Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
1.
J Neurooncol ; 167(3): 415-425, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38441839

RESUMO

PURPOSE: Predicting resistance to first-generation Somatostatin Receptor Ligands (fg-SRL) in Acromegaly patients remains an ongong challenge. Tumor-associated immune components participate in various pathological processes, including drug-resistance. We aimed to identify the immune components involved in resistance of fg-SRL, and to investigate biomarkers that can be targeted to treat those drug-resistant Acromegaly. METHODS: We conducted a retrospective study involving 35 Acromegaly patients with somatotropinomas treated postoperatively with fg-SRL. Gathering clinicopathological data, SSTR2 expression, and immunological profiles, we utilized univariate, binary logistic regression, and ROC analyses to assess their predictive roles in fg-SRL resistance. Spearman correlation analysis further examined interactions among interested characteristics. RESULTS: 19 patients (54.29%) exhibited resistance to postoperative fg-SRL. GH level at diagnosis, preoperative tumor volume, T2WI-MRI intensity, granularity, PD-L1, SSTR2, and CD8 + T cell infiltration showed association with clinical outcomes of fg-SRL. Notably, T2WI-MRI hyperintensity, PD-L1-IRS > 7, CD8 + T cell infiltration < 14.8/HPF, and SSTR2-IRS < 5.4 emerged as reliable predictors for fg-SRL resistance. Correlation analysis highlighted a negative relationship between PD-L1 expression and CD8 + T cell infiltration, while showcasing a positive correlation with preoperative tumor volume of somatotropinomas. Additionally, 5 patients with fg-SRL resistance underwent re-operation were involved. Following fg-SRL treatment, significant increases in PD-L1 and SSTR5 expression were observed, while SSTR2 expression decreased in somatotropinoma. CONCLUSION: PD-L1 expression and CD8 + T cell infiltration, either independently or combined with SSTR2 expression and T2WI-MRI intensity, could form a predictive model guiding clinical decisions on fg-SRL employment. Furthermore, targeting PD-L1 through immunotherapy and embracing second-generations of SRL with higher affinity to SSTR5 represent promising strategies to tackle fg-SRL resistance in somatotropinomas.


Assuntos
Acromegalia , Receptores de Somatostatina , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Acromegalia/metabolismo , Acromegalia/cirurgia , Acromegalia/tratamento farmacológico , Acromegalia/imunologia , Acromegalia/sangue , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Resistencia a Medicamentos Antineoplásicos , Seguimentos , Adenoma Hipofisário Secretor de Hormônio do Crescimento/metabolismo , Adenoma Hipofisário Secretor de Hormônio do Crescimento/cirurgia , Adenoma Hipofisário Secretor de Hormônio do Crescimento/patologia , Adenoma Hipofisário Secretor de Hormônio do Crescimento/tratamento farmacológico , Ligantes , Octreotida/uso terapêutico , Prognóstico , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Estudos Retrospectivos
2.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768949

RESUMO

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.


Assuntos
Síndrome de Cushing/diagnóstico , Síndrome de Cushing/terapia , Neoplasias das Glândulas Suprarrenais/complicações , Síndrome de Cushing/complicações , Desenvolvimento de Medicamentos , Humanos , Hidrocortisona/metabolismo , Modelos Biológicos , Receptores Dopaminérgicos/efeitos dos fármacos , Receptores de Glucocorticoides/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos , Esteroides/biossíntese
3.
Mol Brain ; 14(1): 130, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34429141

RESUMO

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.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Interneurônios/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Somatostatina/fisiologia , Sinapses/efeitos dos fármacos , Animais , Proteínas de Bactérias , Cisteamina/farmacologia , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/metabolismo , Técnicas de Introdução de Genes , Genes Reporter , Humanos , Interneurônios/metabolismo , Proteínas Luminescentes , Masculino , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Peptídeos Cíclicos/farmacologia , Receptores de Glutamato Metabotrópico/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/fisiologia , Somatostatina/farmacologia , Sinapses/fisiologia
4.
Front Endocrinol (Lausanne) ; 12: 793262, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35058882

RESUMO

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).


Assuntos
Adenoma Hipofisário Secretor de ACT/tratamento farmacológico , Adenoma/tratamento farmacológico , Hormônio Adrenocorticotrópico/metabolismo , Neoplasias Brônquicas/tratamento farmacológico , Isoquinolinas/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Pirazóis/farmacologia , Piridinas/farmacologia , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Adenoma Hipofisário Secretor de ACT/diagnóstico por imagem , Adenoma Hipofisário Secretor de ACT/metabolismo , Adenoma/diagnóstico por imagem , Adenoma/metabolismo , Adulto , Idoso , Animais , Neoplasias Brônquicas/diagnóstico por imagem , Neoplasias Brônquicas/metabolismo , Linhagem Celular Tumoral , Dexametasona/farmacologia , Regulação para Baixo , Feminino , Glucocorticoides/farmacologia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Pessoa de Meia-Idade , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/metabolismo , Compostos Organometálicos , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Somatostatina/análogos & derivados
5.
Radiographics ; 39(7): 2069-2084, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31697628

RESUMO

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.


Assuntos
Carcinoma de Célula de Merkel/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Neoplasias Cutâneas/diagnóstico por imagem , Anticorpos Antivirais/sangue , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/secundário , Carcinoma de Célula de Merkel/secundário , Carcinoma de Célula de Merkel/virologia , Humanos , Metástase Linfática/diagnóstico por imagem , Poliomavírus das Células de Merkel/isolamento & purificação , Estadiamento de Neoplasias , Proteínas Oncogênicas/imunologia , Infecções por Polyomavirus/diagnóstico por imagem , Infecções por Polyomavirus/virologia , Prognóstico , Compostos Radiofarmacêuticos/análise , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Somatostatina/efeitos dos fármacos , Biópsia de Linfonodo Sentinela , Neoplasias Cutâneas/virologia , Infecções Tumorais por Vírus/diagnóstico por imagem , Infecções Tumorais por Vírus/virologia , Proteínas Virais/imunologia
6.
J Med Chem ; 62(5): 2708-2719, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30735385

RESUMO

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.


Assuntos
Descoberta de Drogas , Maitansina/farmacologia , Receptores de Somatostatina/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Células CHO , Linhagem Celular , Cricetulus , Cães , Humanos , Imunoconjugados/química , Imunoconjugados/farmacologia , Maitansina/química , Maitansina/farmacocinética , Camundongos , Receptores de Somatostatina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Endocrinol Metab Clin North Am ; 47(3): 615-625, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30098719

RESUMO

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.


Assuntos
Tumores Neuroendócrinos/radioterapia , Compostos Radiofarmacêuticos/uso terapêutico , Radioterapia/métodos , Receptores de Peptídeos , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico , Humanos , Compostos Radiofarmacêuticos/efeitos adversos , Radioterapia/tendências , Receptores de Somatostatina/efeitos dos fármacos , Somatostatina/efeitos adversos
8.
Endocrine ; 58(1): 124-133, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28822091

RESUMO

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.


Assuntos
Glucose/metabolismo , Receptores de Somatostatina/efeitos dos fármacos , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Animais , Teste de Tolerância a Glucose , Crescimento/efeitos dos fármacos , Hormônio do Crescimento/sangue , Hormônios/metabolismo , Infusões Subcutâneas , Insulina/sangue , Masculino , Ratos , Ratos Sprague-Dawley
9.
PLoS One ; 12(1): e0170536, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28107508

RESUMO

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.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Receptores da Bombesina/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos , Autorradiografia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Técnicas In Vitro , Cintilografia/métodos , Receptores da Bombesina/metabolismo , Receptores de Somatostatina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Oncotarget ; 7(50): 83451-83461, 2016 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-27825139

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Octreotida/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Taxoides/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Theranostics ; 6(11): 1821-32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27570553

RESUMO

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.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/radioterapia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/radioterapia , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Radiossensibilizantes/uso terapêutico , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Terapia Combinada , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Sinergismo Farmacológico , Instabilidade Genômica , Humanos , Lutécio , Radioisótopos , Radioterapia/métodos , Receptores de Somatostatina/efeitos dos fármacos
12.
Drug Deliv ; 23(1): 285-96, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-24865288

RESUMO

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.


Assuntos
Acrilamidas/farmacologia , Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Metacrilatos/química , Octreotida/química , Receptores de Somatostatina/efeitos dos fármacos , Acrilamidas/farmacocinética , Animais , Antibióticos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Sistemas de Liberação de Medicamentos , Humanos , Masculino , Metacrilatos/farmacocinética , Camundongos , Octreotida/farmacocinética , Receptores de Somatostatina/metabolismo , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Nucl Med ; 57(4): 503-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26678617

RESUMO

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.


Assuntos
Octreotida/análogos & derivados , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/uso terapêutico , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/diagnóstico por imagem , Simulação por Computador , Feminino , Câmaras gama , Humanos , Rim/metabolismo , Masculino , Meningioma/radioterapia , Pessoa de Meia-Idade , Modelos Biológicos , Modelos Teóricos , Tumores Neuroendócrinos/radioterapia , Octreotida/administração & dosagem , Octreotida/farmacocinética , Octreotida/uso terapêutico , Compostos Organometálicos/farmacocinética , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Somatostatina/efeitos dos fármacos , Fluxo Sanguíneo Regional , Distribuição Tecidual , Radioisótopos de Ítrio/uso terapêutico
14.
Endocrinology ; 157(2): 692-704, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26671185

RESUMO

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.


Assuntos
Mucosa Gástrica/metabolismo , Grelina/metabolismo , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Agonistas Adrenérgicos beta/farmacologia , Animais , Linhagem Celular Tumoral , Colforsina/farmacologia , Dinoprostona/farmacologia , Mucosa Gástrica/citologia , Mucosa Gástrica/efeitos dos fármacos , Perfilação da Expressão Gênica , Grelina/efeitos dos fármacos , Hormônios/farmacologia , Imuno-Histoquímica , Isoproterenol/farmacologia , Ácido Láctico/farmacologia , Camundongos , Camundongos Transgênicos , Muscarina/farmacologia , Agonistas Muscarínicos/farmacologia , Ocitócicos/farmacologia , Ocitocina/farmacologia , Palmitatos/farmacologia , Receptor Muscarínico M4/agonistas , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ocitocina/efeitos dos fármacos , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Receptores de Prostaglandina E Subtipo EP4/agonistas , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Análise de Sequência de RNA , Somatostatina/farmacologia , Triptofano/farmacologia
15.
J Nucl Med ; 56(10): 1487-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26251419

RESUMO

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.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/terapia , Receptores da Bombesina/efeitos dos fármacos , Receptores CXCR4/efeitos dos fármacos , Receptores de Somatostatina/efeitos dos fármacos , Adulto , Idoso , Antineoplásicos Hormonais/administração & dosagem , Antineoplásicos Hormonais/uso terapêutico , Autorradiografia , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Cintilografia , Compostos Radiofarmacêuticos , Análise de Sobrevida , Tamoxifeno/administração & dosagem , Tamoxifeno/uso terapêutico
16.
J Nucl Med ; 56(11): 1748-51, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26294303

RESUMO

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.


Assuntos
Meningioma/radioterapia , Octreotida/análogos & derivados , Compostos Radiofarmacêuticos/uso terapêutico , Adulto , Idoso , Intervalo Livre de Doença , Relação Dose-Resposta à Radiação , Determinação de Ponto Final , Feminino , Humanos , Rim/efeitos da radiação , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Octreotida/efeitos adversos , Octreotida/uso terapêutico , Compostos Radiofarmacêuticos/efeitos adversos , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Resultado do Tratamento
17.
PET Clin ; 9(3): 323-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25030395

RESUMO

68Ga-DOTA-SSTRTs PET/CT has become the most promising noninvasive procedure to study well-differentiated NET. Although the excellent diagnostic accuracy of the procedure is well known, its use is limited to specialized centers in Europe as parts of clinical trials. Literature reports confirm the superiority of 68Ga-DOTA-SSTRTs PET/CT for the assessment of well-differentiated NET over morphologic imaging procedures, SRS, and even PET/CT using metabolic radiotracers. 68Ga-DOTA-SSTRTs provide good visualization of NET lesions at both the primary and the metastatic sites (node, bone, liver, and unusual localizations). The advantages of their use over metabolic tracers (18F-DOPA, 18F-FDG) are not only limited to a better overall detection rate but also to the fact that they also provide data on SSTR expression on target lesions, resulting a fundamental procedure before starting therapy with either hot or cold somatostatin analogues. Moreover, they can be used also in centers without an on-site cyclotron. To interpret 68Ga-DOTA-SSTRTs images correctly, it is crucial to understand the tracer's biodistribution as well as the conditions that may alter tracer uptake. Considering that SSTR are expressed on activated lymphocytes, all areas of inflammation show 68Ga-DOTA-SSTRTs uptake. Areas of increased uptake in frequent sites of inflammation (eg, thyroid, mediastinal nodes, inguinal nodes, and nodes adjacent to areas of recent surgery/trauma) should be interpreted with care. A detailed clinical history with particular attention to concomitant disorders (eg, sarcoidosis, chronic gastritis, chronic thryoiditis) and recent invasive procedures or trauma may often help image interpretation. The presence of uptake in the head of the pancreas should always be carefully evaluated because it may often be benign. Otherwise, because the pancreas is also a frequent site of NET onset, particular attention should be devoted to the evaluation of the uptake pattern (diffuse more likely to be benign) and to the comparison with other imaging techniques.


Assuntos
Radioisótopos de Gálio/farmacocinética , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Somatostatina/efeitos dos fármacos , Tomografia Computadorizada por Raios X/métodos , Diagnóstico Diferencial , Humanos , Octreotida/análogos & derivados , Octreotida/farmacocinética , Compostos Organometálicos/farmacocinética , Distribuição Tecidual
18.
Pituitary ; 17(3): 227-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23740146

RESUMO

Non-functioning pituitary adenoma (NFPA) with higher proliferation index (WHO II) are often a therapeutical challenge. Low somatostatin receptor expression in these tumors usually prevents a treatment with somatostatin analogs. In 1996, a 55-year-old patient was referred due to right-sided headache. A pituitary macroadenoma with infiltration into the right cavernous sinus was diagnosed. There was no visual field deficit and the clinical and biochemical work up was consistent with a NFPA. The patient underwent transsphenoidal surgery. Residual adenoma remained in the right cavernous sinus. Histologically, a null-cell adenoma with a high proliferation index was documented (MIB-1: 11.6%, WHO II). Somatostatin receptor autoradiography was performed in the surgical specimen showing a homogenous expression of sst2 receptors. Radiosurgery was completed with stable disease for 8 years. In 2004, the patient was diagnosed with an incomplete palsy of the right oculomotorius nerve and a significant increase in the volume of the adenoma in the right cavernous sinus. After a positive Octreoscan(®) the patient consented to an experimental therapy approach using Lutetium DOTATOC (3 × 200 mCi). The palsy of the oculomotorius nerve improved and remained stable until today (March 2013), the follow-up MRI scans demonstrated stable disease. This is the first case of a patient with a NFPA (WHO II) in whom PRRT successfully improved the local complications of the tumor for more than 8 years after ineffective surgery and gamma knife therapy. The determination of sst2 in vitro using autoradiography and in vivo by Octreoscan was instrumental to administer this therapy in a challenging situation.


Assuntos
Neoplasias Hipofisárias/radioterapia , Compostos Radiofarmacêuticos/uso terapêutico , Receptores de Peptídeos/efeitos dos fármacos , Adulto , Terapia Combinada , Humanos , Masculino , Octreotida/análogos & derivados , Octreotida/uso terapêutico , Oftalmoplegia/etiologia , Oftalmoplegia/radioterapia , Neoplasias Hipofisárias/cirurgia , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/efeitos da radiação , Somatostatina/análogos & derivados
19.
Expert Opin Ther Targets ; 17(11): 1329-43, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23991721

RESUMO

INTRODUCTION: Neuroendocrine and pituitary tumors are uncommon tumors that develop from cells of the (neuro-)endocrine system. They can secrete hormones, leading to typical symptoms and syndromes. The cornerstone of antisecretory treatment for neuroendocrine and growth hormone-secreting pituitary tumors consists of somatostatin analogs, which target the somatostatin receptors that are expressed on the tumor cell membrane. Somatostatin analogs activate the second messenger pathways that inhibit hormone secretion and may also delay tumor growth. AREAS COVERED: Recent developments in the field of somatostatin analogs and promising new angles in neuroendocrine tumor treatment are discussed. The recently approved somatostatin analog pasireotide and promising new analogs KE108 and somatoprim are reviewed. Further, innovative developments in the field of receptor manipulation, such as epigenetic manipulation and viral somatostatin receptor subtype-2 expression vectors, are discussed, as well as oncolytic viruses specifically targeting neuroendocrine tumor cells. EXPERT OPINION: In addition to the development of novel somatostatin analogs and refining treatment with existing somatostatin analogs, alternative treatments targeting the somatostatin receptors that aim at increasing the number of somatostatin receptors should be explored as well, thereby broadening treatment perspectives and increasing options for prolonging survival.


Assuntos
Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Hipofisárias/tratamento farmacológico , Receptores de Somatostatina/efeitos dos fármacos , Sequência de Aminoácidos , Sistemas de Liberação de Medicamentos , Humanos , Tumores Neuroendócrinos/metabolismo , Neoplasias Hipofisárias/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo
20.
Front Neuroendocrinol ; 34(3): 228-52, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23872332

RESUMO

Somatostatin is a peptide with a potent and broad antisecretory action, which makes it an invaluable drug target for the pharmacological management of pituitary adenomas and neuroendocrine tumors. Somatostatin receptors (SSTR1, 2A and B, 3, 4 and 5) belong to the G protein coupled receptor family and have a wide expression pattern in both normal tissues and solid tumors. Investigating the function of each SSTR in several tumor types has provided a wealth of information about the common but also distinct signaling cascades that suppress tumor cell proliferation, survival and angiogenesis. This provided the rationale for developing multireceptor-targeted somatostatin analogs and combination therapies with signaling-targeted agents such as inhibitors of the mammalian (or mechanistic) target of rapamycin (mTOR). The ability of SSTR to internalize and the development of rabiolabeled somatostatin analogs have improved the diagnosis and treatment of neuroendocrine tumors.


Assuntos
Carcinoma Neuroendócrino/tratamento farmacológico , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/fisiologia , Animais , Carcinoma Neuroendócrino/diagnóstico , Proliferação de Células/efeitos dos fármacos , Dopamina/análogos & derivados , Dopamina/uso terapêutico , Humanos , Octreotida/uso terapêutico , Peptídeos Cíclicos/uso terapêutico , Compostos Radiofarmacêuticos , Transdução de Sinais/efeitos dos fármacos , Somatostatina/efeitos adversos , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA