Germ-Line Mutation , Haploinsufficiency , Humans , Hyperplasia/metabolism , Male , Mice , Mice, Knockout , Mutation , Pituitary ACTH Hypersecretion/metabolism , beta Catenin/genetics , beta Catenin/metabolism
2.
Ann Oncol ; 26(10): 2119-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26392430

ABSTRACT

BACKGROUND: The clinical course of advanced adrenocortical carcinoma (ACC) is heterogeneous. Our study aimed primarily to refine and make headway in the prognostic stratification of advanced ACC. PATIENTS AND METHODS: Patients with advanced ENSAT ACC (stage III or stage IV) at diagnosis registered between 2000 and 2009 in the ENSAT database were enrolled. The primary end point was overall survival (OS). Parameters of potential prognostic relevance were selected. Univariate and multivariate analyses were carried out: model 1 'before surgery'; model 2 'post-surgery'. RESULTS: Four hundred and forty-four patients with advanced ENSAT ACC (stage III: 210; stage IV: 234) were analyzed. After a median follow-up of 55.2 months, the median OS was 24 months. A modified ENSAT (mENSAT) classification was validated: stage III (invasion of surrounding tissues/organs or the vena renalis/cava) and stage IVa, IVb, IVc (2, 3 or >3 metastatic organs, including N, respectively). Two- or 5-year OS was 73%, 46%, 26% and 15% or 50%, 15%, 14% and 2% for stages III, IVa, IVb and IVc, respectively. In the multivariate analysis, mENSAT stages (stages IVa, IVb, or IVc, respectively) were significantly correlated with OS (P < 0.0001), as well as additional parameters: age ≥ 50 years (P < 0.0001), tumor- or hormone-related symptoms (P = 0.01 and 0.03, respectively) in model 1 but also the R status (P = 0.001) and Grade (Weiss >6 and/or Ki67 ≥ 20%, P = 0.06) in model 2. CONCLUSION: The mENSAT classification and GRAS parameters (Grade, R status, Age and Symptoms) were found to best stratify the prognosis of patients with advanced ACC.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Bone Neoplasms/secondary , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Neoplasm Recurrence, Local/pathology , Adrenal Cortex Neoplasms/mortality , Adrenocortical Carcinoma/mortality , Bone Neoplasms/mortality , Europe , Female , Follow-Up Studies , Humans , Liver Neoplasms/mortality , Lung Neoplasms/mortality , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local/mortality , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate
3.
Horm Metab Res ; 46(12): 869-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25105543

ABSTRACT

Stimulation of the cAMP pathway by adrenocorticotropin (ACTH) is essential for adrenal cortex maintenance, glucocorticoid and adrenal androgens synthesis, and secretion. Various molecular and cellular alterations of the cAMP pathway have been observed in endocrine tumors. Protein kinase A (PKA) is a central key component of the cAMP pathway. Molecular alterations of PKA subunits have been observed in adrenocortical tumors. PKA molecular defects can be germline in hereditary disorders or somatic in sporadic tumors. Heterozygous germline inactivating mutations of the PKA regulatory subunit RIα gene (PRKAR1A) can be observed in patients with ACTH-independent Cushing's syndrome (CS) due to primary pigmented nodular adrenocortical disease (PPNAD). PRKAR1A is considered as a tumor suppressor gene. Interestingly, these mutations can also be observed as somatic alterations in sporadic cortisol-secreting adrenocortical adenomas. Germline gene duplication of the catalytic subunits Cα (PRKACA) has been observed in patients with PPNAD. Furthermore, exome sequencing revealed recently activating somatic mutations of PRKACA in about 40% of cortisol-secreting adrenocortical adenomas. In vitro and in vivo functional studies help in the progress to understand the mechanisms of adrenocortical tumors development due to PKA regulatory subunits alterations. All these alterations are observed in benign oversecreting tumors and are mimicking in some way cAMP pathway constitutive activation. On the long term, unraveling these alterations will open new strategies of pharmacological treatment targeting the cAMP pathway in adrenal tumors and cortisol-secretion disorders.


Subject(s)
Adrenal Cortex Neoplasms/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Adrenal Cortex Neoplasms/pathology , Animals , Biocatalysis , Humans , Models, Biological , Protein Subunits/metabolism
4.
Horm Metab Res ; 46(12): 876-82, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25054435

ABSTRACT

Illegitimate G-protein coupled receptors are known to control cortisol secretion in adrenal adenomas and bilateral macronodular adrenal hyperplasias (BMAHs) causing Cushing's syndrome. In the present study, we have evaluated the role of glucagon in the regulation of cortisol secretion in 13 patients with BMAH or adrenocortical adenoma causing subclinical or overt Cushing's syndrome. Injection of glucagon provoked an increase in plasma cortisol in 2 patients. After surgery, immunohistochemical studies showed the presence of glucagon receptor-like immunoreactivity in clusters of spongiocytic cells in adrenal tissues from patients who were sensitive in vivo to glucagon. We also observed an in vitro cortisol response to vasoactive intestinal peptide from an adenoma, which was insensitive to glucagon and pituitary adenylate cyclase-activating peptide. Altogether, our data show that ectopic glucagon receptors are expressed in some adrenal cortisol-producing benign lesions. Our results also indicate that circulating glucagon may influence cortisol release under fasting conditions.


Subject(s)
Adrenal Glands/pathology , Cushing Syndrome/pathology , Glucagon/pharmacology , Peptides/pharmacology , Adrenal Glands/drug effects , Adrenocortical Adenoma/blood , Adult , Aged , Cushing Syndrome/blood , Female , Humans , Hydrocortisone/blood , Hydrocortisone/metabolism , Hyperplasia , Immunohistochemistry , Kinetics , Male , Middle Aged , Receptors, Glucagon/metabolism , Young Adult
5.
Horm Metab Res ; 46(12): 883-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25268545

ABSTRACT

The cyclic AMP/protein kinase A signaling cascade is one of the main pathways involved in the pathogenesis of adrenocortical tumors. The PKA R1A and R2B proteins are the most abundant regulatory subunits in endocrine tissues. Inactivating mutations of PRKAR1A are associated with Carney complex and a subset of sporadic tumors and the abundance of R2B protein is low in a subset of secreting adrenocortical adenomas. We previously showed that PRKAR1A and PRKAR2B inactivation have anti-apoptotic effects on the adrenocortical carcinoma cell line H295R. The aim of this study was to compare the effects of PRKAR1A and PRKAR2B depletion on cell proliferation, apoptosis, cell signaling pathways, and cell cycle regulation. We found that PRKAR2B depletion is compensated by an upregulation of R1A protein, whereas PRKAR1A depletion has no effect on the production of R2B. The depletion of either PRKAR1A or PRKAR2B promotes the expression of Bcl-xL and resistance to apoptosis; and is associated with a high percentage of cells in S and G2 phase, activates PKA and MEK/ERK pathways, and impairs the expression of IkB leading to activate the NF-κB pathway. However, we observed differences in the regulation of cyclins. The depletion of PRKAR1A leads to the accumulation of cyclin D1 and p27kip, whereas the depletion of PRKAR2B promotes the accumulation of cyclin A, B, cdk1, cdc2, and p21Cip. In conclusion, although the depletion of PRKAR1A and PRKAR2B in adrenocortical cells has similar effects on cell proliferation and apoptosis; loss of these PKA subunits differentially affects cyclin expression.


Subject(s)
Adrenal Glands/cytology , Adrenal Glands/enzymology , Cell Cycle Checkpoints , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/metabolism , Signal Transduction , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Humans , MAP Kinase Signaling System , NF-kappa B/metabolism , Protein Subunits/metabolism
6.
Pituitary ; 17(4): 320-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23943009

ABSTRACT

In a previous 15-day, Phase II study of patients with de novo or persistent/recurrent Cushing's disease (core study), treatment with pasireotide 600 µg sc bid reduced urinary free cortisol (UFC) levels in 76% of patients and normalized UFC in 17%. The objective of this study was to evaluate the efficacy and safety of extended treatment with pasireotide. This was a planned, open-ended, single-arm, multicenter extension study (primary endpoint: 6 months). Patients aged ≥18 years with Cushing's disease who completed the core study could enter the extension if they achieved UFC normalization at core study end and/or obtained significant clinical benefit. Of the 38 patients who completed the core study, 19 entered the extension and 18 were included in the efficacy analyses (three responders, 11 reducers, four non-reducers in the core study). At data cut-off, median treatment duration in the extension was 9.7 months (range: 2 months to 4.8 years). At extension month 6, 56% of the 18 patients had lower UFC than at core baseline and 22% had normalized UFC. Of the four patients who remained on study drug at month 24, one had normalized UFC. Reductions in serum cortisol, plasma adrenocorticotropic hormone, body weight and diastolic blood pressure were observed. The most common adverse events were mild-to-moderate gastrointestinal disorders and hyperglycemia. Pasireotide offers a tumor-directed medical therapy that may be effective for the extended treatment of some patients with Cushing's disease.


Subject(s)
Pituitary ACTH Hypersecretion/blood , Pituitary ACTH Hypersecretion/drug therapy , Somatostatin/analogs & derivatives , Adrenocorticotropic Hormone/blood , Adult , Aged , Female , Humans , Hydrocortisone/blood , Male , Middle Aged , Somatostatin/therapeutic use , Young Adult
7.
J Endocrinol Invest ; 36(11): 1000-3, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23812285

ABSTRACT

BACKGROUND: Wnt/ß-catenin signaling pathway activation plays an important role in adrenocortical tumorigenesis, but is only in part related to ß-catenin activating somatic mutations. Recently, genetic alteration in AXIN2, a key component of the Wnt/ß-catenin signaling pathway, has been described in adrenocortical tumors and specifically in adrenocortical carcinoma (ACC). AIM: To assess frequency and consequences of AXIN genes alteration on a large cohort of ACC. PATIENTS AND METHODS: Forty-nine adult sporadic ACC, with expression data available, in addition to both ACC cell lines H295 and H295R were studied. AXIN2 exon 8 hot-spot sequencing was performed on the entire cohort. AXIN1 entire coding region was studied on the 8 ACC with nuclear ß-catenin staining. RESULTS: The previously described AXIN2 in-frame heterozygous 12bp deletion c2013_2024del12 was found in 1 of the 49 ACC studied (2%), in a tumor with pSer45del activating CTNNB1 mutation and nuclear ß-catenin staining. This heterozygous deletion was also found in the patient's germline DNA, extracted from peripheral blood leukocytes. This genetic alteration was also present in H295 and H295R cell lines. The single-nucleotide polymorphism rs35415678 was found with an allele frequency similar to those found in reference populations. No correlation between AXIN2 expression, AXIN2 genetic variant or nuclear ß- catenin staining was observed. No AXIN1 alterations were found in the 8 ACC studied. CONCLUSIONS: AXIN genes do not play a major role in ACC tumorigenesis and Wnt/ß-catenin signaling pathway activation. AXIN2 germline variant c2013_2024del12 is likely to be a non-pathogenic polymorphism.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Axin Protein/genetics , Wnt Signaling Pathway/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Transcriptome
8.
Endocr Connect ; 12(12)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37855374

ABSTRACT

Although hyperandrogenism is a frequent cause of consultation in adolescent girls, more severe forms with virilization must lead to suspicion of an adrenal or ovarian tumor. However, they may also reveal a 46,XY disorder of sexual development (DSD). Here, we describe four adolescent girls referred for pubertal virilization and in whom we diagnosed a 46,XY DSD. We performed gene mutation screening by Sanger sequencing (all patients) and by next-generation sequencing (NGS) in patient #4. We identified new heterozygous NR5A1 gene variants in patients #1 and #2 and a homozygous SRD5A2 gene deletion in patient #3. Patient #4 received a diagnosis of complete androgen insensitivity in childhood; however, due the unusual pubertal virilization, we completed the gene analysis by NGS that revealed two heterozygous HSD17B3 variants. This work underlines the importance of considering the hypothesis of 46,XY DSD in adolescent girls with unexplained virilization at puberty.

9.
Horm Metab Res ; 44(10): 741-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22752956

ABSTRACT

Various molecular and cellular alterations of the cyclic adenosine monophosphate (cAMP) pathway have been observed in endocrine tumors. Since protein kinase A (PKA) is a central key component of the cAMP pathway, studies of the alterations of PKA subunits in endocrine tumors reveal new aspects of the mechanisms of cAMP pathway alterations in human diseases. So far, most alterations have been observed for the regulatory subunits, mainly PRKAR1A and to a lower extent, PRKAR2B. One of the best examples of such alteration today is the multiple neoplasia syndrome Carney complex (CNC). The most common endocrine gland manifestations of CNC are pituitary GH-secreting adenomas, thyroid tumors, testicular tumors, and ACTH-independent Cushing's syndrome due to primary pigmented nodular adrenocortical disease (PPNAD). Heterozygous germline inactivating mutations of the PKA regulatory subunit RIα gene (PRKAR1A) are observed in about two-third of CNC patients, and also in patients with isolated PPNAD. PRKAR1A is considered as a tumor suppressor gene. Interestingly, these mutations can also be observed as somatic alterations in sporadic endocrine tumors. More than 120 different PRKAR1A mutations have been found today. Most of them lead to an unstable mutant mRNA, which will be degraded by nonsense mediated mRNA decay. In vitro and in vivo functional studies are in progress to understand the mechanisms of endocrine tumor development due to PKA regulatory subunits inactivation. PRKAR1A mutations stimulate in most models PKA activity, mimicking in some way cAMP pathway constitutive activation. Cross-talks with other signaling pathways summarized in this review have been described and might participate in endocrine tumorigenesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Endocrine Gland Neoplasms/enzymology , Endocrine Gland Neoplasms/genetics , Mutation/genetics , Adrenal Cortex/pathology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endocrine Gland Neoplasms/drug therapy , Endocrine Gland Neoplasms/pathology , Humans , Molecular Targeted Therapy , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics
10.
Endocrine ; 78(1): 201-204, 2022 10.
Article in English | MEDLINE | ID: mdl-35925470

ABSTRACT

The most frequent endocrine Carney complex manifestation is a bilateral primary pigmented nodular adrenocortical disease and bilateral adrenalectomy (BA) is therefore its main treatment. In this study, a 40 years follow-up of six members of the same family with heterozygous PRKAR1A germline mutation, is reported over two generations. The first cases, two sisters with severe hyperandrogenism and Cushing syndrome (CS) diagnosed in 1972 at age 14 and 25, were successfully treated with unilateral adrenalectomy (UA). Their two brothers were then diagnosed, one with a CS-related severe osteoporosis treated with BA and the other with CS treated with UA. The second generation was diagnosed with CS signs at 7 and 21 years of age and were treated with BA and UA respectively. Out of the four patients treated with UA, the only event possibly related to CS was spontaneous episode of pulmonary embolism, 30 years after surgery. Hormonal evaluation revealed either eucortisolism in one patient or partial adrenal deficiency in two and mild hypercortisolism in one patient. For the two patients with BA, one of them accidentally died. The second one, surprisingly, recovered progressively normal cortisol secretion and circadian variation. Steroid substitution was stopped 6 years after her surgery and we demonstrated by iodocholesterol scintigraphy the presence of bilateral adrenal remnants. In conclusion, our results of long term evolution of PPNAD patients show that UA in this subset of patients could be considered to treat CS.


Subject(s)
Adrenal Cortex Diseases , Adrenal Hyperplasia, Congenital , Carney Complex , Cushing Syndrome , Adolescent , Adrenal Cortex Diseases/diagnosis , Adrenal Hyperplasia, Congenital/surgery , Adrenalectomy , Adult , Carney Complex/genetics , Carney Complex/surgery , Cushing Syndrome/diagnosis , Female , Humans , Male , Radionuclide Imaging , Young Adult
11.
Br J Surg ; 98(10): 1392-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21618212

ABSTRACT

BACKGROUND: Laparoscopic adrenalectomy (LA) is the procedure of choice for surgical management of most benign adrenal tumours, with a reported overall complication rate around 10 per cent. The aim of this study was to determine predictive factors for postoperative complications and conversion to open surgery after unilateral LA. METHODS: From 1994 to 2009, consecutive patients undergoing unilateral LA by the lateral transabdominal approach were analysed from a prospectively maintained database. A mass larger than 12 cm in diameter and suspected primary adrenal carcinoma were considered contraindications to LA. Predictive factors for postoperative complications and conversion to open surgery were analysed. RESULTS: Some 462 patients were analysed. There were no postoperative deaths. Postoperative complications occurred in 53 patients (11·5 per cent), medical complications in 28, and surgical complications in 33 patients. Six patients underwent reoperation for complications. Multivariable logistic regression analysis showed that conversion to open surgery (odds ratio (OR) 6·20, 95 per cent confidence interval 2·08 to 18·53; P = 0·001) and left-sided tumour (OR 1·89, 1·02 to 3·52; P = 0·044) were independent predictive factors for overall complications. Conversion to open surgery was the only independent predictive factor for medical complications (OR 12·88, 4·21 to 39·41; P = 0·001), and left-sided LA was the only predictive factor for surgical complications (OR 2·22, 1·01 to 4·89; P = 0·047). No factor was predictive of conversion to open surgery. CONCLUSION: In this single-institution study, conversion to open surgery and left-sided tumours were independent predictive factors for overall complications, but none of the variables analysed was predictive of conversion.


Subject(s)
Adrenal Gland Neoplasms/surgery , Adrenalectomy/methods , Laparoscopy/methods , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Postoperative Complications/etiology , Reoperation/statistics & numerical data , Risk Factors , Treatment Outcome , Young Adult
12.
Eur J Endocrinol ; 186(1): 25-36, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34709200

ABSTRACT

OBJECTIVE: Adrenocortical carcinoma (ACC) has an aggressive but variable clinical course. Prognostic stratification based on the European Network for the Study of Adrenal Tumours stage and Ki67 index is limited. We aimed to demonstrate the prognostic role of a points-based score (S-GRAS) in a large cohort of patients with ACC. DESIGN: This is a multicentre, retrospective study on ACC patients who underwent adrenalectomy. METHODS: The S-GRAS score was calculated as a sum of the following points: tumour stage (1-2 = 0; 3 = 1; 4 = 2), grade (Ki67 index 0-9% = 0; 10-19% = 1; ≥20% = 2 points), resection status (R0 = 0; RX = 1; R1 = 2; R2 = 3), age (<50 years = 0; ≥50 years = 1), symptoms (no = 0; yes = 1), and categorised, generating four groups (0-1, 2-3, 4-5, and 6-9). Endpoints were progression-free survival (PFS) and disease-specific survival (DSS). The discriminative performance of S-GRAS and its components was tested by Harrell's Concordance index (C-index) and Royston-Sauerbrei's R2D statistic. RESULTS: We included 942 ACC patients. The S-GRAS score showed superior prognostic performance for both PFS and DSS, with best discrimination obtained using the individual scores (0-9) (C-index = 0.73, R2D = 0.30, and C-index = 0.79, R2D = 0.45, respectively, all P < 0.01vs each component). The superiority of S-GRAS score remained when comparing patients treated or not with adjuvant mitotane (n = 481 vs 314). In particular, the risk of recurrence was significantly reduced as a result of adjuvant mitotane only in patients with S-GRAS 4-5. CONCLUSION: The prognostic performance of S-GRAS is superior to tumour stage and Ki67 in operated ACC patients, independently from adjuvant mitotane. S-GRAS score provides a new important guide for personalised management of ACC (i.e. radiological surveillance and adjuvant treatment).


Subject(s)
Adrenal Cortex Neoplasms/diagnosis , Adrenocortical Carcinoma/diagnosis , Diagnostic Techniques, Endocrine , Adrenal Cortex Neoplasms/mortality , Adrenal Cortex Neoplasms/pathology , Adrenal Cortex Neoplasms/surgery , Adrenalectomy , Adrenocortical Carcinoma/mortality , Adrenocortical Carcinoma/pathology , Adrenocortical Carcinoma/surgery , Adult , Aged , Aged, 80 and over , Disease Progression , Humans , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Research Design , Retrospective Studies , Survival Analysis
13.
Horm Metab Res ; 42(2): 93-101, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19882499

ABSTRACT

Storage and tissue handling of surgical tumor specimen have been recognized as critical steps that can potentially affect reproducibility and comparability of molecular endpoints between laboratories. In the preparation of adrenal tumor tissue banking, three different protocols that simulate warm ischemia upon tumor removal (protocol I), thawing and refreezing cycles (protocol II), as well as storage of vital tumor samples (protocol III) were applied. For the first two protocols, samples were subdivided and either snap frozen or treated with a RNA preserving agent (RPA) while in protocol III different storage media were compared. Following these procedures, recovery and integrity of DNA, RNA, and protein by means of pulsed field electrophoresis, long-range PCR, real-time PCR, immunoblot, and immunohistochemistry (protocol I and II) as well as cell viability and steroidogenic capacity (protocol III) were investigated. While DNA integrity was not influenced by different treatment modalities, expression levels of adrenal marker genes were more affected in samples after snap freezing in comparison to RPA pretreatment. Moreover, storage at room temperature before and after freezing could be demonstrated to decrease the relative amount of protein phosphorylation (ERK) and enzymatic activity (succinate cytochrome c reductase) while overall protein levels were not significantly affected. Similarly, morphological or immunohistochemical evaluation was comparable between groups. For primary cell cultures generated after storage of tumor samples similar rates of viability were observable while steroid output varied between the groups. Overall, on the basis of the presented endpoints standardized operational procedures can be defined for a proposed European adrenal tumor biobank.


Subject(s)
Adrenal Gland Neoplasms/pathology , Adrenal Glands/pathology , Biological Specimen Banks/organization & administration , Pheochromocytoma/pathology , Specimen Handling/standards , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Animals , Cell Survival , Cryopreservation , DNA/analysis , Europe , Humans , Immunoenzyme Techniques , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Mitochondrial Proteins/metabolism , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Quality Control , RNA/analysis , Tissue Preservation , Transplantation, Heterologous
14.
J Clin Endocrinol Metab ; 94(1): 115-22, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18957506

ABSTRACT

CONTEXT: There is currently no medical therapy for Cushing's disease that targets the pituitary adenoma. Availability of such a medical therapy would be a valuable therapeutic option for the management of this disorder. OBJECTIVE: Our objective was to evaluate the short-term efficacy of the novel multireceptor ligand somatostatin analog pasireotide in patients with de novo, persistent, or recurrent Cushing's disease. DESIGN: We conducted a phase II, proof-of-concept, open-label, single-arm, 15-d multicenter study. PATIENTS: Thirty-nine patients with either de novo Cushing's disease who were candidates for pituitary surgery or with persistent or recurrent Cushing's disease after surgery without having received prior pituitary irradiation. INTERVENTION: Patients self-administered sc pasireotide 600 microg twice daily for 15 d. MAIN OUTCOME MEASURE: Normalization of urinary free cortisol (UFC) levels after 15 d treatment was the main outcome measure. RESULTS: Of the 29 patients in the primary efficacy analysis, 22 (76%) showed a reduction in UFC levels, of whom five (17%) had normal UFC levels (responders), after 15 d of treatment with pasireotide. Serum cortisol levels and plasma ACTH levels were also reduced. Steady-state plasma concentrations of pasireotide were achieved within 5 d of treatment. Responders appeared to have higher pasireotide exposure than nonresponders. CONCLUSIONS: Pasireotide produced a decrease in UFC levels in 76% of patients with Cushing's disease during the treatment period of 15 d, with direct effects on ACTH release. These results suggest that pasireotide holds promise as an effective medical treatment for this disorder.


Subject(s)
Oligopeptides/therapeutic use , Pituitary ACTH Hypersecretion/drug therapy , Adrenocorticotropic Hormone/blood , Adult , Aged , Blood Glucose/analysis , Female , Glucagon/blood , Humans , Hydrocortisone/urine , Insulin/blood , Male , Middle Aged , Oligopeptides/adverse effects , Oligopeptides/pharmacokinetics , Pituitary ACTH Hypersecretion/metabolism , Somatostatin/analogs & derivatives
16.
Acta Anaesthesiol Scand ; 53(4): 522-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19239408

ABSTRACT

BACKGROUND: Pre-operative hypotensive drugs are assumed to have dramatically decreased operative mortality and morbidity in patients undergoing phaeochromocytoma removal only in non-controlled studies. We evaluated the predictive value of pre-operative high systolic arterial pressure (SAP) on intra- and post-operative haemodynamic instability, in 96 patients undergoing laparoscopic adrenalectomy for phaeochromocytoma. METHODS: Ninety-six consecutive patients underwent laparoscopic adrenalectomy for phaeochromocytoma. Pre-operative SAP was not systematically normalised, provided that increased SAP was clinically tolerated. Intravenous nicardipine, esmolol and norepinephrine were intraoperatively titrated to treat SAP increase >150 mmHg, tachycardia >90-110/min, arrhythmia or SAP decrease under 90 mmHg, respectively. Volume expanders were not systematically administered. Patients with increased and normal pre-operative SAP were compared with respect to (a) nicardipine, esmolol and norepinephrine requirement, (b) highest intraoperative SAP and heat rate, (c) lowest intraoperative SAP, (d) duration of surgery and (e) norepinephrine requirement following tumour removal. RESULTS: Groups did not differ significantly with respect to data defined as being indicative of perioperative haemodynamic instability (all P values>0.05). DISCUSSION: As previously demonstrated, in patients undergoing phaeochromocytoma removal, perioperative haemodynamic changes are mainly due to catecholamine release during tumour manipulation, and to the decrease in catecholamine level following tumour removal. Whether pre-operative hypotensive drugs are likely to alter these changes remains questionable. CONCLUSION: For most patients scheduled for laparoscopic phaeochromocytoma removal, surgery can be carried out without systematic pre-operative arterial pressure normalisation.


Subject(s)
Adrenalectomy , Blood Pressure , Pheochromocytoma/surgery , Adult , Aged , Catecholamines/metabolism , Female , Humans , Male , Middle Aged , Pheochromocytoma/physiopathology , Systole
17.
J Radiol ; 90(3 Pt 2): 422-5, 2009 Mar.
Article in French | MEDLINE | ID: mdl-19421133

ABSTRACT

Adrenal tumors diagnosed after work-up of an incidentaloma can be responsible for excessive secretion of steroids or catecholamines. On the other hand, an infiltrative process (metastases, infection...) involving both adrenals can cause adrenal insufficiency. Clinical and biological work-up is performed to detect hormonal imbalances requiring treatment, and further characterize the nature of the lesion. Screening for pheochromocytoma should be systematic with metanephrine assays on urine or plasma. Screening for cortisol secreting tumors, often responsible for Cushing's syndrome of moderate severity (pre-clinical) in patients with incidentaloma, will be performed using the overnight dexamethasone suppression test (1 mg). Depending on results from the imaging and clinical work-up, excessive secretion of aldosterone, androgens or steroid precursors may sometimes be investigated. In patients with bilateral incidentalomas, the normal synacthen test and ACTH assay are also performed to exclude adrenal insufficiency.


Subject(s)
Adrenal Gland Neoplasms/diagnosis , Cushing Syndrome/diagnosis , Incidental Findings , Pheochromocytoma/diagnosis , Adrenal Gland Neoplasms/blood , Adrenal Gland Neoplasms/diagnostic imaging , Adrenal Gland Neoplasms/surgery , Adrenal Gland Neoplasms/urine , Adrenocorticotropic Hormone/blood , Dexamethasone , Diagnosis, Differential , Humans , Male , Metanephrine/blood , Metanephrine/urine , Middle Aged , Pheochromocytoma/blood , Pheochromocytoma/diagnostic imaging , Pheochromocytoma/surgery , Pheochromocytoma/urine , Radionuclide Imaging , Tomography, X-Ray Computed
18.
Endocr Relat Cancer ; 26(2): G1-G18, 2019 02.
Article in English | MEDLINE | ID: mdl-30400055

ABSTRACT

The management of cancer patients has changed due to the considerably more frequent use of immune checkpoint inhibitors (ICPIs). However, the use of ICPI has a risk of side effects, particularly endocrine toxicity. Since the indications for ICPI are constantly expanding due to their efficacy, it is important that endocrinologists and oncologists know how to look for this type of toxicity and how to treat it when it arises. In view of this, the French Endocrine Society initiated the formulation of a consensus document on ICPI-related endocrine toxicity. In this paper, we will introduce data on the general pathophysiology of endocrine toxicity, and we will then outline expert opinion focusing primarily on methods for screening, management and monitoring for endocrine side effects in patients treated by ICPI. We will then look in turn at endocrinopathies that are induced by ICPI including dysthyroidism, hypophysitis, primary adrenal insufficiency and fulminant diabetes. In each chapter, expert opinion will be given on the diagnosis, management and monitoring for each complication. These expert opinions will also discuss the methodology for categorizing these side effects in oncology using 'common terminology criteria for adverse events' (CTCAE) and the difficulties in applying this to endocrine side effects in the case of these anti-cancer therapies. This is shown in particular by certain recommendations that are used for other side effects (high-dose corticosteroids, contraindicated in ICPI for example) and that cannot be considered as appropriate in the management of endocrine toxicity, as it usually does not require ICPI withdrawal or high-dose glucocorticoid intake.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Endocrine System Diseases/chemically induced , Immunotherapy/adverse effects , France , Humans , Immunotherapy/methods
19.
J Clin Endocrinol Metab ; 93(7): 2454-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18413427

ABSTRACT

OBJECTIVE: Our objective was to evaluate the published literature and reach a consensus on the treatment of patients with ACTH-dependent Cushing's syndrome, because there is no recent consensus on the management of this rare disorder. PARTICIPANTS: Thirty-two leading endocrinologists, clinicians, and neurosurgeons with specific expertise in the management of ACTH-dependent Cushing's syndrome representing nine countries were chosen to address 1) criteria for cure and remission of this disorder, 2) surgical treatment of Cushing's disease, 3) therapeutic options in the event of persistent disease after transsphenoidal surgery, 4) medical therapy of Cushing's disease, and 5) management of ectopic ACTH syndrome, Nelson's syndrome, and special patient populations. EVIDENCE: Participants presented published scientific data, which formed the basis of the recommendations. Opinion shared by a majority of experts was used where strong evidence was lacking. CONSENSUS PROCESS: Participants met for 2 d, during which there were four chaired sessions of presentations, followed by general discussion where a consensus was reached. The consensus statement was prepared by a steering committee and was then reviewed by all authors, with suggestions incorporated if agreed upon by the majority. CONCLUSIONS: ACTH-dependent Cushing's syndrome is a heterogeneous disorder requiring a multidisciplinary and individualized approach to patient management. Generally, the treatment of choice for ACTH-dependent Cushing's syndrome is curative surgery with selective pituitary or ectopic corticotroph tumor resection. Second-line treatments include more radical surgery, radiation therapy (for Cushing's disease), medical therapy, and bilateral adrenalectomy. Because of the significant morbidity of Cushing's syndrome, early diagnosis and prompt therapy are warranted.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Cushing Syndrome/therapy , ACTH Syndrome, Ectopic/therapy , Adrenal Insufficiency/therapy , Adrenalectomy , Humans , Hypophysectomy , Metyrapone/therapeutic use , Mitotane/therapeutic use , Nelson Syndrome/therapy
20.
Gastroenterol Clin Biol ; 32(11): 910-3, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18467057

ABSTRACT

Pancreatic lesions in von Hippel Lindau disease (VHLD) are frequent and mainly consist of cystic lesions, which should not be resected because of their benign evolution. Solid lesions, mostly pancreatic endocrine tumors (PET), are rare and usually occur in combination with cystic lesions. We report a case of a patient with VHLD who underwent PET enucleation in a polycystic pancreas requiring fenestration of multiple adjacent cysts, to ensure complete resection with free resection margins. The postoperative course was complicated by massive ascitic fluid effusion, probably related to pancreatic-cyst fenestration. Although this complication is well-known after liver-cyst fenestration, it has not been reported after pancreatic-cyst fenestration. This observation emphasizes that morbidity from surrounding pancreatic polycystic disease should not be underestimated in pancreatic surgery for VHLD.


Subject(s)
Ascites/etiology , Pancreatic Cyst/surgery , Postoperative Complications/etiology , von Hippel-Lindau Disease/complications , Humans
SELECTION OF CITATIONS
SEARCH DETAIL