Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 142
1.
BMC Cancer ; 22(1): 133, 2022 Feb 02.
Article En | MEDLINE | ID: mdl-35109816

BACKGROUND: Gonadotropin-releasing hormone (GnRH) receptor, a rhodopsin-like G-protein coupled receptor (GPCR) family member involved in GnRH signaling, is reported to be expressed in several tumors including glioblastoma multiforme (GBM), one of the most malignant and aggressive forms of primary brain tumors. However, the molecular targets associated with GnRH receptor are not well studied in GBM or in other cancers. The present study aims at investigating the effect of GnRH agonist (Gosarelin acetate) on cell proliferation and associated signaling pathways in GBM cell line, LN229. METHODS: LN229 cells were treated with different concentrations of GnRH agonist (10-10 M to 10-5 M) and the effect on cell proliferation was analyzed by cell count method. Further, total protein was extracted from control and GnRH agonist treated cells (with maximum reduction in cell proliferation) followed by trypsin digestion, labeling with iTRAQ reagents and LC-MS/MS analysis to identify differentially expressed proteins. Bioinformatic analysis was performed for annotation of proteins for the associated molecular function, altered pathways and network analysis using STRING database. RESULTS: The treatment with different concentrations of GnRH agonist showed a reduction in cell proliferation with a maximum reduction of 48.2% observed at 10-6 M. Quantitative proteomic analysis after GnRH agonist treatment (10-6 M) led to the identification of a total of 29 differentially expressed proteins with 1.3-fold change (23 upregulated, such as, kininogen-1 (KNG1), alpha-2-HS-glycoprotein (AHSG), alpha-fetoprotein (AFP), and 6 downregulated, such as integrator complex subunit 11 (CPSF3L), protein FRG1 (FRG1). Some of them are known [KNG1, AHSG, AFP] while others such as inter-alpha-trypsin inhibitor heavy chain H2 (ITIH2), ITIH4, and LIM domain-containing protein 1 (LIMD1) are novel to GnRH signaling pathway. Protein-protein interaction analysis showed a direct interaction of KNG1, a hub molecule, with GnRH, GnRH receptor, EGFR and other interactors including ITIH2, ITIH4 and AHSG. Overexpression of KNG1 after GnRH agonist treatment was validated using Western blot analysis, while a significant inhibition of EGFR was observed after GnRH agonist treatment. CONCLUSIONS: The study suggests a possible link of GnRH signaling with EGFR signaling pathways likely via KNG1. KNG1 inhibitors may be investigated independently or in combination with GnRH agonist for therapeutic applications.


Brain Neoplasms/metabolism , Cell Proliferation/drug effects , Glioblastoma/metabolism , Gonadotropin-Releasing Hormone/biosynthesis , Receptors, LHRH/biosynthesis , Animals , Antineoplastic Agents, Hormonal/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Chromatography, Liquid , Computational Biology , Glioblastoma/genetics , Glioblastoma/pathology , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/genetics , Goserelin/pharmacology , Humans , Proteomics/methods , Receptors, LHRH/genetics , Signal Transduction/drug effects , Tandem Mass Spectrometry
2.
J Neurosci Res ; 98(7): 1283-1292, 2020 07.
Article En | MEDLINE | ID: mdl-32399989

Sex hormone transition may trigger severe depressive episodes in some women. In order to map mechanisms related to such phenomena we developed a pharmacological preclinical human model using sex hormone manipulation with gonadotropin releasing hormone agonist (GnRHa) in a placebo-controlled design. Here the findings from this model is synthesized and discussed in the context of related literature on hormonal contributions to reproductive mental health disorders. The GnRha model work points to an estradiol-dependent depressive response in healthy women undergoing short-term sex hormone manipulation with GnRHa, which is linked to serotonin transporter changes (a key regulator of synaptic serotonin), a disengagement of hippocampus, and overengagement of brain networks recruited when processing emotional salient information. Further, the GnRHa model suggest that key brain regions in the reward circuit are less engaged in positive stimuli when undergoing sex hormone manipulation, which may underlie anhedonia. Also, the work supports that enhanced sensitivity to estrogen signaling at the level of gene expression may drive increased risk for depressive symptoms when exposed to sex steroid hormone fluctuations. In conclusion, the GnRHa model work highlights the brain signatures of rapid and profound changes in sex steroid hormone milieu, which reflect plausible mechanisms by which risk for mood disorders works. This model points to the role of estrogen dynamics and sensitivity, and offers a rationale for personalized prevention in hormonal transition phases, for example pregnancy to postpartum transition, perimenopause, and hormone treatments, which now can move into clinical translation and ideally pave the way for protecting mental and cognitive health.


Depression/psychology , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Adult , Emotions/drug effects , Estradiol , Female , Humans , Menstrual Cycle/drug effects , Menstrual Cycle/psychology , Models, Theoretical
3.
J Pharm Biomed Anal ; 180: 113044, 2020 Feb 20.
Article En | MEDLINE | ID: mdl-31865209

The purpose of this reported study was to develop and validate an LC-MS/MS method for the quantification of goserelin in a Pheroid® formulation simulated intestinal fluid. Biopharmaceuticals are formulated in drug delivery systems to improve their gastrointestinal stability. Goserelin, a peptide drug was formulated in Pheroid® delivery system and its gastrointestinal stability assessed using simulated intestinal fluid, which required an assay to determine the varying amounts of goserelin remaining after a specific time. Several extraction methods and solvents investigated to extract goserelin from complex matrix led to either poor recovery, peak shape or high background interference. A rapid gradient reversed-phase method coupled to tandem mass spectrometry detection was optimized for the separation and quantification of the extracted peptide. A simple, reproducible and good recovery extraction procedure for goserelin quantification was achieved through simultaneous acetonitrile protein precipitation and water-saturated n-butanol liquid-liquid extraction with water dilution. The method was found to be rapid, specific, precise and accurate, and successfully applied to determine goserelin remaining content in a simulated intestinal fluid, with potential use in other lipid-based formulation evaluated in simulated intestinal fluids.


Biomimetic Materials/metabolism , Drug Carriers/chemistry , Extracellular Fluid/metabolism , Goserelin/chemistry , Goserelin/pharmacology , Tandem Mass Spectrometry/methods , Biosensing Techniques , Chromatography, High Pressure Liquid , Drug Compounding , Drug Liberation , Limit of Detection , Linear Models , Liquid-Liquid Extraction , Reproducibility of Results , Sensitivity and Specificity , Solvents/chemistry
4.
Psychoneuroendocrinology ; 108: 70-77, 2019 10.
Article En | MEDLINE | ID: mdl-31229635

Chronic gonadotropin-releasing hormone agonist (GnRHa) treatment is effective for the medical suppression of the hypothalamic-pituitary-gonadal axis in situations like central precocious puberty and gender dysphoria. However, its administration during the peripubertal period could influence normal brain development and function because GnRH receptors are expressed in brain regions that regulate emotions, cognition, motivation and memory. This study used an ovine model to determine whether chronic peripubertal GnRHa-treatment affected the developmental shift from preference of familiarity to novelty. Experimental groups included Controls and GnRHa-treated rams. To differentiate between effects of altered GnRH signaling and those associated with the loss of sex steroids, a group was also included that received testosterone replacement as well as GnRHa (GnRHa + T). Preference for a novel versus familiar object was assessed during 5-min social isolation at 8, 28 and 46 weeks of age. Approach behavior was measured as interactions with and time spent near the objects, whereas avoidance behavior was measured by time spent in the entrance zone and attempts to escape the arena via the entry point. Emotional reactivity was measured by the number of vocalizations, escape attempts and urinations. As Control and GnRHa-treated rams aged, their approach behaviors showed a shift from preference for familiarity (8 weeks) to novelty (46 weeks). In contrast, relative to the Controls the GnRHa + T rams exhibited more approach behaviors towards both objects, at 28 and 46 weeks of age and preferred familiarity at 46 weeks of age. Vocalisation rate was increased in GnRHa treated rams in late puberty (28 weeks) compared to both Control and GnRHa + T rams but this effect was not seen in young adulthood (46 weeks). These results suggest that the specific suppression of testosterone during a developmental window in late puberty may reduce emotional reactivity and hamper learning a flexible adjustment to environmental change. The results also suggest that disruption of either endogenous testosterone signalling or a synergistic action between GnRH and testosterone signalling, may delay maturation of cognitive processes (e.g. information processing) that affects the motivation of rams to approach and avoid objects.


Exploratory Behavior/drug effects , Goserelin/pharmacology , Sexual Maturation/physiology , Animals , Gonadal Steroid Hormones/pharmacology , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/physiology , Male , Recognition, Psychology/drug effects , Sex Characteristics , Sheep, Domestic/physiology , Testis/drug effects , Testosterone/pharmacology
5.
Acta Psychiatr Scand ; 140(1): 77-84, 2019 07.
Article En | MEDLINE | ID: mdl-31099405

OBJECTIVES: Sensitivity to sex-steroid hormone fluctuations may increase risk for perinatal depression. We aimed to identify genome-wide biological profiles in women demonstrating sensitivity to pharmacological sex-hormone manipulation with gonadotrophin-releasing hormone agonist (GnRHa). METHODS: Longitudinal gene expression (Illumina Human HT12.v4) and DNA methylation data (Infinium HumanMethylation450K BeadChip) from 60 women (30 GnRHa, 30 placebo) were generated (Trial ID: NCT02661789). Differences between baseline and two follow-up points (initial stimulation- and subsequent early suppression phase) in the biphasic ovarian hormone response to GnRHa were assessed using linear mixed effects models. RESULTS: Genome-wide analysis revealed 588 probes differentially expressed from GnRHa intervention to first stimulatory phase follow-up (intervention group × time) after 10% fdr multiple testing correction. Of these, 54% genes were also significantly associated with estradiol changes over time (proxy for GnRHa response magnitude), 9.5% were associated with changes in depressive symptoms, and 38% were associated with changes in neocortical serotonin transporter binding. The genes were implicated in TGF beta signaling, adipogenesis, regulation of actin cytoskeleton, and focal adhesion pathways and enriched for DNA methylation changes (P = 0.006). CONCLUSIONS: These findings point toward an altered peripheral blood transcriptomic landscape in a pharmacological model of sex-hormone-induced depressive symptoms.


DNA Methylation , Depression/metabolism , Estradiol/metabolism , Gene Expression , Genome, Human , Gonadotropin-Releasing Hormone/pharmacology , Adult , Biomarkers/metabolism , Double-Blind Method , Female , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Humans , Longitudinal Studies , Models, Biological
6.
J Am Coll Cardiol ; 73(11): 1288-1296, 2019 03 26.
Article En | MEDLINE | ID: mdl-30898204

BACKGROUND: Circulating natriuretic peptide (NP) levels are markedly lower in healthy men than women. A relative NP deficiency in men could contribute to their higher risk of hypertension and cardiovascular disease. Epidemiological studies suggest testosterone may contribute to sex-specific NP differences. OBJECTIVES: This study aimed to determine the effect of testosterone administration on NP levels using a randomized, placebo-controlled design. METHODS: One hundred and fifty-one healthy men (20 to 50 years of age) received goserelin acetate to suppress endogenous production of gonadal steroids, and anastrazole to suppress conversion of testosterone to estradiol. Subjects were randomized to placebo gel or 4 different doses of testosterone (1%) gel for 12 weeks. Serum N-terminal-pro-B-type natriuretic peptide (NT-proBNP) and total testosterone levels were measured at baseline and follow-up. RESULTS: Men who did not receive testosterone replacement (placebo gel group) after suppression of endogenous gonadal steroid production experienced a profound decrease in serum testosterone (median 540 to 36 ng/dl; p < 0.0001). This was accompanied by an increase in median NT-proBNP (+8 pg/ml; p = 0.02). Each 1-g increase in testosterone dose was associated with a 4.3% lower NT-proBNP at follow-up (95% confidence interval: -7.9% to -0.45%; p = 0.029). An individual whose serum testosterone decreased by 500 ng/dl had a 26% higher predicted follow-up NT-proBNP than someone whose serum testosterone remained constant. CONCLUSIONS: Suppression of testosterone production in men led to increases in circulating NT-proBNP, which were attenuated by testosterone replacement. Inhibition of NP production by testosterone may partly explain the lower NP levels in men. (Dose-Response of Gonadal Steroids and Bone Turnover in Men; NCT00114114).


Anastrozole/pharmacology , Goserelin/pharmacology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Testosterone , Administration, Topical , Adult , Androgens/administration & dosage , Antineoplastic Agents, Hormonal/pharmacology , Aromatase Inhibitors/pharmacology , Correlation of Data , Drug Monitoring/methods , Estradiol/metabolism , Healthy Volunteers , Humans , Male , Middle Aged , Natriuretic Peptide, Brain/metabolism , Peptide Fragments/metabolism , Testosterone/administration & dosage , Testosterone/blood , Testosterone/metabolism
7.
Urol J ; 16(2): 157-161, 2019 05 05.
Article En | MEDLINE | ID: mdl-30345497

AIM: The complaints of lower urinary tract symptoms (LUTS) in cases with Prostate carcinoma (Pca) depend on coexisting benign prostate hyperplasia (BPH) or aging bladder. We aimed to investigate and compare the effect of goserelin acetate with leuprolide acetate on total prostate volume (TPV), post voiding residue (PVR), International Prostate Symptom Score (IPSS) and maximum flow rate (Qmax) reduction on cases of advanced Pca. METHODS: Patients with advanced Pca were treated with goserelin acetate (10.8 mg/3 months) or leuprolide acetate (22.5 mg/3 months) for 6 months. Changes in Prostate specific antigen (PSA), testesterone level, TPV, IPSS, PVR, and Qmax were assessed every 3 months. RESULTS: Fifty-one patients analyzed in this study. Mean percent decrease in PSA and testesterone from baseline to 6th month was not significantly difference between two groups (respectively; p = 0.9, p = 0.15) but TPV was reduced by -20.2 % ± 4.8 and -15.6 % ± 1.04,  the median total IPSS score was decreased by -34.77 % ± 8.8 and -19.77 % ± 6.1, median Qmax increased by 45.34 % ± 10.16 and 23.21 % ± 6.93, median PVR decreased by -31.54 % ± 8.4 and -19.23 % ± 5.5, respectively for two groups (all parameters (p < 0.05))Conclusion. In this study, we observed that the improvement of voiding parameters goserelin acetate was beter than leuprolide acetate. Especially it was detected the superiority of goserelin acetate group on the reduction of TPV, PVR and IPSS. Oncological outcomes were not different in both groups.


Antineoplastic Agents, Hormonal/therapeutic use , Goserelin/therapeutic use , Leuprolide/therapeutic use , Lower Urinary Tract Symptoms/drug therapy , Prostatic Neoplasms/drug therapy , Urination Disorders/drug therapy , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/pharmacology , Goserelin/pharmacology , Humans , Leuprolide/pharmacology , Lower Urinary Tract Symptoms/etiology , Male , Middle Aged , Prostatic Neoplasms/complications , Retrospective Studies , Tumor Burden/drug effects , Urination Disorders/etiology
8.
Breast Cancer ; 26(3): 339-348, 2019 May.
Article En | MEDLINE | ID: mdl-30367358

BACKGROUND: Ovarian function suppression (OFS) plus other endocrine treatment was recommended to hormone receptor (HR)-positive breast cancer by some guidelines recently. We performed this study to validate the survival benefits of OFS plus aromatase inhibitors (AI) or selective estrogen receptor modulators (SERM) in the real world. METHODS: All premenopausal, HR-positive breast cancer patients diagnosed between 1996 and 2017 were identified. Eligible patients were classified into three groups according to their adjuvant endocrine treatment, including OFS plus AI, OFS plus SERM and SERM alone. The primary outcome is invasive disease-free survival (iDFS), whereas the secondary outcome is overall survival (OS). Cox proportional hazards models and propensity score adjusted models were used to compare the survival benefits in three groups. RESULTS: We included 2838 patients, of which 246 received OFS plus AI, 175 received OFS plus SERM, and 2417 received SERM alone. Compared with SERM alone, OFS plus AI was associated with an improved iDFS (HR 0.59, 95% CI 0.36-0.96) and OS (HR 0.26, 95% CI 0.08-0.85). OFS plus SERM, however, was not significantly associated with extended iDFS or OS. Among patients older than 40 years old, OFS plus AI was more effective than OFS plus SERM (HR 0.38, 95% CI 0.17-0.88) or SERM alone (HR 0.44, 95% CI 0.23-0.84) in terms of iDFS. CONCLUSIONS: Our findings suggest that OFS plus AI treatment may extend both iDFS and OS among premenopausal patients with hormone receptor-positive breast cancer in the real world.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Ovary/physiopathology , Adult , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Combined Modality Therapy , Disease-Free Survival , Female , Goserelin/pharmacology , Goserelin/therapeutic use , Humans , Middle Aged , Ovariectomy , Ovary/drug effects , Ovary/surgery , Premenopause , Prospective Studies , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Survival Rate
9.
Eur J Cancer ; 103: 127-136, 2018 11.
Article En | MEDLINE | ID: mdl-30223226

BACKGROUND: We investigated the efficacy and safety of fulvestrant plus goserelin (F + G) versus anastrozole plus goserelin (A + G) in comparison with goserelin (G) alone in premenopausal women with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-), tamoxifen-pretreated metastatic breast cancer (MBC). PATIENTS AND METHODS: In this multicentre, open-label, randomised phase II study, premenopausal women aged ≥18 years with HR+, HER2-, tamoxifen-pretreated MBC were randomly assigned (1:1:1) to F + G, A + G or G alone. The primary end-point was time to progression (TTP). Secondary end-points included overall survival, overall response rate, clinical benefit rate and toxicity. RESULTS: Of 138 eligible patients, 44 were randomly assigned to receive F + G, 47 to A + G and 47 to G alone. The median follow-up duration was 32.2 months (interquartile range: 23.69-40.86) and the median age was 43.0 years (range 23.0-55.0). The median TTP was 16.3 months (95% confidence interval [CI] 7.5-25.1) for F + G, 14.5 months (95% CI 11.0-18.0) for A + G and 13.5 months (95% CI 10.3-16.8) for G alone. Compared with G alone, the hazard ratios were 0.608 for F + G (95% CI, 0.370-0.998; p = 0.049) and 0.982 for A + G (95% CI, 0.624-1.546; p = 0.937). In terms of visceral metastasis, a stratification factor, there were no TTP differences according to treatment arm. Grade III or IV toxicities were rarely observed. Of the common adverse events, grade I arthralgia and joint stiffness were more frequently observed in the F + G than in the A + G or G-alone groups (p < 0.05, respectively). CONCLUSIONS: F + G provides a promising new option for the treatment of premenopausal women with HR+, HER2-, tamoxifen-pretreated MBC. TRIAL REGISTRATION: ClinicalTrials.gov number NCT01266213 and Korean Cancer Study Group (KCSG) Breast cancer protocol number BR10-04.


Anastrozole/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Fulvestrant/therapeutic use , Goserelin/therapeutic use , Tamoxifen/therapeutic use , Adult , Anastrozole/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Female , Fulvestrant/pharmacology , Goserelin/pharmacology , Humans , Neoplasm Metastasis , Premenopause , Tamoxifen/pharmacology
10.
Oncol Rep ; 39(3): 1034-1042, 2018 Mar.
Article En | MEDLINE | ID: mdl-29286125

Gonadotropins, including luteinizing hormone (LH) and follicle stimulating hormone (FSH), are conducive to the growth of ovarian cancer based on the 'gonadotropin theory' and are regulated by gonadotropin-releasing hormone (GnRH). The present study was carried out to investigate the effect of goserelin, a GnRH agonist, on the apoptosis of epithelial ovarian cancer (EOC) cells and the underlying in vitro and in vivo mechanisms. Through flow cytometry, Hoechst staining and TUNEL staining, we demonstrated that goserelin promoted the apoptosis of EOC cells both in vitro and in vivo. Through human apoptosis gene PCR array, we verified that the promotion of EOC cell apoptosis by goserelin was linked to the upregulation of members of the tumor necrosis factor (TNF) and TNF receptor superfamilies, which have been identified as downstream targets of forkhead box O1 (FOXO1). Goserelin enhanced FOXO1 expression, and siRNA-mediated knockdown of FOXO1 abrogated the induction of apoptosis by goserelin. Moreover, goserelin decreased AKT activity, and FOXO1 upregulation by goserelin was dependent on the phosphatidylinositol 3-kinase (PI3K)/AKT pathway. In vivo, the expression of key factors in the PI3K/AKT/FOXO1 pathway was consistent with that observed in vitro. In conclusion, our data suggested that goserelin may promote EOC cell apoptosis by upregulating FOXO1 through the PI3K/AKT signaling pathway. We believe that GnRH agonists may be potential antitumor agents, and key factors in the PI3K/AKT-FOXO1 pathway may also be novel therapeutic targets for the treatment of EOC.


Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Forkhead Box Protein O1/metabolism , Goserelin/pharmacology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial , Cell Proliferation/drug effects , Female , Forkhead Box Protein O1/genetics , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
11.
J Inorg Biochem ; 175: 167-178, 2017 10.
Article En | MEDLINE | ID: mdl-28755574

Goserelin acetate (Gos) as a synthetic analog of the mammalian gonadotropin-releasing hormone (GnRH) is widely used in the treatment of sex hormone-related conditions. In this paper we present the chemical and biological aspects of its interaction with Cu(II) ions. The mode of Cu(II) binding and the thermodynamic stability of the obtained complexes were characterized by potentiometry, UV-Vis and CD spectroscopic methods. The DFT calculations were applied in order to investigate and confirm the molecular structure of the studied systems. The experimental and theoretical results clearly indicated the involvement of three nitrogens from the peptide and two oxygens from the acetate moieties in the Cu(II) coordination under physiological conditions. The investigated metallopeptide caused single- and/or double cleavage of the sugar-phosphate backbone of the plasmid DNA in the reaction accompanied by endogenous substances such as hydrogen peroxide or ascorbic acid. The degradation of the DNA molecule occurred via the free radical mechanism. Calculations based on measured spectra allowed determining the kinetic parameters of OH formation. The cytotoxic effects of Gos and its metallo-derivative were tested in vitro towards two cancer cell lines (A549 - human lung adenocarcinoma, CT26 - mouse colon carcinoma).


Coordination Complexes , Cytotoxins , DNA, Neoplasm/chemistry , Goserelin , A549 Cells , Animals , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Copper , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , DNA, Neoplasm/metabolism , Goserelin/chemistry , Goserelin/pharmacology , Humans , Mice , Oxidation-Reduction
12.
Oncologist ; 22(9): 1028-1038, 2017 09.
Article En | MEDLINE | ID: mdl-28652278

BACKGROUND: The efficacy and safety of palbociclib, a cyclin-dependent kinase 4/6 inhibitor, combined with fulvestrant and goserelin was assessed in premenopausal women with advanced breast cancer (ABC) who had progressed on prior endocrine therapy (ET). PATIENTS AND METHODS: One hundred eight premenopausal endocrine-refractory women ≥18 years with hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) ABC were among 521 women randomized 2:1 (347:174) to fulvestrant (500 mg) ± goserelin with either palbociclib (125 mg/day orally, 3 weeks on, 1 week off) or placebo. This analysis assessed whether the overall tolerable safety profile and significant progression-free survival (PFS) improvement extended to premenopausal women. Potential drug-drug interactions (DDIs) and ovarian suppression with goserelin were assessed via plasma pharmacokinetics and biochemical analyses, respectively. (ClinicalTrials.gov identifier: NCT01942135) RESULTS: Median PFS for premenopausal women in the palbociclib (n = 72) versus placebo arm (n = 36) was 9.5 versus 5.6 months, respectively (hazard ratio, 0.50, 95% confidence interval: 0.29-0.87), and consistent with the significant PFS improvement in the same arms for postmenopausal women. Any-grade and grade ≤3 neutropenia, leukopenia, and infections were among the most frequent adverse events reported in the palbociclib arm with concurrent goserelin administration. Hormone concentrations were similar between treatment arms and confirmed sustained ovarian suppression. Clinically relevant DDIs were not observed. CONCLUSION: Palbociclib combined with fulvestrant and goserelin was an effective and well-tolerated treatment for premenopausal women with prior endocrine-resistant HR+/HER2- ABC. Inclusion of both premenopausal and postmenopausal women in pivotal combination ET trials facilitates access to novel drugs for young women and should be considered as a new standard for clinical trial design. IMPLICATIONS FOR PRACTICE: PALOMA-3, the first registrational study to include premenopausal women in a trial investigating a CDK4/6 inhibitor combined with endocrine therapy, has the largest premenopausal cohort reported in an endocrine-resistant setting. In pretreated premenopausal women with hormone receptor-positive advanced breast cancer, palbociclib plus fulvestrant and goserelin (luteinizing hormone-releasing hormone [LHRH] agonist) treatment almost doubled median progression-free survival (PFS) and significantly increased the objective response rate versus endocrine monotherapy, achieving results comparable to those reported for chemotherapy without apparently interfering with LHRH agonist-induced ovarian suppression. The significant PFS gain and tolerable safety profile strongly support use of this regimen in premenopausal women with endocrine-resistant disease who could possibly delay chemotherapy.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Estradiol/analogs & derivatives , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Disease-Free Survival , Drug Interactions , Drug Resistance, Neoplasm , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Fulvestrant , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Goserelin/therapeutic use , Humans , Middle Aged , Piperazines/pharmacology , Premenopause , Pyridines/pharmacology , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Treatment Outcome
13.
JCI Insight ; 2(4): e89703, 2017 02 23.
Article En | MEDLINE | ID: mdl-28239652

The immunologic potency of IgG is modulated by glycosylation, but mechanisms regulating this process are undefined. A role for sex hormones is suggested by differences in IgG glycans between women and men, most prominently with respect to galactose. We therefore assessed IgG galactosylation in 713 healthy adults from 2 cohorts as well as in 159 subjects from 4 randomized controlled studies of endocrine manipulation: postmenopausal women receiving conjugated estrogens, raloxifene, or placebo; premenopausal women deprived of gonadal hormones with leuprolide and treated with estradiol or placebo; men deprived of gonadal hormones with goserelin and given testosterone or placebo; and men deprived of gonadal hormones with goserelin and given testosterone or placebo together with anastrozole to block conversion of testosterone to estradiol. Menopause was associated with an increase in agalactosylated IgG glycans, particularly in the most abundant fucosylated nonbisected (G0F) glycoform. Conjugated estrogens and raloxifene reduced G0F glycans in postmenopausal women, while in premenopausal women leuprolide increased G0F glycans in a manner reversed by estradiol. Among men, goserelin increased G0F glycans, an effect blocked by testosterone through conversion to estradiol. These results establish estrogens as an in vivo modulator of IgG galactosylation in both women and men, defining a pathway by which sex modulates immunity.


Estrogens/metabolism , Galactose/metabolism , Glycosylation , Immunoglobulin G/metabolism , Polysaccharides/metabolism , Adult , Anastrozole/pharmacology , Androgens/pharmacology , Aromatase Inhibitors/pharmacology , Estradiol/pharmacology , Estrogens/pharmacology , Estrogens, Conjugated (USP)/pharmacology , Female , Fertility Agents, Female/pharmacology , Glycosylation/drug effects , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Healthy Volunteers , Humans , Immunoglobulin G/drug effects , Leuprolide/pharmacology , Male , Middle Aged , Postmenopause , Premenopause , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Sex Factors , Testosterone/pharmacology
14.
Psychoneuroendocrinology ; 77: 1-8, 2017 03.
Article En | MEDLINE | ID: mdl-27987429

Chronic gonadotropin-releasing hormone agonist (GnRHa) administration is used where suppression of hypothalamic-pituitary-gonadal axis activity is beneficial, such as steroid-dependent cancers, early onset gender dysphoria, central precocious puberty and as a reversible contraceptive in veterinary medicine. GnRH receptors, however, are expressed outside the reproductive axis, e.g. brain areas such as the hippocampus which is crucial for learning and memory processes. Previous work, using an ovine model, has demonstrated that long-term spatial memory is reduced in adult rams (45 weeks of age), following peripubertal blockade of GnRH signaling (GnRHa: goserelin acetate), and this was independent of the associated loss of gonadal steroid signaling. The current study investigated whether this effect is reversed after discontinuation of GnRHa-treatment. The results demonstrate that peripubertal GnRHa-treatment suppressed reproductive function in rams, which was restored after cessation of GnRHa-treatment at 44 weeks of age, as indicated by similar testes size (relative to body weight) in both GnRHa-Recovery and Control rams at 81 weeks of age. Rams in which GnRHa-treatment was discontinued (GnRHa-Recovery) had comparable spatial maze traverse times to Controls, during spatial orientation and learning assessments at 85 and 99 weeks of age. Former GnRHa-treatment altered how quickly the rams progressed beyond a specific point in the spatial maze at 83 and 99 weeks of age, and the direction of this effect depended on gonadal steroid exposure, i.e. GnRHa-Recovery rams progressed quicker during breeding season and slower during non-breeding season, compared to Controls. The long-term spatial memory performance of GnRHa-Recovery rams remained reduced (P<0.05, 1.5-fold slower) after discontinuation of GnRHa, compared to Controls. This result suggests that the time at which puberty normally occurs may represent a critical period of hippocampal plasticity. Perturbing normal hippocampal formation in this peripubertal period may also have long lasting effects on other brain areas and aspects of cognitive function.


Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Orientation, Spatial/drug effects , Spatial Learning/drug effects , Spatial Memory/drug effects , Animals , Body Weight/drug effects , Male , Organ Size/drug effects , Sexual Maturation/drug effects , Sexual Maturation/physiology , Sheep , Testis/drug effects , Testis/growth & development
15.
Acta Vet Hung ; 64(1): 116-9, 2016 03.
Article En | MEDLINE | ID: mdl-26919148

The aim of this study was to evaluate the ability of two synthetic GnRH analogues, goserelin and leuprolide, to induce ovulation in rabbit does using intravaginal administration. A total of 252 primiparous lactating does were randomly divided into five groups that, at the time of insemination, received the following treatments for ovulation induction: 1 µg of buserelin administered intramuscularly (control group), 5 µg of goserelin added to the semen dose (Group G5), 10 µg of goserelin added to the semen dose (Group G10), 5 µg of leuprolide added to the semen dose (Group L5), and 10 µg of leuprolide added to the semen dose (Group L10). The kindling rate was 80.5% in Group G10 and 75.0% in Group L10; these values are comparable to the kindling rate obtained in the control group (85.9%). The kindling rates in Groups G5 and L5 were significantly lower than in the control group (60.0%, 54.2% and 85.9%, respectively). The number of live-born rabbits was not significantly affected by the ovulation induction treatment. As regards the total number of rabbits born the only significant difference was observed between Groups G5 and L5. This study shows the possibility of inducing ovulation in rabbits by adding goserelin and leuprolide directly to the semen dose.


Goserelin/pharmacology , Leuprolide/pharmacology , Ovulation Induction/methods , Rabbits , Semen/drug effects , Animals , Female , Gonadotropin-Releasing Hormone , Insemination, Artificial , Lactation , Pregnancy , Pregnancy Rate , Semen/physiology
16.
Reprod Biol Endocrinol ; 13: 130, 2015 Dec 01.
Article En | MEDLINE | ID: mdl-26621118

BACKGROUND: XM17 is a recombinant human follicle-stimulating hormone (rhFSH) intended mainly for use in controlled ovarian hyperstimulation and the treatment of anovulation. The purpose of the current study was to establish bioequivalence, safety and tolerability of single 300-IU subcutaneous (sc) doses of XM17 to that of the reference follitropin alfa (Gonal-f(®)) in healthy young women. METHODS: This open-label, Phase I, single-dose, single-center, two-way crossover study was conducted from February to May 2009. Thirty-six women aged 18-39 years were included, with a study duration of ~27 days per participant. After endogenous FSH downregulation with goserelin (3.6 mg) on study Day 0, XM17 and Gonal-f(®) were administered on Days 11 and 19 in random sequence. Frequent serum samples were drawn for standard pharmacokinetics until 168 h postdosing. Laboratory values, adverse events (AEs) and local tolerability were assessed throughout the study period. Primary endpoints included Cmax and AUC0-t. Secondary endpoints included additional pharmacokinetic (PK) parameters, safety and tolerability. RESULTS: Ratios of XM17 to Gonal-f(®) for Cmax and AUC0-t equaled 1.017 (90 % confidence interval [CI]: 0.958, 1.080) and 1.028 (90 % CI: 0.931, 1.134), respectively, with the CIs contained within the predefined interval (0.8, 1.25). Ratios for AUC0-168h, AUC0-∞ and t1/2 were also ~1, and no difference in tmax was detected. Both XM17 and Gonal-f(®) were well tolerated, with no detectable anti-FSH antibodies, serious AEs or AEs leading to discontinuation or dose reduction. CONCLUSIONS: PK bioequivalence of single 300-IU sc doses of XM17 to the reference product Gonal-f® was statistically demonstrated. XM17 was well tolerated both systemically and locally. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02592031 ; date of registration: 28 October, 2015.


Follicle Stimulating Hormone, Human/pharmacokinetics , Adolescent , Adult , Cross-Over Studies , Female , Follicle Stimulating Hormone, Human/adverse effects , Follicle Stimulating Hormone, Human/pharmacology , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Humans , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Therapeutic Equivalency , Treatment Outcome , Young Adult
17.
Horm Metab Res ; 47(12): 925-31, 2015 Nov.
Article En | MEDLINE | ID: mdl-26197852

Gonadotropin-releasing hormone receptors (GnRHR) have been found in extrapituitary tissues, including the prostate, where they might exert a local effect on tissue growth. Degarelix is a GnRHR antagonist approved for use in patients with prostate cancer (PCa) who need androgen deprivation therapy. The slowing of prostate cell growth is a common goal shared by PCa and benign prostate hyperplasia (BPH) patients, and the effect of degarelix on BPH cells has not yet been investigated. We wanted to evaluate the direct effect of degarelix on human BPH primary cell growth. Gene expression studies performed with BPH (n=11), stage 0 (n=15), and PCa (n=65) human specimens demonstrated the presence of GNRHR1 and GNRHR2 and their respective endogenous peptide ligands. BPH-isolated epithelial and stromal cells were either cultured alone or co-cultured (1:4 or 4:1 ratio of epithelial to stromal cells) and subsequently treated with increasing concentrations of degarelix. Degarelix treatment induced a decrease in cell viability and cell proliferation rates, which occurred in parallel to an increase in apoptosis. Both epithelial and stromal BPH cells are sensitive to degarelix treatment and, interestingly, degarelix is also effective when the cells were growing in a co-culture microenvironment. In contrast to degarelix, the GnRHR agonists, leuprolide and goserelin, exerted no effect on the viability of BPH epithelial or stromal cells. In conclusion, (i) prostate tissues express GNRHR and are a potential target for degarelix; and (ii) degarelix directly inhibits BPH cell growth through a decrease in cell proliferation and an increase in apoptosis. Supporting information for this article is available online at http://www.thieme-connect.de/products.


Gonadotropin-Releasing Hormone/antagonists & inhibitors , Oligopeptides/pharmacology , Prostatic Hyperplasia/drug therapy , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Gonadotropin-Releasing Hormone/genetics , Goserelin/pharmacology , Humans , Leuprolide/pharmacology , Male , Oligopeptides/therapeutic use , Prostatic Hyperplasia/pathology
18.
PLoS One ; 10(3): e0120670, 2015.
Article En | MEDLINE | ID: mdl-25811175

Degarelix is a gonadrotropin-releasing hormone (GnRH) receptor (GnRHR) antagonist used in patients with prostate cancer who need androgen deprivation therapy. GnRHRs have been found in extra-pituitary tissues, including prostate, which may be affected by the GnRH and GnRH analogues used in therapy. The direct effect of degarelix on human prostate cell growth was evaluated. Normal prostate myofibroblast WPMY-1 and epithelial WPE1-NA22 cells, benign prostatic hyperplasia (BPH)-1 cells, androgen-independent PC-3 and androgen-dependent LNCaP prostate cancer cells, as well as VCaP cells derived from a patient with castration-resistant prostate cancer were used. Discriminatory protein and lipid fingerprints of normal, hyperplastic, and cancer cells were generated by matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS). The investigated cell lines express GNRHR1 and GNRHR2 and their endogenous ligands. Degarelix treatment reduced cell viability in all prostate cell lines tested, with the exception of the PC-3 cells; this can be attributed to increased apoptosis, as indicated by increased caspase 3/7, 8 and 9 levels. WPE1-NA22, BPH-1, LNCaP, and VCaP cell viability was not affected by treatment with the GnRH agonists leuprolide and goserelin. Using MALDI MS, we detected changes in m/z signals that were robust enough to create a complete discriminatory profile induced by degarelix. Transcriptomic analysis of BPH-1 cells provided a global map of genes affected by degarelix and indicated that the biological processes affected were related to cell growth, G-coupled receptors, the mitogen-activated protein kinase (MAPK) pathway, angiogenesis and cell adhesion. Taken together, these data demonstrate that (i) the GnRH antagonist degarelix exerts a direct effect on prostate cell growth through apoptosis; (ii) MALDI MS analysis provided a basis to fingerprint degarelix-treated prostate cells; and (iii) the clusters of genes affected by degarelix suggest that this compound, in addition to its known use in the treatment of prostate cancer, may be efficacious in BPH.


Gonadotropin-Releasing Hormone/metabolism , Oligopeptides/pharmacology , Prostate/drug effects , Prostate/metabolism , Apoptosis/drug effects , Caspases/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Ontology , Gonadotropin-Releasing Hormone/genetics , Goserelin/pharmacology , Humans , Leuprolide/pharmacology , Male , Prostate/cytology , Proteomics/methods , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/genetics , Receptors, LHRH/metabolism
19.
Psychoneuroendocrinology ; 40: 130-9, 2014 Feb.
Article En | MEDLINE | ID: mdl-24485485

This study investigated the actions of blocking the GnRH receptor using a specific agonist on the response of male and female sheep to a novel object placed in their pen. The study is part of a series performed on 46 same sex twin animals. One of the pair received a subcutaneous implant of the GnRH agonist Goserelin acetate every four weeks while the other remained untreated. Implantation began immediately prior to puberty; at 8 weeks in the males and 28 weeks in the females (as timing of puberty is sex specific). To determine the effects of agonist treatment on the reproductive axis blood samples were collected for measurement of testosterone in the males and progesterone in the females. In addition the volume of the scrotum was determined. The present study aimed to determine whether there are sexually differentiated behavioural responses to a novel object at different stages of brain development (8, 28 and 48 weeks of age) and whether these responses are altered by GnRHa treatment. Approach behaviour towards and interactions with the novel object were monitored as was the number of vocalisations per unit time during the test period. GnRHa treatment suppressed testosterone concentrations and testicular growth in the males and progesterone release in the females. Sheep vocalised significantly more prior to weaning (8 weeks of age) than post weaning (28 and 48 weeks of age) suggesting stress on separation from their dams. Our current study shows that males are more likely to leave their conspecifics to approach a novel object than females. As this behaviour was not altered by suppression of the reproductive axis we suggest that, although sex differences are more obviously expressed in the phenotype after puberty, these may be developed during adolescence but not primarily altered during puberty by sex hormones.


Behavior, Animal/physiology , Exploratory Behavior/physiology , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Sheep/psychology , Age Factors , Animals , Behavior, Animal/drug effects , Exploratory Behavior/drug effects , Female , Gonadotropin-Releasing Hormone/metabolism , Goserelin/pharmacology , Hydrocortisone/blood , Male , Progesterone/blood , Sex Characteristics , Sheep/blood , Sheep/growth & development , Stress, Psychological/blood
20.
Psychoneuroendocrinology ; 38(12): 3115-27, 2013 Dec.
Article En | MEDLINE | ID: mdl-24103890

The nature of hormonal involvement in pubertal brain development has attracted wide interest. Structural changes within the brain that occur during pubertal development appear mainly in regions closely linked with emotion, motivation and cognitive functions. Using a sheep model, we have previously shown that peri-pubertal pharmacological blockade of gonadotropin releasing hormone (GnRH) receptors, results in exaggerated sex-differences in cognitive executive function and emotional control, as well as sex and hemisphere specific patterns of expression of hippocampal genes associated with synaptic plasticity and endocrine signaling. In this study, we explored effects of this treatment regime on the gene expression profile of the ovine amygdala. The study was conducted with 30 same-sex twin lambs (14 female and 16 male), half of which were treated with the GnRH agonist (GnRHa) goserelin acetate every 4th week, beginning before puberty, until approximately 50 weeks of age. Gene expression profiles of the left and right amygdala were measured using 8×15 K Agilent ovine microarrays. Differential expression of selected genes was confirmed by qRT-PCR (Quantitative real time PCR). Networking analyses and Gene Ontology (GO) Term analyses were performed with Ingenuity Pathway Analysis (IPA), version 7.5 and DAVID (Database for Annotation, Visualization and integrated Discovery) version 6.7 software packages, respectively. GnRHa treatment was associated with significant sex- and hemisphere-specific differential patterns of gene expression. GnRHa treatment was associated with differential expression of 432 (|logFC|>0.3, adj. p value <0.05) and 46 (p value <0.0.5) genes in the left and right amygdala, respectively, of female animals, relative to the reference sample which consisted of all a pooled sample from control and treated animals of both sexes. No genes were found to be differentially expressed as a result of GnRHa treatment in the male animals. The results indicated that GnRH may, directly and/or indirectly, be involved in the regulation of sex- and hemisphere-specific differential expression of genes in the amygdala. This finding should be considered when long-term peri-pubertal GnRHa treatment is used in children.


Amygdala/metabolism , Gene Expression/drug effects , Gonadotropin-Releasing Hormone/agonists , Goserelin/pharmacology , Amygdala/drug effects , Animals , Female , Functional Laterality/physiology , Gene Expression Profiling , Male , Microarray Analysis , RNA/biosynthesis , RNA/genetics , Real-Time Polymerase Chain Reaction , Sex Characteristics , Sheep
...