Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
J Clin Endocrinol Metab ; 106(3): e1111-e1120, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33347565

ABSTRACT

OBJECTIVE: To evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of TU2670, a novel orally active, nonpeptide gonadotropin-releasing hormone (GnRH) antagonist administered to healthy female participants. METHODS: This was a first-in-human, multicenter, phase 1, randomized, double-blind, placebo-controlled, single-dose ascending trial that took place in multiple medical centers. A total of 16 healthy premenopausal women (23 to 45 years of age) were randomized and received 20, 40, 80, and 160 mg TU2670 (GnRH antagonist) or placebo 7 days (±1 day) after the onset of menstrual bleeding. We performed a noncompartmental analysis for pharmacokinetic parameters and calculated relative minimum concentration values (Cmin, % Baseline) of serum pharmacodynamic (PD) markers (luteinizing hormone [LH], follicle-stimulating hormone [FSH], and estradiol). RESULTS: There were no significant differences among treatments with respect to vital signs, electrocardiography, adverse events, ovulation test results, and ultrasonography. The median Tmax of TU2670 occurred 0.75 to 1.00 hours after dosing, and concentrations then declined, with a mean apparent half-life (t1/2) of 3.0 to 5.9 hours. AUClast (17.7-417.9 ng·h/mL) and Cmax (8.1-95.4 ng/mL) increased in a dose-dependent manner. The PD analysis after a single administration of TU2670 revealed dose-dependent suppression of LH, FSH, and estradiol. Maximal suppression of the pre-dose baseline (%) was 58% to 82% at 6 to 8 hours for LH, 28% to 39% at 6 to 12 hours for FSH, and 34% to 82% at 12 to 24 hours for estradiol. CONCLUSION: The single administration of TU2670 in healthy premenopausal women was well tolerated and resulted in the dose-dependent suppression of LH, FSH, and estradiol, suggesting rapid and significant inhibition of pituitary and ovarian hormones.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/administration & dosage , Organic Chemicals/administration & dosage , Administration, Oral , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Healthy Volunteers , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Luteinizing Hormone/blood , Organic Chemicals/adverse effects , Organic Chemicals/pharmacokinetics , Ovulation/drug effects , Premenopause/blood , Premenopause/drug effects , Republic of Korea , Young Adult
3.
J Clin Endocrinol Metab ; 105(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-32946574

ABSTRACT

CONTEXT: SJX-653 is a novel neurokinin 3 receptor (NK3R) antagonist. The NK3 pathway is a central regulator of gonadotropin releasing hormone (GnRH) secretion and has also been implicated in the generation of hot flashes. Therefore, decreases of luteinizing hormone (LH) and testosterone in men serve as sensitive pharmacodynamic (PD) markers of central NK3 antagonism. OBJECTIVE: To characterize the safety, tolerability, pharmacokinetics, and pharmacodynamic activity of SJX-653 in healthy men. DESIGN: A randomized, placebo-controlled, double-blind, single ascending dose study. SETTING: Phase 1 unit. PATIENTS OR OTHER PARTICIPANTS: Seven cohorts of 6 healthy men 18-45 years of age (4:2 randomization to SJX-653/placebo per cohort). INTERVENTION(S): Single oral doses of 0.5-90 mg SJX-653. MAIN OUTCOME MEASURE(S): Safety assessments and serial pharmacokinetic (PK)/PD measurements. RESULTS: SJX-653 was well tolerated at all dose levels. Cmax and AUC0-24 increased in a dose-proportional manner. The terminal elimination half-life ranged between 9.8 and 12.5 hours independent of dose. A statistically significant, dose-dependent, reversible reduction of LH and testosterone was observed with near maximal effect after 15 mg and little to no effect at 4.5 mg. Maximal LH reduction was 70 ±â€…7% (mean ±â€…sd) at 6 hours after 30 mg SJX-653 versus 10 ±â€…43% for placebo (P = 0.0006); maximal T reduction was of 68 ±â€…5% at 8 hours after 60 mg SJX-653 versus 18 ±â€…11% for placebo (P < 0.0001). The plasma IC50 for LH reduction was 33 ng/mL. CONCLUSIONS: These data demonstrate clinical proof-of-mechanism for SJX-653 as a potent centrally-acting NK3R antagonist.


Subject(s)
Hormone Antagonists/pharmacokinetics , Organic Chemicals/pharmacokinetics , Receptors, Neurokinin-3/antagonists & inhibitors , Adolescent , Adult , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Humans , Male , Middle Aged , Organic Chemicals/administration & dosage , Organic Chemicals/adverse effects , Young Adult
4.
Expert Opin Pharmacother ; 21(12): 1419-1430, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32401547

ABSTRACT

INTRODUCTION: Uterine myomas represents a widespread gynecological disease of women in reproductive age. Although surgery remains the first choice for treating most patients, in the last years, new medical approaches have been considered in order to ameliorate heavy menstrual bleeding (HMB) related to their presence. Elagolix is a second-generation gonadotropin-releasing hormone (GnRH) antagonist under investigation for the long-term treatment of uterine myomas. AREAS COVERED: The aim of this drug evaluation is to give a complete overview of pharmacokinetic and pharmacodynamic data on elagolix for treating HMB related to uterine myomas and to report the results of the current clinical trials in this setting. EXPERT OPINION: In two previous phase II studies, this drug succeeded in ameliorating blood loss and quality of life of patients affected by uterine myomas with a good safety profile. Three phase III trials (ELARIS UF-I, UF-II, and EXTEND) investigated the efficacy, tolerability, and safety of elagolix at 300 mg twice daily with add-back therapy. The primary endpoint, consisting in the reduction in HMB compared to placebo, was met in the majority of patients under treatment. Currently, elagolix is under investigation in two other ongoing multicenter phase III clinical studies.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Hydrocarbons, Fluorinated/therapeutic use , Leiomyoma/drug therapy , Pyrimidines/therapeutic use , Uterine Hemorrhage/drug therapy , Uterine Neoplasms/drug therapy , Clinical Trials as Topic , Female , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/adverse effects , Hydrocarbons, Fluorinated/pharmacokinetics , Leiomyoma/complications , Menorrhagia/drug therapy , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Quality of Life , Treatment Outcome , Uterine Hemorrhage/etiology , Uterine Neoplasms/complications
5.
Steroids ; 159: 108649, 2020 07.
Article in English | MEDLINE | ID: mdl-32389717

ABSTRACT

Mifepristone is one of potent anti-progesterone agents, which binds to progesterone receptors and glucocorticoid receptors. Until now, there are a lot of research focusing on enhancing the solubility and oral bioavailability of Mifepristone. However, poor solubility and oral bioavailability has some undesirable consequences. In this work, Mifepristone in form D was discovered for the first time and characterized by PXRD, TGA, DSC, FT-IR, SEM and SS NMR. Form D was a metastable crystal type which manifested favorable stability under ambient conditions. Form D had better dissolution characteristic compared with commercial Mifepristone in 0.5% SDS solution. In addition, Mifepristone in form D exhibited a 1.43-fold higher peak plasma concentration (Cmax) and 1.46-fold higher area under the curve (AUC) in rats. The work in this paper is a complement to the present understanding of drug polymorphism on the in vitro and in vivo behavior, and establishes the ground work for future development of Mifepristone in form D as a promising drug for the market.


Subject(s)
Hormone Antagonists/chemistry , Hormone Antagonists/pharmacokinetics , Mifepristone/chemistry , Mifepristone/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Female , Hormone Antagonists/administration & dosage , Mifepristone/administration & dosage , Molecular Conformation , Rats , Rats, Sprague-Dawley , Solubility
6.
Clin Pharmacokinet ; 59(3): 297-309, 2020 03.
Article in English | MEDLINE | ID: mdl-31749075

ABSTRACT

The clinical pharmacology of elagolix was extensively evaluated in clinical studies in healthy subjects and in women with endometriosis. Elagolix pharmacokinetics (PK) show significant population variability, however they are minimally affected by patients' baseline characteristics and demographics, except for clinically relevant extrinsic and intrinsic factors such as coadministrated strong organic anion transporting polypeptide (OATP) 1B1 inhibitors and severe hepatic impairment, which are contraindications for the use of elagolix. These studies enabled a comprehensive understanding of elagolix mechanism of action and the downstream pharmacodynamic (PD) effects on gonadotropin and ovarian hormones, as well as full characterization of the PK/PD (PKPD) relationships of elagolix at various dosages, including the approved 150 mg once daily and 200 mg twice daily dosing regimens for the management of moderate to severe pain associated with endometriosis. Several model-based analyses have contributed to understanding of the benefit-risk profile of elagolix in patients with endometriosis, through characterization of the exposure relationship with responder rates, with changes in bone mineral density over time, as well as the interaction with coadministered drugs. Collectively, these studies and analyses served as supportive evidence for the effectiveness of the approved dosages and provided general dosing instructions of the first approved oral gonadotropin-releasing hormone receptor antagonist.


Subject(s)
Endometriosis/drug therapy , Hormone Antagonists/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Organic Anion Transporters/antagonists & inhibitors , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , Administration, Oral , Bone Density/drug effects , Drug Interactions/physiology , Endometriosis/complications , Endometriosis/metabolism , Female , Gonadotropin-Releasing Hormone/drug effects , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacology , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacology , Liver Diseases/complications , Organic Anion Transporters/metabolism , Pain/drug therapy , Pain/etiology , Pharmacogenetics , Pharmacology, Clinical , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Treatment Outcome
7.
Drugs ; 79(6): 675-679, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30937733

ABSTRACT

The orally active nonpeptide gonadotropin-releasing hormone (GnRH)-receptor antagonist relugolix (Relumina) is being developed by Takeda and ASKA Pharmaceutical as a treatment for various sex hormone related disorders. Relugolix was recently approved for marketing in Japan as a treatment for symptoms associated with uterine fibroids, and studies evaluating the efficacy of the drug as treatment for endometriosis-associated pain and prostate cancer are currently underway. This article summarizes the milestones in the development of relugolix leading to this first approval for the treatment of symptoms associated with uterine fibroids.


Subject(s)
Hormone Antagonists/therapeutic use , Leiomyoma/drug therapy , Phenylurea Compounds/therapeutic use , Pyrimidinones/therapeutic use , Receptors, LHRH/antagonists & inhibitors , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Drug Approval , Endometriosis/drug therapy , Female , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Japan , Male , Middle Aged , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Phenylurea Compounds/pharmacokinetics , Prostatic Neoplasms/drug therapy , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Treatment Outcome
8.
J Pharm Biomed Anal ; 164: 725-733, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30472591

ABSTRACT

Cligosiban is a highly-affinity nonpeptide oxytocin receptor antagonist. In this study, a simple an sensitive LC-MS/MS method was developed and validated for the determination of cligosiban in rat plasma. The plasma samples were pretreated with acetonitrile as precipitant and then separated on an ACQUITY BEH C18 column (2.1 × 50 mm, 1.7 µm) with 0.1% formic acid in water and acetonitrile as mobile phase. The analytes were monitored using selected reaction monitoring (SRM) mode with transitions at m/z 420.1→248.1 for cligosiban and m/z 304.1→161.1 for IS. The developed method showed good linearity over the concentration range of 1-1000 ng/mL with coefficient of correlation > 0.996. The lower limit of quantification (LLOQ) is 1 ng/mL. The method was validated for selectivity, precision, accuracy, recovery, and stability in accordance with FDA's guidance. The validated assay has been successfully applied to the pharmacokinetic study of cligosiban in rat plasma after intravenous and oral administration. According to the current results, the oral bioavailability of cligosiban was 63.82%. Furthermore, the metabolites present in rat liver microsomes (RLM), human liver microsomes (HLM) and rat plasma were analyzed by UHPLC-LTQ-Orbitrap-MS method, and four metabolites structurally identified based on their accurate masses, and fragment ions. The proposed metabolic pathways of cligosiban were demethylation and glucuronidation. This study is the first report on the pharmacokinetic and metabolic information of cligosiban, which would provide insights into the effectiveness and toxicity of cligosiban.


Subject(s)
Hormone Antagonists/pharmacokinetics , Pyridines/pharmacokinetics , Receptors, Oxytocin/antagonists & inhibitors , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Triazoles/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid/methods , Demethylation , Hormone Antagonists/administration & dosage , Hormone Antagonists/blood , Humans , Male , Microsomes, Liver , Pyridines/administration & dosage , Pyridines/blood , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Triazoles/administration & dosage , Triazoles/blood
9.
Acta Obstet Gynecol Scand ; 98(2): 250-261, 2019 02.
Article in English | MEDLINE | ID: mdl-30325501

ABSTRACT

INTRODUCTION: Women with hereditary mutation in breast cancer-associated genes (BRCA1-/2- ) have a higher lifetime risk of developing ovarian cancer. Here, we aimed to investigate the effect of mifepristone, a selective progesterone receptor modulator of ovarian mesenchymal stem/stromal cells (MSC) from BRCA1-/2- carriers. MATERIAL AND METHODS: Ovarian BRCA1-/2- MSC were positively selected using the markers CD90, CD73 and CD105 from nine healthy women. The effect of dose response and combination treatment with mifepristone and analogs of progesterone- or glucocorticoid-receptors were investigated on BRCA1-/2- MSC in vitro using a panel of markers for proliferation (ki67, BrdU, CDK2, p21CIP ), apoptosis (BAX, BCL2, CASPASE3), tumor suppression (TP53, PTEN) and cell survival (PI3KCA, MAPK3, mTOR). RESULTS: The dose response with mifepristone treatment suggested an optimal effect with 10 µm mifepristone, exhibiting >90% viability and significantly reducing growth signaling markers (TP53 and MAPK3). Furthermore, combined treatment with progesterone plus mifepristone (PG+MIFE) gave an enhanced anti-proliferative effect in comparison with hydrocortisone plus mifepristone (HC+MIFE) by significantly reducing markers of proliferation (BrdU+ and Ki67 expression) and tumor suppressors (PTEN, TP53), and increasing the percentage of pro-apoptotic cells. Consequently, accumulation of p21CIP together with reduced levels of CDK2 confirms growth inhibition by reversibly arresting cell-cycle progression at the G1-S phase, not by inducing apoptosis. CONCLUSIONS: Our study showed an anti-proliferative effect on ovarian BRCA1-/2- MSC on in vitro combined treatment with mifepristone and progesterone. These findings suggest that mifepristone or other selective progesterone receptor modulators could be developed as a preventive treatment and postpone early use of prophylactic salpingo-oophorectomy as well as reduce the risk of ovarian cancer.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cell Proliferation/drug effects , Mesenchymal Stem Cells , Mifepristone/pharmacology , Ovary , Stromal Cells , Cell Survival/drug effects , Female , Hormone Antagonists/pharmacokinetics , Hormone Antagonists/pharmacology , Humans , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Stromal Cells/pathology , Stromal Cells/physiology , Tumor Cells, Cultured
10.
J Clin Endocrinol Metab ; 104(3): 883-891, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30371791

ABSTRACT

Context: A somatostatin-dopamine chimera (BIM23B065) was under investigation to reduce GH secretion for the treatment of pituitary adenomas. Objective: To determine pharmacokinetics, safety, and tolerability and to monitor hormonal changes after single and multiple subcutaneous BIM23B065 administrations. Design: Randomized, double-blind, placebo-controlled, parallel-group design with five single and three 13-day multiple ascending-dose cohorts. Patients: A total of 63 healthy male white volunteers were enrolled (47 active, 16 placebo). Main Outcome Measures: Pharmacokinetics, GH, prolactin (PRL), IGF-1, GH after GHRH administration, and general clinical safety criteria. Results: The maximum dosage of BIM23B065 administered in this study was 1.5 mg. BIM23B065 reduced the mean GH concentrations after 8 and 13 days of treatment. A decrease in GH release after GHRH administration indicated inhibition of the hypothalamic-pituitary-somatotropic axis. IGF-1 was not altered after single doses but showed a significant change from baseline after multiple dosing. PRL secretion was reduced in all subjects who were treated. Orthostatic hypotension and injection site reactions were commonly observed at high dosages. A 6-day uptitration period was included to successfully lower the cardiovascular effects in the multiple ascending dose part of the study. Conclusions: Proof of pharmacology of BIM23B065 was shown by a reduction in GH, IGF-1, and PRL concentrations in healthy male volunteers, supporting activity of the somatostatin analog and dopamine agonist moieties. The safety and tolerability of the higher dosing regions was limited mainly by orthostatic hypotension.


Subject(s)
Dopamine/administration & dosage , Hormone Antagonists/administration & dosage , Human Growth Hormone/antagonists & inhibitors , Pituitary Gland/drug effects , Somatostatin/administration & dosage , Adolescent , Adult , Dopamine/adverse effects , Dopamine/analogs & derivatives , Dopamine/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Healthy Volunteers , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Human Growth Hormone/blood , Human Growth Hormone/metabolism , Humans , Injections, Subcutaneous , Insulin-Like Growth Factor I/analysis , Male , Middle Aged , Pituitary Gland/metabolism , Pituitary Neoplasms/drug therapy , Prolactin/blood , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Somatostatin/pharmacokinetics , Young Adult
11.
Clin Pharmacol Drug Dev ; 8(8): 1053-1061, 2019 11.
Article in English | MEDLINE | ID: mdl-30570832

ABSTRACT

The aim of these studies was to assess the safety and pharmacokinetics of elagolix, an oral nonpeptide gonadotropin-releasing hormone antagonist following oral administration in women with renal or hepatic impairment. Two phase 1 studies were conducted in adult women with normal renal function versus renal impairment (reduced study), and normal hepatic function versus hepatic impairment (full study design). All women received a single dose of elagolix 200 mg (renal) or 150 mg (hepatic). Intensive pharmacokinetic blood samples were collected. Elagolix exposures were comparable in women with normal renal function and those with moderate/severe renal impairment or end-stage renal disease. Elagolix exposures also appeared to be similar in women with normal hepatic function and women with mild hepatic impairment. Elagolix area under the curve in women with moderate hepatic impairment and with severe hepatic impairment was approximately 3-fold and 7-fold higher than in women with normal hepatic function. The adverse event incidence was low, with the main events being mild nausea and headache. No dosage adjustment was needed in women with renal impairment or women with mild hepatic impairment. Although an elagolix dose of 150 mg once daily may be used in women with moderate hepatic impairment for up to 6 months, this elagolix dose should not be used in women with severe hepatic impairment.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Kidney Diseases/blood , Liver Diseases/blood , Pyrimidines/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Dose-Response Relationship, Drug , Female , Glomerular Filtration Rate , Hormone Antagonists/administration & dosage , Hormone Antagonists/adverse effects , Hormone Antagonists/blood , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/adverse effects , Hydrocarbons, Fluorinated/blood , Liver Function Tests , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/blood , Young Adult
12.
Drugs ; 78(14): 1501-1508, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30194661

ABSTRACT

Elagolix (ORILISSA™), an orally bioavailable, second-generation, non-peptide gonadotropin-releasing hormone (GnRH) receptor antagonist, is being developed AbbVie and Neurocrine Biosciences for the treatment of reproductive hormone-dependent disorders in women. In July 2018, the US FDA approved elagolix tablets for the management of moderate to severe pain associated with endometriosis. This approval was based on positive results in two replicate phase III trials; additional phase III trials in the USA, Canada and Puerto Rico are currently evaluating elagolix as both monotherapy and in combination with low-dose hormone add-back therapy in the same indication. Elagolix with and without low-dose hormone add-back therapy is also undergoing phase III clinical development for heavy menstrual bleeding associated with uterine fibroids in the aforementioned locations. This article summarizes the milestones in the development of elagolix leading to its first approval for the management of moderate to severe pain associated with endometriosis.


Subject(s)
Hormone Antagonists/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Pain/drug therapy , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , Drug Approval , Drug Therapy, Combination , Endometriosis/physiopathology , Female , Hormone Antagonists/administration & dosage , Hormone Antagonists/therapeutic use , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/therapeutic use , Pain Management/methods , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Treatment Outcome , United States , United States Food and Drug Administration
13.
J Med Chem ; 61(17): 7767-7784, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30091920

ABSTRACT

The glucocorticoid receptor (GR) has been linked to therapy resistance across a wide range of cancer types. Preclinical data suggest that antagonists of this nuclear receptor may enhance the activity of anticancer therapy. The first-generation GR antagonist mifepristone is currently undergoing clinical evaluation in various oncology settings. Structure-based modification of mifepristone led to the discovery of ORIC-101 (28), a highly potent steroidal GR antagonist with reduced androgen receptor (AR) agonistic activity amenable for dosing in androgen receptor positive tumors and with improved CYP2C8 and CYP2C9 inhibition profile to minimize drug-drug interaction potential. Unlike mifepristone, 28 could be codosed with chemotherapeutic agents readily metabolized by CYP2C8 such as paclitaxel. Furthermore, 28 demonstrated in vivo antitumor activity by enhancing response to chemotherapy in the GR+ OVCAR5 ovarian cancer xenograft model. Clinical evaluation of safety and therapeutic potential of 28 is underway.


Subject(s)
Drug Discovery , Hormone Antagonists/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Female , Hormone Antagonists/chemistry , Hormone Antagonists/pharmacokinetics , Humans , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rats , Swine , Swine, Miniature , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
J Pharm Biomed Anal ; 154: 108-115, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29544105

ABSTRACT

Mifepristone (RU486) is developed originally as a contraceptive used by hundreds of millions of women world-wide, and also reported as a safe and long-term psychotic depressant, or as a cancer chemotherapeutic agent used by both sexes. In our preliminary study aimed at developing mifepristone as a cancer metastatic chemopreventive, we coincidentally observed that blood mifepristone concentrations in female rats seem to be higher than those in male ones post administration. To substantiate if the pharmacokinetic differences between sexes exist, we established a fast UPLC-MS/MS method to determine mifepristone concentrations in plasma, and analyzed blood concentrations of mifepristone over time in rats and dogs of both sexes. Mifepristone in plasma or incubation liquid was recovered by liquid-liquid extraction using 1 mL of ethyl acetate. Chromatographic separation was performed on a C18 column at 35 °C, with a gradient elution consisting of methanol and water containing 0.1% (v/v) formic acid at a flow rate of 0.3 mL/min. And pharmacokinetic parameters such as elimination half-life, and mean residence time were calculated by using the non-compartmental pharmacokinetics data analysis software. In this work, administrations of mifepristone to rats and beagle dogs revealed that the plasma concentrations of mifepristone (AUC, Cmax) were significantly higher (P < 0.05) in females than that in males. In vitro liver microsomal incubation experiments showed that the metabolic rate of mifepristone in males was higher than that in females, which was consistent with the results of in vivo experiments. In general, we first found the sex-related differences about pharmacokinetic properties of mifepristone and revealed the metabolism difference of hepatic microsomal enzyme is the main reason.


Subject(s)
Antipsychotic Agents/pharmacokinetics , Hormone Antagonists/pharmacokinetics , Mifepristone/pharmacokinetics , Animals , Antipsychotic Agents/metabolism , Chromatography, High Pressure Liquid/methods , Dogs , Female , Half-Life , Hormone Antagonists/metabolism , Male , Microsomes, Liver , Mifepristone/metabolism , Models, Animal , Rats , Rats, Sprague-Dawley , Sex Factors , Tandem Mass Spectrometry/methods
15.
Sci Rep ; 7(1): 17190, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215040

ABSTRACT

Metapristone is the primary metabolite of the abortifacient mifepristone (RU486), and is being developed as a safe and effective cancer metastatic chemopreventive agent for both sexes. Here, we systematically investigated the sex-related pharmacokinetics of metapristone in both rats and dogs, and explored the related mechanisms of actions. Administration of metapristone to rats and dogs showed that plasma concentrations of metapristone (AUC, C max ) were significantly higher in female dogs and rats than in males. The sex-related differences in pharmacokinetics become more significant after ten consecutive days of oral administration. Female liver microsomes metabolized metapristone significantly slower than the male ones. The results from P450 reaction phenotyping using recombinant cDNA-expressed human CYPs in conjunction with specific CYP inhibitors suggested that CYP1A2 and CYP3A4 are the predominant CYPs involved in the metapristone metabolism, which were further confirmed by the enhanced protein levels of CYP1A2 and CYP3A4 induced by 1-week oral administration of metapristone to rats. The highest tissue concentration of metapristone was found in the liver. The study demonstrates, for the first time, the sex-related pharmacokinetics of metapristone, and reveals that activities of liver microsomal CYP1A2 and CYP3A4 as well as the renal clearance are primarily responsible for the sex-related pharmacokinetics.


Subject(s)
Hormone Antagonists/pharmacokinetics , Microsomes, Liver/metabolism , Mifepristone/pharmacokinetics , Animals , Cell Proliferation , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Dogs , Female , Hormone Antagonists/pharmacology , Male , Microsomes, Liver/drug effects , Mifepristone/pharmacology , Rats , Rats, Sprague-Dawley , Sex Factors , Tissue Distribution
16.
J Clin Endocrinol Metab ; 101(2): 417-26, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26653113

ABSTRACT

CONTEXT: Women's health disorders are commonly treated by agents that suppress the hypothalamic-pituitary-gonadal axis. NK3 receptor antagonism modulates this axis with distinct pharmacology compared to existing therapies. OBJECTIVE: The study aim was to evaluate safety, pharmacokinetics, and pharmacodynamics on gonadotropins and sex hormones after single- and multiple-dose administration of an NK3R antagonist to healthy men and women. DESIGN AND SETTING: This was a first-in-human, double-blind, placebo-controlled, combined single and multiple ascending dose trial. PARTICIPANTS: Forty-one men and 24 regularly cycling women participated in the study. INTERVENTION(S): In part 1 of the study, men received single oral doses of 3-180 mg or placebo. In part 2, men received placebo or 20, 60, or 180 mg each day for 10 days. In part 3, women received placebo or 20, 60, or 180 mg each day for 21 days, where dosing was initiated on day 3 ± 2 after menses. MAIN OUTCOME MEASURE(S): Safety, tolerability, pharmacokinetics, and pharmacodynamics on circulating levels of LH, FSH, testosterone, estradiol, and progesterone, in addition to physiological biomarkers of endometrial thickening, follicle growth, and the duration of the menstrual cycle were evaluated. RESULTS: ESN364 was well-tolerated and rapidly bioavailable with linear pharmacokinetics and no drug accumulation with repeated, daily oral administration. Drug treatment dose-dependently decreased basal LH, but not FSH, and consequently decreased estradiol and progesterone (in women) as well as testosterone (in men). The hormonal changes in women corresponded to delayed ovulation, decreased endometrial thickening, impeded follicular maturation, and prolongation of the menstrual cycle. Drug effects were rapidly reversible. CONCLUSIONS: Oral administration of the NK3R antagonist, ESN364, suppressed the hypothalamic-pituitary-gonadal axis in healthy volunteers by selective modulation of gonadotropin secretion, leading to a restrained decrease in ovarian hormone levels in women. These results suggest that ESN364 may offer therapeutic benefit in the treatment of women's health disorders with a mitigated risk of menopausal-like adverse events.


Subject(s)
Gonadal Steroid Hormones/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Hormone Antagonists/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Thiadiazoles/pharmacology , Adolescent , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Endometrium/drug effects , Female , Gonadal Steroid Hormones/metabolism , Gonadotropins/blood , Gonads/drug effects , Heterocyclic Compounds, 2-Ring/adverse effects , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Hormone Antagonists/adverse effects , Hormone Antagonists/pharmacokinetics , Humans , Hypothalamo-Hypophyseal System/drug effects , Male , Menstrual Cycle/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Ovulation/drug effects , Thiadiazoles/adverse effects , Thiadiazoles/pharmacokinetics , Young Adult
17.
Clin Pharmacol Drug Dev ; 4(6): 418-26, 2015 11.
Article in English | MEDLINE | ID: mdl-27137713

ABSTRACT

An open-label single- and repeat-dose study was conducted to investigate the pharmacokinetics, safety, and tolerability of ascending doses of epelsiban in healthy female volunteers (n = 48). The pharmacokinetics of the epelsiban metabolite, GSK2395448, were also assessed. Epelsiban was readily absorbed and parent and metabolite readily appeared in plasma. The parent drug's median tmax was approximately 0.5 hours, and the metabolite's median tmax ranged from 0.5 to 1.0 hours post-parent dosing. Both epelsiban and GSK2395448 had rapid elimination half-lives, ranging between 2.66 and 4.85 hours. The metabolite:parent ratios for exposure (AUC and Cmax ) ranged from approximately 70% to greater than 100%, and therefore, GSK2395448 is considered a major metabolite of epelsiban. Mean epelsiban and GSK2395448 AUC values increased in a dose-proportional manner following both single-dose administration from 10 to 200 mg and repeat administration from 10 to 150 mg following twice daily or 4-times-daily dosing. Single-dose epelsiban pharmacokinetics in women was similar to single-dose pharmacokinetics previously observed in men. Epelsiban was generally well tolerated, and no events of clinical concern were observed in volunteers dosed in this study. The safety findings were consistent with the previous study in men, with headache the most commonly reported adverse effect.


Subject(s)
Diketopiperazines/administration & dosage , Diketopiperazines/pharmacokinetics , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacokinetics , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Adolescent , Adult , Area Under Curve , Baltimore , Biotransformation , Diketopiperazines/adverse effects , Diketopiperazines/blood , Drug Administration Schedule , Female , Half-Life , Healthy Volunteers , Hormone Antagonists/adverse effects , Hormone Antagonists/blood , Humans , Metabolic Clearance Rate , Middle Aged , Models, Biological , Morpholines/adverse effects , Morpholines/blood , Young Adult
18.
Curr Pharm Biotechnol ; 16(2): 187-93, 2015.
Article in English | MEDLINE | ID: mdl-25391244

ABSTRACT

In this study, sustained-release of GnRH antagonist peptide LXT-101 was realized through oil formulation, and their releasing characteristics in vitro and in vivo were investigated. In this formulation, the static interaction between cationic charged peptide LXT-101 and the negative charged phospholipid led to the formation of the phospholipid-peptide complex, by which LXT-101 was completely dissolved in oils. This formulation was prepared by mixing an aqueous solution of LXT-101 and empty SUV (small unilamellar liposomes) containing EPC (phosphatidylcholine) and DPPG (1, 2-dipalmitog-sn-glycero-3- phosphoglycerol) at an appropriate ratio, the mixture was subsequently lyophilized, and the resultant was dissolved in the oil to form a clear oily solution containing solubilized peptide LXT-101. With atomic force microscopy combined with Langmuir-Blodgett technology, the morphology of the particles in the oily solution were examined to be oval-shaped and the mean particle size was 150 nm in diameter. In pure water at 37°C, about 70~90 % of LXT-101 was released slowly from the oily formulation over 7 days. An effective sustained suppression of testosterone in beagle dogs could be achieved over a period of seven days with this LXT-101 oily formulation, by i.m. at a dose of 0.2 mg/kg (2 mg/ml). This formulation dramatically improved the bioactivity of LXT-101 compared to its aqueous solution. It was also found that when the concentration of peptide LXT-101 was up to or over 10 mg/ml in aqueous solution, there was no significant difference between the oily formulation and aqueous solution. This fact meant that LXT-101 itself could conduct sustained release in vivo by self-assembly of nanofibers.


Subject(s)
Hormone Antagonists/administration & dosage , Oligopeptides/administration & dosage , Animals , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Dogs , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/blood , Hormone Antagonists/chemistry , Hormone Antagonists/pharmacokinetics , Injections , Liposomes , Male , Oleic Acid/chemistry , Oligopeptides/blood , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Phosphatidylcholines/chemistry , Phosphatidylglycerols/chemistry , Soybean Oil/chemistry , Testosterone/blood , Triglycerides/chemistry , Water/chemistry
19.
J Pept Sci ; 20(11): 868-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25053524

ABSTRACT

It is well known that GnRH analogs can self-assemble into amyloid fibrils and that the duration of action of GnRH analogs depends on the ability of the amyloid to slowly release active peptides. The aim of this study was to investigate the influence of the amino acid residues at position 7 of GnRH analogues on peptide self-assembly. It was found that the dominant shape of the nanostructure can be changed when the structures of the residues at position 7 differ significantly from that of leucine in Degarelix. When the backbone length was extended (peptide 9), or the side chain of the residue at position 7 was replaced by an aromatic ring (peptide 6), or the rotation of the amide bond was restricted (peptide 8), the nanostructure changed from fibrils to vesicles. The results also indicate that the increasing hydrophilicity had little influence on the nanostructure morphology. In addition, a suitable release rate was found to play a more important role for the duration of the peptide action by maintaining the equilibrium between the drug concentration and the persistent release time, while the nanostructure shape was found to exert little influence on the duration of the peptide action.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Amino Acid Sequence , Animals , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacokinetics , Hormone Antagonists/chemistry , Hormone Antagonists/pharmacokinetics , Humans , Hydrophobic and Hydrophilic Interactions , Male , Microscopy, Electron, Transmission , Nanostructures/chemistry , Nanostructures/ultrastructure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Prostatic Neoplasms/drug therapy , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
20.
Am J Hum Genet ; 94(4): 533-46, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24680886

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a severe hypomyelinating disease, characterized by ataxia, intellectual disability, epilepsy, and premature death. In the majority of cases, PMD is caused by duplication of PLP1 that is expressed in myelinating oligodendrocytes. Despite detailed knowledge of PLP1, there is presently no curative therapy for PMD. We used a Plp1 transgenic PMD mouse model to test the therapeutic effect of Lonaprisan, an antagonist of the nuclear progesterone receptor, in lowering Plp1 mRNA overexpression. We applied placebo-controlled Lonaprisan therapy to PMD mice for 10 weeks and performed the grid slip analysis to assess the clinical phenotype. Additionally, mRNA expression and protein accumulation as well as histological analysis of the central nervous system were performed. Although Plp1 mRNA levels are increased 1.8-fold in PMD mice compared to wild-type controls, daily Lonaprisan treatment reduced overexpression at the RNA level to about 1.5-fold, which was sufficient to significantly improve the poor motor phenotype. Electron microscopy confirmed a 25% increase in the number of myelinated axons in the corticospinal tract when compared to untreated PMD mice. Microarray analysis revealed the upregulation of proapoptotic genes in PMD mice that could be partially rescued by Lonaprisan treatment, which also reduced microgliosis, astrogliosis, and lymphocyte infiltration.


Subject(s)
Estrenes/therapeutic use , Hormone Antagonists/therapeutic use , Pelizaeus-Merzbacher Disease/drug therapy , Progesterone/antagonists & inhibitors , Animals , Disease Models, Animal , Estrenes/pharmacokinetics , Estrenes/pharmacology , Gene Expression Regulation/drug effects , Hormone Antagonists/pharmacokinetics , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Phenotype , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL