Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Endocrine ; 75(2): 537-548, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34741720

ABSTRACT

PURPOSE: To assess the pharmacokinetics, pharmacodynamics and tolerability of different doses of octreotide and pasireotide (subcutaneous [sc] and long-acting release [LAR]) when co-administered in healthy volunteers. METHODS: This was an exploratory, Phase I, single-centre study. Healthy adults were enrolled in a staggered approach into seven cohorts to receive octreotide and pasireotide (sc and LAR formulations), alone or in combination. Plasma drug concentrations, growth hormone (GH), insulin-like growth factor I (IGF-I), and plasma glucose were assessed at baseline, immediately after sc treatment, and 21 and 28 days after LAR treatment. RESULTS: Of 88 enrolled subjects, 52 and 82 participated in sc and LAR dosing phases, respectively. There were no relevant pharmacokinetic interactions between octreotide and pasireotide. In combination, pasireotide sc (150 µg) and octreotide sc (100/300 µg) resulted in numerically greater reductions in insulin levels and a higher incidence of AEs than either single agent; the rapid (within 1 h) increase in plasma glucose after pasireotide was delayed with combination treatment. Octreotide sc and pasireotide sc, alone or in combination, reduced IGF-I levels and led to undetectable GH levels in most subjects. During the LAR phase, addition of a low dose of pasireotide (5 mg) to a standard dose of octreotide (20 mg) resulted in an ~2-fold reduction in median IGF-I versus octreotide 20 mg 21 days post-dose; this effect was numerically greater than seen for pasireotide 20 mg alone. Peak plasma glucose was substantially lower after LAR than sc dosing. Interestingly, glucose levels were also numerically lower in the pasireotide 5 mg plus octreotide 20 mg group than for 20 mg of octreotide or pasireotide alone. AEs were less frequent after LAR than sc dosing. CONCLUSIONS: Combined low doses of pasireotide LAR (5 mg) and octreotide LAR (10-30 mg) provided greater suppression of IGF-I than either single agent and did not increase blood glucose or incidence of AEs versus either agent alone.


Subject(s)
Acromegaly , Octreotide , Acromegaly/drug therapy , Adult , Blood Glucose , Delayed-Action Preparations/therapeutic use , Growth Hormone , Healthy Volunteers , Humans , Insulin-Like Growth Factor I/metabolism , Octreotide/adverse effects , Somatostatin/analogs & derivatives
2.
Cancer Lett ; 524: 232-244, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34637845

ABSTRACT

Increasingly common, neuroendocrine tumors (NETs) are regarded nowadays as neoplasms potentially causing debilitating symptoms and life-threatening medical conditions. Pheochromocytoma is a NET that develops from chromaffin cells of the adrenal medulla, and is responsible for an excessive secretion of catecholamines. Consequently, patients have an increased risk for clinical symptoms such as hypertension, elevated stroke risk and various cardiovascular complications. Somatostatin analogues are among the main anti-secretory medical drugs used in current clinical practice in patients with NETs. However, their impact on pheochromocytoma-associated catecholamine hypersecretion remains incompletely explored. This study investigated the potential efficacy of octreotide and pasireotide (SOM230) on human tumor cells directly cultured from freshly resected pheochromocytomas using an implemented catecholamine secretion measurement by carbon fiber amperometry. SOM230 treatment efficiently inhibited nicotine-induced catecholamine secretion both in bovine chromaffin cells and in human tumor cells whereas octreotide had no effect. Moreover, SOM230 specifically decreased the number of exocytic events by impairing the stimulation-evoked calcium influx as well as the nicotinic receptor-activated inward current in human pheochromocytoma cells. Altogether, our findings indicate that SOM230 acts as an inhibitor of catecholamine secretion through a mechanism involving the nicotinic receptor and might be considered as a potential anti-secretory treatment for patients with pheochromocytoma.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Pheochromocytoma/drug therapy , Somatostatin/analogs & derivatives , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Catecholamines/biosynthesis , Catecholamines/metabolism , Cell Line, Tumor , Humans , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Somatostatin/pharmacology
3.
Cell Mol Gastroenterol Hepatol ; 11(5): 1405-1436, 2021.
Article in English | MEDLINE | ID: mdl-33482394

ABSTRACT

BACKGROUND & AIMS: Cancer-associated fibroblasts (CAFs) from pancreatic adenocarcinoma (PDA) present high protein synthesis rates. CAFs express the G-protein-coupled somatostatin receptor sst1. The sst1 agonist SOM230 blocks CAF protumoral features in vitro and in immunocompromised mice. We have explored here the therapeutic potential of SOM230, and underlying mechanisms, in immunocompetent models of murine PDA mimicking the heavy fibrotic and immunosuppressive stroma observed in patient tumors. METHODS: Large-scale mass spectrometry analyses were performed on media conditioned from 9 patient PDA-derived CAF primary cultures. Spontaneous transgenic and experimental (orthotopic co-graft of tumor cells plus CAFs) PDA-bearing mice were longitudinally ultrasound-monitored for tumor and metastatic progression. Histopathology and flow cytometry analyses were performed on primary tumors and metastases. Stromal signatures were functionally validated through bioinformatics using several published, and 1 original, PDA database. RESULTS: Proteomics on the CAF secretome showed that SOM230 controls stromal activities including inflammatory responses. Among the identified secreted proteins, we validated that colony-stimulating factor 1 (CSF-1) (a macrophage growth factor) was reduced by SOM230 in the tumor and plasma of PDA-harboring mice, alongside intratumor stromal normalization (reduced CAF and macrophage activities), and dramatic metastasis reduction. In transgenic mice, these SOM230 benefits alleviate the chemotherapy-induced (gemcitabine) immunosuppressive stroma reshaping. Mechanistically, SOM230 acts in vivo on CAFs through sst1 to disrupt prometastatic CAF production of CSF-1 and cross-talk with macrophages. We found that in patients, stromal CSF-1 was associated with aggressive PDA forms. CONCLUSIONS: We propose SOM230 as an antimetastatic therapy in PDA for its capacity to remodel the fibrotic and immunosuppressive myeloid stroma. This pharmacotherapy should benefit PDA patients treated with chemotherapies.


Subject(s)
Cancer-Associated Fibroblasts/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Macrophages/drug effects , Pancreatic Neoplasms/drug therapy , Secretome/drug effects , Somatostatin/analogs & derivatives , Aged , Aged, 80 and over , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Female , Hormones/pharmacology , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Somatostatin/pharmacology
4.
Can Vet J ; 59(10): 1089-1093, 2018 10.
Article in English | MEDLINE | ID: mdl-30510314

ABSTRACT

An 8-year-old castrated male border terrier dog was diagnosed with acromegaly resulting from a growth hormone secreting pituitary tumor. Sixteen daily fractions of radiation therapy were delivered followed, approximately 1 year later, by administration of pasireotide. The aforementioned treatment was considered effective and should be further evaluated in similar cases.


Radiothérapie et traitement au pasiréotide pour une tumeur pituitaire produisant une hormone de croissance chez un chien diabétique. Un chien Terrier-Border castré âgé de 8 ans a été diagnostiqué avec de l'acromégalie découlant d'une tumeur pituitaire secrétant une hormone de croissance. Seize fractions quotidiennes de radiothérapie ont été administrées et ont été suivies, environ un an plus tard, de l'administration du pasiréotide. Le traitement précédemment mentionné a été considéré efficace et devrait être étudié de plus près dans des cas similaires.(Traduit par Isabelle Vallières).


Subject(s)
Dog Diseases/radiotherapy , Growth Hormone-Secreting Pituitary Adenoma/veterinary , Hormones/therapeutic use , Somatostatin/analogs & derivatives , Acromegaly/etiology , Acromegaly/veterinary , Adenoma/drug therapy , Adenoma/radiotherapy , Adenoma/veterinary , Animals , Diabetes Mellitus/drug therapy , Diabetes Mellitus/veterinary , Dog Diseases/drug therapy , Dogs , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/radiotherapy , Male , Somatostatin/therapeutic use , Treatment Outcome
5.
Stress ; 21(4): 370-375, 2018 07.
Article in English | MEDLINE | ID: mdl-29661114

ABSTRACT

Pasireotide is a new-generation somatostatin analog that acts through binding to multiple somatostatin receptor subtypes. Studies have shown that pasireotide induces hyperglycemia, reduces glucocorticoid secretion, alters neurotransmission, and potentially affects stress responses typically manifested as hyperglycemia and increased corticosterone secretion. This study specifically aimed to evaluate whether pasireotide treatment modifies glucose and costicosterone secretion in response to acute restraint stress. Male Holtzman rats of 150-200 g were treated with pasireotide (10 µg/kg/day) twice-daily for two weeks or vehicle for the same period. Blood samples were collected at baseline and after 5, 10, 30, and 60 min of restraint stress. The three experimental groups comprised of vehicle + restraint (VEHR), pasireotide + restraint (PASR), and pasireotide + saline (PASNR). Following pasireotide treatment, no significant differences in baseline glucose and corticosterone levels were observed among the three groups. During restraint, hyperglycemia was observed at 10 min (p < .01 for both comparisons), peaked at 30 min (p < .01 for both comparisons) and showed higher 60 min areas under glucose curves in the VEHR and PASR stressed groups when compared to the non-stressed PASNR group (p < .05 for both comparisons). Restraint also increased corticosterone secretion in the VEHR and PASR stressed groups at 5 min (p < .01 for both comparisons), and peaked at 30 min (p < .01 for both comparisons) with corresponding higher 60 min areas under corticosterone curves when compared to the non-stressed PASNR group (p < .01 for both comparisons). In conclusion, pasireotide treatment does not modify hyperglycemic- and corticosterone-restraint stress responses, thus preserving acute stress regulation.


Subject(s)
Blood Glucose/analysis , Corticosterone/blood , Somatostatin/analogs & derivatives , Stress, Physiological/drug effects , Animals , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical , Somatostatin/pharmacology , Synaptic Transmission
6.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29025873

ABSTRACT

The recent demonstration that pancreatic α cells can be continuously regenerated and converted into ß-like cells upon ectopic expression of Pax4 opened new avenues of research in the endocrine cell differentiation and diabetes fields. To determine whether such plasticity was also shared by δ cells, we generated and characterized transgenic animals that express Pax4 specifically in somatostatin-expressing cells. We demonstrate that the ectopic expression of Pax4 in δ cells is sufficient to induce their conversion into functional ß-like cells. Importantly, this conversion induces compensatory mechanisms involving the reactivation of endocrine developmental processes that result in dramatic ß-like cell hyperplasia. Importantly, these ß-like cells are functional and can partly reverse the consequences of chemically induced diabetes.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Ectopic Gene Expression , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Cell Proliferation , Cell Transdifferentiation/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Genetic Therapy/methods , Glucagon/biosynthesis , Glucagon/genetics , Homeodomain Proteins/metabolism , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Transgenic , Paired Box Transcription Factors/metabolism , Somatostatin/biosynthesis , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Streptozocin
7.
J Endocrinol ; 231(2): 135-145, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27587848

ABSTRACT

Somatostatin analogs (SSA) are the mainstay of pharmacological treatment for pituitary adenomas. However, some patients escape from therapy with octreotide, a somatostatin receptor 2 (sst2)-preferring SSA, and pasireotide, a novel multi-sst-preferring SSA, may help to overcome this problem. It has been proposed that correspondence between sst1-sst5 expression pattern and SSA-binding profile could predict patient's response. To explore the cellular/molecular features associated with octreotide/pasireotide response, we performed a parallel comparison of their in vitro effects, evaluating sst1-sst5 expression, intracellular Ca2+ signaling ([Ca2+]i), hormone secretion and cell viability, in a series of 85 pituitary samples. Somatotropinomas expressed sst5>sst2, yet octreotide reduced [Ca2+]i more efficiently than pasireotide, while both SSA similarly decreased growth hormone release/expression and viability. Corticotropinomas predominantly expressed sst5, but displayed limited response to pasireotide, while octreotide reduced functional endpoints. Non-functioning adenomas preferentially expressed sst3 but, surprisingly, both SSA increased cell viability. Prolactinomas mainly expressed sst1 but were virtually unresponsive to SSA. Finally, both SSA decreased [Ca2+]i in normal pituitaries. In conclusion, both SSA act in vitro on pituitary adenomas exerting both similar and distinct effects; however, no evident correspondence was found with the sst1-sst5 profile. Thus, it seems plausible that additional factors, besides the simple abundance of a given sst, critically influence the SSA response.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Neoplasm Proteins/agonists , Octreotide/pharmacology , Pituitary Gland/drug effects , Pituitary Neoplasms/drug therapy , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , ACTH-Secreting Pituitary Adenoma/drug therapy , ACTH-Secreting Pituitary Adenoma/metabolism , ACTH-Secreting Pituitary Adenoma/pathology , Adenoma/drug therapy , Adenoma/metabolism , Adenoma/pathology , Antineoplastic Agents, Hormonal/adverse effects , Calcium Signaling/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Growth Hormone-Secreting Pituitary Adenoma/pathology , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Octreotide/adverse effects , Pituitary Gland/metabolism , Pituitary Gland/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prolactinoma/drug therapy , Prolactinoma/metabolism , Prolactinoma/pathology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/adverse effects , Somatostatin/pharmacology , Tumor Cells, Cultured
8.
Endocrinology ; 157(5): 1789-98, 2016 05.
Article in English | MEDLINE | ID: mdl-26990064

ABSTRACT

Pasireotide, a somatostatin analog, is reported to have anti-proliferative effects in neuroendocrine tumors (NETs). We therefore assessed the efficacy of pasireotide for treating pancreatic and pituitary NETs that develop in a mouse model of multiple endocrine neoplasia type 1 (MEN1). Men1(+/-) mice were treated from age 12 mo with 40 mg/kg pasireotide long-acting release formulation, or PBS, intramuscularly monthly for 9 mo. The Men1(+/-) mice had magnetic resonance imaging at 12 and 21 mo, and from 20 mo oral 5-bromo-2-deoxyuridine for 1 mo, to assess tumor development and proliferation, respectively. NETs were collected at age 21 mo, and proliferation and apoptosis assessed by immunohistochemistry and TUNEL assays, respectively. Pasireotide-treated Men1(+/-) mice had increased survival (pasireotide, 80.9% vs PBS, 65.2%; P < .05), with fewer mice developing pancreatic NETs (pasireotide, 86.9% vs PBS, 96.9%; P < .05) and smaller increases in pituitary NET volumes (pre-treated vs post-treated, 0.803 ± 0.058 mm(3) vs 2.872 ± 0.728 mm(3) [pasireotide] compared with 0.844 ± 0.066 mm(3) vs 8.847 ±1.948 mm(3) [PBS]; P < .01). In addition, pasireotide-treated mice had fewer pancreatic NETs compared with PBS-treated mice (2.36 ± 0.25 vs 3.72 ± 0.32, respectively; P < .001), with decreased proliferation in pancreatic NETs (pasireotide, 0.35 ± 0.03% vs PBS, 0.78 ± 0.08%; P < .0001) and pituitary NETs (pasireotide, 0.73 ±0.07% vs PBS, 1.81 ± 0.15%; P < .0001), but increased apoptosis in pancreatic NETs (pasireotide, 0.42 ± 0.05% vs PBS, 0.19 ± 0.03%; P < .001) and pituitary NETs (pasireotide, 14.75 ± 1.58% vs PBS, 2.35 ± 0.44%; P < .001). Thus, pasireotide increased survival and inhibited pancreatic and pituitary NET growth, thereby indicating its potential as an anti-proliferative and pro-apoptotic therapy.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia/drug therapy , Neuroendocrine Tumors/drug therapy , Somatostatin/analogs & derivatives , Alleles , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Female , Mice , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/pathology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Somatostatin/pharmacology , Somatostatin/therapeutic use
9.
Endocrine ; 53(1): 210-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26906713

ABSTRACT

The purpose of this study was to gain more insight into the mechanism of action of pasireotide in patients who completed the PAOLA study. PAOLA was a 24-week, Phase III, randomized, three-arm study of pasireotide LAR 40 and 60 mg versus octreotide LAR 30 mg or lanreotide Autogel 120 mg in patients with inadequately controlled acromegaly. The current work was a planned exploratory objective of the PAOLA study that evaluated changes in levels of growth hormone (GH), insulin-like growth factor 1 (IGF-1), IGF-binding proteins (IGFBP-2, IGFBP-3), glycated haemoglobin (HbA1c) and fasting plasma glucose (FPG) in each treatment arm. Responders to pasireotide LAR (mean GH levels <2.5 µg/L and normal IGF-1 levels at 24 weeks) had lower GH and IGF-1 levels at baseline (GH 5.1 ng/mL, IGF-1 519 ng/mL) than non-responders (GH 7.9 ng/mL, IGF-1 672 ng/mL). Frequency of hyperglycaemia after pasireotide treatment was similar in responders and non-responders and depended more on the baseline FPG level. 47 % of all patients treated with pasireotide LAR (40 or 60 mg) did not receive antidiabetic medication at any time during this study. This is the first study to evaluate the treatment effect of pasireotide on key hormonal and glycaemic biomarkers and to identify potential predictors of pasireotide-associated hyperglycaemia. Pre-treatment glucose status may be predictive of the development of pasireotide-associated hyperglycaemia. A large subset of patients with acromegaly does not experience major disturbances in glucose homeostasis while receiving pasireotide LAR.


Subject(s)
Acromegaly/drug therapy , Blood Glucose , Human Growth Hormone/blood , Insulin-Like Growth Factor Binding Protein 2/blood , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Somatostatin/analogs & derivatives , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Double-Blind Method , Female , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Octreotide/therapeutic use , Peptides, Cyclic/therapeutic use , Retreatment , Somatostatin/pharmacology , Somatostatin/therapeutic use , Treatment Failure , Treatment Outcome , Young Adult
10.
Am J Physiol Regul Integr Comp Physiol ; 309(6): R668-74, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26157058

ABSTRACT

Siberian hamsters (Phodopus sungorus) show spontaneous daily torpor only after ∼2 mo in winter-like short photoperiods (SP). Although some SP-induced hormonal changes have been demonstrated to be necessary for the occurrence of seasonal torpor, the whole set of preconditions is still unknown. Recent findings provide evidence that the hypothalamic pituitary growth axis is involved in endocrine responses to SP exposure in the photoperiodic hamsters. To examine whether suppression of growth hormone (GH) and insulin-like growth factor-1 (IGF-1) secretion affects the incidence of daily torpor, we used two somatostatin receptor agonists, pasireotide (SOM230) and octreotide, with different affinity profiles for receptor subtypes. Pasireotide strikingly increased the torpor frequency in male hamsters compared with sham-treated controls, and torpor duration was often increased, which in some cases exceeded 12 h. In contrast, administration of octreotide reduced the body weight of SP hamsters but had only a marginal effect on torpor frequency in males and no effect in females. Together with measured concentrations of circulating IGF-1, the present results strongly suggest that reduced activity of the GH/IGF-1 axis is not critical for stimulation of torpor expression but activation of specific somatostatin receptors is critical. This putative role for certain somatostatin receptor subtypes in torpor induction provides a promising new approach to unravel the endocrine mechanisms of torpor regulation.


Subject(s)
Receptors, Somatostatin/drug effects , Torpor/drug effects , Animals , Body Temperature , Body Weight , Cricetinae , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Octreotide/pharmacology , Phodopus , RNA, Messenger/biosynthesis , Seasons , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
11.
Toxicol Appl Pharmacol ; 286(3): 224-33, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25981165

ABSTRACT

The somatostatin analog pasireotide and the 11ß-hydroxylase inhibitor osilodrostat (LCI699) reduce cortisol levels by distinct mechanisms of action. There exists a scientific rationale to investigate the clinical efficacy of these two agents in combination. This manuscript reports the results of a toxicology study in rats, evaluating different doses of osilodrostat and pasireotide alone and in combination. Sixty male and 60 female rats were randomized into single-sex groups to receive daily doses of pasireotide (0.3mg/kg/day, subcutaneously), osilodrostat (20mg/kg/day, orally), osilodrostat/pasireotide in combination (low dose, 1.5/0.03mg/kg/day; mid-dose, 5/0.1mg/kg/day; or high dose, 20/0.3mg/kg/day), or vehicle for 13weeks. Mean body-weight gains from baseline to Week 13 were significantly lower in the pasireotide-alone and combined-treatment groups compared to controls, and were significantly higher in female rats receiving osilodrostat monotherapy. Osilodrostat and pasireotide monotherapies were associated with significant changes in the histology and mean weights of the pituitary and adrenal glands, liver, and ovary/oviduct. Osilodrostat alone was associated with adrenocortical hypertrophy and hepatocellular hypertrophy. In combination, osilodrostat/pasireotide did not exacerbate any target organ changes and ameliorated the liver and adrenal gland changes observed with monotherapy. Cmax and AUC0-24h of osilodrostat and pasireotide increased in an approximately dose-proportional manner. In conclusion, the pasireotide and osilodrostat combination did not exacerbate changes in target organ weight or toxicity compared with either monotherapy, and had an acceptable safety profile; addition of pasireotide to the osilodrostat regimen may attenuate potential adrenal gland hyperactivation and hepatocellular hypertrophy, which are potential side effects of osilodrostat monotherapy.


Subject(s)
Imidazoles/administration & dosage , Pyridines/administration & dosage , Somatostatin/analogs & derivatives , Somatostatin/administration & dosage , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Drug Combinations , Drug Delivery Systems/methods , Enzyme Inhibitors/administration & dosage , Female , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Organ Size/drug effects , Organ Size/physiology , Rats , Rats, Wistar , Steroid 11-beta-Hydroxylase/metabolism
12.
Chimia (Aarau) ; 68(7-8): 483-4, 2014.
Article in English | MEDLINE | ID: mdl-25437387

ABSTRACT

A rational drug design approach involving transposition of functional groups from SRIF into a reduced size cyclohexapeptide template has led to the discovery of SOM230, a novel, stable cyclohexapeptide somatostatin mimic which exhibits unique high affinity binding to human somatostatin receptors (sst1-5). This unique receptor subtype binding profile, in particular the exceptional high affinity binding to sst5, led to SOM230 being approved by EMEA and FDA in 2012 as the first effective pituitary directed therapeutic modality for Cushing's disease.


Subject(s)
Cushing Syndrome/drug therapy , Somatostatin/analogs & derivatives , Humans , Models, Molecular , Somatostatin/chemical synthesis , Somatostatin/chemistry , Somatostatin/therapeutic use
13.
Am J Physiol Lung Cell Mol Physiol ; 306(12): L1064-77, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24727584

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive and lethal disease, characterized by loss of lung elasticity and alveolar surface area, secondary to alveolar epithelial cell injury, reactive inflammation, proliferation of fibroblasts, and deposition of extracellular matrix. The effects of oropharyngeal aspiration of bleomycin in Sprague-Dawley rats and C57BL/6 mice, as well as of intratracheal administration of ovalbumin to actively sensitized Brown Norway rats on total lung volume as assessed noninvasively by magnetic resonance imaging (MRI) were investigated here. Lung injury and volume were quantified by using nongated or respiratory-gated MRI acquisitions [ultrashort echo time (UTE) or gradient-echo techniques]. Lung function of bleomycin-challenged rats was examined additionally using a flexiVent system. Postmortem analyses included histology of collagen and hydroxyproline assays. Bleomycin induced an increase of MRI-assessed total lung volume, lung dry and wet weights, and hydroxyproline content as well as collagen amount. In bleomycin-treated rats, gated MRI showed an increased volume of the lung in the inspiratory and expiratory phases of the respiratory cycle and a temporary decrease of tidal volume. Decreased dynamic lung compliance was found in bleomycin-challenged rats. Bleomycin-induced increase of MRI-detected lung volume was consistent with tissue deposition during fibrotic processes resulting in decreased lung elasticity, whereas influences by edema or emphysema could be excluded. In ovalbumin-challenged rats, total lung volume quantified by MRI remained unchanged. The somatostatin analog, SOM230, was shown to have therapeutic effects on established bleomycin-induced fibrosis in rats. This work suggests MRI-detected total lung volume as readout for tissue-deposition in small rodent bleomycin models of pulmonary fibrosis.


Subject(s)
Bleomycin/pharmacology , Lung/pathology , Pulmonary Fibrosis/drug therapy , Somatostatin/analogs & derivatives , Animals , Disease Models, Animal , Extracellular Matrix/pathology , Hydroxyproline/metabolism , Inflammation/metabolism , Inflammation/pathology , Lung/drug effects , Lung/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Somatostatin/therapeutic use
14.
Regul Pept ; 187: 35-41, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-24188818

ABSTRACT

INTRODUCTION: The expression and reliable detection of somatostatin receptor subtypes (SSTR1-5) is a prerequisite for the successful use of somatostatin analogs in neuroendocrine tumors (NETs). Two sets of monoclonal antibodies (mAbs) against human SSTR1, 2A, 3 and 5 have recently been developed by two independent laboratories using rabbit and mouse hybridomas. Our aim was to evaluate the usefulness of both sets of mAbs for detection of SSTRs in NET samples as they are routinely collected in clinical practice. METHODS: Mouse and rabbit mAbs were characterized in SSTR1, 2A, 3 and 5-transfected HEK293 cells and human archival samples of pancreatic tissue and NET. Comparative analysis of mAbs was also conducted by immunostaining of a tissue microarray composed of 75 cores of NET. RESULTS: Immunohistochemical analysis of HEK293 cells showed that both rabbit and mouse mAbs specifically detect their cognate receptor subtype, with mild cytoplasmic cross-reactivity observed for rabbit mAbs. Both sets of mAbs labeled normal pancreatic islets and showed similar patterns of immunoreactivity in NET controls. Direct comparison of mAb sets using a NET tissue microarray revealed strong correlation between rabbit and mouse mAbs against SSTR1 and 5, and moderate correlation for SSTR3. The rabbit mAb against SSTR2A showed higher affinity for its cognate receptor than the corresponding mouse mAb, resulting in a more reliable detection of this SSTR. CONCLUSIONS: mAbs from both sets are reliable tools for the detection of SSTR1, 3 and 5, whereas the rabbit mAb against SSTR2A is recommended for use in routine clinical testing due to its superior binding affinity.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Gastrointestinal Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Animals , Antibody Specificity , Fixatives/chemistry , Formaldehyde/chemistry , Gastrointestinal Neoplasms/pathology , HEK293 Cells , Humans , Immunohistochemistry , Mice , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Paraffin Embedding , Rabbits , Receptors, Somatostatin/immunology , Tissue Array Analysis , Tissue Fixation
15.
Surgery ; 152(6): 1068-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23102680

ABSTRACT

BACKGROUND: Pasireotide (SOM230), a long-acting somatostatin analogue (LAR), has improved agonist activity at somatostatin receptors. We tested the effect of SOM230 on insulin secretion, serum glucose concentrations, tumor growth, and survival using an MEN1 transgenic mouse model. METHODS: Eight 12-month-old conditional Men1 knockout mice with insulinoma were assessed. The treatment (n = 4) and control groups (n = 4) received monthly subcutaneous injections of SOM230 or PBS. Serum insulin and glucose levels were determined by enzyme-linked immunosorbent assay and enzymatic colorimetric assay, respectively. Tumor activity, growth, and apoptosis were determined by microPET/CT scan and histologic analysis. RESULTS: On day 7, there was a decrease in serum insulin levels from 1.06 ± 0.28 µg/L to 0.37 ± 0.17 µg/L (P = .0128) and a significant increase in serum glucose from 4.2 ± 0.45 mmol/L to 7.12 ± 1.06 mmol/L (P = .0075) in the treatment group but no change in the control group. Tumor size was less in the treatment group (2,098 ± 388 µm(2)) compared with the control group (7,067 ± 955 µm(2); P = .0024). Furthermore, apoptosis was increased in the treatment group (6.9 ± 1.23%) compared with the control group (0.29 ± 0.103%; P = .002). CONCLUSION: SOM230 demonstrates antisecretory, antiproliferative, and proapoptotic activity in our MEN1 model of insulinoma. Further studies of the effects of SOM230 in PNET patients with MEN1 mutations are warranted.


Subject(s)
Multiple Endocrine Neoplasia Type 1/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Animals , Apoptosis , Blood Glucose/analysis , Insulin/blood , Mice , Mice, Knockout , Multimodal Imaging , Multiple Endocrine Neoplasia Type 1/genetics , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Positron-Emission Tomography , Somatostatin/therapeutic use , Tomography, X-Ray Computed
16.
J Endocrinol ; 212(1): 49-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21987782

ABSTRACT

Pasireotide (SOM230) is a multireceptor-targeted somatostatin analog with high binding affinity for sstr(1,2,3) and sstr(5). The effects of pasireotide and octreotide on blood glucose, insulin, and glucagon levels in rats were evaluated alone and in combination. Single-dose s.c. pasireotide acutely elevated plasma glucose, whereas single-dose s.c. octreotide had no or a small hypoglycemic effect. Glucose elevation with s.c. pasireotide was transient with tachyphylaxis after repeated or continuous administration. Pasireotide and octreotide caused similar inhibitory effects on insulin secretion, whereas pasireotide had a weaker inhibitory effect on glucagon secretion than octreotide. Continuous infusion of pasireotide or injection of pasireotide long-acting release (LAR) resulted in only small and transient elevations of plasma glucose. Based on these results, and differences in the sstr binding affinity of pasireotide vs octreotide, it was hypothesized that the sstr(5) vs sstr(2) receptor activation ratio is the main driver of hyperglycemia after pasireotide. The results also suggest that stronger activation of sstr(2) may counteract the hyperglycemic effect. Indeed, co-administration of octreotide, which has a high affinity for sstr(2), with a hyperglycemic dose of pasireotide did not cause significant changes in plasma glucose levels. In conclusion, although pasireotide and octreotide inhibited insulin to a similar degree, only pasireotide administration was associated with hyperglycemia. The strong glucagon inhibitory effect exhibited by octreotide but not pasireotide may explain this observation. The lack of hyperglycemia during co-administration of pasireotide and octreotide may be explained by the greater activation of sstr(2) compared with pasireotide alone, causing the insulin-glucagon balance to shift within the normoglycemic range. Extrapolation of these data to humans must account for species differences in islet cell sstr expression.


Subject(s)
Blood Glucose/drug effects , Hyperglycemia/chemically induced , Octreotide/administration & dosage , Receptors, Somatostatin/agonists , Somatostatin/analogs & derivatives , Animals , Glucagon/blood , Homeostasis/drug effects , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Rats , Rats, Inbred Lew , Somatostatin/administration & dosage
17.
Neuroendocrinology ; 95(3): 232-47, 2012.
Article in English | MEDLINE | ID: mdl-22156600

ABSTRACT

BACKGROUND: Activation of somatostatin receptors (sstr1-5) by somatostatin and its analogues exerts an inhibitory effect on hormone secretion and provides the basis for the treatment of a range of endocrine diseases such as acromegaly, Cushing's disease and neuroendocrine tumors (NET). The lack of well-characterized commercially available sstr subtype-specific antibodies prevents routine identification of the sstr expression profile in patients. METHODS: We generated and characterized new mouse monoclonal antibodies (mAbs) targeting the five human sstr subtypes using ELISA and immunohistochemistry, and tested their suitability in formalin-fixed and paraffin-embedded (FFPE) human tissues and archival samples of normal pancreatic tissue and NET. RESULTS: All mAbs were highly specific with no cross-reactivity. The sstr1-5 immunoreactivity in gastrointestinal NET (n=67) was correlated with clinicopathologic data. With the exception of sstr3, NET were highly positive for all receptor subtypes (42, 63, 6, 32 and 65% of tumors were positive for sstr1, sstr2a, sstr3, sstr4 and sstr5, respectively). sstr1, sstr2a and sstr5 were present at the plasma membrane and in the cytoplasm of tumor cells, whereas sstr3 and sstr4 were almost exclusively cytoplasmic. Immunoreactivity of sstr1, sstr2a and sstr4 tended to decrease as tumor aggressiveness increased. sstr5 showed an opposite pattern, with higher staining in well-differentiated carcinomas compared with well-differentiated tumors. sstr5 immunoreactivity was correlated with the presence of metastases and angioinvasion, suggesting a possible association with more aggressive behavior. CONCLUSION: Determination of the sstr1-5 by immunohistochemistry using subtype-specific mAbs is feasible in FFPE tissue and may provide a tool for routine clinical practice.


Subject(s)
Antibodies, Monoclonal , Gastrointestinal Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Somatostatin/immunology , Receptors, Somatostatin/metabolism , Adult , Animals , Arabidopsis Proteins/metabolism , Cell Line, Transformed , Enzyme-Linked Immunosorbent Assay , Female , Gastrointestinal Neoplasms/diagnosis , Humans , Immunohistochemistry , Intramolecular Transferases/metabolism , Male , Mice , Middle Aged , Neuroendocrine Tumors/diagnosis , Protein Binding , Receptors, Somatostatin/genetics , Transfection
18.
Future Oncol ; 7(7): 895-913, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21732759

ABSTRACT

Somatostatin receptors (sstrs) are G-protein-coupled receptors that mediate various physiological effects when activated by the neuropeptide somatostatin or its synthetic analogs. In addition to the well-documented antisecretory effects of sstr(2)-preferential somatostatin analogs octreotide and lanreotide, ligand binding to sstr initiates an inhibitory action on tumor growth. This effect may result from both indirect actions (suppression of growth factors and growth-promoting hormones [e.g., GH/IGF-1 axis] and inhibition of angiogenesis) and direct actions (activation of antigrowth activities [e.g., apoptosis]). As solid tumor cells express multiple sstrs, there is a rationale to evaluate the potential antitumor effects of pasireotide (SOM230), a multireceptor-targeted somatostatin analog with high binding affinity for sstr(1-3) and sstr(5). Pasireotide reduces systemic IGF-1 levels more potently than currently available somatostatin analogs and has been well tolerated in clinical trials.


Subject(s)
Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/drug therapy , Somatostatin/therapeutic use
19.
Radiat Res ; 175(6): 728-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21529145

ABSTRACT

The somatostatin analog SOM230 has potent radioprophylactic and radiation mitigating properties that are unrelated to cytoprotection but appear to be due to suppression of secretion of pancreatic enzymes into the intestinal lumen. To determine the maximal postirradiation time window for administration, male CD2F1 mice were exposed to 8.5-11 Gy total-body radiation; SOM230 (0.5, 2 or 5 mg/kg) or vehicle was given by twice daily subcutaneous injections for 14 days, beginning 24-72 h after irradiation, and 30-day animal survival was recorded. The contribution of the gut to systemic cytokine levels was estimated by analyzing plasma samples obtained simultaneously from the portal vein and carotid artery. The effect of SOM230 on cell trypsin secretion was assessed in vitro and intestinal proteolytic activity was measured in vivo. SOM230 was associated with a 40-60% absolute improvement in overall postirradiation survival when treatment was started 48 h after irradiation and even exhibited a statistically significant survival benefit when started at 72 h. SOM230 ameliorated the radiation-induced decrease in chemokine (C-X-C motif) ligand 9 (CXCL9). SOM230 inhibited pancreatic acinar cell trypsin secretion in vitro in a dose-dependent fashion and reduced intraluminal and intestinal tissue proteolytic activity in vivo. SOM230 is an excellent radiation mitigator with a postirradiation time window in excess of 48 h. The mechanism likely involves preservation of intestinal barrier function due to decreased secretion of pancreatic enzymes into the bowel lumen.


Subject(s)
Radiation-Protective Agents/administration & dosage , Somatostatin/analogs & derivatives , Animals , Chemokines/biosynthesis , Dose-Response Relationship, Drug , Interferon-gamma/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Mice , Pancreas/drug effects , Rats , Somatostatin/administration & dosage , Somatostatin/pharmacology , Time Factors , Trypsin/biosynthesis
20.
Neuroendocrinology ; 94(2): 124-36, 2011.
Article in English | MEDLINE | ID: mdl-21525729

ABSTRACT

SOM230 (pasireotide) is a multiligand somatostatin (SRIF) analog able to bind to somatostatin receptor (SSTR) subtypes 1, 2, 3 and 5, and trigger antisecretory and antiproliferative signaling cascades. Canines have become in vivo models to test the pharmacological treatment of corticotropinomas because they frequently develop Cushing's disease in a spontaneous manner, due to adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. Different levels of expression of SSTR2 and SSTR5 have been shown in both mouse AtT20 cells and canine tumoral corticotropinoma cells. The objective of this study was to evaluate whether SOM230 controls both tumor cell growth and hormone synthesis, therefore controlling the disease. SOM230 was tested in dogs suffering from Cushing's disease (10 animals were treated continuously during 6 months, and another 10 were treated with 3 cycles consisting of 2 months of treatment followed by a 2-month rest period). A significant decrease in ACTH, urinary cortisol creatinine ratio, adenoma size (magnetic nuclear resonance) and improvement of clinical signs were obtained, without side effects. AtT20 cells treated with SOM230 suppressed pro-opiomelanocortin (POMC) promoter activity through SSTR2, via the G(i) α-subunit, and reduced Nur77/Nurr1 transcriptional activity. We conclude that SOM230, in addition to its well-described antisecretory effects, inhibits, as shown in AtT20 cells, ACTH synthesis at the POMC transcriptional level, an effect mediated mainly through SSTR2, and limits tumor growth. The controlled Cushing's disease in the dogs that received the treatment indicates that SOM230 has a potential therapeutic use in humans suffering from Cushing's disease.


Subject(s)
Adrenocorticotropic Hormone/biosynthesis , Corticotrophs/drug effects , Corticotrophs/metabolism , Pituitary ACTH Hypersecretion/metabolism , Somatostatin/analogs & derivatives , Adrenocorticotropic Hormone/blood , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Blood Glucose/metabolism , Cell Line, Tumor , Cholesterol/blood , Creatinine/urine , Dogs , Female , Hydrocortisone/urine , Liver Function Tests , Magnetic Resonance Imaging , Male , Pituitary ACTH Hypersecretion/pathology , Somatostatin/pharmacology , Triglycerides/blood , alpha-MSH/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...