Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Ital J Pediatr ; 48(1): 184, 2022 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371229

RESUMO

BACKGROUND: Congenital hypogonadotropic hypogonadism (CHH) is a clinically and genetically heterogeneous disease characterized by absent or incomplete puberty and infertility. Clinical characteristics are secondary to insufficient gonadotropin secretion, caused by deficient gonadotropin-releasing hormone (GnRH) production, secretion, or action. Loss-of-function variants of the gonadotropin-releasing hormone receptor (GNRHR) are associated with CHH without anosmia. CHH was previously considered a permanent condition, but in the past two decades, cases of spontaneous recovery of CHH have been reported. The reversal of hypogonadism in CHH is currently unpredictable, and can happen unnoticed. CASE PRESENTATION: The male proband was diagnosed with CHH due to compound heterozygosity for two previously reported pathogenic missense variants in the GNRHR gene, NM_000406.2:c.416G > A (NP_000397.1:p.Arg139His) and c.785G > A (p.Arg262Gln) at 16 years of age. In addition to arrested partial puberty, he had a low testosterone level, gonadotropins in the range of early puberty, and a normal inhibin B level. A therapy with increasing doses of intramuscular testosterone undecanoate was received for 2.5 years, while there was no change in testicular volume. At the age of 19 years, testosterone supplementation was interrupted. During the next two years, he had spontaneous pubertal development to achieve a testicular volume of 20 mL, with normal adult levels of gonadotropins and testosterone. CONCLUSIONS: Genetic diagnostics can help discriminate congenital hypogonadotropic hypogonadism, deserving therapeutic intervention, from the self-limited constitutional delay of growth and puberty (CDGP). Patients with GNRHR associated hypogonadism can experience spontaneous recovery of the hypothalamic-pituitary-gonadal axis. Spontaneous testis enlargement in patients with central hypogonadism not taking gonadotropins or pulsatile GnRH therapy can indicate recovery of hypogonadism.


Assuntos
Hipogonadismo , Receptores LHRH , Adulto , Humanos , Masculino , Adulto Jovem , Receptores LHRH/genética , Receptores LHRH/uso terapêutico , Hipogonadismo/diagnóstico , Hipogonadismo/tratamento farmacológico , Hipogonadismo/genética , Hormônio Liberador de Gonadotropina/uso terapêutico , Hormônio Liberador de Gonadotropina/genética , Gonadotropinas/uso terapêutico , Testosterona/uso terapêutico
2.
Am Fam Physician ; 106(4): 397-404, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36260896

RESUMO

Endometriosis is an inflammatory condition caused by the presence of endometrial tissue in extra-uterine locations and can involve bowel, bladder, and all peritoneal structures. It is one of the most common gynecologic disorders, affecting up to 10% of people of reproductive age. Presentation of endometriosis can vary widely, from infertility in asymptomatic people to debilitating pelvic pain, dysmenorrhea, and period-related gastrointestinal or urinary symptoms. Diagnosis of endometriosis in the primary care setting is clinical and often challenging, frequently resulting in delayed diagnosis and treatment. Although transvaginal ultrasonography is used to evaluate endometriosis of deep pelvic sites to rule out other causes of pelvic pain, magnetic resonance imaging is preferred if deep infiltrating endometriosis is suspected. Laparoscopy with biopsy remains the definitive method for diagnosis, although several gynecologic organizations recommend empiric therapy without immediate surgical diagnosis. Combined hormonal contraceptives with or without nonsteroidal anti-inflammatory drugs are first-line options in managing symptoms and have a tolerable adverse effect profile. Second-line treatments include gonadotropin-releasing hormone (GnRH) receptor agonists with add-back therapy, GnRH receptor antagonists, and danazol. Aromatase inhibitors are reserved for severe disease. All of these treatments are effective but may cause additional adverse effects. Referral to gynecology for surgical management is indicated if empiric therapy is ineffective, immediate diagnosis and treatment are necessary, or patients desire pregnancy. Alternative treatments have limited benefit in alleviating pain symptoms but may warrant further investigation.


Assuntos
Endometriose , Feminino , Humanos , Gravidez , Anti-Inflamatórios não Esteroides/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Anticoncepcionais , Danazol/uso terapêutico , Endometriose/terapia , Endometriose/tratamento farmacológico , Hormônio Liberador de Gonadotropina/uso terapêutico , Hormônio Liberador de Gonadotropina/agonistas , Dor Pélvica/terapia , Dor Pélvica/tratamento farmacológico , Receptores LHRH/uso terapêutico
3.
Lancet ; 400(10356): 896-907, 2022 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-36116480

RESUMO

BACKGROUND: Uterine fibroids are common non-cancerous neoplasm that cause heavy menstrual bleeding and other signs. Linzagolix is an oral gonadotropin-releasing hormone receptor antagonist taken once per day that dose-dependently suppresses gonadal steroids and might reduce uterine-fibroid-associated signs. Two phase 3 trials were conducted to confirm the efficacy and safety of linzagolix at full-suppression (200 mg) and partial-suppression (100 mg) doses with or without hormonal add-back therapy (1 mg oestradiol and 0·5 mg norethisterone acetate) compared with placebo for the treatment of symptomatic uterine fibroids. METHODS: PRIMROSE 1 and PRIMROSE 2 were identical 52-week, randomised, parallel, double-blind, placebo-controlled, phase 3 trials conducted at clinics in the USA (PRIMROSE 1) and Europe and the USA (PRIMROSE 2). Eligible women with uterine fibroid-associated heavy menstrual bleeding (menstrual blood loss >80 mL per cycle) were randomly assigned in a 1:1:1:1:1 ratio to one of five masked treatments: (1) placebo, (2) 100 mg linzagolix per day alone, (3) 100 mg linzagolix per day with once-per-day hormonal add-back therapy (1 mg oestradiol and 0·5 mg norethisterone acetate), (4) 200 mg linzagolix per day alone, or (5) 200 mg linzagolix per day with once-per-day hormonal add-back therapy (1 mg oestradiol and 0·5 mg norethisterone acetate). The primary endpoint was a response (menstrual blood loss ≤80 mL and ≥50% reduction from baseline) at 24 weeks in women who received at least one dose of treatment and did not meet any exclusion criteria based on predosing assessments. These trials are registered with ClinicalTrials.gov (NCT03070899 and NCT03070951). The trials have been completed. FINDINGS: Between May, 2017, and October, 2020, in PRIMROSE 1, 574 women were enrolled, of which 48 discontinued and 15 were excluded; therefore, 511 women were included in the full analysis set; and in PRIMROSE 2, 535 women were enrolled, of which 24 did not receive the study drug and ten women were excluded from the study, resulting in 501 women being included in the full analysis set. In both trials, a significantly higher proportion of women had a reduction in heavy menstrual bleeding in all linzagolix (with or without add-back therapy) treatment groups compared with the placebo group (p≤0·003). In PRIMROSE 1, the response rates were 56·4% (95% CI 45·8-66·6%) in the 100 mg group, 66·4% (56·6-75·2%) in the 100 mg plus add-back therapy group, 71·4% (61·8-79·8%) in the 200 mg group, and 75·5% (66·0-83·5%) in the 200 mg plus add-back therapy group, compared with 35·0% (25·8-45·0%) in the placebo group. In PRIMROSE 2, the response rates were 56·7% (46·3-66·7%) in the 100 mg group, 77·2% (67·8-85·0%) in the 100 mg plus add-back therapy group, 77·7% (68·4-85·3%) in the 200 mg group, and 93·9% (87·1-97·7%) in the 200 mg plus add-back therapy group, compared with 29·4% (20·8-39·3%) with placebo. The most common adverse events up to 24 weeks were hot flushes (35% of participants in PRIMROSE 1 and 32% in PRIMROSE 2 with linzagolix [200 mg] alone and 3-14% in all other groups). INTERPRETATION: Linzagolix (100 mg or 200 mg) with or without add-back therapy significantly reduced heavy menstrual bleeding. Partial suppression with once-per-day linzagolix (100 mg) without add-back therapy potentially provides a unique option for the chronic treatment of symptomatic uterine fibroids in women who cannot or do not want to take concomitant hormonal add-back therapy. FUNDING: ObsEva.


Assuntos
Leiomioma , Menorragia , Neoplasias Uterinas , Ácidos Carboxílicos , Estradiol , Feminino , Humanos , Leiomioma/tratamento farmacológico , Menorragia/complicações , Menorragia/etiologia , Acetato de Noretindrona , Pirimidinas , Receptores LHRH/uso terapêutico , Neoplasias Uterinas/complicações , Neoplasias Uterinas/tratamento farmacológico
4.
Appl Immunohistochem Mol Morphol ; 30(7): 509-516, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35777983

RESUMO

Luteinizing hormone-releasing hormone receptor (LHRHR) expression has been reported in various cancers, including endometrial neoplasms. Thus, LHRHR provides a potential point for therapeutic approach using LHRH analogs as carrier molecules for chemotherapeutic agents in this cancer population. However, clinical data did not prove any potential benefits for patients. We decided to assess LHRHR expression in patients with endometrial cancer to explain possible lack of efficacy in previous clinical reports. LHRHR expression was assessed immunohistochemically in different anatomic and histogenetic compartments of female genital tract of patients with endometrial cancer. The study sample consisted of paraffin tissue blocks obtained from patients who has undergone primary surgery owing to endometrial cancer. Strong LHRHR expression was found in endometrial cancer, fallopian tube, and concurrent atypical hyperplasia. Interestingly, LHRHR expression showed significant differences depending on the respective compartment of the ovary analyzed. Level of LHRHR expression in patients with primary advanced and unresectable disease, particularly in certain ovarian compartments may be substantially lower, which may influence the use of new targeted therapy regimens. The studies on secondary Müllerian system compartment and its hormonal receptor status may be crucial to understand mechanisms of lack of efficacy of LHRH hybrid molecules anti-cancer treatment.


Assuntos
Antineoplásicos , Neoplasias do Endométrio , Antineoplásicos/uso terapêutico , Neoplasias do Endométrio/metabolismo , Feminino , Genitália Feminina/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Receptores LHRH/metabolismo , Receptores LHRH/uso terapêutico
5.
Int J Mol Sci ; 23(9)2022 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-35563462

RESUMO

The human gonadotropin releasing hormone (GnRH-I) and its sea lamprey analogue GnRH-III specifically bind to GnRH receptors on cancer cells and can be used as targeting moieties for targeted tumor therapy. Considering that the selective release of drugs in cancer cells is of high relevance, we were encouraged to develop cleavable, self-immolative GnRH-III-drug conjugates which consist of a p-aminobenzyloxycarbonlyl (PABC) spacer between a cathepsin B-cleavable dipeptide (Val-Ala, Val-Cit) and the classical anticancer drugs daunorubicin (Dau) and paclitaxel (PTX). Alongside these compounds, non-cleavable GnRH-III-drug conjugates were also synthesized, and all compounds were analyzed for their antiproliferative activity. The cleavable GnRH-III bioconjugates revealed a growth inhibitory effect on GnRH receptor-expressing A2780 ovarian cancer cells, while their activity was reduced on Panc-1 pancreatic cancer cells exhibiting a lower GnRH receptor level. Moreover, the antiproliferative activity of the non-cleavable counterparts was strongly reduced. Additionally, the efficient cleavage of the Val-Ala linker and the subsequent release of the drugs could be verified by lysosomal degradation studies, while radioligand binding studies ensured that the GnRH-III-drug conjugates bound to the GnRH receptor with high affinity. Our results underline the high value of GnRH-III-based homing devices and the application of cathepsin B-cleavable linker systems for the development of small molecule drug conjugates (SMDCs).


Assuntos
Hormônio Liberador de Gonadotropina , Terapia de Alvo Molecular , Neoplasias Ovarianas , Receptores LHRH , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Catepsina B/química , Catepsina B/uso terapêutico , Linhagem Celular Tumoral , Daunorrubicina/química , Daunorrubicina/uso terapêutico , Feminino , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Terapia de Alvo Molecular/métodos , Paclitaxel/química , Paclitaxel/uso terapêutico , Petromyzon , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/uso terapêutico , Receptores LHRH/uso terapêutico
6.
Br J Clin Pharmacol ; 88(5): 2359-2371, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34904270

RESUMO

AIMS: To identify linzagolix doses, an oral GnRH receptor antagonist, that effectively lower oestradiol (E2) to relieve endometriosis-related pelvic pain without compromising bone health. METHODS: Integrated statistical, pharmacokinetic-pharmacodynamic and systems pharmacology models were developed from Phase 1 and 2 clinical trial data in healthy volunteers and patients, receiving linzagolix 25-200 mg daily or placebo, and analysed simultaneously. The main outcome measures were pelvic pain scores for dysmenorrhoea, nonmenstrual pelvic pain (NMPP), uterine bleeding and lumbar spine bone mineral density (BMD). RESULTS: Linzagolix pharmacokinetics were described by a 2-compartment model with sequential zero/first-order absorption process (CL/F: 0.422 L/h). E2 changes over time were well described as a function of linzagolix 24-hour AUC (AUC50 : 1.68 × 105 ng h/mL). For a Caucasian reference patient, a change in E2 from 50-20 pg/mL at 24 weeks increased the odds of relief of dysmenorrhoea 1.33-fold and NMPP 1.07-fold (95% CI: 1.22-1.47 and 1.02-1.12, respectively) and decreased bleeding days by 1.55 (95% CI: 1.39-1.72). A previously validated quantitative systems pharmacology BMD model was adjusted to the clinical data. The mean week 24 lumbar spine BMD change from baseline ranged from -0.092% in the 50 mg dose, -1.30% in the 100 mg dose group and -2.67% in the 200 mg dose group. DISCUSSION: The previously-reported E2 target range (20-50 pg/mL) to balance efficacy and safety endpoints was confirmed. Linzagolix once daily doses between 75-125 mg daily were expected to meet endometriosis-associated pain, efficacy, and BMD loss targets in Caucasian patients.


Assuntos
Endometriose , Receptores LHRH , Densidade Óssea , Ácidos Carboxílicos , Dismenorreia/tratamento farmacológico , Endometriose/tratamento farmacológico , Feminino , Antagonistas de Hormônios/farmacologia , Antagonistas de Hormônios/uso terapêutico , Humanos , Dor Pélvica/tratamento farmacológico , Pirimidinas , Receptores LHRH/uso terapêutico
7.
Molecules ; 26(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673080

RESUMO

This study investigated the effect of type 1 gonadotropin releasing hormone receptor (GnRH-R) localization within lipid rafts on the properties of plasma membrane (PM) nanodomain structure. Confocal microscopy revealed colocalization of PM-localized GnRH-R with GM1-enriched raft-like PM subdomains. Electron paramagnetic resonance spectroscopy (EPR) of a membrane-partitioned spin probe was then used to study PM fluidity of immortalized pituitary gonadotrope cell line αT3-1 and HEK-293 cells stably expressing GnRH-R and compared it with their corresponding controls (αT4 and HEK-293 cells). Computer-assisted interpretation of EPR spectra revealed three modes of spin probe movement reflecting the properties of three types of PM nanodomains. Domains with an intermediate order parameter (domain 2) were the most affected by the presence of the GnRH-Rs, which increased PM ordering (order parameter (S)) and rotational mobility of PM lipids (decreased rotational correlation time (τc)). Depletion of cholesterol by methyl-ß-cyclodextrin (methyl-ß-CD) inhibited agonist-induced GnRH-R internalization and intracellular Ca2+ activity and resulted in an overall reduction in PM order; an observation further supported by molecular dynamics (MD) simulations of model membrane systems. This study provides evidence that GnRH-R PM localization may be related to a subdomain of lipid rafts that has lower PM ordering, suggesting lateral heterogeneity within lipid raft domains.


Assuntos
Lipídeos de Membrana/química , Microdomínios da Membrana/química , Receptores LHRH/química , Colesterol/química , Colesterol/genética , Espectroscopia de Ressonância de Spin Eletrônica , Células HEK293 , Humanos , Lipídeos de Membrana/genética , Microdomínios da Membrana/genética , Microdomínios da Membrana/ultraestrutura , Domínios Proteicos/genética , Receptores LHRH/genética , Receptores LHRH/uso terapêutico , Receptores LHRH/ultraestrutura , Transdução de Sinais/genética
9.
Actas urol. esp ; 42(4): 249-255, mayo 2018. tab, graf
Artigo em Espanhol | IBECS | ID: ibc-172888

RESUMO

Introducción y objetivos: El manejo del cáncer de próstata (CP) en receptores de trasplante renal (RTR) es un reto y muchos aspectos siguen siendo controvertidos. Actualmente no hay consensos sobre el manejo de esta enfermedad. El objetivo del estudio fue analizar nuestra experiencia en el diagnóstico y tratamiento del CP en RTR. Método: Estudio monocéntrico retrospectivo de una base de datos prospectiva y consecutiva del 2003-2017. Inclusión de RTR diagnosticados de CP. Estadificación y tratamiento según guías contemporáneas. Los principales parámetros evaluados incluyeron estadificación clínica, tipo de tratamiento, resultados oncológicos y seguimiento. Resultados: De 1.330 trasplantes renales (787 varones), se diagnosticó CP en 33 RTR (4,2%), con una edad media de 66 años ± 6,3 (51-78). La media de PSA fue de 8,8 ng/ml y PSA ratio de 0,19. El tiempo medio entre el trasplante renal y el diagnóstico de CP fue de 130 meses ± 90 (2-236). Tratamiento: prostatectomía radical (PR) (n = 22; 66,7%), radioterapia (RT) más terapia de privación androgénica (TDA) (n = 7; 21,2%), vigilancia activa (n = 3; 9,1%) y TDA (n = 1; 3%). No se reportaron pérdidas del injerto ni deterioro de la función renal atribuibles al tratamiento del CP. Tras PR, 2 pacientes (9,1%) presentaron recurrencia bioquímica, tratados con RT. La remisión fue del 100% y el seguimiento medio, de 61 meses ± 37 (6-132). Conclusiones: El CP en trasplantados renales puede ser manejado con las mismas opciones terapéuticas que en la población general. La vigilancia activa debería también considerarse en los RTR a pesar de estar bajo tratamiento inmunosupresor


Introduction and objectives: The management of Prostate cancer (PCa) in renal transplant recipients (RTR) is challenging and remain controversial. Currently there is no consensus about this condition. The aim of the study was to analyse our experience in the diagnosis and management of PCa in RTR. Method: Retrospective monocentric study of a prospective and consecutive database from 2003-2017. Inclusion of RTR diagnosed of PCa. Staging and treatment in agreement with the contemporary guidelines. The main outcome measures included clinical staging, type of treatment, oncological outcomes and follow-up. Results: 1,330 renal transplants were performed (787 males), diagnosed of PCa in 33 RTR (4.2%), mean age 66 years ± 6.3 (51-78). Median PSA was 8.8 ng/ml and PSA ratio 0.19. Mean time between renal transplantation and PCa diagnosis 130 months ± 90 (2-236). Treatments: Radical prostatectomy (RP) (n = 22; 66.7%), Radiation therapy (RT) with Androgen deprivation therapy (ADT) (n = 7; 21.2%), Active surveillance (n = 3; 9.1%), ADT (n = 1; 3%). No graft loss neither impaired renal function due to PCa treatment was reported. After RP two patients (9.1%) presented biochemical recurrence treated with RT. Remission of the 100%. Mean follow-up was 61months ± 37 (6-132). Conclusions: PCa in renal transplant patients can be managed with the same therapeutic options as in the general population. Active surveillance should also be provided in RTR despite being under immunosuppressive therapy


Assuntos
Humanos , Masculino , Pessoa de Meia-Idade , Idoso , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Transplante de Rim/métodos , Prostatectomia/métodos , Biópsia , Estudos Retrospectivos , Estadiamento de Neoplasias/métodos , Antígeno Prostático Específico/análise , Neoplasias da Próstata/radioterapia , Receptores LHRH/uso terapêutico , Imunossupressores/uso terapêutico
10.
Vitam Horm ; 107: 27-66, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29544634

RESUMO

Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Modelos Biológicos , Células Neuroendócrinas/fisiologia , Ovário/fisiologia , Precursores de Proteínas/metabolismo , Receptores LHRH/agonistas , Reprodução , Animais , Endométrio/efeitos dos fármacos , Endométrio/crescimento & desenvolvimento , Endométrio/fisiologia , Endométrio/fisiopatologia , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Fármacos para a Fertilidade Feminina/uso terapêutico , Preservação da Fertilidade/tendências , Hormônio Liberador de Gonadotropina/química , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/fisiopatologia , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/patologia , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/terapia , Ciclo Menstrual/efeitos dos fármacos , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Ovário/fisiopatologia , Gravidez , Insuficiência Ovariana Primária/tratamento farmacológico , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/fisiopatologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/química , Puberdade/efeitos dos fármacos , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo , Receptores LHRH/uso terapêutico , Reprodução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
11.
Psychoneuroendocrinology ; 82: 164-172, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28550793

RESUMO

Leuprolide acetate (LEU), also known as Lupron, is commonly used to treat prostate cancer in men. As a gonadotropin-releasing hormone (GnRH) receptor agonist, it initially stimulates the release of gonadal hormones, testosterone (T) and estradiol. This surge eventually suppresses these hormones, preventing the further growth and spread of cancer cells. Individuals receiving this treatment often report anxiety and cognitive changes, but LEU's effects on the neural mechanisms that are involved in anxiety during the trajectory of treatment are not well known. In this study, we examined the acute effects of LEU on fear extinction, hypothesizing that increased T levels following a single administration of LEU will facilitate extinction recall by altering neuronal activity within the fear extinction circuitry. Two groups of naïve adult male rats underwent a 3-day fear conditioning, extinction, and recall experiment. The delayed group (n=15) received a single injection of vehicle or LEU (1.2mg/kg) 3weeks before behavioral testing. The acute group (n=25) received an injection one day after fear conditioning, 30min prior to extinction training. Following recall, the brains for all animals were collected for c-fos immunohistochemistry. Blood samples were also collected and assayed for T levels. Acute administration of LEU increased serum T levels during extinction training and enhanced extinction recall 24h later. This enhanced extinction memory was correlated with increased c-fos activity within the infralimbic cortex and amygdala, which was not observed in the delayed group. These results suggest that the elevation in T induced by acute administration of LEU can influence extinction memory consolidation, perhaps through modification of neuronal activity within the infralimbic cortex and amygdala. This may be an important consideration in clinical applications of LEU and its effects on anxiety and cognition.


Assuntos
Extinção Psicológica/efeitos dos fármacos , Leuprolida/farmacologia , Memória/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Condicionamento Clássico/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário , Leuprolida/metabolismo , Masculino , Memória/fisiologia , Neurônios/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores LHRH/agonistas , Receptores LHRH/uso terapêutico , Testosterona/sangue
12.
Rev. iberoam. fertil. reprod. hum ; 33(3): 33-41, jul.-sept. 2016. ilus
Artigo em Espanhol | IBECS | ID: ibc-156071

RESUMO

OBJETIVO: Revisar y actualizar la fisiología de la regulación de fertilidad y reproducción, poniendo especial énfasis en los factores neuroendocrinos que controlan la secreción pulsátil de GnRH. MATERIAL Y MÉTODOS: Revisión bibliográfica, utilizando como palabras clave «GnRH», «Kisspeptin», «GnIH», «RF-amide» y RPRF-3, entre otras. RESULTADOS: Siendo la secreción pulsátil de GnRH el elemento clave para el control de la secreción de gonadotrofinas, se describen los mecanismos neuroendocrinos (Kisspeptina, GnIH) que regulan la actividad de las neuronas GnRH


OBJECTIVE: To Review and update the physiology regulating fertility and reproduction, with particular emphasis on neuroendocrine factors controlling the pulsatile secretion of GnRH. MATERIAL AND METHODS: Review of the literature, using as key words «GnRH», «Kisspeptin», «GnIH», «RF-amide» and RPRF-3, among others. RESULTS: Being the pulsatile secretion of GnRH the key element for the control of gonadotrophin secretion, the neuroendocrine mechanisms (Kisspeptine, GnIH) that regulate the activity of GnRH neurons are described


Assuntos
Humanos , Masculino , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/uso terapêutico , Receptores LHRH/uso terapêutico , Neuroendocrinologia/métodos , Neuropeptídeos/uso terapêutico , Receptores de Neuropeptídeos/uso terapêutico , Fertilidade , Fármacos para a Fertilidade/uso terapêutico , Receptores da Gonadotropina , Sistema Hipotálamo-Hipofisário
13.
Urol Oncol ; 33(6): 270-4, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25512159

RESUMO

BACKGROUND: Androgen deprivation therapy (ADT) has been the standard of care for treating patients with hormone-sensitive advanced prostate cancer (PCa) for 3 decades. The agonists of luteinizing hormone-releasing hormone (LHRH), also called gonadotropin-releasing hormone, are still the most frequently used form of medical ADT. ADT AND LHRH ANALOGS: The application of agonists of LHRH has improved and modernized the treatment of advanced PCa; millions of patients have benefited from therapy with LHRH agonists as a preferred alternative to surgical castration, as the psychological effects and perpetuity of orchiectomy are undesirable for most men. Despite their efficacy, agonists of LHRH have several shortcomings, including initial surge in testosterone, producing exacerbation of clinical symptoms, and microsurges in testosterone that might occur after each administration. A new, alternate approach to ADT is emerging with the improvements in antagonists of LHRH. This class of LHRH analogues produces a direct and immediate blockade of pituitary LHRH receptors and leads to a more rapid suppression of testosterone without an initial surge or subsequent microsurges. Degarelix, a third-generation LHRH antagonist, is the only antagonist with a low histamine-releasing activity that is currently on the market for clinical use in advanced PCa with improved testosterone suppression, better control of follicle-stimulating hormone and prostate-specific antigen, and which offers a prolonged delay to progression and more favorable effects on serum alkaline phosphatase. CONCLUSIONS: Although LHRH agonists are still the mainstay for treatment of advanced PCa, antagonists of LHRH offer an alternative as a pharmacological approach.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/uso terapêutico , Humanos , Masculino , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Receptores LHRH/agonistas
14.
Cancer Chemother Pharmacol ; 73(5): 931-41, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24610297

RESUMO

PURPOSE: To conduct a phase I study determining the safety, pharmacokinetics and preliminary efficacy of EP-100, a novel anticancer drug consisting of natural luteinizing-hormone-releasing hormone (LHRH) ligand linked to a cationic membrane-disrupting peptide. METHODS: Patients with advanced, solid tumors, positive for LHRH receptor by immunohistochemistry (IHC), received EP-100 weekly or twice weekly for 3 of 4 weeks in a 28 day cycle. A modified Fibonacci 3 + 3 dose-escalation schema was used. Initial cohorts received EP-100 once weekly (cohorts 1-7, 0.6-7.8 mg/m(2), n = 21). Later cohorts received doses twice weekly (cohorts 7-11, 7.8-40 mg/m(2), n = 16). RESULTS: LHRH-receptor expression was confirmed by IHC in 52 of 89 consented patients; 37 patients received at least 1 dose. Cohorts receiving doses of 5.2 mg/m(2) and above achieved therapeutic levels from in vitro studies Clearance was rapid (mean half-life 7.1 ± 3.8 to 15.9 ± 3.6 min). The maximum-tolerated dose was not reached at the highest dose evaluated (40 mg/m(2) twice weekly). Grade 2 increase in alanine aminotransferase/serum aspartate aminotransferase in one patient resolved, did not recur upon re-treatment, and was not observed in other patients. The only drug-related adverse event was transient infusion-related dermatologic reactions (10 patients). No complete or partial tumor responses were observed; seven patients had stable disease of 16 weeks. CONCLUSIONS: EP-100 was well tolerated in patients with advanced, LHRH-receptor-expressing solid tumors. The recommended phase 2 dose is 40 mg/m(2) twice weekly for 3 of 4 weeks per cycle.


Assuntos
Neoplasias/tratamento farmacológico , Receptores LHRH/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Estudos de Coortes , Feminino , Meia-Vida , Humanos , Masculino , Dose Máxima Tolerável , Neoplasias/metabolismo , Neoplasias/patologia , Receptores LHRH/administração & dosagem , Resultado do Tratamento
15.
JAAPA ; 26(7): 51-2, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23923287

RESUMO

GnRH receptor antagonists can reduce testosterone levels without the adverse reactions caused by other drugs used to treat prostate cancer. These drugs also offer hope for prolonged control of metastasis.


Assuntos
Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/uso terapêutico , Humanos , Masculino
16.
Acta méd. peru ; 28(4): 118-193, oct.-dic. 2011. tab, graf
Artigo em Espanhol | LILACS, LIPECS | ID: lil-645841

RESUMO

Objetivo: Evaluación farmacoeconómica de dos tratamientos con drogas de distinto mecanismo de acción: Degarelix y triptorelina en el manejo de pacientes con cáncer de próstata avanzado hormonodependiente. Material y método: Se realizó una revisión de la literatura sobre el tratamiento estándar de estos pacientes, efectos tempranos y tardíos de las terapias existentes y además una valoración de Costo Integral del Tratamiento usando el tarifario de Essalud. Resultados: El Costo Integral del Tratamiento, es S/ 10 793 para un paciente que usa Degarelix y S/ 12 251 para un paciente que usa triptorelina genérica; es decir, la terapia con el antagonista de la GnRH genera un ahorro de S/ 1 458 por paciente. Conclusiones: Este ahorro representa S/ 1 008 017 para el total de pacientes con cáncer de próstata avanzado hormonodependiente que se atienden en Essalud, a nivel nacional, con la ventaja adicional que Degarelix no genera costos adicionales por complicaciones producto del efecto Flare.


Objective: This is a pharmacoeconomic evaluation of two therapy schedules using drugs with different modes of action: Degarelix and triptorelin in the treatment of patients with advanced hormone-dependent prostate cancer. Methods: We reviewed the literature on the standard treatment for these patients, early and late effects of existing therapies, and we also performed a valuation using the Comprehensive Cost Treatment EsSalud (Peruvian Social Security) rates. Results:The Comprehensive Cost Treatment is S/. 10 793 for a patient using Degarelix and S/. 12 251 for a patient using generic triptorelin, so the therapy with the GnRH antagonist generates S/. 1 458 savings per patient. Conclusions: This represents S/. 1,008,017 savings for all patients with advanced hormone-dependent prostate who attend to EsSalud, with the added advantage that there are no extra costs with the use of Degarelix because of the absence of complications due to any flare effect.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Próstata , Neoplasias da Próstata/economia , Neoplasias da Próstata/terapia , Pamoato de Triptorrelina/uso terapêutico , Receptores LHRH/uso terapêutico , Testosterona
17.
Actas Fund. Puigvert ; 22(1): 18-23, ene. 2003. ilus
Artigo em Es | IBECS | ID: ibc-30505

RESUMO

La evolución a lo largo de 13 años de un adenocarcinoma de próstata, tratado con hormonoterapia, radioterapia y cirugía nos muestra el espectro fenotípico que puede adquirir. Esta observación se puede explicar a través del modelo histogénico del cáncer de próstata, a partir de la célula basal, que sería una auténtica stem cell, y justificaría la posibilidad de formas celulares intermedias (o en transformación) que tendrían su contrapartida neoplásica (AU)


Assuntos
Masculino , Pessoa de Meia-Idade , Humanos , Fenótipo , Neoplasias da Próstata/patologia , Adenocarcinoma/patologia , Receptores LHRH/uso terapêutico , Prostatectomia , Radioterapia , Transformação Celular Neoplásica/patologia , Neoplasias da Próstata/terapia
18.
Mol Cells ; 9(1): 31-6, 1999 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-10102568

RESUMO

Human prostate and breast tumor cells produce luteinizing hormone-releasing hormone (LHRH) receptors on their cell surface even when they have lost dependency on sex steroid hormones for growth. To investigate whether LHRH can be used as a cell-binding moiety to deliver toxin molecules into prostate and breast tumor cells, LHRH-bovine RNase A conjugates were constructed using the chemical cross-linking method. The treatment of the LHRH receptor-positive cells such as prostate LNCapFGC and breast MCF7 tumor cells with LHRH-RNase A conjugates resulted in a dose-dependent inhibition of growth. The cytotoxic activities of these conjugates were effectively reduced by the presence of exogenous LHRH. Either free RNase A or LHRH alone did not affect the proliferation of these cells. The LHRH-RNase A conjugates did not show cytotoxicity against FRTL5 and TM4 cells which do not express the LHRH receptors. These results suggest that LHRH can be used as a cell-binding molecule for the specific delivery of toxin molecules into the cells which express LHRH receptors on their surface. Thus, a new class of biomedicines that act as fusion proteins between LHRH and toxins will give us a new avenue for the treatment of human prostate and breast cancers, regardless of their steroid hormone dependency.


Assuntos
Neoplasias da Mama/metabolismo , Imunotoxinas/farmacologia , Neoplasias da Próstata/metabolismo , Ribonuclease Pancreático/farmacologia , Neoplasias da Mama/tratamento farmacológico , Relação Dose-Resposta a Droga , Hormônio Liberador de Gonadotropina/imunologia , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Imunotoxinas/uso terapêutico , Masculino , Especificidade de Órgãos , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/imunologia , Receptores LHRH/uso terapêutico , Ribonuclease Pancreático/uso terapêutico , Fatores de Tempo
19.
Obstet Gynecol Clin North Am ; 16(1): 105-22, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2546112

RESUMO

Hormonal therapy has long been the mainstay of the medical management of endometriosis. However, there is considerable confusion regarding the appropriate application of hormones in the treatment of endometriosis. This article critically reviews the current status of the steroidal therapy of endometriosis.


Assuntos
Endometriose/tratamento farmacológico , Hormônios Esteroides Gonadais/uso terapêutico , Cetosteroides/uso terapêutico , Androgênios/uso terapêutico , Danazol/uso terapêutico , Estrogênios/uso terapêutico , Feminino , Gestrinone/uso terapêutico , Humanos , Noretinodrel/uso terapêutico , Progesterona/uso terapêutico , Receptores LHRH/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...