Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 22(1): 133, 2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35109816

RESUMO

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.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células/efeitos dos fármacos , Glioblastoma/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Receptores LHRH/biossíntese , Animais , Antineoplásicos Hormonais/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cromatografia Líquida , Biologia Computacional , Glioblastoma/genética , Glioblastoma/patologia , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/genética , Gosserrelina/farmacologia , Humanos , Proteômica/métodos , Receptores LHRH/genética , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Massas em Tandem
2.
J Neurosci Res ; 98(7): 1283-1292, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32399989

RESUMO

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.


Assuntos
Depressão/psicologia , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Adulto , Emoções/efeitos dos fármacos , Estradiol , Feminino , Humanos , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/psicologia , Modelos Teóricos
3.
Acta Psychiatr Scand ; 140(1): 77-84, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31099405

RESUMO

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.


Assuntos
Metilação de DNA , Depressão/metabolismo , Estradiol/metabolismo , Expressão Gênica , Genoma Humano , Hormônio Liberador de Gonadotropina/farmacologia , Adulto , Biomarcadores/metabolismo , Método Duplo-Cego , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Humanos , Estudos Longitudinais , Modelos Biológicos
4.
Oncologist ; 22(9): 1028-1038, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28652278

RESUMO

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.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Estradiol/análogos & derivados , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Intervalo Livre de Doença , Interações Medicamentosas , Resistencia a Medicamentos Antineoplásicos , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Fulvestranto , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Gosserrelina/uso terapêutico , Humanos , Pessoa de Meia-Idade , Piperazinas/farmacologia , Pré-Menopausa , Piridinas/farmacologia , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Resultado do Tratamento
5.
Acta Vet Hung ; 64(1): 116-9, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26919148

RESUMO

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.


Assuntos
Gosserrelina/farmacologia , Leuprolida/farmacologia , Indução da Ovulação/métodos , Coelhos , Sêmen/efeitos dos fármacos , Animais , Feminino , Hormônio Liberador de Gonadotropina , Inseminação Artificial , Lactação , Gravidez , Taxa de Gravidez , Sêmen/fisiologia
6.
Reprod Biol Endocrinol ; 13: 130, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26621118

RESUMO

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.


Assuntos
Hormônio Foliculoestimulante Humano/farmacocinética , Adolescente , Adulto , Estudos Cross-Over , Feminino , Hormônio Foliculoestimulante Humano/efeitos adversos , Hormônio Foliculoestimulante Humano/farmacologia , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Humanos , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Equivalência Terapêutica , Resultado do Tratamento , Adulto Jovem
7.
Horm Metab Res ; 47(12): 925-31, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26197852

RESUMO

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.


Assuntos
Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Hiperplasia Prostática/tratamento farmacológico , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Hormônio Liberador de Gonadotropina/genética , Gosserrelina/farmacologia , Humanos , Leuprolida/farmacologia , Masculino , Oligopeptídeos/uso terapêutico , Hiperplasia Prostática/patologia
8.
Gynecol Endocrinol ; 29(6): 608-10, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23656392

RESUMO

The effect of long-acting GnRHa, in the luteal phase, during ART cycles varies from one patient to another. The aim of this study was to evaluate whether the effect of long-acting GnRHa in the luteal phase, in ART cycles, affects pregnancy rates according to the duration of its action in such phase. This is a retrospective study of 367 patients submitted to ovulation induction for in vitro fertilization/intracytoplasmic sperm injection procedures that used long-acting depot GnRHa for pituitary suppression. Patients were stratified according to the period of action of the agonist in the luteal phase: group 1, ≤ 6 days; group 2, 7 to 12 days; and group 3, >12 days. The following variables were analyzed: ovarian response, age, infertility causes and pregnancy rates. Group 1 (n = 53) had a mean age of 33.8 ± 4.55 years (23-44 years) and a pregnancy rate of 45.2%. In group 2 (n = 118), mean age was 33.7 ± 4.5 years (24-44 years) and the pregnancy rate was 38.9%. In group 3 (n = 196), mean age was 33.7 ± 4.4 years (23-43 years) and the pregnancy rate was 47.4%. Regardless of the duration of depot GnRHa action in the luteal phase, no significant association with pregnancy rates was found.


Assuntos
Fármacos para a Fertilidade Feminina/administração & dosagem , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/administração & dosagem , Fase Luteal/efeitos dos fármacos , Técnicas de Reprodução Assistida , Adulto , Preparações de Ação Retardada , Esquema de Medicação , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Gosserrelina/farmacologia , Humanos , Indução da Ovulação/métodos , Gravidez , Taxa de Gravidez , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
9.
Arch Gynecol Obstet ; 287(2): 369-73, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22987256

RESUMO

PURPOSE: The aim of this study was to evaluate the use of Doppler velocimetry of the uterine arteries and its association to endometrial thickness as a method to confirm pituitary suppression after administration of gonadotropin-releasing hormone analogues in assisted reproduction treatment cycles. METHODS: A total of 70 patients using gonadotropin-releasing hormone analogues for pituitary suppression for in vitro fertilization treatment were studied. To confirm down-regulation, serum estradiol levels and endometrial thickness were evaluated 10 days after gonadotropin-releasing hormone analogues administration. When estradiol was <30 pg/ml and endometrial thickness was <3 mm, pituitary suppression was confirmed. Doppler velocimetric measurements were performed at the same day to study the pulsatility index of the uterine arteries, until pituitary suppression was confirmed. RESULTS: All 70 patients had normal ovarian morphology. For the patients who had estradiol levels ≤30 pg/ml, the mean pulsatility index of the uterine arteries was 2.95 ± 0.79 and for those who had levels >30 pg/ml the mean PI was 2.22 ± 0.8 (p = 0.005). For the patients who had endometrial thickness ≤5 mm the mean PI was 2.86 ± 0.82 and for those with endometrial thickness >5 mm the mean PI was 2.17 ± 0.79 (p = 0.004). Using a cut-off point of 2.51 for the pulsatility index, to compare to estradiol levels, we observed a sensitivity of 72.7 % and specificity of 71 %. The combination of Doppler velocimetric and endometrial thickness showed a sensitivity of 94 % and specificity of 82.3 %. CONCLUSIONS: Doppler velocimetric analysis of the uterine arteries can be an important tool in the diagnosis of the down-regulation after the use of gonadotropin-releasing hormone analogues and might help simplify assisted reproduction programmes.


Assuntos
Monitoramento de Medicamentos/métodos , Endométrio/efeitos dos fármacos , Fármacos para a Fertilidade Feminina/farmacologia , Fertilização in vitro/métodos , Gosserrelina/farmacologia , Fluxometria por Laser-Doppler , Artéria Uterina/diagnóstico por imagem , Adulto , Biomarcadores/sangue , Regulação para Baixo , Esquema de Medicação , Endométrio/diagnóstico por imagem , Endométrio/fisiologia , Estradiol/sangue , Feminino , Fármacos para a Fertilidade Feminina/administração & dosagem , Gosserrelina/administração & dosagem , Humanos , Injeções Subcutâneas , Pessoa de Meia-Idade , Indução da Ovulação/métodos , Estudos Prospectivos , Fluxo Pulsátil/efeitos dos fármacos , Sensibilidade e Especificidade , Ultrassonografia , Artéria Uterina/efeitos dos fármacos , Artéria Uterina/fisiologia
10.
Horm Behav ; 59(1): 22-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20934426

RESUMO

In mammals, sex specialization is reflected by differences in brain anatomy and function. Measurable differences are documented in reproductive behavior, cognition, and emotion. We hypothesized that gonadotropin-releasing hormone (GnRH) plays a crucial role in controlling the extent of the brain's sex specificity and that changes in GnRH action during critical periods of brain development, such as puberty, will result in altered sex-specific behavioral and physiological patterns. We blocked puberty in half of the 48 same-sex Scottish mule Texel cross sheep twins with GnRH analog (GnRHa) goserelin acetate every 3 weeks, beginning just before puberty. To determine the effects of GnRHa treatment on sex-specific behavior and emotion regulation in different social contexts, we employed the food acquisition task (FAT) and measurement of heart rate variability (HRV). ANOVA revealed significant sex and sex×treatment interaction effects, suggesting that treated males were more likely to leave their companions to acquire food than untreated, while the opposite effect was observed in females. Concordant results were seen in HRV; treated males displayed higher HRV than untreated, while the reverse pattern was found in females, as shown by significant sex and sex×treatment interaction effects. We conclude that long-term prepubertal GnRHa treatment significantly affected sex-specific brain development, which impacted emotion and behavior regulation in sheep. These results suggest that GnRH is a modulator of cognitive function in the developing brain and that the sexes are differentially affected by GnRH modulation.


Assuntos
Comportamento Animal/efeitos dos fármacos , Emoções/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Gosserrelina/farmacologia , Análise de Variância , Animais , Feminino , Frequência Cardíaca/efeitos dos fármacos , Masculino , Fatores Sexuais , Ovinos , Comportamento Social
11.
Clin Obstet Gynecol ; 54(1): 150-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21278514

RESUMO

The mortality rate due to breast cancer has declined over the preceding decades to a great extent, secondary to the development and use of effective adjuvant therapy. Tamoxifen remains the standard of care in premenopausal women, whereas aromatase inhibitors have become standard therapy after menopause for women with hormone-sensitive disease. Tumor gene profiling assays are being increasingly used to identify women with hormone-sensitive disease, who would benefit from adjuvant chemotherapy. For those women with hormone negative cancer, systemic chemotherapy provides substantial reduction in the risk of disease recurrence and death.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Tamoxifeno/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos Hormonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimioterapia Adjuvante , Ciclofosfamida/farmacologia , Ciclofosfamida/uso terapêutico , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Gosserrelina/farmacologia , Gosserrelina/uso terapêutico , Humanos , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Ovário/efeitos dos fármacos , Tamoxifeno/farmacologia , Taxoides/uso terapêutico , Trastuzumab
12.
Zhonghua Fu Chan Ke Za Zhi ; 46(12): 892-7, 2011 Dec.
Artigo em Zh | MEDLINE | ID: mdl-22333277

RESUMO

OBJECTIVE: To investigate the influence of gonadotropin-releasing hormone (GnRH) analogues on ovarian cancer and ovarian function in vivo. METHODS: ES-2 cells were cultured and xenotransplanted into 36 nude mice, which were divided into 6 groups: normal saline (NS) group: NS 0.1 ml/day subcutaneous injection, and then NS 0.2 ml/week peritoneal injection; cisplatin (DDP) group: NS 0.1 ml/day subcutaneous injection, and then DDP 5 mg/kg (diluted to 0.2 ml) per week peritoneal injection; goserelin group: 100 µg goserelin (diluted to 0.1 ml) per day subcutaneous injection, and then NS 0.2 ml/week peritoneal injection; goserelin + DDP group: 100 µg goserelin (diluted to 0.1 ml) per day subcutaneous injection, and DDP 5 mg/kg (diluted to 0.2 ml) per week peritoneal injection; cetrorelix group:100 µg cetrorelix (diluted to 0.1 ml) per day subcutaneous injection and NS 0.2 ml/week peritoneal injection; cetrorelix + DDP group: 100 µg cetrorelix (diluted to 0.1 ml) per day subcutaneous injection and DDP 5 mg/kg (diluted to 0.2 ml) per week peritoneal injection. All the peritoneal injection started from subcutaneous injection one week later. To compare the weight of nude mice, the volumes of transplanted tumors, the expression of Ki-67 antigen in transplanted tumors, the estrus, the ratio of atretic follicles, the ratio of primary and preantral follicles, the levels of serum anti-Mullerian hormone (AMH), follicle-stimulating hormone (FSH), estradio (E(2)) and progesterone (P) in each group. RESULTS: There were no significant difference in the weight of nude mice among 6 groups (P > 0.05), which on day 29 in NS group was (19.8 ± 2.2) g, DDP group (20.5 ± 1.4) g, gosereline group (19.6 ± 0.9) g, goserelin + DDP group (19.7 ± 1.6) g, cetrorelix group (20.7 ± 2.2) g, and cetrorelix + DDP group (19.0 ± 1.7) g. The tumor volumes of different groups on the 12(th) day: NS group (241 ± 179) mm(3), DDP group (78 ± 20) mm(3), gosereline group (78 ± 55) mm(3), goserelin + DDP group (64 ± 48) mm(3), cetrorelix group (78 ± 64) mm(3), or cetrorelix + DDP group (70 ± 19) mm(3), in which there were significant difference between NS group and the other groups (P < 0.05); and the same result was obtained on day 15, 19, 22, 26 and 29 (P < 0.05). The expression of Ki-67 in NS group was (33 ± 10)%, in which it was higher than those in DDP group 3.5%, goserelin group 8.8%, goserelin + DDP group 1.5%, cetrorelix group (23 ± 11)%, or cetrorelix + DDP group (8 ± 6)% (P < 0.05). The ratio of primary and preantral follicles in goserelin group was (71.5 ± 8.1)%, in goserelin + DDP group was (62.4 ± 4.1)%, in cetrorelix group was (71.2 ± 7.4)%, and in cetrorelix + DDP group was (63.8 ± 3.1)%, in which they were much higher than that in DDP group (47.0 ± 4.8)% (P < 0.05). The level of AMH in goserelin group was (98 ± 27) ng/ml, which was much higher than that in NS group (66.2 ± 17.4) ng/ml (P < 0.05), while there were no difference in the levels of FSH, E(2) or P among different groups (P > 0.05). CONCLUSION: GnRH analogues could inhibit the growth of transplanted tumors in nude mice, meanwhile increase the secretion of AMH, decrease the frequencies and prolong the lasting time of estrus, decrease the ratio of atretic follicles, raise the ratio of primary and preantral follicles, which may be protect the ovarian function of nude mice.


Assuntos
Antineoplásicos Hormonais/farmacologia , Antineoplásicos/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Folículo Ovariano/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Animais , Hormônio Antimülleriano , Antineoplásicos/efeitos adversos , Antineoplásicos Hormonais/efeitos adversos , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Gosserrelina/efeitos adversos , Gosserrelina/farmacologia , Antagonistas de Hormônios/administração & dosagem , Humanos , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Nus , Progesterona , Células Tumorais Cultivadas
13.
Stat Med ; 29(22): 2310-24, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20552577

RESUMO

Breast cancer is the leading cancer in women of reproductive age; more than a quarter of women diagnosed with breast cancer in the US are premenopausal. A common adjuvant treatment for this patient population is chemotherapy, which has been shown to cause premature menopause and infertility with serious consequences to quality of life. Luteinizing-hormone-releasing hormone (LHRH) agonists, which induce temporary ovarian function suppression (OFS), has been shown to be a useful alternative to chemotherapy in the adjuvant setting for estrogen-receptor-positive breast cancer patients. LHRH agonists have the potential to preserve fertility after treatment, thus, reducing the negative effects on a patient's reproductive health. However, little is known about the association between a patient's underlying degree of OFS and disease-free survival (DFS) after receiving LHRH agonists. Specifically, we are interested in whether patients with lower underlying degrees of OFS (i.e. higher estrogen production) after taking LHRH agonists are at a higher risk for late breast cancer events. In this paper, we propose a latent class joint model (LCJM) to analyze a data set from International Breast Cancer Study Group (IBCSG) Trial VIII to investigate the association between OFS and DFS. Analysis of this data set is challenging due to the fact that the main outcome of interest, OFS, is unobservable and the available surrogates for this latent variable involve masked event and cured proportions. We employ a likelihood approach and the EM algorithm to obtain parameter estimates and present results from the IBCSG data analysis.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Modelos Biológicos , Modelos Estatísticos , Adulto , Amenorreia/induzido quimicamente , Antineoplásicos Hormonais/uso terapêutico , Intervalo Livre de Doença , Feminino , Gosserrelina/uso terapêutico , Humanos , Pré-Menopausa , Ensaios Clínicos Controlados Aleatórios como Assunto
14.
Cell Biochem Funct ; 28(4): 293-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20517893

RESUMO

Kisspeptin is a recently discovered hypothalamic peptide which plays an important role in the central control of reproductive functions. We have investigated direct and indirect effects of kisspeptin on the liver oxidative stress in young male rats. Twenty-four rats were divided into four groups (n = 6/group). First group served as control and received saline. Kisspeptin-10 was administered to the animals in the second group (20 nmol/rat/day), for a period of 7 days. Rats were given only one dose gosereline (0.9 mg/rat), a GnRH agonist in the third group. The last group received kisspeptin-10 with gosereline. The activities of catalase, superoxide dismutase (SOD), xanthine oxidase (XO), adenosine deaminase (AD) and level of malondialdehyde were studied in liver tissue. Serum samples were separated for total antioxidant capacity (TAC), total oxidant status (TOS), alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase, blood urea nitrogen (BUN), colesterol, high-density lipoprotein (HDL) and triglyceride. Kisspeptin increased the activities of SOD and catalase (p < 0.05). When compared to the control group, the levels of malondialdehyde, TOS and AST were lower, but levels of BUN, cholesterole, HDL and AD were higher in the other three groups (p < 0.05). In conclusion, our findings suggest that kisspeptin may have antioxidant and thus protective effects on the liver tissue.


Assuntos
Fígado/metabolismo , Oligopeptídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Adenosina Desaminase/metabolismo , Alanina Transaminase/sangue , Fosfatase Alcalina/sangue , Animais , Antineoplásicos Hormonais/farmacologia , Aspartato Aminotransferases/sangue , Nitrogênio da Ureia Sanguínea , Catalase/metabolismo , Colesterol/sangue , Gosserrelina/farmacologia , Kisspeptinas , Lipoproteínas HDL/sangue , Fígado/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Triglicerídeos/sangue , Xantina Oxidase/metabolismo
15.
Gynecol Endocrinol ; 26(9): 669-72, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20236028

RESUMO

Most surgical procedures performed by obstetrician-gynecologists are associated with pelvic adhesions that cause subsequent serious sequelae, including small bowel obstruction, infertility, chronic pelvic pain, and difficulty in postoperative treatment, including complexity during subsequent surgical procedures. This study was conducted to determine if gonadotropin-releasing hormone analogues (GnRHa) affect the expressing tissue-type plasminogen activator (t-PA) and its inhibitor-1 (PAI-1) in peritoneal cells in culture. Human peritoneal Met5A cells were used to examine the effects of GnRHa leuprolide, buserelin and goserelin on the levels of t-PA and PA-1. Antigen concentrations were measured in conditioned media and cell lysates by real-time PCR and ELISA. GnRH receptor (GnRHR) mRNA was determined by RT-PCR. GnRHR mRNA was detected in Met5A cells. Exposure of Met5A cells to GnRHa induced a rapid decrease of PAI-1 level in cultured medium but not in cell lysate (protein and mRNA). These effects of GnRHa on PAI-1 were not associated with any changes in t-PA level. These results suggest that GnRHa may be an effective stimulator of local peritoneal fibrinolytic activity, as it decreases PAI-1 secretion in peritoneal Met5A cells by a mechanism linked to GnRHR.


Assuntos
Fibrinolisina/metabolismo , Peritônio/metabolismo , Receptores LHRH/fisiologia , Busserrelina/farmacologia , Células Cultivadas , Ativação Enzimática , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Gosserrelina/farmacologia , Humanos , Leuprolida/farmacologia , Peritônio/enzimologia , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo
16.
Zhonghua Fu Chan Ke Za Zhi ; 45(4): 247-51, 2010 Apr.
Artigo em Zh | MEDLINE | ID: mdl-20646533

RESUMO

OBJECTIVE: To compare clinical effect of gonadotropin releasing hormone agonist (GnRH-a) alone and GnRH-a combined with low-dose dydrogesteronea and estradiol valerate on sex hormone, hypoestrogenic symptoms, quality of life and bone mineral density (BMD) in treatment of endometriosis. METHODS: Seventy patients with moderate or severe endometriosis, who were diagnosed by laparotomy or laparoscopic surgery within two months, were randomly assigned into two groups. 35 patients in GnRH-a group were treated by goserelin (3.6 mg) for three months, and 35 patients in add-back group were treated by goserelin (3.6 mg) combined with estradiol valerate 0.5 mg and dydrogesteronea 5 mg daily. Before and after the treatment, clinical parameters were recorded and analyzed, including visual analog scale (VAS), medical outcomes survey short form 36 (SF-36), Kupperman menopausal index (KMI), BMD, the serum level of follicle stimulating hormone (FSH), estradiol (E2) and bone gla-protein (BGP). The first menstruation and VAS were also followed up after treatment. RESULTS: Every 3 cases in two groups lost follow-up. (1) Reproductive hormone: the level of E2 in add-back group [(94+/-71) pmol/L] was significantly higher than (54+/-52) pmol/L in GnRH-a group (P<0.01). The level of FSH in add-back group [(3.0+/-1.9) U/L] was significantly lower than (5.7+/-2.9) U/L in GnRH-a group (P<0.05). (2) VAS: after treatment, VAS in both group decreased significantly when compared with that before treatment (P<0.05), and remained until menstruated. (3) KMI: KMI in add back-group (10+/-8)was significantly lower than (14+/-6) in GnRH-a group (P<0.05). (4) BMD: compared with that before treatment, BMD decreased significantly after treatment in GnRH-a group (P<0.05), no remarkable difference of BMD was observed before and after treatment in add-back group. Before treatment, serum BGP in both groups did not show statistical difference. After treatment, the level of BGP in GnRH-a group [(7932+/-5206) ng/L] was significantly higher than (5419+/-2917) ng/L in add-back group (P<0.05). CONCLUSIONS: GnRH-a combined with estrogen-progesterone regimen could relieve pain from endometriosis as effectively as GnRH-a alone and reduce hypoestrogenic symptoms and bone loss. Therefore, it is a safe and effective treatment.


Assuntos
Didrogesterona/administração & dosagem , Endometriose/tratamento farmacológico , Estradiol/análogos & derivados , Hormônio Liberador de Gonadotropina/agonistas , Adolescente , Adulto , Densidade Óssea/efeitos dos fármacos , Esquema de Medicação , Didrogesterona/uso terapêutico , Endometriose/sangue , Endometriose/patologia , Estradiol/administração & dosagem , Estradiol/sangue , Estradiol/uso terapêutico , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/administração & dosagem , Gosserrelina/farmacologia , Gosserrelina/uso terapêutico , Humanos , Pessoa de Meia-Idade , Osteocalcina/sangue , Dor/tratamento farmacológico , Dor/patologia , Qualidade de Vida , Índice de Gravidade de Doença , Resultado do Tratamento , Adulto Jovem
17.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 35(5): 409-18, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20543462

RESUMO

OBJECTIVE: To determine the effect of GnRH I and GnRH II on the secretion of VEGF by eutopic and ectopic endometrial stromal cells cultured in vitro, and to provide theoretical basis for exploring new treatments for endometriosis (EMs). METHODS: Eutopic and ectopic endometrium stromal cells cultured in vitro were treated with different concentrations of GnRH II and a GnRH I (goserelin), and a control group was not treated by GnRH II and GnRH I. Enzyme linked immunosorbent assay (ELISA) was used to measure the content of vascular endothelial growth factor (VEGF) protein in the medium of the above 2 groups. RESULTS: (1) There was no difference in the VEGF protein secreted by eutopic and ectopic stromal cells in the medium after being cultured in vitro for 48 h (P>0.05). (2) 10(-10), 10(-8), and 10(-6) mol/L GnRH II dose-dependently reduced VEGF protein secreted by endometrial stromal cells (P<0.05),and the inhibition effect was stronger than that of GnRH I (P<0.05).(3)The inhibition effect of GnRH II on VEGF in ectopic stromal cells was stronger than that of eutopic stromal cells (P<0.05). CONCLUSION: (1) Ectopic stromal cells cultured in vitro can secrete VEGF,which has no difference from the eutopic stromal cells, and which may play an important role in the formation and development of EMs. (2) GnRH II can dose-dependently reduce VEGF protein secreted by ectopic and eutopic endometrial stromal cells cultured in vitro,and the inhibition effect is stronger than that of GnRH I, providing theoretical basis for exploring new treatments for EMs.


Assuntos
Endometriose/patologia , Endométrio/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Gosserrelina/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Células Cultivadas , Endométrio/efeitos dos fármacos , Endométrio/patologia , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Pessoa de Meia-Idade , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Adulto Jovem
18.
J Pharm Biomed Anal ; 180: 113044, 2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-31865209

RESUMO

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.


Assuntos
Materiais Biomiméticos/metabolismo , Portadores de Fármacos/química , Líquido Extracelular/metabolismo , Gosserrelina/química , Gosserrelina/farmacologia , Espectrometria de Massas em Tandem/métodos , Técnicas Biossensoriais , Cromatografia Líquida de Alta Pressão , Composição de Medicamentos , Liberação Controlada de Fármacos , Limite de Detecção , Modelos Lineares , Extração Líquido-Líquido , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Solventes/química
19.
Oncol Rep ; 21(5): 1277-82, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19360304

RESUMO

Malignant glioblastoma is one of the highest proliferative and invasive tumors within the central nervous system (CNS); the therapeutical options for this disease are still very poor. Receptors for gonadotropin-releasing hormone (GnRH) have been reported to be present in glioblastoma tissues. This study aimed to determine the role of these receptors in the control of glioma growth. In two human glioblastoma cell lines, U87MG and U373, we demonstrated the expression of GnRH receptors, both at mRNA and protein levels. We also found that GnRH receptor is expressed in glioblastoma tissues from tumor patients as shown by Western blotting. In U87MG and U373 cell lines, we found the expression of mRNA for GnRH, indicating the presence of an autocrine GnRH-based system in these cell lines. Treatment of the two cell lines with a GnRH agonist resulted in a significant decrease of cell proliferation. Moreover, the GnRH agonist significantly counteracted the forskolin-induced increase of intracellular cAMP levels in these cells. These findings suggest that the GnRH receptor might represent a molecular target for an endocrine therapeutical approach against gliomas.


Assuntos
Glioblastoma/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Receptores LHRH/biossíntese , Antineoplásicos Hormonais/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Glioblastoma/genética , Glioblastoma/patologia , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/genética , Gosserrelina/farmacologia , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores LHRH/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Thromb Thrombolysis ; 27(2): 172-4, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18183354

RESUMO

BACKGROUND: Gonadotropin releasing hormone (GnRH) agonists are the cornerstone of metastatic prostate cancer treatment. Cardiovascular effects of GnRH agonists are unclear. In this study, we investigated the short term effects of GnRH agonists on plasma fibrinolytic parameters in patients with metastatic prostate cancer. METHODS: Eleven patients (mean age 69.3 +/- 6.5) with metastatic prostate cancer and a clinical indication for GnRH agonist therapy were selected. Plasma plasminogen activator inhibitor (PAI-1) antigen (Ag), tissue plasminogen activator (t-PA) Ag and thrombin-activatable fibrinolysis inhibitor (TAFI) activity levels were measured at baseline and at 4 weeks after the first dose of GnRH agonist, Goserelin Acetate (Zoladex, subcutaneous administration, 10.8 mg). RESULTS: Serum prostate specific antigen (PSA) levels significantly decreased from 36.6 +/- 19.3 to 1.1 +/- 0.3 ng/ml after Goserelin acetate treatment (P = 0.005). Significant changes occurred in the fibrinolytic parameters. GnRH agonists decreased plasma t-PA Ag levels (16.3 +/- 4.9 vs. 12.2 +/- 2.8 ng/ml, P = 0.047) and increased PAI-1/t-PA molar ratio (4.8 +/- 3.6 vs. 6.6 +/- 3.4, P = 0.16), on the other hand, plasma PAI-1 Ag (59.0 +/- 48.5 vs. 56.4 +/- 30.5 ng/ml, P = 0.8), and TAFI levels (130.6 +/- 9.5 vs. 124.2 +/- 26.5% activity, P = 0.3) did not change significantly. CONCLUSION: This study provides evidence that GnRH agonists may inhibit fibrinolytic system by decreasing t-PA levels.


Assuntos
Fibrinólise/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/agonistas , Gosserrelina/farmacologia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/tratamento farmacológico , Idoso , Biomarcadores/sangue , Gosserrelina/uso terapêutico , Hemostasia/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Antígeno Prostático Específico/sangue , Ativador de Plasminogênio Tecidual/sangue , Ativador de Plasminogênio Tecidual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA