Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
Rev Endocr Metab Disord ; 25(2): 383-398, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38051470

RESUMO

As the incidence of neuroendocrine tumors has been rising, gender differences in epidemiology and clinical behavior have emerged, and interest into a gender-driven management of these tumors has grown with the aim to improve survival and quality of life of these patients. Somatostatin Analogues represent the first line of systemic treatment of both functional and non-functional neuroendocrine tumors, through the expression of somatostatin receptors (SSTRs) in the tumor cells, and proved effective in controlling hormonal hypersecretion and inhibiting tumor growth, improving progression-free survival and overall survival of these patients. Aim of the present review is to investigate any differences by gender in efficacy and safety of SSTS-targeted therapies, that represent the mainstay treatment of neuroendocrine tumors, as they emerge from studies of varying design and intent. Although preclinical studies have provided evidence in favor of differences by gender in tumor expression of SSTR, as well as of the role of sex hormones and related receptors in modulating SSTRs expression and function, the clinical studies conducted so far have not shown substantial differences between males and females in either efficacy or toxicity of SSTR-targeted therapies, even if with sometimes inconsistent results. Moreover, in most studies gender was not a predictor of response to treatment. Studies specifically designed to address this issue are needed to develop gender-specific therapeutic algorithms, improving patients' prognosis and quality of life.


Assuntos
Tumores Neuroendócrinos , Somatostatina , Masculino , Feminino , Humanos , Somatostatina/uso terapêutico , Tumores Neuroendócrinos/patologia , Qualidade de Vida , Receptores de Somatostatina/metabolismo
2.
Medicina (Kaunas) ; 60(7)2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-39064468

RESUMO

Background and Objectives: High-grade malignant neuroendocrine tumors (G3 NETs) and neuroendocrine carcinomas (NECs) are characterized by rapid proliferation, high metastatic capacity, and strong expression of somatostatin receptors (SSTRs). We aimed to analyze the presence of SSTRs in NET G3 and NEC, and to correlate their expression with the use of octreotide and pasireotide. Materials and Methods: For this purpose, we first performed a retrospective study of G3 NET and NEC patients, which included the determination of SSTR expression and response to octreotide treatment. Second, we selected the H69 small cell lung cancer cell line to determine the effect of octreotide and pasireotide. Results: Our results showed the traditional somatostatin analog (SSA) octreotide was ineffective in patients with NET G3 and NEC. On the other hand, RT-qPCR showed a high expression of SSTR2 and SSTR5 in H69 cells. Interestingly, while octreotide did not modify H69 cell proliferation, a strong inhibition of proliferation was detected with the use of pasireotide. Conclusions: In view of these results, a clinical trial in NET G3 and NEC patients using pasireotide is necessary to determine the usefulness of this drug in improving patient treatment.


Assuntos
Tumores Neuroendócrinos , Octreotida , Receptores de Somatostatina , Somatostatina , Humanos , Octreotida/uso terapêutico , Octreotida/farmacologia , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico , Somatostatina/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Receptores de Somatostatina/análise , Estudos Retrospectivos , Masculino , Pessoa de Meia-Idade , Feminino , Idoso , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Adulto , Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos Hormonais/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia
3.
Int J Mol Sci ; 25(1)2023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-38203605

RESUMO

Somatostatin (SST), a growth hormone inhibitory peptide, is expressed in endocrine and non-endocrine tissues, immune cells and the central nervous system (CNS). Post-release from secretory or immune cells, the first most appreciated role that SST exhibits is the antiproliferative effect in target tissue that served as a potential therapeutic intervention in various tumours of different origins. The SST-mediated in vivo and/or in vitro antiproliferative effect in the tumour is considered direct via activation of five different somatostatin receptor subtypes (SSTR1-5), which are well expressed in most tumours and often more than one receptor in a single cell. Second, the indirect effect is associated with the regulation of growth factors. SSTR subtypes are crucial in tumour diagnosis and prognosis. In this review, with the recent development of new SST analogues and receptor-specific agonists with emerging functional consequences of signaling pathways are promising therapeutic avenues in tumours of different origins that are discussed.


Assuntos
Neoplasias , Receptores de Somatostatina , Humanos , Somatostatina , Hormônio do Crescimento , Neoplasias/tratamento farmacológico , Biologia
4.
Curr Treat Options Oncol ; 23(7): 1001-1013, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35501552

RESUMO

OPINION STATEMENT: Neuroendocrine tumors (NET) represent a complex and heterogeneous group of malignancies arising from the diffuse endocrine cells and other cells derived from the neural crest. Advanced disease is observed at diagnosis in more than one-third of patients. Somatostatin analogs (SSA) are the cornerstone in advanced well-differentiated NET treatment. Unfortunately, most patients will eventually develop resistance to SSA treatment by different mechanisms that are not fully understood. In some cases of refractory carcinoid syndrome or progressive disease, the increase of SSA dose may help to reach out a symptomatic and/or tumor growth control. The clinical evidence behind above-label SSA administration is limited and should be individualized and discussed patient by patient. Some questions regarding high-dose SSA use are unsolved, such as the optimal dose to use, the frequency of administration, or the need of deepen molecular understanding that could help to adequately select patients for this approach.


Assuntos
Síndrome do Carcinoide Maligno , Tumores Neuroendócrinos , Humanos , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/patologia , Somatostatina/efeitos adversos
5.
Int J Mol Sci ; 23(6)2022 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-35328517

RESUMO

Somatostatin is an inhibitory peptide, which regulates the release of several hormones, and affects neurotransmission and cell proliferation via its five Gi protein-coupled receptors (SST1-5). Although its endocrine regulatory and anti-tumour effects have been thoroughly studied, little is known about its effect on the vascular system. The aim of the present study was to analyse the effects and potential mechanisms of somatostatin on endothelial barrier function. Cultured human umbilical vein endothelial cells (HUVECs) express mainly SST1 and SST5 receptors. Somatostatin did not affect the basal HUVEC permeability, but primed HUVEC monolayers for thrombin-induced hyperpermeability. Western blot data demonstrated that somatostatin activated the phosphoinositide 3-kinases (PI3K)/protein kinase B (Akt) and p42/44 mitogen-activated protein kinase (MAPK) pathways by phosphorylation. The HUVEC barrier destabilizing effects were abrogated by pre-treating HUVECs with mitogen-activated protein kinase kinase/extracellular signal regulated kinase (MEK/ERK), but not the Akt inhibitor. Moreover, somatostatin pre-treatment amplified vascular endothelial growth factor (VEGF)-induced angiogenesis (3D spheroid formation) in HUVECs. In conclusion, the data demonstrate that HUVECs under quiescence conditions express SST1 and SST5 receptors. Moreover, somatostatin primes HUVECs for thrombin-induced hyperpermeability mainly via the activation of MEK/ERK signalling and promotes HUVEC proliferation and angiogenesis in vitro.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Fator A de Crescimento do Endotélio Vascular , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Somatostatina/metabolismo , Somatostatina/farmacologia , Trombina/metabolismo , Trombina/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Rev Endocr Metab Disord ; 22(3): 581-594, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32495250

RESUMO

Neuroendocrine neoplasms (NENs) comprise a heterogeneous group of tumors, mainly localized in the gastrointestinal system. What characterizes NENs is the expression of hormone receptors on the tumor cell surface, making them accessible for diagnostic and therapeutic approaches (theranostics) using radiolabelled peptides. Somatostatin receptors subtype-two (SST2) play an important role in NENs since they are overexpressed and homogeneously distributed at the surface of the majority of NENs. Accordingly, targeting SST2 for diagnostic and therapeutic purposes has been established. Current research aims at upregulating its expression by epigenetic treatment or improving its targeting via use of alternative radioligands. In addition, recent data suggest a future role of SST antagonists as a diagnostic tool and a potential therapeutic option. Another promising target is the glucagon-like peptide-1 (GLP-1) receptor. Targeting GLP-1R using exendin-4 (GLP-1 analogue) has a high sensitivity for the localization of the often SST2-negative sporadic insulinomas and insulinomas in the context of multiple endocrine neoplasia type-1. Further options for patients with insufficient expression of SST2 involve metaiodobenzylguanidine (MIBG) and the molecular target C-X-C motif chemokine receptor-4 (CXCR4), which have been evaluated for potential theranostic approach in symptomatic NENs or dedifferentiated tumors. Recently, new targets such as the glucose-dependent insulinotropic polypeptide receptor (GIPR) and the fibroblast activation protein (FAP) have been identified in NENs. Finally, minigastrin - a ligand targeting the cholecystokinin-2 (CCK2) receptors in medullary thyroid carcinoma and foregut neuroendocrine tumors - may improve future management of these diseases with currently limited therapeutic options. This review summarises the current approaches and future challenges of diagnostic and therapeutic evaluations in neuroendocrine neoplasms.


Assuntos
Insulinoma , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Humanos , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Medicina de Precisão , Receptores de Somatostatina
7.
BMC Cancer ; 21(1): 950, 2021 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433438

RESUMO

BACKGROUND: Neuroblastoma (NB) is one of the most frequently diagnosed tumors in infants. NB is a neuroendocrine tumor type with various characteristics and features, and with diverse outcome. The most malignant NBs have a 5-year survival rate of only 40-50%, indicating the need for novel and improved treatment options. 177Lu-octreotate is routinely administered for treatment of neuroendocrine tumors overexpressing somatostatin receptors (SSTR). The aim of this study was to examine the biodistribution of 177Lu-octreotate in mice bearing aggressive human NB cell lines, in order to evaluate the potential usefulness of 177Lu-octreotate for treatment of NB. METHODS: BALB/c nude mice bearing CLB-BAR, CLB-GE or IMR-32 tumor xenografts (n = 5-7/group) were i.v. injected with 0.15 MBq, 1.5 MBq or 15 MBq 177Lu-octreotate and sacrificed 1 h, 24 h, 48 h and 168 h after administration. The radioactivity concentration was determined for collected tissue samples, tumor-to-normal-tissue activity concentration ratios (T/N) and mean absorbed dose for each tissue were calculated. Immunohistochemical (IHC) staining for SSTR1-5, and Ki67 were carried out for tumor xenografts from the three cell lines. RESULTS: High 177Lu concentration levels and T/N values were observed in all NB tumors, with the highest for CLB-GE tumor xenografts (72%IA/g 24 h p.i.; 1.5 MBq 177Lu-octreotate). The mean absorbed dose to the tumor was 6.8 Gy, 54 Gy and 29 Gy for CLB-BAR, CLB-GE and IMR-32, respectively, p.i. of 15 MBq 177Lu-octreotate. Receptor saturation was clearly observed in CLB-BAR, resulting in higher concentration levels in the tumor when lower activity levels where administered. IHC staining demonstrated highest expression of SSTR2 in CLB-GE, followed by CLB-BAR and IMR-32. CONCLUSION: T/N values for all three human NB tumor xenograft types investigated were high relative to previously investigated neuroendocrine tumor types. The results indicate a clear potential of 177Lu-octreotate as a therapeutic alternative for metastatic NB.


Assuntos
Lutécio/uso terapêutico , Neuroblastoma/radioterapia , Octreotida/análogos & derivados , Radioisótopos/uso terapêutico , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Eur Radiol ; 31(10): 7734-7745, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33787974

RESUMO

OBJECTIVES: To investigate an additional value of [68Ga]Ga-DOTA-TOC PET/CT for characterizing suspected pancreatic neuroendocrine neoplasms (NENs) in a large study cohort. METHODS: This retrospective study included 167 patients who underwent [68Ga]Ga-DOTA-TOC PET/CT for suspected pancreatic NENs detected by contrast-enhanced CT (n = 153) and/or MRI (n = 85). Two board-certified radiologists independently reviewed CT and/or MRI as well as [68Ga]Ga-DOTA-TOC PET/CT and scored the probability of NEN on a 5-point scale. Radiologists' diagnostic performances with and without [68Ga]Ga-DOTA-TOC PET/CT were compared using pathologic findings as the standard of reference. RESULTS: All 167 patients were pathologically diagnosed with NENs (n = 131) or non-NENs (n = 36) by surgery (n = 93) or biopsy (n = 74). The non-NEN group included focal pancreatitis (n = 7), gastrointestinal stromal tumor (n = 6), serous cystadenoma (n = 5), metastatic renal cell carcinoma (n = 4), intrapancreatic accessory spleen (n = 4), ductal adenocarcinoma (n = 3), solid pseudopapillary neoplasm (n = 2), intraductal papillary mucinous carcinoma (n = 1), adenosquamous carcinoma (n = 1), schwannoma (n = 1), paraganglioma (n = 1), and solitary fibrous tumor (n = 1). Radiologists' diagnostic performance significantly improved after the addition of [68Ga]Ga-DOTA-TOC PET/CT (AUC of CT: 0.737 vs. 0.886 for reviewer 1 [p = 0.0004]; 0.709 vs. 0.859 for reviewer 2 [p = 0.0002], AUC of MRI: 0.748 vs. 0.872 for reviewer 1 [p = 0.023]; 0.670 vs. 0.854 for reviewer 2 [p = 0.001]). [68Ga]Ga-DOTA-TOC PET/CT significantly improved sensitivity (CT: 87.4% vs. 96.6% for reviewer 1 [p = 0.001]; 74.8% vs. 92.5% for reviewer 2 [p = 0.0001], MRI: 86.9% vs. 98.4% for reviewer 1 [p = 0.016]; 70.5% vs. 91.8% for reviewer 2 [p = 0.002]). CONCLUSIONS: [68Ga]Ga-DOTA-TOC PET/CT provided an additional value over conventional CT or MRI for the characterization of suspected pancreatic NENs. KEY POINTS: • [68Ga]Ga-DOTA-TOC PET/CT could provide additional value over conventional CT and/or MRI for the exact characterization of suspected pancreatic NENs by increasing AUC values and sensitivity. • Diagnostic improvement was significant, especially in NENs showing an atypical enhancement pattern. • The inter-observer agreement was improved when [68Ga]Ga-DOTA-TOC PET/CT was added to CT and/or MRI.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Tumores Neuroendócrinos , Compostos Organometálicos , Neoplasias Pancreáticas , Radioisótopos de Gálio , Humanos , Imageamento por Ressonância Magnética , Tumores Neuroendócrinos/diagnóstico por imagem , Octreotida , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Tomografia por Emissão de Pósitrons , Estudos Retrospectivos
9.
J Pept Sci ; 27(12): e3361, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34291534

RESUMO

Somatostatin analogues play an important role in the therapy of neuroendocrine tumors by binding to somatostatin receptors on the surface of cancer cells. In this work, we analyze the receptor-binding affinity and in vitro stability of a novel ultra-short somatostatin analogue Thz-Phe-D-Trp-Lys-Thr-DOTA (DOTA-P4). This conjugate is successfully radiolabeled with 44 Sc, 90 Y, 152 Eu, and 207 Bi, characterized and validated by thin layer and high-performance liquid chromatography. The optimum conditions for M-DOTA-P4 labeling are found. In vitro stability studies are performed in saline, in the presence of serum proteins, and with biologically relevant metal cations. All complexes demonstrate no cation release in vitro within 4-24 h. The conformations of DOTA-conjugates are studied by circular dichroism spectroscopy. The circular dichroism spectra of DOTA-P4 conjugates show a negative peak at 225 nm, which may correspond to the required ß-sheet conformation. The binding to somatostatin receptors of types 2 and 5 is performed with the IMR-32 cells at 4°C, with non-specific binding representing 26% of the total binding. A two-line approximation of the Scatchard plot results in the apparent dissociation constants of 0.10 and 2.25 nM. It is shown that the chelator position with respect to the amino acid sequence significantly affects the labeling conditions with cations of different ionic radii. For the first time, the binding of a linear type ultra-short peptide conjugate with DOTA to somatostatin receptors is demonstrated. The obtained results are promising for experiments with DOTA-P4 in vivo in mice with inoculated tumors.


Assuntos
Receptores de Somatostatina , Somatostatina , Animais , Compostos Heterocíclicos com 1 Anel , Camundongos , Peptídeos
10.
Curr Oncol Rep ; 23(7): 80, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33937962

RESUMO

PURPOSE OF REVIEW: Pancreatic neuroendocrine neoplasms (panNENs) often present as advanced disease and there is little data to guide treatment sequencing in the advance disease setting. Therefore, we aim to provide a comprehensive summary of the current evidence supporting the use of systemic treatment for patients with diagnosis of advanced and metastatic panNENs, as well as to provide strategies for treatment selection and address challenges for treatment selection and sequencing of therapy. RECENT FINDINGS: Substantial advances have been made and many clinical trials have been performed over the past two decades expanding therapeutic options available for patients with advanced panNETs. Available systemic treatments for patients with well-differentiated pancreatic neuroendocrine tumors include somatostatin receptors ligands (SRLs), traditional cytotoxic chemotherapy regimens, peptide receptor radiotherapy (PRRT), and biologically targeted therapies, whereas patients with poorly differentiated neurodocrine carcinomas have more limited treatment options. Despite these advances, no clear guidelines exist to support the best sequence of treatments, not only the first-line, but also subsequent lines of therapy in patients with panNENs. Advances in molecular research and discovery of biomarkers for response allowing a more personalized approach to the multimodality therapy of panNENs are still limited. Understanding the impact of previous therapies on subsequent treatment efficacy and toxicity is also an ongoing research question. In the absence of definite predictive markers and paucity of comparative randomized trials, along with the heterogeneity of this patient population, systemic therapy selection in advanced non-resectable disease should be patient centered and often require evaluation within a multidisciplinary setting. The specific clinical context of the patient, with assessment of individual patient clinical and pathological features, somatostatin receptors imaging, and goals of treatment must all be considered when deciding on systemic therapy in the patient. Additional research is needed to address the gap in knowledge regarding optimal sequencing and timing of therapies and to provide individual care.


Assuntos
Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Humanos , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Somatostatina/fisiologia
11.
Pharm Res ; 37(11): 217, 2020 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-33037505

RESUMO

BACKGROUND: Leukaemia is the most prevalent form of cancer-causing death in a large number of populations and needs prompt and effective treatment. Chemotherapeutics can be used to treat leukaemia, but their pronounced killing effects to other living cells is still an issue. Active targeting to certain specific receptors in leukaemic cells is the best way to avoid damage to other living cells. Leukaemic cells can be targeted using novel nanoparticles (NPs) coated with a specific ligand, such as octreotide (OCD), to target somatostatin receptor type 2 (SSTR2), which is expressed in leukaemic cells. METHODS: Amino-PEGylated quantum dots (QDs) were chosen as model NPs. The QDs were first succinylated using succinic anhydride and then coated with OCD. The reactivity and selectivity of the formulated QDs-OCD were studied in cell lines with well-expressed SSTR2, while fluorescence was detected using confocal laser scanning microscopy (CLSM) and flow cytometry (FACS). Conclusively, QD-OCD targeting to blood cells was studied in vivo in mice and detected using inductively coupled plasma mass spectrometry and CLSM in tissues. RESULTS: Highly stable QDs coated with OCD were prepared. FACS and CLSM showed highly definite interactions with overexpressed SSTR2 in the investigated cell lines. Moreover, the in vivo results revealed a higher concentration of QDs-OCD in blood cells. The fluorescence intensity of the QDs-OCD was highly accumulated in blood cells, while the unmodified QDs did not accumulate significantly in blood cells. CONCLUSION: The formulated novel QDs-OCD can target SSTR2 overexpressed in blood cells with great potential for treating blood cancer.


Assuntos
Antineoplásicos/metabolismo , Corantes Fluorescentes/química , Leucemia/metabolismo , Monócitos/metabolismo , Octreotida/metabolismo , Pontos Quânticos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Composição de Medicamentos , Citometria de Fluxo , Células HeLa , Humanos , Leucemia/tratamento farmacológico , Leucemia/patologia , Masculino , Camundongos , Microscopia Confocal , Octreotida/química , Octreotida/farmacologia
12.
Curr Treat Options Oncol ; 21(4): 25, 2020 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-32172368

RESUMO

OPINION STATEMENT: Neuroendocrine tumors (NETs) are a heterogenous group of neoplasms characterized by varied biological hallmarks and behavior, ranging from indolent to aggressive. For many decades, somatostatin analogues and few targeted therapies were available for NETs and these therapies had minimal response rates. However, there have been a number of recent treatment advances. Peptide receptor radionuclide therapy (PRRT) is a novel approach to treatment of NETs and has changed the landscape of treatment for NETs. It is a form of targeted therapy in which a radiolabeled somatostatin analogue delivers radiation specifically to tumor cells expressing the somatostatin receptor.


Assuntos
Braquiterapia/efeitos adversos , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/terapia , Seleção de Pacientes , Compostos Radiofarmacêuticos/efeitos adversos , Receptores de Peptídeos/antagonistas & inibidores , Pesquisa Biomédica , Braquiterapia/métodos , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Gerenciamento Clínico , Humanos , Metástase Neoplásica , Estadiamento de Neoplasias , Tumores Neuroendócrinos/etiologia , Guias de Prática Clínica como Assunto , Doses de Radiação , Compostos Radiofarmacêuticos/administração & dosagem , Resultado do Tratamento
13.
Bioorg Chem ; 94: 103381, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31662215

RESUMO

Somatostatin receptor-targeted radionuclide therapy has become an effective treatment in patients with neuroendocrine tumors. Recently, investigations on the development of antagonistic peptides are increasing with possible superior biological properties as opposed to the agonists. Herein, we have reported the development of a new somatostatin receptor peptide ligand labeled with 177Lu to achieve a therapeutic ligand for tumor treatment. The interactions of selected and drown ligands using Avogadro software were docked on somatostatin receptor by Dink algorithm. The best docked peptide-chelator conjugate (DOTA-p-Cl-Phe-Cyclo(d-Cys-l-BzThi-d-Aph-Lys-Thr-Cys)-d-Tyr-NH2) (DOTA-Peptide 2) was synthesized using the Fmoc solid-phase method. DOTA-Peptide 2 was radiolabeled with the 177Lu Trichloride (177LuCl3) solution at 95 °C for 30 min and radiochemical purity (RCP) of 177Lu-DOTA-Peptide 2 solution was monitored by radio-HPLC and radio-TLC procedures. The new radiolabeled peptide was evaluated for stability, receptor binding, internalization, biodistribution and single-photon emission computed tomography (SPECT) imaging using C6 glioma cells and C6 tumor-bearing rats. DOTA-Peptide 2 was obtained with 98% purity and efficiently labeled with 177Lu (RCP > 99%). 177Lu-DOTA-Peptide 2 showed a high value of stability in acetate buffer (91.4% at 312 h) and human plasma (>97% at 24 h). Radioconjugate exhibited low internalization (<5%) and high affinity for somatostatin receptors (Kd = 12.06 nM, Bmax = 0.20 pmol/106 cells) using saturation binding assay. Effective tumor uptake of 7.3% ID/g (percentage of injected dose per gram of tumor) at 4 h post-injection and fast clearance of radiopeptide from blood and other organs led to a high tumor-to-normal organ ratios. SPECT/CT imaging clearly showed the activity localization in tumor. The favorable antagonistic properties of 177Lu-DOTA-Peptide 2 on the somatostatin receptors can make it a suitable candidate for peptide receptor radionuclide therapy (PRRT). In the future study, the therapeutic application of this radiopeptide will be evaluated.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Tumores Neuroendócrinos/tratamento farmacológico , Octreotida/análogos & derivados , Compostos Organometálicos/química , Peptídeos/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptores de Somatostatina/antagonistas & inibidores , Antineoplásicos/síntese química , Antineoplásicos/química , Relação Dose-Resposta a Droga , Humanos , Ligantes , Simulação de Acoplamento Molecular , Estrutura Molecular , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/metabolismo , Octreotida/química , Peptídeos/síntese química , Peptídeos/química , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Receptores de Somatostatina/metabolismo , Relação Estrutura-Atividade , Tomografia Computadorizada de Emissão de Fóton Único
14.
J Endocrinol Invest ; 43(11): 1543-1553, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32557353

RESUMO

BACKGROUND: Somatostatin receptors (SSTs) are widely co-expressed in pituitary tumors. SST2 and SST5 are the most represented SST subtypes. First-generation somatostatin receptor ligands (SRLs) mainly target SST2, while pasireotide, a multi-receptor ligand, shows high binding affinity for both SST5 and SST2. Therefore, SRLs are routinely used as medical treatment for GH-, TSH-, and ACTH-secreting pituitary tumors. METHODS: Critical revision of literature data correlating SST expression with patients' response to SRLs. RESULTS: SST2 expression in somatroph tumors directly correlates with GH and IGF-1 decrease after first-generation SRL treatment. SST2 immunohistochemistry represents a valuable tool to predict biochemical response to first-generation SRLs in acromegalic patients. Pasireotide seems to exert its biological effects via SST2 in unselected patients. However, in those subjects resistant to first-generation SRLs, harbouring tumors with negligible SST2 expression, pasireotide can act throughout SST5. More than somatotroph tumors, TSH-omas represent the paradigm of tumors showing a satisfactory response to SRLs. This is probably due to the high SST2 expression observed in nearly 100% of cases, as well as to the balanced amount of SST5. In corticotroph tumors, pasireotide mainly act via SST5, although there is a need for translational studies correlating its efficacy with SST expression in this peculiar tumor histotype. CONCLUSIONS: The assumption "more target receptor, more drug efficacy" is not straightforward for SRLs. The complex pathophysiology of SSTs, and the technical challenges faced to translate research findings into clinical practice, still need our full commitment to make receptor evaluation a worthwhile procedure for individualizing treatment decisions.


Assuntos
Adenoma , Terapia de Alvo Molecular , Neoplasias Hipofisárias , Receptores de Somatostatina/genética , Adenoma/diagnóstico , Adenoma/genética , Adenoma/metabolismo , Adenoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Hormônio do Crescimento Humano/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Octreotida/uso terapêutico , Neoplasias Hipofisárias/diagnóstico , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/terapia , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/metabolismo , Somatostatina/uso terapêutico
15.
Drug Dev Ind Pharm ; 46(5): 744-750, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32250174

RESUMO

Even so, the metal nanoparticles (metal NPs) have attractive optical and biomedical applications, the translation of metal NPs into the clinical practice remains a challenge due to their severe accumulation in the body. Active targeting to renal podocytes opens the door for enhancing kidney targeting and clearance. The goal of this study was to assess the excretion of larger particle size through kidney podocyte via active targeting. To reach this goal, PEGylated quantum dots (QDs) were coated with vapreotide (VAP) for selectively reaching somatostatin receptors (SSTRs) expressed in the podocyte cells. This QDs-VAP was tested on isolated primary podocytes, while the flow cytometry (FACS), confocal microscopy (CLSM), and inductively coupled plasma mass spectrometry (ICP-MS) were used to confirm this hypothesis. The results showed highly specific interactions with podocyte cells as detected by FACS, and CLSM. Moreover, ICP-MS demonstrated higher amount of QDs in the podocyte cells one-hour post-incubation (67.99% ID/g tissue), while the unmodified QDs did not accumulate. This study confirmed that QDs-VAP can target the podocyte's SSTRs then can be cleared via podocyte cells. Moreover, these results are considered as a highly promising approach for future therapy, targeting, clearance, and diagnosis of podocyte-associated diseases.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Vias de Eliminação de Fármacos/efeitos dos fármacos , Nanopartículas Metálicas , Podócitos/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Analgésicos/administração & dosagem , Analgésicos/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Vias de Eliminação de Fármacos/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Podócitos/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Pontos Quânticos/administração & dosagem , Pontos Quânticos/metabolismo , Somatostatina/administração & dosagem , Somatostatina/metabolismo
16.
Pol J Pathol ; 71(1): 30-37, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32429652

RESUMO

Most neuroendocrine neoplasms (NEN) are characterized by the presence of somatostatin receptors (SSTR) which we use in location diagnostics and treatment. The aim of this study was to evaluate the expression of somatostatin receptors by immunohistochemistry in tissue obtained after surgery of the primary focus in the small intestine. The group of patients consisted of 41 people, in 18 cases the primary tumor was in the jejunum and in 23 in the ileum. The immunohistochemical method was used to visualize the receptors, using polyclonal antibodies in a two-stage peroxidase method. In patients with NEN of the small intestine, the SSTR2a and SSTR5 receptors are most commonly expressed, followed by SSTR2b and 3. In statistical analysis, it was shown that the expression of somatostatin receptors was not dependent on the primary site of the tumor (p > 0.05). The dependence of SSTR expression on histological maturity is evident. SSTR1, SSTR2b, SSTR3 and SSTR5 are more common in tumors with grading G1 (p < 0.05). In the study group, the exception was SSTR2a, whose incidence was comparable in both groups (p = 0.35). In NEN of the small intestine, the expression SSTR2a and SSTR5 is the most common.


Assuntos
Neoplasias Intestinais/diagnóstico , Intestino Delgado/patologia , Tumores Neuroendócrinos/diagnóstico , Receptores de Somatostatina/genética , Humanos , Imuno-Histoquímica , Neoplasias Intestinais/genética , Tumores Neuroendócrinos/genética
17.
Int J Mol Sci ; 21(7)2020 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272767

RESUMO

Obesity is one of the major social and health problems globally and often associated with various other pathological conditions. In addition to unregulated eating behaviour, circulating peptide-mediated hormonal secretion and signaling pathways play a critical role in food intake induced obesity. Amongst the many peptides involved in the regulation of food-seeking behaviour, somatostatin (SST) is the one which plays a determinant role in the complex process of appetite. SST is involved in the regulation of release and secretion of other peptides, neuronal integrity, and hormonal regulation. Based on past and recent studies, SST might serve as a bridge between central and peripheral tissues with a significant impact on obesity-associated with food intake behaviour and energy expenditure. Here, we present a comprehensive review describing the role of SST in the modulation of multiple central and peripheral signaling molecules. In addition, we highlight recent progress and contribution of SST and its receptors in food-seeking behaviour, obesity (orexigenic), and satiety (anorexigenic) associated pathways and mechanism.


Assuntos
Regulação do Apetite/fisiologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Somatostatina/metabolismo , Animais , Apetite/fisiologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Humanos , Peptídeos/metabolismo
18.
Molecules ; 25(23)2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33297556

RESUMO

Hematological and oncological disorders represent leading causes of childhood mortality. Neuropeptide somatostatin (SST) has been previously demonstrated in various pediatric tumors, but limited information exists on the expression and characteristics of SST receptors (SSTR) in hematological and oncological disorders of children. We aimed to investigate the expression of mRNA for SSTR subtypes (SSTR-1-5) in 15 pediatric hematological/oncological specimens by RT-PCR. The presence and binding characteristics of SSTRs were further studies by ligand competition assay. Our results show that the pediatric tumor samples highly expressed mRNA for the five SSTR subtypes with various patterns. The mRNA for SSTR-2 was detected in all specimens independently of their histological type. A Hodgkin lymphoma sample co-expressed mRNA for all five SSTR subtypes. SSTR-3 and SSTR-5 were detected only in malignant specimens, such as rhabdomyosarcoma, Hodgkin lymphoma, acute lymphoblastic leukemia, and a single nonmalignant condition, hereditary spherocytosis. The incidence of SSTR-1 and SSTR-4 was similar (60%) in the 15 specimens investigated. Radioligand binding studies demonstrated the presence of specific SSTRs and high affinity binding of SST analogs in pediatric solid tumors investigated. The high incidence of SSTRs in hematological and oncological disorders in children supports the merit of further investigation of SSTRs as molecular targets for diagnosis and therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Hematológicas/genética , Neoplasias/genética , Receptores de Somatostatina/genética , Adolescente , Fatores Etários , Criança , Pré-Escolar , Feminino , Seguimentos , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Lactente , Masculino , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Receptores de Somatostatina/metabolismo
19.
Am J Physiol Endocrinol Metab ; 317(6): E1081-E1093, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31503512

RESUMO

DPP-4 inhibitors, used for treatment of type 2 diabetes, act by increasing the concentrations of intact glucagon-like peptide-1 (GLP-1), but at the same time, they inhibit secretion of GLP-1, perhaps by a negative feedback mechanism. We hypothesized that GLP-1 secretion is feedback regulated by somatostatin (SS) from neighboring D-cells, and blocking this feedback circuit results in increased GLP-1 secretion. We used a wide range of experimental techniques, including gene expression analysis, immunohistochemical approaches, and the perfused mouse intestine to characterize the paracrine circuit controlling GLP-1 and SS. We show that 1) antagonizing the SS receptor (SSTr) 2 and SSTr5 led to increased GLP-1 and SS secretion in the mouse, 2) SS exhibits strong tonic inhibition of GLP-1 secretion preferentially through SSTr5, and 3) the secretion of S was GLP-1 receptor dependent. We conclude that SS is a tonic inhibitor of GLP-1 secretion, and interventions in the somatostain-GLP-1 paracrine loop lead to increased GLP-1 secretion.


Assuntos
Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Mucosa Intestinal/metabolismo , Comunicação Parácrina , Células Secretoras de Somatostatina/metabolismo , Somatostatina/metabolismo , Animais , Inibidores da Dipeptidil Peptidase IV/farmacologia , Células Enteroendócrinas/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/efeitos dos fármacos , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Intestinos , Camundongos , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/metabolismo , Somatostatina/farmacologia , Somatostatina-28/farmacologia , Células Secretoras de Somatostatina/efeitos dos fármacos
20.
Pituitary ; 22(1): 70-78, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30607746

RESUMO

PURPOSE: Spindle cell oncocytomas (SCOs) are very rare tumors of the posterior pituitary with potential for locally aggressive behaviour. Their treatment includes surgery and possibly radiotherapy, however other options are lacking. Somatostatin receptors (SSTs) are a possible therapeutic target for somatostatin analogues and their expression has been demonstrated recently in closely related pituicytomas, but there are no data about their presence in SCOs. METHODS: We collected five cases of SCO from four patients including one recurrent case. Immunohistochemical detection of TTF1, GFAP, CD68, SST1, SST2, SST3, SST5 and D2 dopamine receptor (D2DR) was performed. Intensity, percentage of positive cells and pattern of expression was evaluated in semiquantitative fashion. Protein expression of SST1-5 and D2DR was further evaluated by western blot. RESULTS: Mean patient age was 61.8 years (range 47-71 years) with male to female ratio 1:1. In one patient, samples from the original tumor and its recurrence 16 years later were assessed. TTF1 was positive in all five cases, no expression of GFAP and CD68 was seen. Immunohistochemical expression of SST1 was noted in 1/5 cases, SST2 in 2/5 cases, including recurrent case but not the original case. SST3 was expressed in 3/5 tumors and D2 dopamine receptor in 4/5 cases. Western blot was successfully performed in four samples. SST2, SST3 and D2DR expression was identified in all the samples, including two cases originally negative for SST2 and one case negative for SST3 by immunohistochemistry. The number of positive cells and level of expression varied among different areas of the same tumors. No expression of SST5 was observed. In the patient with the recurrent tumor, intensity of SST2, SST3 and D2DR expression varied between original tumor and its recurrence. CONCLUSIONS: We demonstrated presence of different SST subtypes and D2DR in spindle cell oncocytomas. The most commonly expressed subtype was SST2 and SST3, while no expression of SST5 was observed. Expression showed spatial heterogeneity and temporal changes as seen in the recurrent case. The biological meaning of SSTs expression in SCOs is unclear as well as whether it may be exploited in treatment of selected cases.


Assuntos
Adenoma Oxífilo/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Acromegalia/metabolismo , Acromegalia/patologia , Adenoma Oxífilo/patologia , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Câncer Papilífero da Tireoide/metabolismo , Câncer Papilífero da Tireoide/patologia , Doenças da Glândula Tireoide/metabolismo , Doenças da Glândula Tireoide/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA