Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 524: 232-244, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34637845

RESUMEN

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.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Feocromocitoma/tratamiento farmacológico , Somatostatina/análogos & derivados , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/patología , Catecolaminas/biosíntesis , Catecolaminas/metabolismo , Línea Celular Tumoral , Humanos , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Octreótido/farmacología , Feocromocitoma/metabolismo , Feocromocitoma/patología , Somatostatina/farmacología
2.
Endocrine ; 75(2): 537-548, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34741720

RESUMEN

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.


Asunto(s)
Acromegalia , Octreótido , Acromegalia/tratamiento farmacológico , Adulto , Glucemia , Preparaciones de Acción Retardada/uso terapéutico , Hormona del Crecimiento , Voluntarios Sanos , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Octreótido/efectos adversos , Somatostatina/análogos & derivados
3.
Cancers (Basel) ; 13(12)2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-34205778

RESUMEN

Invasive nonfunctioning pituitary tumors (NFPTs) are non-resectable neoplasms associated with frequent relapse and significant comorbidities. Current treatments, including somatostatin receptor 2 (SSTR2)-directed somatostatin analogs (SSAs), often fail against NFPTs. Thus, identifying effective therapies is clinically relevant. As NFPTs express SSTR3 at high levels, pasireotide, a multireceptor-targeted SSA, might be beneficial. Here we evaluated pasireotide in the only representative model of spontaneous NFPTs (MENX rats) in vivo. Octreotide long-acting release (LAR), pasireotide LAR, or placebo, were administered to age-matched, tumor-bearing MENX rats of both sexes for 28 d or 56 d. Longitudinal high-resolution magnetic resonance imaging monitored tumor growth. While tumors in placebo-treated rats increased in volume over time, PTs in drug-treated rats displayed significant growth suppression, and occasional tumor shrinkage. Pasireotide elicited stronger growth inhibition. Radiological responses correlated with tumors' proliferation rates. Both SSAs, but especially pasireotide, were more effective in female vs. male rats. Basal Sstr3 expression was significantly higher in the former group. It is noteworthy that female human NFPTs patients also have a trend towards higher SSTR3 expression. Altogether, our studies provide the rationale for testing pasireotide in patients with residual/recurrent NFPTs. If confirmed, the sex-related SSTR3 expression might be used as criteria to stratify NFPTs patients for treatment with pasireotide.

4.
Cell Mol Gastroenterol Hepatol ; 11(5): 1405-1436, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33482394

RESUMEN

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.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Secretoma/efectos de los fármacos , Somatostatina/análogos & derivados , Anciano , Anciano de 80 o más Años , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundario , Femenino , Hormonas/farmacología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Somatostatina/farmacología
5.
Zootaxa ; 5071(4): 579-586, 2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-35390893

RESUMEN

The unpublished formal synonymy of Callichroma chiriquina Bates, 1879, C. opiparum Bates, 1874, C. compressipes Casey, 1912, and C. regalis Casey, 1912 is commented. Schwarzerion carinatum Schmidt, 1924 is removed from the synonym of Callichroma holochlorum holochlorum Bates, 1872, and transferred to the synonymy of Callichroma euthalia Bates, 1879. The species group name euthalium is corrected. The type-localities of Schwarzerion carinatum are reported as Colombia and West Indies, and the institution where one of the syntypes is deposited is corrected. Callichroma collarti Fuchs, 1959 is revalidated, and Callichroma magnificum Napp Martins, 2009 is considered its junior synonym. The status of the specimens of the type series of Callichroma collarti is commented. Additionally, the holotype of Callichroma opiparum Bates, 1874, and the lectotype of Callichroma chiriquina Bates, 1879 are illustrated for the first time.


Asunto(s)
Escarabajos , Animales , Estados Unidos
6.
Can Vet J ; 59(10): 1089-1093, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30510314

RESUMEN

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).


Asunto(s)
Enfermedades de los Perros/radioterapia , Adenoma Hipofisario Secretor de Hormona del Crecimiento/veterinaria , Hormonas/uso terapéutico , Somatostatina/análogos & derivados , Acromegalia/etiología , Acromegalia/veterinaria , Adenoma/tratamiento farmacológico , Adenoma/radioterapia , Adenoma/veterinaria , Animales , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Perros , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Adenoma Hipofisario Secretor de Hormona del Crecimiento/radioterapia , Masculino , Somatostatina/uso terapéutico , Resultado del Tratamiento
7.
ESMO Open ; 3(5): e000388, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30094073

RESUMEN

INTRODUCTION: Somatostatin analogues exert antitumour activity via direct and indirect mechanisms. The present study was designed to assess the safety and efficacy of pasireotide in patients with BRAF-wild type (WT) and NRAS-WT metastatic melanoma. PATIENTS AND METHODS: Patients with unresectable and/or metastatic melanoma or Merkel cell carcinoma were eligible. Pasireotide was administered at different doses for ≤8 weeks in dose-escalation phase, followed by long-acting pasireotide 80 mg or lower dose in case of toxicity in follow-up phase up to six additional months. Primary endpoint was safety in the first 8 weeks of dose-escalation phase. RESULTS: The study was terminated early due to slow recruitment. Of the 10 patients with metastatic melanoma enrolled, only four reached the high dose level: two patients reached 3600 µg in dose-escalation and follow-up phases and two patients reached 3600 µg in dose-escalation and long-acting pasireotide 80 mg in follow-up phases and were stable for >5 months. Most common adverse events (AEs) during dose-escalation phase in ≥2 patients (20%) were: diarrhoea (50%), nausea (50%), fatigue (20%), hyperglycaemia (20%), hypophosphatemia (20%), chills (20%) and tumour pain (20%). Grade 3 or 4 study drug-related AEs were diarrhoea and nausea, reported in one patient. Partial response was documented in one patient and stable disease in another. CONCLUSIONS: Pasireotide was well tolerated, and safety results were similar to those previously reported in other indications. Further studies are needed to evaluate its antitumour activity alone and in combination with other drugs in melanoma.

8.
Stress ; 21(4): 370-375, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29661114

RESUMEN

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.


Asunto(s)
Glucemia/análisis , Corticosterona/sangre , Somatostatina/análogos & derivados , Estrés Fisiológico/efectos de los fármacos , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Restricción Física , Somatostatina/farmacología , Transmisión Sináptica
9.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29025873

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/genética , Expresión Génica Ectópica , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Paired Box/genética , Células Secretoras de Somatostatina/metabolismo , Animales , Proliferación Celular , Transdiferenciación Celular/genética , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Terapia Genética/métodos , Glucagón/biosíntesis , Glucagón/genética , Proteínas de Homeodominio/metabolismo , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Transgénicos , Factores de Transcripción Paired Box/metabolismo , Somatostatina/biosíntesis , Somatostatina/genética , Células Secretoras de Somatostatina/citología , Estreptozocina
10.
J Endocrinol ; 231(2): 135-145, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27587848

RESUMEN

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.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Proteínas de Neoplasias/agonistas , Octreótido/farmacología , Hipófisis/efectos de los fármacos , Neoplasias Hipofisarias/tratamiento farmacológico , Receptores de Somatostatina/agonistas , Somatostatina/análogos & derivados , Adenoma Hipofisario Secretor de ACTH/tratamiento farmacológico , Adenoma Hipofisario Secretor de ACTH/metabolismo , Adenoma Hipofisario Secretor de ACTH/patología , Adenoma/tratamiento farmacológico , Adenoma/metabolismo , Adenoma/patología , Antineoplásicos Hormonales/efectos adversos , Señalización del Calcio/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Humanos , Masculino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Octreótido/efectos adversos , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Prolactinoma/tratamiento farmacológico , Prolactinoma/metabolismo , Prolactinoma/patología , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Somatostatina/efectos adversos , Somatostatina/farmacología , Células Tumorales Cultivadas
11.
Oncotarget ; 7(27): 41584-41598, 2016 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-27177087

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) shows a rich stroma where cancer-associated fibroblasts (CAFs) represent the major cell type. CAFs are master secretors of proteins with pro-tumor features. CAF targeting remains a promising challenge for PDA, a devastating disease where treatments focusing on cancer cells have failed. We previously introduced a novel pharmacological CAF-targeting approach using the somatostatin analog SOM230 (pasireotide) that inhibits protein synthesis in CAFs, and subsequent chemoprotective features of CAF secretome. Using primary cultures of CAF isolated from human PDA resections, we here report that CAF secretome stimulates in vitro cancer cell survival, migration and invasive features, that are abolished when CAFs are treated with SOM230. Mechanistically, SOM230 inhibitory effect on CAFs depends on the somatostatin receptor subtype sst1 expressed in CAFs but not in non-activated pancreatic fibroblasts, and on protein synthesis shutdown through eiF4E-Binding Protein-1 (4E-BP1) expression decrease. We identify interleukin-6 as a SOM230-inhibited CAF-secreted effector, which stimulates cancer cell features through phosphoinositide 3-kinase activation. In vivo, mice orthotopically co-xenografted with the human pancreatic cancer MiaPaCa-2 cells and CAFs develop pancreatic tumors, on which SOM230 treatment does not inhibit growth but abrogates metastasis. Consistently, CAF secretome stimulates epithelial-to-mesenchymal transition in cancer cells, which is reversed upon CAF treatment with SOM230. Our results highlight a novel promising anti-metastatic potential for SOM230 indirectly targeting pancreatic cancer cell invasion through pharmacological inhibition of stromal CAFs.


Asunto(s)
Antineoplásicos/uso terapéutico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Somatostatina/análogos & derivados , Animales , Antineoplásicos/farmacología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/fisiología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Somatostatina/farmacología , Somatostatina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Endocrinology ; 157(5): 1789-98, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26990064

RESUMEN

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.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Somatostatina/análogos & derivados , Alelos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Ratones , Neoplasia Endocrina Múltiple/genética , Neoplasia Endocrina Múltiple/patología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Somatostatina/farmacología , Somatostatina/uso terapéutico
13.
Endocrine ; 53(1): 210-9, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26906713

RESUMEN

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.


Asunto(s)
Acromegalia/tratamiento farmacológico , Glucemia , Hormona de Crecimiento Humana/sangre , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Somatostatina/análogos & derivados , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Método Doble Ciego , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Octreótido/uso terapéutico , Péptidos Cíclicos/uso terapéutico , Retratamiento , Somatostatina/farmacología , Somatostatina/uso terapéutico , Insuficiencia del Tratamiento , Resultado del Tratamiento , Adulto Joven
14.
Am J Physiol Regul Integr Comp Physiol ; 309(6): R668-74, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26157058

RESUMEN

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.


Asunto(s)
Receptores de Somatostatina/efectos de los fármacos , Letargo/efectos de los fármacos , Animales , Temperatura Corporal , Peso Corporal , Cricetinae , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Octreótido/farmacología , Phodopus , ARN Mensajero/biosíntesis , Estaciones del Año , Somatostatina/análogos & derivados , Somatostatina/farmacología
15.
Toxicol Appl Pharmacol ; 286(3): 224-33, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981165

RESUMEN

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.


Asunto(s)
Imidazoles/administración & dosificación , Piridinas/administración & dosificación , Somatostatina/análogos & derivados , Somatostatina/administración & dosificación , Esteroide 11-beta-Hidroxilasa/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Sistemas de Liberación de Medicamentos/métodos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Tamaño de los Órganos/fisiología , Ratas , Ratas Wistar , Esteroide 11-beta-Hidroxilasa/metabolismo
16.
EMBO Mol Med ; 7(6): 735-53, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25834145

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is extremely stroma-rich. Cancer-associated fibroblasts (CAFs) secrete proteins that activate survival and promote chemoresistance of cancer cells. Our results demonstrate that CAF secretome-triggered chemoresistance is abolished upon inhibition of the protein synthesis mTOR/4E-BP1 regulatory pathway which we found highly activated in primary cultures of α-SMA-positive CAFs, isolated from human PDAC resections. CAFs selectively express the sst1 somatostatin receptor. The SOM230 analogue (Pasireotide) activates the sst1 receptor and inhibits the mTOR/4E-BP1 pathway and the resultant synthesis of secreted proteins including IL-6. Consequently, tumour growth and chemoresistance in nude mice xenografted with pancreatic cancer cells and CAFs, or with pieces of resected human PDACs, are reduced when chemotherapy (gemcitabine) is combined with SOM230 treatment. While gemcitabine alone has marginal effects, SOM230 is permissive to gemcitabine-induced cancer cell apoptosis and acts as an antifibrotic agent. We propose that selective inhibition of CAF protein synthesis with sst1-directed pharmacological compounds represents an anti-stromal-targeted therapy with promising chemosensitization potential.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Resistencia a Medicamentos , Fibroblastos/fisiología , Fosfoproteínas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Adenocarcinoma/tratamiento farmacológico , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Proteínas de Ciclo Celular , Células Cultivadas , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Xenoinjertos , Humanos , Ratones Desnudos , Fosfoproteínas/antagonistas & inhibidores , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Resultado del Tratamiento , Gemcitabina
17.
Chimia (Aarau) ; 68(7-8): 483-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25437387

RESUMEN

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.


Asunto(s)
Síndrome de Cushing/tratamiento farmacológico , Somatostatina/análogos & derivados , Humanos , Modelos Moleculares , Somatostatina/síntesis química , Somatostatina/química , Somatostatina/uso terapéutico
18.
Am J Physiol Lung Cell Mol Physiol ; 306(12): L1064-77, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727584

RESUMEN

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.


Asunto(s)
Bleomicina/farmacología , Pulmón/patología , Fibrosis Pulmonar/tratamiento farmacológico , Somatostatina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Hidroxiprolina/metabolismo , Inflamación/metabolismo , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Somatostatina/uso terapéutico
19.
Regul Pept ; 187: 35-41, 2013 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-24188818

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Neoplasias Gastrointestinales/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Especificidad de Anticuerpos , Fijadores/química , Formaldehído/química , Neoplasias Gastrointestinales/patología , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Adhesión en Parafina , Conejos , Receptores de Somatostatina/inmunología , Análisis de Matrices Tisulares , Fijación del Tejido
20.
Small ; 9(7): 1042-6, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23239577

RESUMEN

Aluminium/aluminium oxide wires form under microgravity, earth conditions, and hypergravity in different forms. While under 0.04 G the biphasic wires are predominantly linear, they form bundles of wires of high curvature at 1 G and 1.8 G. The absence (0.04 G) and presence (1 G, 1.8 G) of gradients are reflected by the agglomeration and growth direction of the nanowires.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...