Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 369
Filter
1.
Best Pract Res Clin Endocrinol Metab ; 38(4): 101906, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38845246

ABSTRACT

Injectable first-generation somatostatin receptor ligands (fg-SRLs) are the standard of care of medical treatment for acromegaly. While fg-SRLs control acromegaly in up to 50 % of patients, they may lead to bothersome injection pain and site reactions. Paltusotine is an investigational, highly selective somatostatin receptor subtype 2 agonist, which is administered orally once a day. To date, phase 2 and 3 clinical trials suggest paltusotine treatment can achieve biochemical and symptom control in acromegaly, with a safety profile comparable to those of the fg-SRLs. Since paltusotine is a once-daily oral drug, it may represent a future treatment option for addressing patient preference or improving quality of life.


Subject(s)
Acromegaly , Receptors, Somatostatin , Humans , Receptors, Somatostatin/agonists , Acromegaly/drug therapy
2.
Proc Natl Acad Sci U S A ; 121(26): e2321710121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38885377

ABSTRACT

Somatostatin receptor 5 (SSTR5) is an important G protein-coupled receptor and drug target for neuroendocrine tumors and pituitary disorders. This study presents two high-resolution cryogenicelectron microscope structures of the SSTR5-Gi complexes bound to the cyclic neuropeptide agonists, cortistatin-17 (CST17) and octreotide, with resolutions of 2.7 Å and 2.9 Å, respectively. The structures reveal that binding of these peptides causes rearrangement of a "hydrophobic lock", consisting of residues from transmembrane helices TM3 and TM6. This rearrangement triggers outward movement of TM6, enabling Gαi protein engagement and receptor activation. In addition to hydrophobic interactions, CST17 forms conserved polar contacts similar to somatostatin-14 binding to SSTR2, while further structural and functional analysis shows that extracellular loops differently recognize CST17 and octreotide. These insights elucidate agonist selectivity and activation mechanisms of SSTR5, providing valuable guidance for structure-based drug development targeting this therapeutically relevant receptor.


Subject(s)
Octreotide , Receptors, Somatostatin , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry , Humans , Octreotide/chemistry , Octreotide/pharmacology , Octreotide/metabolism , Neuropeptides/metabolism , Neuropeptides/chemistry , Cryoelectron Microscopy , Protein Binding , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/metabolism , Somatostatin/metabolism , Somatostatin/chemistry , Somatostatin/analogs & derivatives , Models, Molecular , HEK293 Cells
3.
Best Pract Res Clin Endocrinol Metab ; 38(4): 101893, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38575404

ABSTRACT

Although predictors of response to first-generation somatostatin receptor ligands (fg-SRLs), and to a lesser extent to pasireotide, have been studied in acromegaly for many years, their use is still not recommended in clinical guidelines. Is there insufficient evidence to use them? Numerous biomarkers including various clinical, functional, radiological and molecular markers have been identified. The first ones are applicable pre-surgery, while the molecular predictors are utilized for patients not cured after surgery. In this regard, factors predicting a good response to fg-SRLs are specifically: low basal GH, a low GH nadir in the acute octreotide test, T2 MRI hypointensity, a densely granulated pattern, high immunohistochemistry staining for somatostatin receptor 2 (SSTR2), and E-cadherin. However, there is still a lack of consensus regarding which of these biomarkers is more useful or how to integrate them into clinical practice. With classical statistical methods, it is complex to define reliable and generalizable cut-off values for a single biomarker. The potential solution to the limitations of traditional methods involves combining systems biology with artificial intelligence, which is currently providing answers to such long-standing questions that may eventually be finally included into the clinical guidelines and make personalized medicine a reality. The aim of this review is to describe the current knowledge of the main fg-SRLs and pasireotide response predictors, discuss their current usefulness, and point to future directions in the research of this field.


Subject(s)
Acromegaly , Receptors, Somatostatin , Somatostatin , Humans , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/agonists , Acromegaly/drug therapy , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Ligands , Octreotide/therapeutic use , Human Growth Hormone/metabolism , Biomarkers/blood , Treatment Outcome
4.
Best Pract Res Clin Endocrinol Metab ; 38(4): 101875, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38290866

ABSTRACT

"First-generation" somatostatin receptor agonists (SSTRAs) octreotide and lanreotide are the most commonly used first-line pharmacological therapy for patients with acromegaly. A subset of patients respond only partially or not at all to the first-generation SSTRA, necessitating the use of additional pharmacological agents or other modes of therapy. Pasireotide is a "second-generation" SSTRA that has multi-receptor activity. Prospective studies have shown promise in the use of pasireotide in patients with poor response to first-generation SSTRA. Here we elucidate the molecular pathways of resistance to first-generation SSTRA, the mechanism of action, pre-clinical and clinical evidence of the use of pasireotide in patients having incomplete / lack of response to first-generation SSTRA. We also discuss the clinical, pathological, and radiological markers predicting response to pasireotide, and the difference in side-effect profiles of pasireotide, compared to first-generation SSTRA.


Subject(s)
Acromegaly , Somatostatin , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Humans , Acromegaly/drug therapy , Receptors, Somatostatin/agonists , Treatment Outcome
5.
Cell Res ; 32(8): 761-772, 2022 08.
Article in English | MEDLINE | ID: mdl-35739238

ABSTRACT

Somatostatin receptors (SSTRs) play versatile roles in inhibiting the secretion of multiple hormones such as growth hormone and thyroid-stimulating hormone, and thus are considered as targets for treating multiple tumors. Despite great progress made in therapeutic development against this diverse receptor family, drugs that target SSTRs still show limited efficacy with preferential binding affinity and conspicuous side-effects. Here, we report five structures of SSTR2 and SSTR4 in different states, including two crystal structures of SSTR2 in complex with a selective peptide antagonist and a non-peptide agonist, respectively, a cryo-electron microscopy (cryo-EM) structure of Gi1-bound SSTR2 in the presence of the endogenous ligand SST-14, as well as two cryo-EM structures of Gi1-bound SSTR4 in complex with SST-14 and a small-molecule agonist J-2156, respectively. By comparison of the SSTR structures in different states, molecular mechanisms of agonism and antagonism were illustrated. Together with computational and functional analyses, the key determinants responsible for ligand recognition and selectivity of different SSTR subtypes and multiform binding modes of peptide and non-peptide ligands were identified. Insights gained in this study will help uncover ligand selectivity of various SSTRs and accelerate the development of new molecules with better efficacy by targeting SSTRs.


Subject(s)
Neoplasms , Receptors, Somatostatin , Cryoelectron Microscopy , Humans , Ligands , Neoplasms/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Somatostatin/pharmacology , Somatostatin/therapeutic use
6.
Bioorg Med Chem Lett ; 71: 128807, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35605837

ABSTRACT

SST5 receptor activation potently inhibits insulin secretion from pancreatic ß-cells, and an orally available nonpeptide selective SST5 agonist may be used to effectively manage the blood glucose levels of congenital HI patients to avoid severe hypoglycemia. Our medicinal chemistry efforts have led to the discovery of 4-(3-aminopyrrolidinyl)-3-aryl-5-(benzimidazol-2-yl)-pyridine analogs as potent SST5 agonists. This class of molecules exhibits excellent human SST5 potency and selectivity against SST1, SST2, SST3 and SST4 receptors. Leading compound 3-{4-[(3S)-3-aminopyrrolidin-1-yl]-5-(4-methyl-1H-1,3-benzodiazol-2-yl)pyridin-3-yl-5-fluorobenzonitrile (28, CRN02481) showed limited off-target activity and good pharmacokinetic profiles in both male Sprague Dawley rats and Beagle dogs to advance into further preclinical evaluations.


Subject(s)
Congenital Hyperinsulinism , Somatostatin , Animals , Congenital Hyperinsulinism/drug therapy , Dogs , Humans , Male , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/agonists , Somatostatin/pharmacology , Somatostatin/physiology
7.
Elife ; 112022 04 21.
Article in English | MEDLINE | ID: mdl-35446253

ABSTRACT

Somatostatin is a peptide hormone that regulates endocrine systems by binding to G-protein-coupled somatostatin receptors. Somatostatin receptor 2 (SSTR2) is a human somatostatin receptor and is highly implicated in hormone disorders, cancers, and neurological diseases. Here, we report the high-resolution cryo-EM structure of full-length human SSTR2 bound to the agonist somatostatin (SST-14) in complex with inhibitory G (Gi) proteins. Our structural and mutagenesis analyses show that seven transmembrane helices form a deep pocket for ligand binding and that SSTR2 recognizes the highly conserved Trp-Lys motif of SST-14 at the bottom of the pocket. Furthermore, our sequence analysis combined with AlphaFold modeled structures of other SSTR isoforms provide a structural basis for the mechanism by which SSTR family proteins specifically interact with their cognate ligands. This work provides the first glimpse into the molecular recognition mechanism of somatostatin receptors and a crucial resource to develop therapeutics targeting somatostatin receptors.


Subject(s)
Receptors, Somatostatin , Somatostatin , Cryoelectron Microscopy , Humans , Ligands , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Somatostatin/metabolism
8.
Mol Neurobiol ; 59(1): 245-265, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34665407

ABSTRACT

The melanin-concentrating hormone (MCH) system is involved in numerous functions, including energy homeostasis, food intake, sleep, stress, mood, aggression, reward, maternal behavior, social behavior, and cognition. In rodents, MCH acts on MCHR1, a G protein-coupled receptor, which is widely expressed in the brain and abundantly localized to neuronal primary cilia. Cilia act as cells' antennas and play crucial roles in cell signaling to detect and transduce external stimuli to regulate cell differentiation and migration. Cilia are highly dynamic in terms of their length and morphology; however, it is not known if cilia length is causally regulated by MCH system activation in vivo. In the current work, we examined the effects of activation and inactivation of MCH system on cilia lengths by using different experimental models and methodologies, including organotypic brain slice cultures from rat prefrontal cortex (PFC) and caudate-putamen (CPu), in vivo pharmacological (MCHR1 agonist and antagonist GW803430), germline and conditional genetic deletion of MCHR1 and MCH, optogenetic, and chemogenetic (designer receptors exclusively activated by designer drugs (DREADD)) approaches. We found that stimulation of MCH system either directly through MCHR1 activation or indirectly through optogenetic and chemogenetic-mediated excitation of MCH-neuron, caused cilia shortening, detected by the quantification of the presence of ADCY3 protein, a known primary cilia marker. In contrast, inactivation of MCH signaling through pharmacological MCHR1 blockade or through genetic manipulations - germline deletion of MCHR1 and conditional ablation of MCH neurons - induced cilia lengthening. Our study is the first to uncover the causal effects of the MCH system in the regulation of the length of brain neuronal primary cilia. These findings place MCH system at a unique position in the ciliary signaling in physiological and pathological conditions and implicate MCHR1 present at primary cilia as a potential therapeutic target for the treatment of pathological conditions characterized by impaired primary cilia function associated with the modification of its length.


Subject(s)
Caudate Nucleus/metabolism , Cilia/metabolism , Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Prefrontal Cortex/metabolism , Receptors, Somatostatin/metabolism , Animals , Caudate Nucleus/drug effects , Cilia/drug effects , Hypothalamic Hormones/genetics , Melanins/genetics , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Optogenetics , Pituitary Hormones/genetics , Prefrontal Cortex/drug effects , Pyrimidinones/pharmacology , Rats , Rats, Wistar , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Thiophenes/pharmacology
9.
Bioorg Med Chem ; 49: 116424, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34626901

ABSTRACT

Somatostatin receptors are members of G-protein coupled receptor superfamily. Receptors can be classified into five subtypes, SSTR1 to 5. The highly potent and orally active SSTR2 agonist 7, which had been identified by our group, was found out to have toxicological liabilities such as hERG inhibition and phospholipidosis (PLD). We investigated the relationship between in silico physicochemical properties and hERG and PLD, and explored well-balanced agonists to identify amide 19 and benzimidazole 30. As a result of this exploration, we found out that the value of (cLogP) [2] + (pKa) [2] needs to be less than 110 to mitigate the liabilities.


Subject(s)
Amides/pharmacology , Benzimidazoles/pharmacology , Drug Design , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Phospholipids/antagonists & inhibitors , Receptors, Somatostatin/agonists , Amides/chemical synthesis , Amides/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Molecular Structure , Phospholipids/metabolism , Structure-Activity Relationship
10.
Front Endocrinol (Lausanne) ; 12: 677919, 2021.
Article in English | MEDLINE | ID: mdl-34025586

ABSTRACT

Background: First-generation somatostatin receptor ligands (fg-SRLs) represent the first-line medical treatment for acromegaly, recommended in patients with persistent disease after neurosurgery, or when surgical approach is not feasible. Despite the lack of strong recommendations from guidelines and consensus statements, data from national Registries report an increasing use of medical therapy as first-line treatment in acromegaly. Objective: We retrospectively evaluated the potential role of a large number of clinical and radiological parameters in predicting the biochemical response to 6-month treatment with fg-SRLs, in a cohort of naïve acromegaly patients referred to a single tertiary center for pituitary diseases. Methods: Univariable and multivariable logistic regression and linear regression analyses were performed. Biochemical response was defined based on IGF-1 levels, represented as both categorical (tight control, control, >50% reduction) and continuous (linear % reduction) variables. Results: Fifty-one patients (33 females, median age 57 years) were included in the study. At univariable logistic regression analysis, we found that younger age (≤ 40 years; OR 0.04, p=0.045) and higher BMI (OR 0.866, p=0.034) were associated with a lower chance of achieving >50% IGF-1 reduction. On the contrary, higher IGF-1 xULN values at diagnosis (OR 2.304, p=0.007) and a T2-hypointense tumor (OR 18, p=0.017) were associated with a significantly higher likelihood of achieving >50% IGF-1 reduction after SRL therapy. Of note, dichotomized age, IGF1 xULN at diagnosis, and T2-hypointense signal of the tumor were retained as significant predictors by our multivariable logistic regression model. Furthermore, investigating the presence of predictors to the linear % IGF-1 reduction, we found a negative association with younger age (≤ 40 years; ß -0.533, p<0.0001), while a positive association was observed with both IGF-1 xULN levels at diagnosis (ß 0.330, p=0.018) and the presence of a T2-hypointense pituitary tumor (ß 0.466, p=0.019). All these variables were still significant predictors at multivariable analysis. Conclusions: Dichotomized age, IGF-1 levels at diagnosis, and tumor T2-weighted signal are reliable predictors of both >50% IGF-1 reduction and linear % IGF-1 reduction after 6 month fg-SRL treatment in naïve acromegaly patients. These parameters should be considered in the light of an individualized treatment for acromegaly patients.


Subject(s)
Acromegaly/drug therapy , Human Growth Hormone/blood , Insulin-Like Growth Factor I/metabolism , Receptors, Somatostatin/agonists , Acromegaly/blood , Adult , Age Factors , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome
11.
Elife ; 102021 03 02.
Article in English | MEDLINE | ID: mdl-33650969

ABSTRACT

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


Subject(s)
Cilia/drug effects , Pyramidal Cells/physiology , Receptors, Somatostatin/drug effects , Synapses/physiology , Animals , Cells, Cultured , Female , Male , Neocortex/cytology , Patch-Clamp Techniques , Rats, Long-Evans , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Signal Transduction
12.
Exp Eye Res ; 204: 108449, 2021 03.
Article in English | MEDLINE | ID: mdl-33465395

ABSTRACT

Somatostatin plays important roles in modulating neuronal functions by activating the five specific G-protein coupled receptors (sst1-sst5). Previous studies have demonstrated that sst5 were expressed in retinal ganglion cells (RGCs) and sst5 agonist attenuated the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid-induced retinal neurotoxicity. In this study, we investigated effects and underlying mechanisms of the sst5 agonist L-817,818 on RGC injury induced by elevated intraocular pressure (COH) in experimental glaucoma. Our results showed that intraperitoneal administration of L-817,818 significantly reduced RGC loss and decreased the number of terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (TUNEL)-positive RGCs in COH retinas, suggesting that L-817,818 may attenuate RGC apoptosis. Consistently, in COH retinas with L-817,818 administration, both the down-regulated mRNA and protein levels of anti-apoptotic Bcl-2 and the up-regulated mRNA and protein levels of pro-apoptotic Bax were partially reversed. L-817,818 administration downregulated the expression of apoptosis-related proteins caspase-9 and caspase-3 in COH retinas. In addition, L-817,818 administration reduced the concentrations of reactive oxygen species/reactive nitrogen species and malondialdehyde, and ameliorated the functions of mitochondrial respiratory chain complex (MRCC). Our results imply that administration of the sst5 agonist L-817,818 reduces RGC loss in COH rats through decreasing RGC apoptosis, which is mediated by regulating Bcl-2/Bax balance, reducing oxidative stress and rescuing activities of MRCC. Activation of sst5 may provide neuroprotective roles for RGCs in glaucoma.


Subject(s)
Amides/pharmacology , Disease Models, Animal , Glaucoma/pathology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Somatostatin/agonists , Retinal Ganglion Cells/drug effects , Animals , Apoptosis/drug effects , Cell Survival , Hydrogen Peroxide/metabolism , In Situ Nick-End Labeling , Injections, Intraperitoneal , Male , Malondialdehyde/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/genetics
13.
Int J Mol Sci ; 21(20)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096776

ABSTRACT

Somatostatin is an important mood and pain-regulating neuropeptide, which exerts analgesic, anti-inflammatory, and antidepressant effects via its Gi protein-coupled receptor subtype 4 (SST4) without endocrine actions. SST4 is suggested to be a unique novel drug target for chronic neuropathic pain, and depression, as a common comorbidity. However, its neuronal expression and cellular mechanism are poorly understood. Therefore, our goals were (i) to elucidate the expression pattern of Sstr4/SSTR4 mRNA, (ii) to characterize neurochemically, and (iii) electrophysiologically the Sstr4/SSTR4-expressing neuronal populations in the mouse and human brains. Here, we describe SST4 expression pattern in the nuclei of the mouse nociceptive and anti-nociceptive pathways as well as in human brain regions, and provide neurochemical and electrophysiological characterization of the SST4-expressing neurons. Intense or moderate SST4 expression was demonstrated predominantly in glutamatergic neurons in the major components of the pain matrix mostly also involved in mood regulation. The SST4 agonist J-2156 significantly decreased the firing rate of layer V pyramidal neurons by augmenting the depolarization-activated, non-inactivating K+ current (M-current) leading to remarkable inhibition. These are the first translational results explaining the mechanisms of action of SST4 agonists as novel analgesic and antidepressant candidates.


Subject(s)
Analgesics/pharmacology , Brain/metabolism , Neurons/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Affect/physiology , Animals , Brain/cytology , Butanes/pharmacology , CHO Cells , Cricetulus , Electrophysiology/methods , Humans , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Targeted Therapy , Naphthalenes/pharmacology , Neurons/drug effects , Receptors, Somatostatin/agonists , Sulfones/pharmacology , Vesicular Glutamate Transport Protein 1/genetics
14.
Bioconjug Chem ; 31(10): 2431-2438, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33047605

ABSTRACT

Tumor targeting with bivalent radiolabeled ligands for GPCRs is an attractive means for cancer imaging and therapy. Here, we studied and compared the distance dependence of homobivalent ligands for the human gastrin-releasing peptide receptor (hGRP-R) and the somatostatin receptor subtype II (hSstR2a). Oligoprolines were utilized as molecular scaffolds to enable distances of 10, 20, or 30 Å between two identical, agonistic recognition motifs. In vitro internalization assays revealed that ligands with a distance of 20 Å between the recognition motifs exhibit the highest cellular uptake in both ligand series. Structural modeling and molecular dynamics simulations support an optimal distance of 20 Å for accommodating ligand binding to both binding sites of a GPCR dimer. Translation of these findings to the significantly higher complexity in vivo proved difficult and showed only for the hGRP-R increased tumor uptake of the bivalent ligand.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Proline/analogs & derivatives , Proline/pharmacokinetics , Receptors, Bombesin/agonists , Receptors, Somatostatin/agonists , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Ligands , Mice, SCID , Molecular Dynamics Simulation , Neoplasms/metabolism , Oligopeptides/pharmacology , Proline/pharmacology , Receptors, Bombesin/metabolism , Receptors, Somatostatin/metabolism
15.
Res Vet Sci ; 133: 163-173, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33002813

ABSTRACT

We studied the inflammation effect on somatostatin receptors subtypes 2 (sstr2) and 5 (sstr5) expression in myometrium and somatostatin influence alone or with sstr2 and sstr5 antagonists on the contractility of gilt inflamed uterus. On day 3 of the estrous cycle, either E.coli suspension (E.coli group) or saline (SAL group) were injected into uterine horns. In the control pigs (CON group), only laparotomy was performed. Eight days later, in the E.coli group developed severe acute endometritis. In this group, myometrial sstr2 mRNA expression lowered and protein expression increased compared to other groups. Compared to period before somatostatin administration, somatostatin did not change tension in myometrium and endometrium/myometrium of three groups, reduced amplitude and frequency in the CON and SAL groups, and increased amplitude and decreased frequency in the E.coli group. In this group, amplitude was increased by somatostatin compared to other groups. In the CON and SAL groups, sstr2 eliminated inhibitory somatostatin effect on amplitude, while sstr5 antagonist reversed inhibitory somatostatin effect on amplitude. In the E.coli group, sstr2 antagonist reversed stimulatory somatostatin effect on amplitude, while in sstr5 antagonist presence stimulatory somatostatin effect was more deepened compared to somatostatin action alone. After using sstr2 antagonist more deepened inhibitory somatostatin effect on frequency in the CON and E.coli groups was found. Sstr5 antagonist partly eliminated inhibitory somatostatin effect on frequency in the SAL group. Summarizing, the uterine inflammation increases the myometrial sstr2 protein expression; somatostatin raises amplitude of the inflamed uterus acting by sstr2, while drops this parameter by sstr5.


Subject(s)
Escherichia coli Infections/veterinary , Gene Expression Regulation/drug effects , Receptors, Somatostatin/genetics , Somatostatin/pharmacology , Uterine Contraction/drug effects , Uterine Diseases/veterinary , Animals , Anti-Inflammatory Agents/pharmacology , Endometrium/drug effects , Escherichia coli Infections/physiopathology , Female , Hormones/pharmacology , Inflammation/drug therapy , Inflammation/veterinary , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Somatostatin/therapeutic use , Sus scrofa , Swine , Swine Diseases/drug therapy , Swine Diseases/physiopathology , Uterine Contraction/metabolism , Uterine Diseases/physiopathology
16.
Pharm Res ; 37(11): 217, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33037505

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/metabolism , Fluorescent Dyes/chemistry , Leukemia/metabolism , Monocytes/metabolism , Octreotide/metabolism , Quantum Dots , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Compounding , Flow Cytometry , HeLa Cells , Humans , Leukemia/drug therapy , Leukemia/pathology , Male , Mice , Microscopy, Confocal , Octreotide/chemistry , Octreotide/pharmacology
17.
Neuropharmacology ; 178: 108198, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32739276

ABSTRACT

We provided strong proof of concept evidence that somatostatin mediates potent analgesic and anti-inflammatory actions via its receptor subtype 4 (sst4) located both at the periphery and the central nervous system. Therefore, sst4 agonists are promising novel drug candidates for neuropathic pain and neurogenic inflammation, but rational drug design was not possible due to the lack of knowledge about its 3-dimensional structure. We modeled the sst4 receptor structure, described its agonist binding properties, and characterized the binding of our novel small molecule sst4 agonists (4-phenetylamino-7H-pyrrolo[2,3-d]pyrimidine derivatives) using an in silico platform. In addition to the in silico binding data, somatostatin displacement by Compound 1 was demonstrated in the competitive binding assay on sst4-expressing cells. In vivo effects were investigated in rat models of neurogenic inflammation and chronic traumatic neuropathic pain. We defined high- and low-affinity binding pockets of sst4 for our ligands, binding of the highest affinity compounds were similar to that of the reference ligand J-2156. We showed potent G-protein activation with the highest potency of 10 nM EC50 value and highest efficacy of 342%. Oral administration of 100 µg/kg of 5 compounds significantly inhibited acute neurogenic plasma protein extravasation in the paw skin by 40-60%, one candidate abolished and 3 others diminished sciatic nerve-ligation induced neuropathic hyperalgesia by 28-62%. The in silico predictions on sst4-ligands were tested in biological systems. Low oral dose of our novel agonists inhibit neurogenic inflammation and neuropathic pain, which opens promising drug developmental perspectives for these unmet medical need conditions.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Receptors, Somatostatin/agonists , Amino Acid Sequence , Analgesics/chemistry , Analgesics/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , Male , Pain Measurement/drug effects , Pain Measurement/methods , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Wistar , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism
18.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32754748

ABSTRACT

CONTEXT: Combination therapy with somatostatin receptor ligand (SRL) plus pegvisomant for patients with acromegaly is recommended after a maximizing dose on monotherapy. Lower-dose combination regimens are not well studied. OBJECTIVE: To compare cost-effectiveness and efficacy of 3 lower-dose combination regimens in controlled and uncontrolled acromegaly. DESIGN AND SETTING: Prospective, randomized, open-label, parallel arm study at a tertiary referral pituitary center. PATIENTS: Adults with acromegaly regardless of response to prior SRL and biochemical control status at baseline, stratified by an SRL dose required for insulin-like growth factor (IGF)-I normalization during any 3-month period within 12 months preceding enrollment. INTERVENTION: Combination therapy for 24 to 32 weeks on arm A, high-dose SRL (lanreotide 120 mg/octreotide long-acting release [LAR] 30 mg) plus weekly pegvisomant (40-160 mg/week); arm B, low-dose SRL (lanreotide 60 mg/octreotide LAR 10 mg) plus weekly pegvisomant; or arm C, low-dose SRL plus daily pegvisomant (15-60 mg/day). MAIN OUTCOME MEASURE: Monthly treatment cost in each arm in participants completing ≥ 24 weeks of therapy. RESULTS: Sixty patients were enrolled and 52 were evaluable. Fifty of 52 (96%) demonstrated IGF-I control regardless of prior SRL responsiveness (arm A, 14/15 [93.3%]; arm B, 22/23 [95.7%]; arm C, 14/14 [100%]). Arm B was least costly (mean, $9837 ±â€…1375 per month), arm C was most expensive (mean, $22543 ±â€…11158 per month), and arm A had an intermediate cost (mean, $14261 ±â€…1645 per month). Approximately 30% of patients required pegvisomant dose uptitration. Rates of adverse events were all < 10%. CONCLUSIONS: Low-dose SRL plus weekly pegvisomant represents a novel dosing option for achieving cost-effective, optimal biochemical control in patients with uncontrolled acromegaly requiring combination therapy.


Subject(s)
Acromegaly/drug therapy , Acromegaly/economics , Human Growth Hormone/analogs & derivatives , Octreotide/administration & dosage , Peptides, Cyclic/administration & dosage , Somatostatin/analogs & derivatives , Adult , Cost-Benefit Analysis , Delayed-Action Preparations , Dosage Forms , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Costs , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/economics , Female , Human Growth Hormone/administration & dosage , Human Growth Hormone/adverse effects , Human Growth Hormone/economics , Humans , Male , Middle Aged , Octreotide/adverse effects , Octreotide/economics , Peptides, Cyclic/adverse effects , Peptides, Cyclic/economics , Receptors, Somatostatin/agonists , Somatostatin/administration & dosage , Somatostatin/adverse effects , Somatostatin/economics , Therapies, Investigational/adverse effects , Therapies, Investigational/economics , Therapies, Investigational/methods , Treatment Outcome
19.
BMC Endocr Disord ; 20(1): 117, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32736547

ABSTRACT

BACKGROUND: Acromegaly patients managed on Somatostatin receptor ligands (SRLs), the most common first-line pharmacotherapy for acromegaly, may still experience acromegaly symptoms such as headache, sweating, fatigue, soft tissue swelling, and joint pain, even those with normal IGF-1. Additionally, treatment with SRLs may cause injection site reactions and other side effects such as gastro-intestinal (GI) symptoms. This study utilized patient-reported outcome measures to examine the burden associated with acromegaly and its treatment for patients receiving a stable dose of long-acting SRLs in routine clinical practice. METHODS: US acromegaly patients on a stable dose of SRL seen by their treating healthcare provider in the past 12 months completed a one-time online survey including the Acro-TSQ, an acromegaly-specific tool for assessing symptom burden and treatment satisfaction and convenience. RESULTS: One hundred five patients were enrolled (mean age 49.9 years, 79.1% female). Patients experienced numerous symptoms, including > 80% who experienced joint pain, acro-fog, swelling of soft tissue, and fatigue/weakness. Many symptoms occurred constantly, while some occurred at the end of the injection cycle, even among those with IGF-1 < = 1.0 ULN. Injection site reactions were common. Patients were moderately satisfied with their current treatment; symptoms and side effects often affected daily activities. On average, patients reported > 3 acromegaly provider visits/year. CONCLUSIONS: Despite receiving a stable dose of SRL and regular visits with an acromegaly healthcare provider, US acromegaly patients in routine clinical practice, and even the subgroup with normal IGF-1, report significant burden of disease and treatment.


Subject(s)
Acromegaly/drug therapy , Octreotide/administration & dosage , Peptides, Cyclic/administration & dosage , Somatostatin/analogs & derivatives , Acromegaly/epidemiology , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Injections, Intramuscular , Injections, Subcutaneous , Ligands , Male , Middle Aged , Octreotide/adverse effects , Patient Reported Outcome Measures , Peptides, Cyclic/adverse effects , Practice Patterns, Physicians'/statistics & numerical data , Receptors, Somatostatin/agonists , Somatostatin/administration & dosage , Somatostatin/adverse effects , Treatment Outcome , United States/epidemiology , Young Adult
20.
Bioorg Med Chem Lett ; 30(17): 127391, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32738999

ABSTRACT

Nonpeptide sst2 agonists can provide a new treatment option for patients with acromegaly, carcinoid tumors, and neuroendocrine tumors. Our medicinal chemistry efforts have led to the discovery of novel 3,4-dihydroquinazoline-4-carboxamides as sst2 agonists. This class of molecules exhibits excellent human sst2 potency and selectivity against sst1, sst3, sst4 and sst5 receptors. Leading compound 3-(3-chloro-5-methylphenyl)-6-(3-fluoro-2-hydroxyphenyl)-N,7-dimethyl-N-{[(2S)-pyrrolidin-2-yl]methyl}-3,4-dihydroquinazoline-4-carboxamide (28) showed no inhibition of major CYP450 enzymes (2C9, 2C19, 2D6 and 3A4) and weak inhibition of the hERG channel.


Subject(s)
Amides/chemistry , Receptors, Somatostatin/agonists , Amides/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Humans , Protein Isoforms/agonists , Protein Isoforms/metabolism , Receptors, Somatostatin/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL