Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 309
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768949

RESUMEN

Mild hypercortisolism (mHC) is defined as an excessive cortisol secretion, without the classical manifestations of clinically overt Cushing's syndrome. This condition increases the risk of bone fragility, neuropsychological alterations, hypertension, diabetes, cardiovascular events and mortality. At variance with Cushing's syndrome, mHC is not rare, with it estimated to be present in up to 2% of individuals older than 60 years, with higher prevalence (up to 10%) in individuals with uncontrolled hypertension and/or diabetes or with unexplainable bone fragility. Measuring cortisol after a 1 mg overnight dexamethasone suppression test is the first-line test for searching for mHC, and the degree of cortisol suppression is associated with the presence of cortisol-related consequences and mortality. Among the additional tests used for diagnosing mHC in doubtful cases, the basal morning plasma adrenocorticotroph hormone, 24-h urinary free cortisol and/or late-night salivary cortisol could be measured, particularly in patients with possible cortisol-related complications, such as hypertension and diabetes. Surgery is considered as a possible therapeutic option in patients with munilateral adrenal incidentalomas and mHC since it improves diabetes and hypertension and reduces the fracture risk. In patients with mHC and bilateral adrenal adenomas, in whom surgery would lead to persistent hypocortisolism, and in patients refusing surgery or in whom surgery is not feasible, medical therapy is needed. Currently, promising though scarce data have been provided on the possible use of pituitary-directed agents, such as the multi-ligand somatostatin analog pasireotide or the dopamine agonist cabergoline for the-nowadays-rare patients with pituitary mHC. In the more frequently adrenal mHC, encouraging data are available for metyrapone, a steroidogenesis inhibitor acting mainly against the adrenal 11-ßhydroxylase, while data on osilodrostat and levoketoconazole, other new steroidogenesis inhibitors, are still needed in patients with mHC. Finally, on the basis of promising data with mifepristone, a non-selective glucocorticoid receptor antagonist, in patients with mild cortisol hypersecretion, a randomized placebo-controlled study is ongoing for assessing the efficacy and safety of relacorilant, a selective glucocorticoid receptor antagonist, for patients with mild adrenal hypercortisolism and diabetes mellitus/impaired glucose tolerance and/or uncontrolled systolic hypertension.


Asunto(s)
Síndrome de Cushing/diagnóstico , Síndrome de Cushing/terapia , Neoplasias de las Glándulas Suprarrenales/complicaciones , Síndrome de Cushing/complicaciones , Desarrollo de Medicamentos , Humanos , Hidrocortisona/metabolismo , Modelos Biológicos , Receptores Dopaminérgicos/efectos de los fármacos , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Esteroides/biosíntesis
2.
Mol Brain ; 14(1): 130, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429141

RESUMEN

Somatostatin-expressing interneurons (SOM-INs) are a major subpopulation of GABAergic cells in CA1 hippocampus that receive excitation from pyramidal cells (PCs), and, in turn, provide feedback inhibition onto PC dendrites. Excitatory synapses onto SOM-INs show a Hebbian long-term potentiation (LTP) mediated by type 1a metabotropic glutamate receptors (mGluR1a) that is implicated in hippocampus-dependent learning. The neuropeptide somatostatin (SST) is also critical for hippocampal long-term synaptic plasticity, as well as learning and memory. SST effects on hippocampal PCs are well documented, but its actions on inhibitory interneurons remain largely undetermined. In the present work, we investigate the involvement of SST in long-term potentiation of CA1 SOM-IN excitatory synapses using pharmacological approaches targeting the somatostatinergic system and whole cell recordings in slices from transgenic mice expressing eYFP in SOM-INs. We report that application of exogenous SST14 induces long-term potentiation of excitatory postsynaptic potentials in SOM-INs via somatostatin type 1-5 receptors (SST1-5Rs) but does not affect synapses of PC or parvalbumin-expressing interneurons. Hebbian LTP in SOM-INs was prevented by inhibition of SSTRs and by depletion of SST by cysteamine treatment, suggesting a critical role of endogenous SST in LTP. LTP of SOM-IN excitatory synapses induced by SST14 was independent of NMDAR and mGluR1a, activity-dependent, and prevented by blocking GABAA receptor function. Our results indicate that endogenous SST may contribute to Hebbian LTP at excitatory synapses of SOM-INs by controlling GABAA inhibition, uncovering a novel role for SST in regulating long-term synaptic plasticity in somatostatinergic cells that may be important for hippocampus-dependent memory processes.


Asunto(s)
Región CA1 Hipocampal/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Somatostatina/fisiología , Sinapsis/efectos de los fármacos , Animales , Proteínas Bacterianas , Cisteamina/farmacología , Femenino , Antagonistas de Receptores de GABA-A/farmacología , Neuronas GABAérgicas/metabolismo , Técnicas de Sustitución del Gen , Genes Reporteros , Humanos , Interneuronas/metabolismo , Proteínas Luminiscentes , Masculino , Memoria/fisiología , Ratones , Ratones Transgénicos , Péptidos Cíclicos/farmacología , Receptores de Glutamato Metabotrópico/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/fisiología , Somatostatina/farmacología , Sinapsis/fisiología
3.
Sci Rep ; 11(1): 10962, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34040115

RESUMEN

Neuropeptides have been reported to regulate progenitor proliferation and neurogenesis in the central nervous system. However, these studies have typically been conducted using pharmacological agents in ex vivo preparations, and in vivo evidence for their developmental function is generally lacking. Recent scRNA-Seq studies have identified multiple neuropeptides and their receptors as being selectively expressed in neurogenic progenitors of the embryonic mouse and human retina. This includes Sstr2, whose ligand somatostatin is transiently expressed by immature retinal ganglion cells. By analyzing retinal explants treated with selective ligands that target these receptors, we found that Sstr2-dependent somatostatin signaling induces a modest, dose-dependent inhibition of photoreceptor generation, while correspondingly increasing the relative fraction of primary progenitor cells. These effects were confirmed by scRNA-Seq analysis of retinal explants but abolished in Sstr2-deficient retinas. Although no changes in the relative fraction of primary progenitors or photoreceptor precursors were observed in Sstr2-deficient retinas in vivo, scRNA-Seq analysis demonstrated accelerated differentiation of neurogenic progenitors. We conclude that, while Sstr2 signaling may act to negatively regulate retinal neurogenesis in combination with other retinal ganglion cell-derived secreted factors such as Shh, it is dispensable for normal retinal development.


Asunto(s)
Proteínas del Ojo/fisiología , Neurogénesis/fisiología , Neuropéptidos/fisiología , Receptores de Somatostatina/fisiología , Retina/citología , Animales , Relación Dosis-Respuesta a Droga , Proteínas del Ojo/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Edad Gestacional , Humanos , Ligandos , Ratones , Ratones Noqueados , Neuropéptidos/agonistas , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/farmacología , Fenotipo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/efectos de los fármacos , Retina/embriología , Transducción de Señal/fisiología , Análisis de la Célula Individual
4.
Elife ; 102021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33650969

RESUMEN

Primary cilia are compartmentalized sensory organelles present on the majority of neurons in the mammalian brain throughout adulthood. Recent evidence suggests that cilia regulate multiple aspects of neuronal development, including the maintenance of neuronal connectivity. However, whether ciliary signals can dynamically modulate postnatal circuit excitability is unknown. Here we show that acute cell-autonomous knockdown of ciliary signaling rapidly strengthens glutamatergic inputs onto cultured rat neocortical pyramidal neurons and increases spontaneous firing. This increased excitability occurs without changes to passive neuronal properties or intrinsic excitability. Further, the neuropeptide receptor somatostatin receptor 3 (SSTR3) is localized nearly exclusively to excitatory neuron cilia both in vivo and in culture, and pharmacological manipulation of SSTR3 signaling bidirectionally modulates excitatory synaptic inputs onto these neurons. Our results indicate that ciliary neuropeptidergic signaling dynamically modulates excitatory synapses and suggest that defects in this regulation may underlie a subset of behavioral and cognitive disorders associated with ciliopathies.


Asunto(s)
Cilios/efectos de los fármacos , Células Piramidales/fisiología , Receptores de Somatostatina/efectos de los fármacos , Sinapsis/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Neocórtex/citología , Técnicas de Placa-Clamp , Ratas Long-Evans , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inhibidores , Transducción de Señal
5.
Front Endocrinol (Lausanne) ; 12: 793262, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35058882

RESUMEN

Somatostatin exhibits an inhibitory effect on pituitary hormone secretion, including inhibition of growth hormone and adrenocorticotropic hormone (ACTH), and it can have antisecretory and antitumor effects on neuroendocrine tumors (NETs) that express somatostatin receptors. Although the precise mechanism remains unclear, the finding that glucocorticoids downregulate somatostatin receptor subtype 2 (SSTR2) expression has been used to explain the lack of efficacy of traditional SSTR2-targeting analogs in patients with ACTH-secreting NETs. Glucocorticoid receptor (GR) antagonism with mifepristone has been shown to reverse the glucocorticoid-induced downregulation of SSTR2; however, the effects of GR modulation on SSTR2 expression in ACTH-secreting NETs, particularly corticotroph pituitary tumors, are not well known. The current study presents new insight from in vitro data using the highly selective GR modulator relacorilant, showing that GR modulation can overcome dexamethasone-induced suppression of SSTR2 in the murine At-T20 cell line. Additional data presented from clinical case observations in patients with ACTH-secreting NETs suggest that upregulation of SSTR2 via GR modulation may re-sensitize tumors to endogenous somatostatin and/or somatostatin analogs. Clinical, laboratory, and imaging findings from 4 patients [2 ACTH-secreting bronchial tumors and 2 ACTH-secreting pituitary tumors (Cushing disease)] who were treated with relacorilant as part of two clinical studies (NCT02804750 and NCT02762981) are described. In the patients with ectopic ACTH secretion, SSTR2-based imaging (Octreoscan and 68Ga-DOTATATE positron emission tomography) performed before and after treatment with relacorilant showed increased radiotracer uptake by the tumor following treatment with relacorilant without change in tumor size at computed tomography. In the patients with Cushing disease who received relacorilant prior to scheduled pituitary surgery, magnetic resonance imaging after a 3-month course of relacorilant showed a reduction in tumor size. Based on these findings, we propose that GR modulation in patients with ACTH-secreting NETs upregulates previously suppressed SSTR2s, resulting in tumor-specific antisecretory and anti-proliferative effects. The effect of relacorilant on pituitary corticotroph tumors is being investigated in an ongoing phase 3 study (NCT03697109; EudraCT 2018-003096-35).


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/tratamiento farmacológico , Adenoma/tratamiento farmacológico , Hormona Adrenocorticotrópica/metabolismo , Neoplasias de los Bronquios/tratamiento farmacológico , Isoquinolinas/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Pirazoles/farmacología , Piridinas/farmacología , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/metabolismo , Adenoma Hipofisario Secretor de ACTH/diagnóstico por imagen , Adenoma Hipofisario Secretor de ACTH/metabolismo , Adenoma/diagnóstico por imagen , Adenoma/metabolismo , Adulto , Anciano , Animales , Neoplasias de los Bronquios/diagnóstico por imagen , Neoplasias de los Bronquios/metabolismo , Línea Celular Tumoral , Dexametasona/farmacología , Regulación hacia Abajo , Femenino , Glucocorticoides/farmacología , Humanos , Técnicas In Vitro , Masculino , Ratones , Persona de Mediana Edad , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/metabolismo , Compuestos Organometálicos , Tomografía de Emisión de Positrones , Radiofármacos , Somatostatina/análogos & derivados
6.
Radiographics ; 39(7): 2069-2084, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31697628

RESUMEN

Merkel cell carcinoma (MCC) is a rare and aggressive cutaneous neuroendocrine tumor with a higher mortality rate than melanoma. Approximately 40% of MCC patients have nodal or distant metastasis at initial presentation, and one-third of patients will develop distant metastatic disease over their clinical course. Although MCC is rare, its incidence has been steadily increasing. Furthermore, the immunogenicity of MCC and its diagnostic and therapeutic application have made MCC one of the most rapidly developing topics in dermatology and oncology. Owing to the aggressive and complex nature of MCC, a multidisciplinary approach is necessary for management of this tumor, including dermatologists, surgeons, radiation oncologists, medical oncologists, pathologists, radiologists, and nuclear medicine physicians. Imaging plays a crucial role in diagnosis, planning for surgery or radiation therapy, and assessment of treatment response and surveillance. However, MCC is still not well recognized among radiologists and nuclear medicine physicians, likely owing to its rarity. The purpose of this review is to raise awareness of MCC among imaging experts by describing the epidemiology, pathophysiology, and clinical features of MCC and current clinical management with a focus on the role of imaging. The authors highlight imaging findings characteristic of MCC, as well as the clinical significance of CT, MRI, sentinel lymph node mapping, fluorine 18 fluorodeoxyglucose PET/CT, and other nuclear medicine studies such as bone scintigraphy and somatostatin receptor scintigraphy. ©RSNA, 2019.


Asunto(s)
Carcinoma de Células de Merkel/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Neoplasias Cutáneas/diagnóstico por imagen , Anticuerpos Antivirales/sangre , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/secundario , Carcinoma de Células de Merkel/secundario , Carcinoma de Células de Merkel/virología , Humanos , Metástasis Linfática/diagnóstico por imagen , Poliomavirus de Células de Merkel/aislamiento & purificación , Estadificación de Neoplasias , Proteínas Oncogénicas/inmunología , Infecciones por Polyomavirus/diagnóstico por imagen , Infecciones por Polyomavirus/virología , Pronóstico , Radiofármacos/análisis , Radiofármacos/farmacocinética , Receptores de Somatostatina/efectos de los fármacos , Biopsia del Ganglio Linfático Centinela , Neoplasias Cutáneas/virología , Infecciones Tumorales por Virus/diagnóstico por imagen , Infecciones Tumorales por Virus/virología , Proteínas Virales/inmunología
7.
J Med Chem ; 62(5): 2708-2719, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30735385

RESUMEN

Somatostatin receptor 2 (SSTR2) is frequently overexpressed on several types of solid tumors, including neuroendocrine tumors and small-cell lung cancer. Peptide agonists of SSTR2 are rapidly internalized upon binding to the receptor and linking a toxic payload to an SSTR2 agonist is a potential method to kill SSTR2-expressing tumor cells. Herein, we describe our efforts towards an efficacious SSTR2-targeting cytotoxic conjugate; examination of different SSTR2-targeting ligands, conjugation sites, and payloads led to the discovery of 22 (PEN-221), a conjugate consisting of microtubule-targeting agent DM1 linked to the C-terminal side chain of Tyr3-octreotate. PEN-221 demonstrates in vitro activity which is both potent (IC50 = 10 nM) and receptor-dependent (IC50 shifts 90-fold upon receptor blockade). PEN-221 targets high levels of DM1 to SSTR2-expressing xenograft tumors, which has led to tumor regressions in several SSTR2-expressing xenograft mouse models. The safety and efficacy of PEN-221 is currently under evaluation in human clinical trials.


Asunto(s)
Descubrimiento de Drogas , Maitansina/farmacología , Receptores de Somatostatina/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Células CHO , Línea Celular , Cricetulus , Perros , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacología , Maitansina/química , Maitansina/farmacocinética , Ratones , Receptores de Somatostatina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Endocrinol Metab Clin North Am ; 47(3): 615-625, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30098719

RESUMEN

Peptide receptor radionuclide therapy is a form of systemic radiotherapy shown to be effective in treating neuroendocrine tumors expressing somatostatin receptors. The NETTER-1 trial was the first randomized phase III clinical trial evaluating a radiolabeled somatostatin analog, and demonstrated significant improvement in progression-free survival among patients with midgut neuroendocrine tumors treated with 177Lu-DOTATATE versus high-dose octreotide. This article discusses the evolution of peptide receptor radionuclide therapy, side effects, and potential future treatment approaches.


Asunto(s)
Tumores Neuroendocrinos/radioterapia , Radiofármacos/uso terapéutico , Radioterapia/métodos , Receptores de Péptidos , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Humanos , Radiofármacos/efectos adversos , Radioterapia/tendencias , Receptores de Somatostatina/efectos de los fármacos , Somatostatina/efectos adversos
9.
Psychopharmacology (Berl) ; 235(7): 2137-2149, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29713785

RESUMEN

RATIONALE: The role of somatostatin and its receptors for the stress-related neuropsychiatric disorders has been widely raised. Recently, we have also demonstrated the involvement of somatostatin receptor type 2-sst2R and dopamine receptor type 2-D2R in stress. OBJECTIVE: In this context, we decided to find if these receptors are involved in response to antidepressant treatment in animal model of depression-chronic mild stress (CMS). METHODS: Here, we report data obtained following 7-week CMS procedure. The specific binding of [125I]Tyr3-Octreotide to sst2R and [3H]Domperidone to D2R was measured in the rat brain, using autoradiography. Additionally, the level of dopamine and metabolites was measured in the rat brain. RESULTS: In the final baseline test after 7 weeks of stress, the reduced consumption of sucrose solution was observed (controls vs the stressed animals (6.25 0.16 vs. 10.39 0.41; p < 0.05). Imipramine was administered for the next 5 weeks, and it reversed anhedonia in majority of animals (imipramine-reactive); however, in some animals, it did not (imipramine-non-reactive). Two-way repeated measures ANOVA revealed significant effects of stress and treatment and time interaction [F(16, 168) = 3.72; p < 0.0001], n = 10 per groups. We observed decreased binding of [125I]Tyr3-Octreotide in most of rat brain regions in imipramine non-reactive groups of animals. The decrease of D2R after stress in striatum and nucleus accumbens and no effect of imipramine were observed. In the striatum and prefrontal cortex, the significant role of stress and imipramine in dopamine levels was observed. CONCLUSIONS: The results obtained in binding assays, together with dopamine level, indicate the involvement of sst2R receptors for reaction to antidepressant treatment. Besides, the stress context itself changes the effect of antidepressant drug.


Asunto(s)
Encéfalo/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Estrés Psicológico/metabolismo , Anhedonia/efectos de los fármacos , Animales , Antidepresivos/farmacología , Autorradiografía , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Depresión/diagnóstico por imagen , Depresión/metabolismo , Modelos Animales de Enfermedad , Domperidona/metabolismo , Dopamina/metabolismo , Imipramina/farmacología , Radioisótopos de Yodo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Octreótido/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Estrés Psicológico/diagnóstico por imagen , Sacarosa , Tritio
10.
Brain Res Bull ; 135: 92-97, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28987282

RESUMEN

The interaction between the dopaminergic and somatostatinergic systems is considered to play a potential role in mood regulation. Chronic administration of antidepressants influences release of both neurotransmitters. The molecular basis of the functional cooperation may stem from the physical interaction of somatostatin receptor subtypes and dopamine D2 receptors since they colocalize in striatal interneurons and were shown to undergo ligand-dependent heterodimerization in heterologous expression systems. In present study we adapted in situ proximity ligation assay to investigate the occurrence of D2-Sst5 receptor heterocomplexes, and their possible alterations in the striatum of mice treated acutely and repeatedly (21days) with antidepressant drugs of different pharmacological profiles (escitalopram and desipramine). Additionally we analysed number of heterocomplexes in primary striatal neuronal cultures incubated with both antidepressant drugs for 1h and 6days. The studies revealed that antidepressants increase formation of D2-Sst5 receptors heterodimers. These findings provide interesting evidence that dopamine D2 and somatostatin Sst5 heterodimers may be considered as potential mediators of antidepressant effects, since the heterodimerization of these receptors occurs in native brain tissue as well as in primary striatal neuronal cultures where receptors are expressed at physiological levels.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Animales , Antidepresivos/metabolismo , Antidepresivos/farmacología , Técnicas de Cultivo de Célula , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/efectos de los fármacos , Neostriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo
11.
Endocrine ; 58(1): 124-133, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28822091

RESUMEN

PURPOSE: Somatostatin analogs are widely used to treat conditions associated with hormonal hypersecretion such as acromegaly and metastatic neuroendocrine tumors. First generation somatostatin analogs, such as octreotide and lanreotide, have high affinity for somatostatin receptor subtype 2 (SSTR2), but have incomplete efficacy in many patients. Pasireotide targets multiple SSTRs, having the highest affinity for SSTR5, but causes hyperglycemia and diabetes mellitus in preclinical and clinical studies. AP102 is a new somatostatin analogs with high affinity at both SSTR2 and SSTR5. We aimed to characterize the effects of AP102 vs. pasireotide on random and dynamic glucose levels, glucoregulatory hormone concentrations and growth axis measures in healthy Sprague-Dawley rats. METHODS: Three doses of each compound were evaluated under acute conditions (1, 10, and 30 µg/kg s.c.), and two doses during a chronic (4-week) infusion (3 and 10 µg/kg/h s.c.). RESULTS: Neither acute nor chronic AP102 administration altered blood glucose concentrations or dynamic responses following an intraperitoneal glucose tolerance test. In contrast, acute and chronic pasireotide dosing increased random and post-intraperitoneal glucose tolerance test blood glucose measures, compared to vehicle-treated controls. Both AP102 and pasireotide acutely suppressed growth hormone levels, although insulin-like growth factor-1 and somatic growth was suppressed to a greater extent with pasireotide. CONCLUSIONS: AP102 is a new dual SSTR2/SSTR5-specific somatostatin analog that acutely reduces growth hormone but does not cause hyperglycemia during acute or chronic administration in a healthy rat model. Further studies in diabetic animals and in humans are necessary to determine the potential utility of AP102 in the clinical setting.


Asunto(s)
Glucosa/metabolismo , Receptores de Somatostatina/efectos de los fármacos , Somatostatina/análogos & derivados , Somatostatina/farmacología , Animales , Prueba de Tolerancia a la Glucosa , Crecimiento/efectos de los fármacos , Hormona del Crecimiento/sangre , Hormonas/metabolismo , Infusiones Subcutáneas , Insulina/sangre , Masculino , Ratas , Ratas Sprague-Dawley
12.
PLoS One ; 12(1): e0170536, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107508

RESUMEN

BACKGROUND: The gastrin releasing peptide receptor (GRPR) and the somatostatin receptor 2 (SSTR2) are overexpressed on primary breast cancer (BC), making them ideal candidates for receptor-mediated nuclear imaging and therapy. The aim of this study was to determine whether these receptors are also suitable targets for metastatic BC. METHODS: mRNA expression of human BC samples were studied by in vitro autoradiography and associated with radioligand binding. Next, GRPR and SSTR2 mRNA levels of 60 paired primary BCs and metastases from different sites were measured by quantitative reverse transcriptase polymerase chain reaction. Receptor mRNA expression levels were associated with clinico-pathological factors and expression levels of primary tumors and corresponding metastases were compared. RESULTS: Binding of GRPR and SSTR radioligands to tumor tissue correlated significantly with receptor mRNA expression. High GRPR and SSTR2 mRNA levels were associated with estrogen receptor (ESR1)-positive tumors (p<0.001 for both receptors). There was no significant difference in GRPR mRNA expression of primary tumors versus paired metastases. Regarding SSTR2 mRNA expression, there was also no significant difference in the majority of cases, apart from liver and ovarian metastases which showed a significantly lower expression compared to the corresponding primary tumors (p = 0.02 and p = 0.03, respectively). CONCLUSION: Targeting the GRPR and SSTR2 for nuclear imaging and/or treatment has the potential to improve BC care in primary as well as metastatic disease.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Receptores de Bombesina/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Autorradiografía , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Técnicas In Vitro , Cintigrafía/métodos , Receptores de Bombesina/metabolismo , Receptores de Somatostatina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Oncotarget ; 7(50): 83451-83461, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27825139

RESUMEN

Peptide hormone-based targeted therapy to tumors has been studied extensively. Our previous study shows that somatostatin receptor expresses high level on drug-resistant human ovarian cancer. The paclitaxel-octreotide conjugate (POC) exhibits enhanced growth inhibition, as well as reduced toxicity, in paclitaxel-resistant human ovarian cancer cells. The aim of this study was to investigate the effect of targeted cytotoxicity and potential reversal mechanism of resistance in paclitaxel-resistant human ovarian cancer cells xenografted into nude mice. The SSTR2 shows higher expression levels in tumor tissue. Moreover, fluorescein-labeled POC displays favorable targeting in tumor cells. POC presents the perfect efficacy in inhibiting tumor growth and exerts lower or no toxic effects on normal tissues. Real-time PCR and Western Blotting has demonstrated that the mRNA and protein expressions of SSTR2 in POC group were significantly higher, while MDR1, α-tubulin, ßIII-tubulin, VEGF and MMP-9 were significantly lower than in the other treatment groups and controls. Combined with the previous study in vitro, this study evaluates an effective approach on the treatment of paclitaxel-resistant ovarian cancer which expresses somatostatin receptor SSTR. Our investigation has also revealed the possible molecular mechanism of POC in treating the ovarian cancer, and therefore, provided a theoretical basis for the clinical application of this newly-invented compound.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Octreótido/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , Taxoides/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Theranostics ; 6(11): 1821-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27570553

RESUMEN

Metastases expressing tumor-specific receptors can be targeted and treated by binding of radiolabeled peptides (peptide receptor radionuclide therapy or PRRT). For example, patients with metastasized somatostatin receptor-positive neuroendocrine tumors (NETs) can be treated with radiolabeled somatostatin analogues, resulting in strongly increased progression-free survival and quality of life. There is nevertheless still room for improvement, as very few patients can be cured at this stage of disease. We aimed to specifically sensitize replicating tumor cells without further damage to healthy tissues. Thereto we investigated the DNA damaging effects of PRRT with the purpose to enhance these effects through modulation of the DNA damage response. Although PRRT induces DNA double strand breaks (DSBs), a larger fraction of the induced lesions are single strand breaks (expected to be similar to those induced by external beam radiotherapy) that require poly-[ADP-ribose]-polymerase 1 (PARP-1) activity for repair. If these breaks cannot be repaired, they will cause replication fork arrest and DSB formation during replication. Therefore, we used the PARP-1 inhibitor Olaparib to increase the number of cytotoxic DSBs. Here we show that this new combination strategy synergistically sensitized somatostatin receptor expressing cells to PRRT. We observed increased cell death and reduced cellular proliferation compared to the PRRT alone. The enhanced cell death was caused by increased numbers of DSBs that are repaired with remarkably slow kinetics, leading to genome instability. Furthermore, we validated the increased DSB induction after PARP inhibitor addition in the clinically relevant model of living human NET slices. We expect that this combined regimen can thus augment current PRRT outcomes.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/radioterapia , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/radioterapia , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Sinergismo Farmacológico , Inestabilidad Genómica , Humanos , Lutecio , Radioisótopos , Radioterapia/métodos , Receptores de Somatostatina/efectos de los fármacos
15.
Endocrinology ; 157(2): 692-704, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26671185

RESUMEN

To determine the comprehensive G protein-coupled receptor (GPCR) expression profile in ghrelin-producing cells and to elucidate the role of GPCR-mediated signaling in the regulation of ghrelin secretion, we determined GPCR expression profiles by RNA sequencing in the ghrelin-producing cell line MGN3-1 and analyzed the effects of ligands for highly expressed receptors on intracellular signaling and ghrelin secretion. Expression of selected GPCRs was confirmed in fluorescence-activated cell-sorted fluorescently tagged ghrelin-producing cells from ghrelin-promoter CreERT2/Rosa-CAG-LSL-ZsGreen1 mice. Expression levels of GPCRs previously suggested to regulate ghrelin secretion including adrenergic-ß1 receptor, GPR81, oxytocin receptor, GPR120, and somatostatin receptor 2 were high in MGN3-1 cells. Consistent with previous reports, isoproterenol and oxytocin stimulated the Gs and Gq pathways, respectively, whereas lactate, palmitate, and somatostatin stimulated the Gi pathway, confirming the reliability of current assays. Among other highly expressed GPCRs, prostaglandin E receptor 4 agonist prostaglandin E2 significantly stimulated the Gs pathway and ghrelin secretion. Muscarine, the canonical agonist of cholinergic receptor muscarinic 4, stimulated both the Gq and Gi pathways. Although muscarine treatment alone did not affect ghrelin secretion, it did suppress forskolin-induced ghrelin secretion, suggesting that the cholinergic pathway may play a role in counterbalancing the stimulation of ghrelin by Gs (eg, by adrenaline). In addition, GPR142 ligand tryptophan stimulated ghrelin secretion. In conclusion, we determined the comprehensive expression profile of GPCRs in ghrelin-producing cells and identified two novel ghrelin regulators, prostaglandin E2 and tryptophan. These results will lead to a greater understanding of the physiology of ghrelin and facilitate the development of ghrelin-modulating drugs.


Asunto(s)
Mucosa Gástrica/metabolismo , Ghrelina/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Agonistas Adrenérgicos beta/farmacología , Animales , Línea Celular Tumoral , Colforsina/farmacología , Dinoprostona/farmacología , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Perfilación de la Expresión Génica , Ghrelina/efectos de los fármacos , Hormonas/farmacología , Inmunohistoquímica , Isoproterenol/farmacología , Ácido Láctico/farmacología , Ratones , Ratones Transgénicos , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Oxitócicos/farmacología , Oxitocina/farmacología , Palmitatos/farmacología , Receptor Muscarínico M4/agonistas , Receptores Adrenérgicos beta 1/efectos de los fármacos , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Oxitocina/efectos de los fármacos , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Análisis de Secuencia de ARN , Somatostatina/farmacología , Triptófano/farmacología
16.
J Nucl Med ; 57(4): 503-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26678617

RESUMEN

UNLABELLED: In peptide receptor radionuclide therapy with (90)Y-labeled DOTATATE, the kidney absorbed dose limits the maximum amount of total activity that can be safely administered in many patients. A higher tumor-to-kidney absorbed dose ratio might be achieved by optimizing the amount of injected peptide and activity, as recent studies have shown different degrees of receptor saturation for normal tissue and tumor. The aim of this work was to develop and implement a modeling method for treatment planning to determine the optimal combination of peptide amount and pertaining therapeutic activity for each patient. METHODS: A whole-body physiologically based pharmacokinetic (PBPK) model was developed. General physiologic parameters were taken from the literature. Individual model parameters were fitted to a series (n= 12) of planar γ-camera and serum measurements ((111)In-DOTATATE) of patients with meningioma or neuroendocrine tumors (NETs). Using the PBPK model and the individually estimated parameters, we determined the tumor, liver, spleen, and red marrow biologically effective doses (BEDs) for a maximal kidney BED (20 Gy2.5) for different peptide amounts and activities. The optimal combination of peptide amount and activity for maximal tumor BED, considering the additional constraint of a red marrow BED less than 1 Gy15, was individually quantified. RESULTS: The PBPK model describes the biokinetic data well considering the criteria of visual inspection, the coefficients of determination, the relative standard errors (<50%), and the correlation of the parameters (<0.8). All fitted parameters were in a physiologically reasonable range but varied considerably between patients, especially tumor perfusion (meningioma, 0.1-1 mL·g(-1)·min(-1), and NETs, 0.02-1 mL·g(-1)·min(-1)) and receptor density (meningioma, 5-34 nmol·L(-1), and NETs, 7-35 nmol·L(-1)). Using the proposed method, we identified the optimal amount and pertaining activity to be 76 ± 46 nmol (118 ± 71 µg) and 4.2 ± 1.8 GBq for meningioma and 87 ± 50 nmol (135 ± 78 µg) and 5.1 ± 2.8 GBq for NET patients. CONCLUSION: The presented work suggests that to achieve higher efficacy and safety for (90)Y-DOATATE therapy, both the administered amount of peptide and the activity should be optimized in treatment planning using the proposed method. This approach could also be adapted for therapy with other peptides.


Asunto(s)
Octreótido/análogos & derivados , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/uso terapéutico , Radiofármacos/administración & dosificación , Radiofármacos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Médula Ósea/diagnóstico por imagen , Simulación por Computador , Femenino , Cámaras gamma , Humanos , Riñón/metabolismo , Masculino , Meningioma/radioterapia , Persona de Mediana Edad , Modelos Biológicos , Modelos Teóricos , Tumores Neuroendocrinos/radioterapia , Octreótido/administración & dosificación , Octreótido/farmacocinética , Octreótido/uso terapéutico , Compuestos Organometálicos/farmacocinética , Cintigrafía , Radiofármacos/farmacocinética , Receptores de Somatostatina/efectos de los fármacos , Flujo Sanguíneo Regional , Distribución Tisular , Radioisótopos de Itrio/uso terapéutico
17.
Drug Deliv ; 23(1): 285-96, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-24865288

RESUMEN

Somatostatin receptor 2 (SSTR2), specifically over-expressed on many tumor cells, is a potential receipt for active targeting in cancer therapy. In the present study, octreotide (Oct), which had high affinity to SSTR2, was attached to N-(2-hydroxypropyl) methacrylamide (HPMA) polymeric system to enhance the antitumor efficiency of the anticancer drug doxorubicin (DOX). Two kinds of cell lines (HepG2 and A549), which overexpress SSTR2, were chosen as cell models. Compared with non-modified conjugates, Oct-modified conjugates exhibited superior cytotoxicity and intracellular uptake on both HepG2 and A549 cell lines. This might be due to the mechanism of receptor-mediated endocytosis. Subsequently, the in vivo biodistribution and antitumor activity evaluations showed that Oct modification significantly improved the tumor accumulation and antitumor efficacy of HPMA copolymer conjugates in SSTR2 over-expressed Kunming mice bearing H22 tumor xenografts. In summary, Oct-modified HPMA polymer-DOX conjugates might be a promising system for the treatment of SSTR2 over-expressed cancers.


Asunto(s)
Acrilamidas/farmacología , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Metacrilatos/química , Octreótido/química , Receptores de Somatostatina/efectos de los fármacos , Acrilamidas/farmacocinética , Animales , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos , Humanos , Masculino , Metacrilatos/farmacocinética , Ratones , Octreótido/farmacocinética , Receptores de Somatostatina/metabolismo , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Am J Psychiatry ; 172(11): 1131-40, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26238605

RESUMEN

OBJECTIVE: Gene expression dysregulation in the brain has been associated with bipolar disorder through candidate gene and microarray expression studies, but questions remain about isoform-specific dysregulation and the role of noncoding RNAs whose importance in the brain has been suggested recently but not yet characterized for bipolar disorder. METHOD: The authors used RNA sequencing, a powerful technique that captures the complexity of gene expression, in postmortem tissue from the anterior cingulate cortex from 13 bipolar disorder case subjects and 13 matched comparison subjects. Differential expression was computed, and a global pattern of downregulation was detected, with 10 transcripts significant at a false discovery rate ≤5%. Importantly, all 10 genes were also replicated in an independent RNA sequencing data set (N=61) from the anterior cingulate cortex. RESULTS: Among the most significant results were genes coding for class A G protein-coupled receptors: SSTR2 (somatostatin receptor 2), CHRM2 (cholinergic receptor, muscarinic 2), and RXFP1 (relaxin/insulin-like family peptide receptor 1). A gene ontology analysis of the entire set of differentially expressed genes pointed to an overrepresentation of genes involved in G protein-coupled receptor regulation. The top genes were followed up by querying the effect of treatment with mood stabilizers commonly prescribed in bipolar disorder, which showed that these drugs modulate expression of the candidate genes. CONCLUSIONS: By using RNA sequencing in the postmortem bipolar disorder brain, an interesting profile of G protein-coupled receptor dysregulation was identified, several new bipolar disorder genes were indicated, and the noncoding transcriptome in bipolar disorder was characterized. These findings have important implications with regard to fine-tuning our understanding of the bipolar disorder brain, as well as for identifying potential new drug target pathways.


Asunto(s)
Trastorno Bipolar/genética , Regulación de la Expresión Génica/genética , Giro del Cíngulo/metabolismo , ARN Mensajero/metabolismo , Receptor Muscarínico M2/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Receptores de Somatostatina/genética , Adulto , Antimaníacos/farmacología , Trastorno Bipolar/metabolismo , Carbamazepina/farmacología , Estudios de Casos y Controles , Línea Celular , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Compuestos de Litio/farmacología , Masculino , Persona de Mediana Edad , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , ARN Mensajero/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Muscarínico M2/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Péptidos/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Análisis de Secuencia de ARN , Ácido Valproico/farmacología
19.
J Nucl Med ; 56(10): 1487-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26251419

RESUMEN

UNLABELLED: Imaging and therapy using radioligands targeting receptors overexpressed on tumor cells is successfully applied in neuroendocrine tumor patients. Because expression of the gastrin-releasing peptide receptor (GRPR), somatostatin receptor 2 (SSTR2), and chemokine C-X-C motif receptor 4 (CXCR4) has been demonstrated in breast cancer, targeting these receptors using radioligands might offer new imaging and therapeutic opportunities for breast cancer patients. The aim of this study was to correlate messenger RNA (mRNA) expression of GRPR, SSTR2, and CXCR4 with clinicopathologic and biologic factors, and with prognosis and prediction to therapy response, in order to identify specific breast cancer patient groups suited for the application of radioligands targeting these receptors. METHODS: First, we studied GRPR and SSTR2 expression in 13 clinical breast cancer specimens by in vitro autoradiography and correlated this with corresponding mRNA levels to investigate whether mRNA levels reliably represent cell surface expression. Next, GRPR, SSTR2, and CXCR4 mRNA levels were measured by quantitative reverse transcriptase polymerase chain reaction in 915 primary breast cancer tissues and correlated with known clinicopathologic and biologic factors, disease-free survival, distant metastasis-free survival, and overall survival (DFS, MFS, and OS, respectively). In 224 adjuvant hormonal treatment-naïve estrogen receptor (ER, ESR1)-positive patients who received tamoxifen as first-line therapy for recurrent or metastatic disease, the expression levels of the receptors were correlated with progression-free survival. RESULTS: Our results showed a significant positive correlation between GRPR and SSTR2 expression analyzed by in vitro autoradiography and by quantitative reverse transcriptase polymerase chain reaction (Spearman's rank correlation coefficient [Rs]=0.94, P<0.001, and Rs=0.73, P=0.0042, respectively). Furthermore, high GRPR and SSTR2 mRNA levels were observed more frequently in ESR1-positive specimens, whereas high CXCR4 expression was associated with ESR1-negative specimens. Also, high mRNA expression of CXCR4 was associated with a prolonged DFS, MFS, and OS (multivariate hazard ratio MFS=0.76 [95% confidence interval, 0.64-0.90], P=0.001), whereas high mRNA levels of GRPR were associated with a prolonged progression-free survival after the start of first-line tamoxifen treatment (multivariate hazard ratio=0.68 [95% confidence interval, 0.48-0.97], P=0.031). CONCLUSION: Our data indicate that imaging and therapy using GRPR or SSTR2 radioligands might especially be beneficial for ESR1-positive breast cancer and CXCR4 radioligands for ESR1-negative breast cancer.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Receptores de Bombesina/efectos de los fármacos , Receptores CXCR4/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Adulto , Anciano , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/uso terapéutico , Autorradiografía , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Persona de Mediana Edad , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Cintigrafía , Radiofármacos , Análisis de Supervivencia , Tamoxifeno/administración & dosificación , Tamoxifeno/uso terapéutico
20.
J Nucl Med ; 56(11): 1748-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26294303

RESUMEN

UNLABELLED: The standard treatment of meningiomas is surgery or radiotherapy. Complex, especially recurrent or progressive cases, may exhibit tumor growth involving critical neurovascular structures or diffuse growth, resulting in limited efficacy and higher risk of standard treatment. We evaluated whether somatostatin receptor-targeted radionuclide therapy with (90)Y-DOTATOC may be a therapeutic option. METHODS: Fifteen patients with recurrent or progressive meningiomas after multimodal pretreatment or unfavorable medical risk profile were treated with systemic (90)Y-DOTATOC. Endpoints were progression-free survival and toxicity. RESULTS: Usually applied doses were 7,400 MBq/m(2) of (90)Y-DOTATOC in 2 fractions. Mean observation time was 49.7 mo (range, 12-137 mo). Overall median progression-free survival was at least 24 mo. Toxicity was moderate, mostly hematologic (n = 8) and transient. CONCLUSION: (90)Y-DOTATOC therapy is feasible and may represent a promising second- or third-line option for complex meningiomas, which are progressive or otherwise not treatable with a reasonable risk-benefit ratio.


Asunto(s)
Meningioma/radioterapia , Octreótido/análogos & derivados , Radiofármacos/uso terapéutico , Adulto , Anciano , Supervivencia sin Enfermedad , Relación Dosis-Respuesta en la Radiación , Determinación de Punto Final , Femenino , Humanos , Riñón/efectos de la radiación , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Octreótido/efectos adversos , Octreótido/uso terapéutico , Radiofármacos/efectos adversos , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA