Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Eur J Cancer ; 162: 221-236, 2022 02.
Article in English | MEDLINE | ID: mdl-34980502

ABSTRACT

INTRODUCTION: Olfactory neuroblastoma (ONB) is a rare cancer of the sinonasal region. We provide a comprehensive analysis of this malignancy with molecular and clinical trial data on a subset of our cohort to report on the potential efficacy of somatostatin receptor 2 (SSTR2)-targeting imaging and therapy. METHODS: We conducted a retrospective analysis of 404 primary, locally recurrent, and metastatic olfactory neuroblastoma (ONB) patients from 12 institutions in the United States of America, United Kingdom and Europe. Clinicopathological characteristics and treatment approach were evaluated. SSTR2 expression, SSTR2-targeted imaging and the efficacy of peptide receptor radionuclide therapy [PRRT](177Lu-DOTATATE) were reported in a subset of our cohort (LUTHREE trial; NCT03454763). RESULTS: Dural infiltration at presentation was a significant predictor of overall survival (OS) and disease-free survival (DFS) in primary cases (n = 278). Kadish-Morita staging and Dulguerov T-stage both had limitations regarding their prognostic value. Multivariable survival analysis demonstrated improved outcomes with lower stage and receipt of adjuvant radiotherapy. Prophylactic neck irradiation significantly reduces the rate of nodal recurrence. 82.4% of the cohort were positive for SSTR2; treatment of three metastatic cases with SSTR2-targeted peptide-radionuclide receptor therapy (PRRT) in the LUTHREE trial was well-tolerated and resulted in stable disease (SD). CONCLUSIONS: This study presents pertinent clinical data from the largest dataset, to date, on ONB. We identify key prognostic markers and integrate these into an updated staging system, highlight the importance of adjuvant radiotherapy across all disease stages, the utility of prophylactic neck irradiation and the potential efficacy of targeting SSTR2 to manage disease.


Subject(s)
Esthesioneuroblastoma, Olfactory , Neuroblastoma , Nose Neoplasms , Esthesioneuroblastoma, Olfactory/pathology , Esthesioneuroblastoma, Olfactory/therapy , Humans , Nasal Cavity/metabolism , Nasal Cavity/pathology , Neuroblastoma/pathology , Nose Neoplasms/radiotherapy , Positron-Emission Tomography , Radioisotopes , Radionuclide Imaging , Receptors, Somatostatin/metabolism , Retrospective Studies
2.
Int J Cancer ; 149(5): 1129-1136, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33990938

ABSTRACT

Aloe-emodin (1,8-dihydroxy-3-[hydroxymethyl]-anthraquinone), AE, is one of the active constituents of a number of plant species used in traditional medicine. We have previously identified, for the first time, AE as a new antitumor agent and shown that its selective in vitro and in vivo killing of neuroblastoma cells was promoted by a cell-specific drug uptake process. However, the molecular mechanism underlying the cell entry of AE has remained elusive as yet. In this report, we show that AE enters tumor cells via two of the five somatostatin receptors: SSTR2 and SSTR5. This observation was suggested by gene silencing, receptor competition, imaging and molecular modeling experiments. Furthermore, SSTR2 was expressed in all surgical neuroblastoma specimens we analyzed by immunohistochemistry. The above findings have strong implications for the clinical adoption of this natural anthraquinone molecule as an antitumor agent.


Subject(s)
Aloe/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Biomarkers, Tumor/metabolism , Emodin/pharmacology , Neoplasms/drug therapy , Receptors, Somatostatin/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Receptors, Somatostatin/genetics , Tumor Cells, Cultured
3.
Cells ; 10(3)2021 03 06.
Article in English | MEDLINE | ID: mdl-33800837

ABSTRACT

Central actions of leptin and insulin on hepatic lipid metabolism can be opposing and the mechanism underlying this phenomenon remains unclear. Both hormones can modulate the central somatostatinergic system that has an inhibitory effect on growth hormone (GH) expression, which plays an important role in hepatic metabolism. Using a model of chronic central leptin infusion, we evaluated whether an increase in central leptin bioavailability modifies the serum lipid pattern through changes in hepatic lipid metabolism in male rats in response to an increase in central insulin and the possible involvement of the GH axis in these effects. We found a rise in serum GH in leptin plus insulin-treated rats, due to an increase in pituitary GH mRNA levels associated with lower hypothalamic somatostatin and pituitary somatostatin receptor-2 mRNA levels. An augment in hepatic lipolysis and a reduction in serum levels of non-esterified fatty acids (NEFA) and triglycerides were found in leptin-treated rats. These rats experienced a rise in lipogenic-related factors and normalization of serum levels of NEFA and triglycerides after insulin treatment. These results suggest that an increase in insulin in leptin-treated rats can act on the hepatic lipid metabolism through activation of the GH axis.


Subject(s)
Hypothalamus/drug effects , Insulin/pharmacology , Leptin/pharmacology , Lipid Metabolism/drug effects , Liver/drug effects , Pituitary Gland/drug effects , Animals , Fatty Acids, Nonesterified/blood , Gene Expression Regulation , Growth Hormone/genetics , Growth Hormone/metabolism , Hypothalamus/metabolism , Injections, Intravenous , Injections, Intraventricular , Insulin/metabolism , Leptin/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Signal Transduction , Triglycerides/blood
4.
Mol Inform ; 39(6): e2000020, 2020 06.
Article in English | MEDLINE | ID: mdl-32329245

ABSTRACT

Several encouraging pre-clinical results highlight the melanin-concentrating hormone receptor 1 (MCHR1) as promising target for anti-obesity drug development. Currently however, experimentally resolved structures of MCHR1 are not available, which complicates rational drug design campaigns. In this study, we aimed at developing accurate, homologymodel-based 3D pharmacophores against MCHR1. We show that traditional approaches involving docking of known active small molecules are hindered by the flexibility of binding pocket residues. Instead, we derived three-dimensional pharmacophores from molecular dynamics simulations by employing our novel open-source software PyRod. In a retrospective evaluation, the generated 3D pharmacophores were highly predictive returning up to 35 % of active molecules and showing an early enrichment (EF1) of up to 27.6. Furthermore, PyRod pharmacophores demonstrate higher sensitivity than ligand-based pharmacophores and deliver structural insights, which are key to rational lead optimization.


Subject(s)
Drug Evaluation, Preclinical , Software , Structural Homology, Protein , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Receptors, Somatostatin
5.
Clin Cancer Res ; 26(4): 957-969, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31624102

ABSTRACT

PURPOSE: Somatostatin analogues (SSA) are efficacious and safe treatments for a variety of neuroendocrine tumors, especially pituitary neuroendocrine tumors (PitNET). Their therapeutic effects are mainly mediated by somatostatin receptors SST2 and SST5. Most SSAs, such as octreotide/lanreotide/pasireotide, are either nonselective or activate mainly SST2. However, nonfunctioning pituitary tumors (NFPTs), the most common PitNET type, mainly express SST3 and finding peptides that activate this particular somatostatin receptor has been very challenging. Therefore, the main objective of this study was to identify SST3-agonists and characterize their effects on experimental NFPT models. EXPERIMENTAL DESIGN: Binding to SSTs and cAMP level determinations were used to screen a peptide library and identify SST3-agonists. Key functional parameters (cell viability/caspase activity/chromogranin-A secretion/mRNA expression/intracellular signaling pathways) were assessed on NFPT primary cell cultures in response to SST3-agonists. Tumor growth was assessed in a preclinical PitNET mouse model treated with a SST3-agonist. RESULTS: We successfully identified the first SST3-agonist peptides. SST3-agonists lowered cell viability and chromogranin-A secretion, increased apoptosis in vitro, and reduced tumor growth in a preclinical PitNET model. As expected, inhibition of cell viability in response to SST3-agonists defined two NFPT populations: responsive and unresponsive, wherein responsive NFPTs expressed more SST3 than unresponsive NFPTs and exhibited a profound reduction of MAPK, PI3K-AKT/mTOR, and JAK/STAT signaling pathways upon SST3-agonist treatments. Concurrently, SSTR3 silencing increased cell viability in a subset of NFPTs. CONCLUSIONS: This study demonstrates that SST3-agonists activate signaling mechanisms that reduce NFPT cell viability and inhibit pituitary tumor growth in experimental models that expresses SST3, suggesting that targeting this receptor could be an efficacious treatment for NFPTs.


Subject(s)
Neuroendocrine Tumors/drug therapy , Peptides/pharmacology , Pituitary Neoplasms/drug therapy , Receptors, Somatostatin/agonists , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Knockout , Middle Aged , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Peptides/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Signal Transduction , Tumor Cells, Cultured , Young Adult
6.
Int J Mol Sci ; 20(16)2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31412614

ABSTRACT

Endogenous somatostatin shows anti-secretory effects in both physiological and pathological settings, as well as inhibitory activity on cell growth. Since somatostatin is not suitable for clinical practice, researchers developed synthetic somatostatin receptor ligands (SRLs) to overcome this limitation. Currently, SRLs represent pivotal tools in the treatment algorithm of neuroendocrine tumors (NETs). Octreotide and lanreotide are the first-generation SRLs developed and show a preferential binding affinity to somatostatin receptor (SST) subtype 2, while pasireotide, which is a second-generation SRL, has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). A number of studies demonstrated that first-generation and second-generation SRLs show distinct functional properties, besides the mere receptor affinity. Therefore, the aim of the present review is to critically review the current evidence on the biological effects of SRLs in pituitary adenomas and neuroendocrine tumors, by mainly focusing on the differences between first-generation and second-generation ligands.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Receptors, Somatostatin/metabolism , Animals , Clinical Studies as Topic , Drug Evaluation, Preclinical , Humans , Ligands , Neuroendocrine Tumors/etiology , Neuroendocrine Tumors/pathology , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/etiology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Protein Binding , Protein Multimerization , Receptors, Somatostatin/chemistry , Signal Transduction , Treatment Outcome
7.
Nanomedicine ; 21: 102042, 2019 10.
Article in English | MEDLINE | ID: mdl-31247311

ABSTRACT

Targeted, biocompatible, and synergistic "all in one" systems should be designed to combat the heterogeneity of cancer. In this study, we constructed a dual function nanosystem, copper sulfide nanoplatform loaded with the chemotherapeutic drug docetaxel wrapped by a conjugated polymer-peptide for targeted chemo-phototherapy. The nanoconstruct has been successfully designed with a size of 186.1 ±â€¯5.2 nm, a polydispersity index of 0.18 ±â€¯0.01, and zeta potential of -16.4 ±â€¯0.1 mV. The enhanced uptake and near-infrared-responsive behavior of the nanosystem resulted in efficient drug release, photothermal ablation, effective cytotoxic activity, and potentiated reactive oxygen species generation. The induction of apoptotic markers, enhanced accumulation in the tumor site, and maximum tumor growth inhibition were seen during in vivo studies compared to non-targeted nanoformulations and free drug. Cumulatively, our results indicate that, with low systemic toxicity and better biocompatibility, this nanoconstruct could provide a promising strategy for treating prostate cancer.


Subject(s)
Doxorubicin/pharmacology , Drug Delivery Systems , Polymers/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/radiation effects , Cell Proliferation/drug effects , Copper/chemistry , Doxorubicin/chemistry , Drug Liberation/radiation effects , Humans , Hyperthermia, Induced , Male , Nanoparticles/chemistry , Peptides/chemistry , Peptides/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Phototherapy , Polymers/chemistry , Polymers/radiation effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Reactive Oxygen Species/chemistry , Receptors, Somatostatin/genetics , Somatostatin/analogs & derivatives , Somatostatin/chemistry , Somatostatin/pharmacology , Sulfides/chemistry
8.
Article in English, Spanish | MEDLINE | ID: mdl-30442558

ABSTRACT

BACKGROUND AND OBJECTIVES: To prospectively compare the detection rate of 68Ga-DOTATATE versus 11C-choline PET/CT in patients with prostate cancer in biochemical relapse, and to evaluate somatostatin receptor expression in vivo to plan targeted therapies (177Lu-DOTATATE). MATERIAL AND METHODS: We prospectively analysed 64 patients with biochemical relapse (median PSA: 4.25 ng/mL). A PET/CT was performed with 11C-choline, and another with 68Ga-DOTATATE. The SUVmax was measured in all lesions. The correlative images, histopathology and/or clinical and biochemical follow-up were taken as the reference standard. RESULTS: The overall detection rate per patient was 48.43% for 68Ga-DOTATATE and 46.87% for 11C-choline. The results were concordant in 53 cases (82.81%). The maximum SUV of 11C-choline was significantly higher than that of 68Ga-DOTATATE for all the concordant lesions (n=130): 6.17 (1.7-15.5) versus 4.38 (1.37-26.7), median (range) for each radiotracer, respectively (p < .0001). The sensitivity and specificity values per patient were the same for both techniques: 0.82 (0.65-0.93) and 0.9 (0.73-0.98), respectively. Although the difference was not significant, the sensitivity was lower in patients with lower PSA levels: 0.63 vs. 0.89; p=.13. A significant correlation was found between the SUVmax of both tracers (r = 0.41, n = 130, p <.0001). CONCLUSIONS: 68Ga-DOTATATE PET/CT and 11C-choline PET/CT seem to have a high capacity to detect pathological lesions in the assessment of patients with prostate cancer with biochemical relapse. Further studies are required to test the potential complementary value of these PET/CT techniques, and to evaluate the potential role of 8Ga-DOTATATE for planning somostatin receptor-mediated therapies (177Lu-DOTATATE).


Subject(s)
Choline/analogs & derivatives , Neoplasm Recurrence, Local/diagnostic imaging , Organometallic Compounds , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Receptors, Somatostatin/biosynthesis , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Positron Emission Tomography Computed Tomography/methods , Prospective Studies , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood
9.
Acta Biochim Pol ; 65(2): 251-258, 2018.
Article in English | MEDLINE | ID: mdl-29850655

ABSTRACT

We aimed to quantify the gene expression changes of the potent orexigenic melanin-concentrating hormone (MCH) in chicken (Gallus gallus) hypothalamus with quantitative real-time polymerase chain reaction (qPCR), and for the first time determine peptide concentrations with a novel radioimmunoassay (RIA) under different feeding status. Three different experimental conditions, namely ad libitum feeding; fasting for 24 h; fasting for 24 h and then refeeding for 2 h, were applied to study changes of the aforementioned target and its receptor (MCHR4) gene expression under different nutritional status. The relative changes of MCH and MCHR4 were also studied from 7 to 35 days of age. Expression of PMCH and MCHR4 along the gastrointestinal tract (GIT) was also investigated. We found that expression of both targets was significant in the hypothalamus, while only weak expression was detected along the GIT. Different nutritional states did not affect the PMCH and MCHR4 mRNA levels. However, fasting for 24 h had significantly increased the MCH-like immunoreactivity by 25.65%. Fasting for 24 h and then refeeding for 2 h had further significantly increased the MCH peptide concentration by 32.51%, as compared to the ad libitum state. A decreasing trend with age was observable for both, the PMCH and MCHR4 mRNA levels, and also for the MCH-like immunoreactivity. Correlation analysis did not result in a significant correlation between MCH peptide concentration and abdominal fat mass in ad libitum fed birds. In conclusion, MCH peptide concentration altered in response to 24 h fasting, which indicated that this peptide may take part in feed intake regulation of broiler chickens.


Subject(s)
Feeding Behavior , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Animals , Chickens , Fasting , Hypothalamic Hormones/analysis , Melanins/analysis , Pituitary Hormones/analysis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Somatostatin
10.
Genesis ; 56(8): e23217, 2018 08.
Article in English | MEDLINE | ID: mdl-29806135

ABSTRACT

The neuropeptide, melanin concentrating hormone (MCH), and its G protein-coupled receptor, melanin concentrating hormone receptor 1 (Mchr1), are expressed centrally in adult rodents. MCH signaling has been implicated in diverse behaviors such as feeding, sleep, anxiety, as well as addiction and reward. While a model utilizing the Mchr1 promoter to drive constitutive expression of Cre recombinase (Mchr1-Cre) exists, there is a need for an inducible Mchr1-Cre to determine the roles for this signaling pathway in neural development and adult neuronal function. Here, we generated a BAC transgenic mouse where the Mchr1 promotor drives expression of tamoxifen inducible CreER recombinase. Many aspects of the Mchr1-Cre expression pattern are recapitulated by the Mchr1-CreER model, though there are also notable differences. Most strikingly, compared to the constitutive model, the new Mchr1-CreER model shows strong expression in adult animals in hypothalamic brain regions involved in feeding behavior but diminished expression in regions involved in reward, such as the nucleus accumbens. The inducible Mchr1-CreER allele will help reveal the potential for Mchr1 signaling to impact neural development and subsequent behavioral phenotypes, as well as contribute to the understanding of the MCH signaling pathway in terminally differentiated adult neurons and the diverse behaviors that it influences.


Subject(s)
Hypothalamic Hormones/physiology , Melanins/physiology , Pituitary Hormones/physiology , Receptors, Somatostatin/physiology , Animals , Brain/metabolism , Brain/physiology , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Integrases , Melanins/metabolism , Mice , Mice, Transgenic , Models, Animal , Neurons/metabolism , Neuropeptides/metabolism , Pituitary Hormones/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction , Tamoxifen
11.
Brain Behav ; 8(1): e00881, 2018 01.
Article in English | MEDLINE | ID: mdl-29568682

ABSTRACT

Objectives: Bombesin receptor subtype-3 (BRS-3) has been suggested to play a potential role in energy homeostasis. However, the physiological mechanism of BRS-3 on energy homeostasis remains unknown. Thus, we investigated the BRS-3-mediated neuronal pathway involved in food intake and energy expenditure. Materials and Methods: Expression of BRS-3 in the rat brain was histologically examined. The BRS-3 neurons activated by refeeding-induced satiety or a BRS-3 agonist were identified by c-Fos immunostaining. We also analyzed expression changes in feeding-relating peptides in the brain of fasted rats administered with the BRS-3 agonist. Results: In the paraventricular hypothalamic nucleus (PVH), dorsomedial hypothalamic nucleus (DMH), and medial preoptic area (MPA), strong c-Fos induction was observed in the BRS-3 neurons especially in PVH after refeeding. However, the BRS-3 neurons in the PVH did not express feeding-regulating peptides, while the BRS-3 agonist administration induced c-Fos expression in the DMH and MPA, which were not refeeding-sensitive, as well as in the PVH. The BRS-3 agonist administration changed the Pomc and Cart mRNA level in several brain regions of fasted rats. Conclusion: These results suggest that BRS-3 neurons in the PVH are a novel functional subdivision in the PVH that regulates feeding behavior. As the MPA and DMH are reportedly involved in thermoregulation and energy metabolism, the BRS-3 neurons in the MPA/DMH might mediate the energy expenditure control. POMC and CART may contribute to BRS-3 neuron-mediated energy homeostasis regulation. In summary, BRS-3-expressing neurons could regulate energy homeostasis through a novel neuronal pathway.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/metabolism , Neurons/metabolism , Receptors, Bombesin/metabolism , Animals , CHO Cells , Cricetulus , Eating/physiology , Feeding Behavior/physiology , Hypothalamus/drug effects , Male , Mice, Knockout , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Bombesin/agonists , Receptors, Somatostatin/genetics
12.
Acta Biomater ; 68: 154-167, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29292170

ABSTRACT

Lanreotide (LT), a synthetic analog of somatostatin, has been demonstrated to specifically bind to somatostatin receptors (SSTRs), which are widely overexpressed in several types of cancer cells. In this study, we incorporated a chemotherapeutic agent, methotrexate (MTX), and a photosensitizer material, polyaniline (PANI), into hybrid polymer nanoparticles (NPs), which could target cancer cells after conjugation with LT (LT-MTX/PANI NPs). The successful preparation of LT-MTX/PANI NPs was confirmed by a small particle size (187.9 ±â€¯3.2 nm), a polydispersity index of 0.232 ±â€¯0.011, and a negative ζ potential of -14.6 ±â€¯1.0 mV. Notably, LT-MTX/PANI NPs showed a greater uptake into SSTR-positive cancer cells and thereby better inhibited cell viability and induced higher levels of apoptosis than MTX, PANI NP, and MTX/PANI NP treatments did. In addition, the heat associated with the burst drug release induced by near-infrared (NIR) irradiation resulted in remarkably enhanced cell apoptosis, which was confirmed by an increase in the expression levels of apoptotic marker proteins. In agreement with the in vitro results, the administration of the SSTR-targeting NPs, followed by NIR exposure, to xenograft tumor-bearing mice resulted in an improved suppression of tumor development compared to that shown by MTX, PANI NPs, and MTX/PANI NPs, as well as by LT-MTX/PANI NPs without photothermal therapy. Thus, the SSTR-targeting NPs could be a promising delivery system for the effective treatment of SSTR-positive cancers. STATEMENT OF SIGNIFICANCE: Somatostatin receptors are widely overexpressed in several types of cancer cells. In this study, we designed nanoparticles for targeted delivery of chemotherapeutic agents to tumor sites by conjugating hybrid polymers with a synthetic analog of somatostatin, specifically binding to somatostatin receptors. In addition, a photosensitizer material, polyaniline, was incorporated into the nanoparticles for combined chemo-photothermal therapy. The results demonstrated clear advantages of the newly designed targeted nanoparticles over their non-targeted counterparts or a free chemotherapeutic drug in inhibiting the viability of cancer cells in vitro and targeting/suppressing the tumor growth in an animal xenograft model. The study suggests that the designed nanoparticles are a promising delivery system for the effective treatment of somatostatin receptor-positive cancers.


Subject(s)
Aniline Compounds/chemistry , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Hyperthermia, Induced , Methotrexate/therapeutic use , Nanoparticles/chemistry , Phototherapy , Receptors, Somatostatin/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Humans , Methotrexate/pharmacology , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Tissue Distribution/drug effects
13.
Bioorg Med Chem ; 25(15): 4153-4162, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28622905

ABSTRACT

Somatostatin (SST) is a peptide hormone comprising 14 or 28 amino acids that inhibits endocrine and exocrine secretion via five distinct G-protein-coupled receptors (SSTR1-5). SSTR5 has an important role in inhibiting the secretion of pancreatic and gastrointestinal hormones (e.g., insulin, GLP-1, PYY) through the binding of SSTs; hence, SSTR5 antagonists are expected to be novel anti-diabetic drugs. In the course of our lead generation program of SSTR5 antagonists, we have discovered a novel spiroazetidine derivative 3a. However, pharmacological evaluation of 3a revealed that it had to be administered at a high dose (100mg/kg) to show a persistent glucose-lowering effect in an oral glucose tolerance test (OGTT). We therefore initiated an optimization study based on 3a aimed at improving the antagonistic activity and mean residence time (MRT), resulting in the identification of 2-cyclopropyl-5-methoxybiphenyl derivative 3k. However, 3k did not show a sufficient persistent glucose-lowering effect in an OGTT; moreover, hERG inhibition was observed. Hence, further optimization study of the biphenyl moiety of compound 3k, focused on improving the pharmacokinetic (PK) profile and hERG inhibition, was conducted. Consequently, the introduction of a chlorine atom at the 6-position on the biphenyl moiety addressed a putative metabolic soft spot and increased the dihedral angle of the biphenyl moiety, leading to the discovery of 3p with an improved PK profile and hERG inhibition. Furthermore, 3p successfully exhibited a persistent glucose-lowering effect in an OGTT at a dose of 3mg/kg.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Drug Design , Drug Discovery , Glucose Tolerance Test , Humans , Hypoglycemic Agents/chemistry
14.
Zhonghua Wei Chang Wai Ke Za Zhi ; 19(11): 1201-1204, 2016 Nov 25.
Article in Chinese | MEDLINE | ID: mdl-27928785

ABSTRACT

Neuroendocrine neoplasms(NEN) is a rare group of tumors with gastrointestinal tract as one of the most common primary locations. The most commonly used guidelines are proposed by European Neuroendocrine Tumor Society (ENETS) and National Comprehensive Cancer Network(NCCN) respectively, while the management of gastrointestinal NEN is generally identical in these two guidelines. Surgery is still the sole curative method which should be considered as the first choice of locoregional NEN. Otherwise, 40% to 50% of patients manifests metastatic disease when diagnosed. Hence, somatostatin analogs, targeted drugs, chemotherapy, and peptide receptor radionuclide therapy should be used comprehensively, especially for unresectable lesions. Therapeutic decision making should be based on the tumor location, functional status, tumor grade, tumor stage, somatostatin receptor status and adverse effect of drugs, etc.


Subject(s)
Gastrointestinal Neoplasms/therapy , Neuroendocrine Tumors/therapy , Gastrointestinal Neoplasms/chemistry , Humans , Pancreatic Neoplasms , Receptors, Somatostatin
15.
Alcohol Clin Exp Res ; 40(10): 2199-2207, 2016 10.
Article in English | MEDLINE | ID: mdl-27579857

ABSTRACT

BACKGROUND: Reward and energy homeostasis are both regulated by a network of hypothalamic neuropeptide systems. The melanin-concentrating hormone (MCH) and its MCH-1 receptor (MCH1-R) modulate alcohol intake, but it remains unknown to what extent this reflects actions on energy balance or reward. Here, we evaluated the MCH1-R in regulation of caloric intake and motivation to consume alcohol in states of escalated consumption. METHODS: Rats had intermittent access (IA) to alcohol and were divided into high- and low-drinking groups. Food and alcohol consumption was assessed after administration of an MCH1-R antagonist, GW803430. Next, GW803430 was evaluated on alcohol self-administration in protracted abstinence induced by IA in high-drinking rats. Finally, the effect of GW803430 was assessed on alcohol self-administration in acute withdrawal in rats exposed to alcohol vapor. Gene expression of MCH and MCH1-R was measured in the hypothalamus and nucleus accumbens (NAc) in both acute and protracted abstinence. RESULTS: High-drinking IA rats consumed more calories from alcohol than chow and GW803430 decreased both chow and alcohol intake. In low-drinking rats, only food intake was affected. In protracted abstinence from IA, alcohol self-administration was significantly reduced by pretreatment with GW803430 and gene expression of both MCH and the MCH1-R were dysregulated in hypothalamus and NAc. In contrast, during acute withdrawal from vapor exposure, treatment with GW803430 did not affect alcohol self-administration, and no changes in MCH or MCH1-R gene expression were observed. CONCLUSIONS: Our data suggest a dual role of MCH and the MCH1-R in regulation of alcohol intake, possibly through mechanisms involving caloric intake and reward motivation. A selective suppression of alcohol self-administration during protracted abstinence by GW803430 was observed and accompanied by adaptations in gene expression of MCH and MCH1-R. Selective suppression of escalated consumption renders the MCH1-R an attractive target for treatment of alcohol use disorders.


Subject(s)
Alcohol Drinking/physiopathology , Energy Intake/physiology , Hypothalamic Hormones/physiology , Melanins/physiology , Motivation/physiology , Pituitary Hormones/physiology , Receptors, Somatostatin/physiology , Animals , Eating/physiology , Gene Expression/drug effects , Gene Expression/physiology , Hypothalamic Hormones/biosynthesis , Hypothalamus/metabolism , Male , Melanins/biosynthesis , Nucleus Accumbens/metabolism , Pituitary Hormones/biosynthesis , Pyrimidinones/pharmacology , Rats , Receptors, Somatostatin/antagonists & inhibitors , Self Administration , Thiophenes/pharmacology
16.
J Nucl Med ; 57(11): 1805-1810, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27363833

ABSTRACT

Targeted diagnosis and therapy enable precise tumor detection and treatment. Successful examples for precise tumor targeting are diagnostic and therapeutic radioligands. However, patients with tumors expressing low levels of the relevant molecular targets are deemed ineligible for such targeted approaches. METHODS: We performed a screen for drugs that upregulate the somatostatin receptor subtype 2 (sstr2). Then, we characterized the effects of these drugs on transcriptional, translational, and functional levels in vitro and in vivo. RESULTS: We identified 9 drugs that act as epigenetic modifiers, including the inhibitor of DNA methyltransferase decitabine as well as the inhibitors of histone deacetylase tacedinaline and romidepsin. In vitro, these drugs upregulated sstr2 on transcriptional, translational, and functional levels in a time- and dose-dependent manner. Thereby, their combinations revealed synergistic effects. In vivo, drug-based sstr2 upregulation improved the tumor-to-background and tumor-to-kidney ratios, which are the key determinants of successful sstr2-targeted imaging and radiopeptide therapy. CONCLUSION: We present an approach that uses epigenetic modifiers to improve sstr2 targeting in vitro and in vivo. Translation of this method into the clinic may potentially convert patients ineligible for targeted imaging and therapy to eligible candidates.


Subject(s)
Azacitidine/analogs & derivatives , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Organometallic Compounds/pharmacokinetics , Pathology, Molecular/methods , Receptors, Somatostatin/metabolism , Animals , Azacitidine/administration & dosage , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Decitabine , Drug Evaluation, Preclinical/methods , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Positron-Emission Tomography/methods , Reproducibility of Results , Sensitivity and Specificity , Up-Regulation/drug effects
17.
Discov Med ; 21(117): 381-9, 2016 05.
Article in English | MEDLINE | ID: mdl-27355334

ABSTRACT

Neuroendocrine tumors (NETs) comprise a wide range of neoplasms with diverse biological behaviors, often secreting excessive amounts of endocrine-active substances causing hormone syndromes. They are classified according to the location of the primary site and the level of histological differentiation, which has prognostic as well as therapeutic implications. Biotherapy had traditionally a significant role in the treatment of these tumors, when not amenable to surgery or local treatments. Control of carcinoid syndrome with somatostatin analogs (SSAs) significantly contributed to the improvement of the quality of life. Also, interferon has long been administered, but data were based on small studies. In contrast, PROMID and CLARINET randomized phase III trials provided the first strong evidence of significant improvement in progression-free survival in patients with gastroenteropancreatic (GEP)-NETs with octreotide and lanreotide, respectively, validating somatostatin receptors as important targets. Clinical trials testing the role of these SSAs in other primaries, e.g., lung carcinoids, as well as the efficacy of newer analogs are underway.


Subject(s)
Antineoplastic Agents/therapeutic use , Biological Therapy/methods , Biological Therapy/trends , Neuroendocrine Tumors/therapy , Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/analogs & derivatives , Chemoradiotherapy/methods , Clinical Trials as Topic , Disease-Free Survival , Humans , Immunologic Factors/therapeutic use , Interferons/therapeutic use , Neuroendocrine Tumors/mortality , Octreotide/therapeutic use , Peptides, Cyclic/therapeutic use , Prognosis , Quality of Life , Somatostatin/therapeutic use
18.
Mol Endocrinol ; 30(4): 479-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26967369

ABSTRACT

Stable somatostatin analogues and dopamine receptor agonists are the mainstay for the pharmacological treatment of functional pituitary adenomas; however, only a few cellular assays have been developed to detect receptor activation of novel compounds without disrupting cells to obtain the second messenger content. Here, we adapted a novel fluorescence-based membrane potential assay to characterize receptor signaling in a time-dependent manner. This minimally invasive technique provides a robust and reliable read-out for ligand-induced receptor activation in permanent and primary pituitary cells. The mouse corticotropic cell line AtT-20 endogenously expresses both the somatostatin receptors 2 (sst2) and 5 (sst5). Exposure of wild-type AtT-20 cells to the sst2- and sst5-selective agonists BIM-23120 and BIM-23268, respectively, promoted a pertussis toxin- and tertiapin-Q-sensitive reduction in fluorescent signal intensity, which is indicative of activation of G protein-coupled inwardly rectifying potassium (GIRK) channels. After heterologous expression, sst1, sst3, and sst4 receptors also coupled to GIRK channels in AtT-20 cells. Similar activation of GIRK channels by dopamine required overexpression of dopamine D2 receptors (D2Rs). Interestingly, the presence of D2Rs in AtT-20 cells strongly facilitated GIRK channel activation elicited by the sst2-D2 chimeric ligand BIM-23A760, suggesting a synergistic action of sst2 and D2Rs. Furthermore, stable somatostatin analogues produced strong responses in primary pituitary cultures from wild-type mice; however, in cultures from sst2 receptor-deficient mice, only pasireotide and somatoprim, but not octreotide, induced a reduction in fluorescent signal intensity, suggesting that octreotide mediates its pharmacological action primarily via the sst2 receptor.


Subject(s)
Dopamine/physiology , Somatostatin/physiology , Animals , Cell Line , Dopamine/analogs & derivatives , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dopaminergic Neurons/physiology , Drug Evaluation, Preclinical , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Male , Membrane Potentials , Mice, Inbred C57BL , Pituitary Gland/cytology , Primary Cell Culture , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction , Single-Cell Analysis , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Spectrometry, Fluorescence
19.
Brain Struct Funct ; 221(7): 3803-23, 2016 09.
Article in English | MEDLINE | ID: mdl-26843217

ABSTRACT

Neuronal calcium-binding protein 1 and -2 (NECAB1/2) localize to multiple excitatory neuron populations in the mouse spinal cord. Here, we analyzed rat and human spinal cord, combining in situ hybridization and immunohistochemistry, complementing newly collated data on mouse spinal cord for direct comparisons. Necab1/2 mRNA transcripts showed complementary distribution in rodent's spinal cord. Multiple-labeling fluorescence histochemistry with neuronal phenotypic markers localized NECAB1 to a dense fiber plexus in the dorsal horn, to neurons mainly in superficial layers and to commissural interneurons in both rodent species. NECAB1-positive (+) motor neurons were only found in mice. NECAB1 distribution in the human spinal cord was similar with the addition of NECAB1-like immunoreactivity surrounding myelinated axons. NECAB2 was mainly present in excitatory synaptic boutons in the dorsal horn of all three species, and often in calbindin-D28k(+) neuronal somata. Rodent ependymal cells expressed calbindin-D28k. In humans, they instead were NECAB2(+) and/or calretinin(+). Our results reveal that the association of NECAB2 to excitatory neuronal circuits in the spinal cord is evolutionarily conserved across the mammalian species investigated so far. In contrast, NECAB1 expression is more heterogeneous. Thus, our study suggests that the phenotypic segregation of NECAB1 and -2 to respective excitatory and inhibitory spinal systems can underpin functional modalities in determining the fidelity of synaptic neurotransmission and neuronal responsiveness, and might bear translational relevance to humans.


Subject(s)
Calcium-Binding Proteins/metabolism , Eye Proteins/metabolism , Mixed Function Oxygenases/metabolism , Neurons/metabolism , Spinal Cord/metabolism , Animals , Calbindin 1/metabolism , Calbindin 2/metabolism , Glutamate Decarboxylase/metabolism , Humans , Male , Mice, Inbred C57BL , Motor Neurons/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/metabolism , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
20.
Article in Chinese | WPRIM | ID: wpr-323509

ABSTRACT

Neuroendocrine neoplasms(NEN) is a rare group of tumors with gastrointestinal tract as one of the most common primary locations. The most commonly used guidelines are proposed by European Neuroendocrine Tumor Society (ENETS) and National Comprehensive Cancer Network(NCCN) respectively, while the management of gastrointestinal NEN is generally identical in these two guidelines. Surgery is still the sole curative method which should be considered as the first choice of locoregional NEN. Otherwise, 40% to 50% of patients manifests metastatic disease when diagnosed. Hence, somatostatin analogs, targeted drugs, chemotherapy, and peptide receptor radionuclide therapy should be used comprehensively, especially for unresectable lesions. Therapeutic decision making should be based on the tumor location, functional status, tumor grade, tumor stage, somatostatin receptor status and adverse effect of drugs, etc.


Subject(s)
Humans , Gastrointestinal Neoplasms , Chemistry , Therapeutics , Neuroendocrine Tumors , Therapeutics , Pancreatic Neoplasms , Receptors, Somatostatin
SELECTION OF CITATIONS
SEARCH DETAIL