Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Clin Pharmacol Drug Dev ; 13(6): 601-610, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593267

ABSTRACT

Fixed-dose combination (FDC) therapies can enhance patient convenience and adherence to prescribed treatment regimens. Elagolix is a novel oral gonadotropin-releasing hormone receptor antagonist approved for management of moderate to severe pain associated with endometriosis and heavy menstrual bleeding associated with uterine fibroids. Hormonal add-back therapy can attenuate the reversible hypoestrogenic effects of elagolix. An FDC formulation containing elagolix/estradiol (E2)/norethindrone acetate (NETA) 300/1/0.5 mg as the morning dose and an elagolix 300 mg capsule as the evening dose, were evaluated in 2 bioequivalence studies including the effects of food. Study 1 in premenopausal women assessed the bioavailability of the elagolix 300-mg capsule relative to the commercially available elagolix 300-mg tablet. Study 2 in postmenopausal women, elagolix/E2/NETA (300 mg/1 mg/0.5 mg) FDC capsule was assessed relative to the elagolix 300-mg tablet coadministered with E2/NETA 1-mg/0.5-mg tablet, the regimen that was studied in Phase 3 uterine fibroid studies. Under fasting conditions, the test elagolix 300-mg capsule was bioequivalent to the reference elagolix 300-mg tablet. Under fasting conditions, the elagolix/E2/NETA FDC capsule was bioequivalent to the coadministered elagolix 300-mg tablet and E2/NETA 1/0.5-mg tablet. Following administration of elagolix/E2/NETA FDC capsule after a high-fat breakfast, elagolix mean maximum concentration (Cmax) and area under the plasma concentration-time curve (AUC) were 38% and 28% lower, relative to fasting conditions. NETA mean Cmax was 51% lower and AUC from time 0 to infinity was 20% higher, while baseline-adjusted total estrone mean Cmax and AUC were 46% and 14% lower, respectively. No safety concerns were identified. These results enabled bridging the elagolix/E2/NETA FDC capsule.


Subject(s)
Drug Combinations , Estradiol , Hydrocarbons, Fluorinated , Norethindrone Acetate , Postmenopause , Premenopause , Pyrimidines , Therapeutic Equivalency , Humans , Female , Estradiol/pharmacokinetics , Estradiol/administration & dosage , Estradiol/adverse effects , Adult , Middle Aged , Norethindrone Acetate/administration & dosage , Pyrimidines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/adverse effects , Cross-Over Studies , Capsules , Area Under Curve , Biological Availability , Young Adult , Norethindrone/administration & dosage , Norethindrone/pharmacokinetics , Norethindrone/adverse effects , Administration, Oral , Double-Blind Method
3.
CPT Pharmacometrics Syst Pharmacol ; 10(1): 40-47, 2021 01.
Article in English | MEDLINE | ID: mdl-33200566

ABSTRACT

Selection of a personalized dose for an individual patient can be informed by the patient's preferences, translated as weights on each of the clinically relevant safety and efficacy drug attributes, based on results from a brief patient preference elicitation questionnaire. In this analysis, the weighted attributes were simulated to represent various endometriosis patient profiles. Exposure-response simulations were performed for elagolix, a drug approved for management of moderate to severe pain associated with endometriosis, across a range of plasma exposures corresponding to a range of doses. The results were combined to calculate a personalized clinical utility index. An interactive user-friendly online application was developed and envisioned as a physician's desk tool to personalize the dose selection process based on individual patient preferences. This demonstration should serve as an example of how patient/physician conversation can be facilitated with quantitative tools for personalizing the dose.


Subject(s)
Analgesics/administration & dosage , Endometriosis/drug therapy , Hydrocarbons, Fluorinated/administration & dosage , Models, Biological , Pain/drug therapy , Patient Preference , Pyrimidines/administration & dosage , Adolescent , Adult , Analgesics/blood , Analgesics/pharmacokinetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Computer Simulation , Dose-Response Relationship, Drug , Endometriosis/complications , Endometriosis/metabolism , Female , Humans , Hydrocarbons, Fluorinated/blood , Hydrocarbons, Fluorinated/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/genetics , Middle Aged , Pain/blood , Pain/etiology , Precision Medicine , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Surveys and Questionnaires , Young Adult
4.
J Clin Pharmacol ; 60(12): 1606-1616, 2020 12.
Article in English | MEDLINE | ID: mdl-33045114

ABSTRACT

Elagolix is an oral gonadotropin-releasing hormone receptor antagonist indicated for the management of endometriosis-associated pain and in combination with estradiol/norethindrone acetate indicated for the management of heavy menstrual bleeding associated with uterine leiomyomas (fibroids) in premenopausal women. Elagolix coadministered with estradiol/norethindrone acetate is in late-stage development for the management of heavy menstrual bleeding associated with uterine fibroids. Based on the in vitro profile of elagolix metabolism and disposition, 9 drug-drug interaction (DDI) studies evaluating the victim and perpetrator characteristics of elagolix were conducted in 144 healthy volunteers. As a victim of cytochrome P450 (CYPs) and transporter-mediated DDIs, elagolix area under the curve (AUC) increased by ∼2-fold following coadministration with ketoconazole and by ∼5- and ∼2-fold with single and multiple doses of rifampin, respectively. As a perpetrator, elagolix decreased midazolam AUC (90% confidence interval) by 54% (50%-59%) and increased digoxin AUC by 32% (23%-41%). Elagolix decreased rosuvastatin AUC by 40% (29%-50%). No clinically significant changes in exposure on coadministration with sertraline or fluconazole occurred. A elagolix 150-mg once-daily regimen should be limited to 6 months with strong CYP3A inhibitors and rifampin because of the potential increase in bone mineral density loss, as described in the drug label. A 200-mg twice-daily regimen is recommended for no more than 1 month with strong CYP3A inhibitors and not recommended with rifampin. Elagolix is contraindicated with strong organic anion transporter polypeptide B1 inhibitors (eg, cyclosporine and gemfibrozil). Consider increasing the doses of midazolam and rosuvastatin when coadministered with elagolix, and individualize therapy based on patient response. Clinical monitoring is recommended for P-glycoprotein substrates with a narrow therapeutic window (eg, digoxin). Dose adjustments are not required for sertraline, fluconazole, bupropion (or any CYP2B6 substrate), or elagolix when coadministered.


Subject(s)
Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2B6 Inducers/administration & dosage , Cytochrome P-450 CYP2B6 Inducers/pharmacokinetics , Cytochrome P-450 CYP2C9 Inhibitors/administration & dosage , Cytochrome P-450 CYP2C9 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Administration Schedule , Drug Interactions , Female , Healthy Volunteers , Humans , Hydrocarbons, Fluorinated/blood , Hydrocarbons, Fluorinated/pharmacology , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Premenopause , Pyrimidines/blood , Pyrimidines/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Young Adult
5.
Theranostics ; 10(23): 10548-10562, 2020.
Article in English | MEDLINE | ID: mdl-32929365

ABSTRACT

The use of reporter genes to non-invasively image molecular processes inside cells has significant translational potential, particularly in the context of systemically administered gene therapy vectors and adoptively administered cells such as immune or stem cell based therapies. Bacterial nitroreductase enzymes possess ideal properties for reporter gene imaging applications, being of non-human origin and possessing the ability to metabolize a range of clinically relevant nitro(hetero)cyclic substrates. Methods: A library of eleven Escherichia coli nitroreductase candidates were screened for the ability to efficiently metabolize 2-nitroimidazole based positron emission tomography (PET) probes originally developed as radiotracers for hypoxic cell imaging. Several complementary methods were utilized to detect formation of cell-entrapped metabolites, including various in vitro and in vivo models to establish the capacity of the 2-nitroimidazole PET agent EF5 to quantify expression of a nitroreductase candidate. Proof-of-principle PET imaging studies were successfully conducted using 18F-HX4. Results: Recombinant enzyme kinetics, bacterial SOS reporter assays, anti-proliferative assays and flow cytometry approaches collectively identified the major oxygen-insensitive nitroreductase NfsA from E. coli (NfsA_Ec) as the most promising nitroreductase reporter gene. Cells expressing NfsA_Ec were demonstrably labelled with the imaging agent EF5 in a manner that was quantitatively superior to hypoxia, in monolayers (2D), multicellular layers (3D), and in human tumor xenograft models. EF5 retention correlated with NfsA_Ec positive cell density over a range of EF5 concentrations in 3D in vitro models and in xenografts in vivo and was predictive of in vivo anti-tumor activity of the cytotoxic prodrug PR-104. Following PET imaging with 18F-HX4, a significantly higher tumor-to-blood ratio was observed in two xenograft models for NfsA_Ec expressing tumors compared to the parental tumors thereof, providing verification of this reporter gene imaging approach. Conclusion: This study establishes that the bacterial nitroreductase NfsA_Ec can be utilized as an imaging capable reporter gene, with the ability to metabolize and trap 2-nitroimidazole PET imaging agents for non-invasive imaging of gene expression.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Escherichia coli Proteins/administration & dosage , Genes, Reporter , Neoplasms/diagnostic imaging , Nitroreductases/administration & dosage , Positron-Emission Tomography/methods , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Drug Resistance, Neoplasm , Escherichia coli Proteins/genetics , Etanidazole/administration & dosage , Etanidazole/analogs & derivatives , Etanidazole/pharmacokinetics , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacokinetics , HCT116 Cells , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacokinetics , Imidazoles/administration & dosage , Indicators and Reagents/administration & dosage , Indicators and Reagents/pharmacokinetics , Mice , Molecular Imaging/methods , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Nitrogen Mustard Compounds/pharmacology , Nitrogen Mustard Compounds/therapeutic use , Nitroreductases/genetics , Precision Medicine/methods , Proof of Concept Study , Radiopharmaceuticals/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Triazoles/administration & dosage , Tumor Hypoxia , Xenograft Model Antitumor Assays
6.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 639-648, 2020 11.
Article in English | MEDLINE | ID: mdl-32945631

ABSTRACT

Elagolix is a novel oral gonadotropin releasing hormone receptor antagonist, that can suppress estradiol in a dose-dependent manner. It is indicated for management of moderate-to-severe pain associated with endometriosis. A population exposure-response model describing the relationship between elagolix exposure and changes in bone mineral density (BMD) was developed using data from four phase III studies in premenopausal women with endometriosis-associated pain. Elagolix pharmacokinetic exposure-dependent changes in BMD were described by an indirect-response maximum effect (Emax ) model through stimulation of bone resorption. African American race, higher body mass index (BMI), and lower type-I collagen C-telopeptide concentrations were significantly associated with higher baseline BMD. Higher BMI was significantly associated with higher bone formation rates. Simulations using the final model demonstrated that elagolix 150 mg q.d. dosing for 24 months is predicted to result in -1.45% (-2.04 to -0.814) decrease from baseline in BMD and were used to support corresponding dosing recommendations in the label.


Subject(s)
Bone Density/drug effects , Hydrocarbons, Fluorinated/adverse effects , Hydrocarbons, Fluorinated/pharmacokinetics , Pain/drug therapy , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , Absorptiometry, Photon/methods , Administration, Oral , Adult , Black or African American/ethnology , Biological Variation, Population , Body Mass Index , Case-Control Studies , Collagen Type I/analysis , Computer Simulation , Drug Labeling/standards , Endometriosis/complications , Female , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/therapeutic use , Middle Aged , Pain/etiology , Peptides/analysis , Predictive Value of Tests , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Safety
7.
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
8.
Invest New Drugs ; 38(6): 1763-1773, 2020 12.
Article in English | MEDLINE | ID: mdl-32377978

ABSTRACT

5-fluorouracil (5-FU) and 5-FU derivatives, such as capecitabine, UFT, and S-1, are the mainstay of chemotherapy treatment for gastrointestinal cancers, and other solid tumors. Compared with other cytotoxic chemotherapies, these drugs generally have a favorable safety profile, but hematologic and gastrointestinal toxicities remain common. DFP-11207 is a novel oral cytotoxic agent that combines a 5-FU pro-drug with a reversible DPD inhibitor and a potent inhibitor of OPRT, resulting in enhanced pharmacological activity of 5-FU with decreased gastrointestinal and myelosuppressive toxicities. In this Phase I study (NCT02171221), DFP-11207 was administered orally daily, in doses escalating from 40 mg/m2/day to 400 mg/m2/day in patients with esophageal, colorectal, gastric, pancreatic or gallbladder cancer (n = 23). It was determined that DFP-11207 at the dose of 330 mg/m2/day administered every 12 hours was well-tolerated with mild myelosuppressive and gastrointestinal toxicities. The pharmacokinetic analysis determined that the 5-FU levels were in the therapeutic range at this dose. In addition, fasted or fed states had no influence on the 5-FU levels (patients serving as their own controls). Among 21 efficacy evaluable patients, 7 patients had stable disease (33.3%), of which two had prolonged stable disease of >6 months duration. DFP-11207 can be explored as monotherapy or easily substitute 5-FU, capecitabine, or S-1 in combination regimens.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Hydrocarbons, Fluorinated/administration & dosage , Neoplasms/drug therapy , Prodrugs/administration & dosage , Pyrimidines/administration & dosage , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Female , Fluorouracil/blood , Food-Drug Interactions , Humans , Hydrocarbons, Fluorinated/adverse effects , Hydrocarbons, Fluorinated/pharmacokinetics , Male , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Treatment Outcome
9.
J Pharmacol Exp Ther ; 373(2): 261-268, 2020 05.
Article in English | MEDLINE | ID: mdl-32127372

ABSTRACT

In vitro approaches for predicting drug-drug interactions (DDIs) caused by alterations in transporter protein regulation are not well established. However, reports of transporter regulation via nuclear receptor (NR) modulation by drugs are increasing. This study examined alterations in transporter protein levels in sandwich-cultured human hepatocytes (SCHH; n = 3 donors) measured by liquid chromatography-tandem mass spectrometry-based proteomic analysis after treatment with N-[4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl]-N-(2,2,2-trifluoroethyl)benzenesulfonamide (T0901317), the first described synthetic liver X receptor agonist. T0901317 treatment (10 µM, 48 hours) decreased the levels of organic cation transporter (OCT) 1 (0.22-, 0.43-, and 0.71-fold of control) and organic anion transporter (OAT) 2 (0.38-, 0.38-, and 0.53-fold of control) and increased multidrug resistance protein (MDR) 1 (1.37-, 1.48-, and 1.59-fold of control). The induction of NR downstream gene expression supports the hypothesis that T0901317 off-target effects on farnesoid X receptor and pregnane X receptor activation are responsible for the unexpected changes in OCT1, OAT2, and MDR1. Uptake of the OCT1 substrate metformin in SCHH was decreased by T0901317 treatment. Effects of decreased OCT1 levels on metformin were simulated using a physiologically-based pharmacokinetic (PBPK) model. Simulations showed a clear decrease in metformin hepatic exposure resulting in a decreased pharmacodynamic effect. This DDI would not be predicted by the modest changes in simulated metformin plasma concentrations. Altogether, the current study demonstrated that an approach combining SCHH, proteomic analysis, and PBPK modeling is useful for revealing tissue concentration-based DDIs caused by unexpected regulation of hepatic transporters by NR modulators. SIGNIFICANCE STATEMENT: This study utilized an approach combining sandwich-cultured human hepatocytes, proteomic analysis, and physiologically based pharmacokinetic modeling to evaluate alterations in pharmacokinetics (PK) and pharmacodynamics (PD) caused by transporter regulation by nuclear receptor modulators. The importance of this approach from a mechanistic and clinically relevant perspective is that it can reveal drug-drug interactions (DDIs) caused by unexpected regulation of hepatic transporters and enable prediction of altered PK and PD changes, especially for tissue concentration-based DDIs.


Subject(s)
Hepatocytes/drug effects , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors/agonists , Proteomics/methods , Sulfonamides/pharmacology , ATP Binding Cassette Transporter, Subfamily B/analysis , Adult , Cells, Cultured , Drug Interactions , Female , Hepatocytes/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacokinetics , Middle Aged , Models, Biological , Octamer Transcription Factor-1/analysis , Organic Anion Transporters, Sodium-Independent/analysis , Sulfonamides/pharmacokinetics
10.
J Orthop Res ; 38(6): 1316-1326, 2020 06.
Article in English | MEDLINE | ID: mdl-31903618

ABSTRACT

Mechanical overloading of the temporomandibular joint (TMJ) and biochemical changes, like inflammation and hypoxia, contribute to cartilage degeneration and pain associated with osteoarthritis (OA). Yet, how overloading contributes to early dysregulation of chondrocytes is not understood, limiting the development of diagnostics and treatments for TMJ OA. Hypoxia-inducible factors (HIF)-1α/2α in chondrocytes were evaluated at Days 8 and 15 in a rat TMJ pain model induced by jaw loading (1 h/day for 7 days) using immunohistochemistry and compared between cases that induce persistent (3.5 N), acute (2 N), or no (0 N) sensitivity. Hypoxia was measured on Day 8 by immunolabeling of the tracer EF5 and 18 F-EF5 PET imaging. To assess the role of tumor necrosis factor (TNF) in painful TMJ loading, intra-articular etanercept was given before loading. Orofacial sensitivity was evaluated during and after loading. Facial grimace, TNF-α, HIF-2α, and hypoxia levels in the TMJ were measured after loading. HIF-2α was elevated (P = .03) after 3.5 N loading at Day 8, but HIF-1α was unchanged. EF5 uptake increased on Day 8 in the 3.5 N group (P < .048) by tissue assay and 18 F-EF5 PET. At Day 8, both HIF-2α (P = .01) and EF5 uptake (P = .005) were correlated with loading magnitude. Etanercept attenuated sensitivity (P < .01) and the facial grimace on Day 7 (P = .01). It also reduced (P < .01) HIF-2α and EF5 uptake on Day 8; but TNF-α levels were not different from controls at that time. Findings suggest that TMJ loading that induces persistent sensitivity upregulates the catabolic factor HIF-2α and reduces oxygen levels in the cartilage, which may be TNF-driven.


Subject(s)
Etanercept/administration & dosage , Hypoxia/etiology , Osteoarthritis/drug therapy , Pain Management/methods , Temporomandibular Joint , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Etanidazole/analogs & derivatives , Etanidazole/pharmacokinetics , Female , Hydrocarbons, Fluorinated/pharmacokinetics , Injections, Intra-Articular , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/physiology
11.
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
12.
Clin Pharmacokinet ; 59(5): 617-627, 2020 05.
Article in English | MEDLINE | ID: mdl-31713224

ABSTRACT

INTRODUCTION: Elagolix is approved for the management of moderate-to-severe pain associated with endometriosis. The aim of this analysis was to develop a physiologically based pharmacokinetic (PBPK) model that describes the enzyme-transporter interplay involved in the disposition of elagolix and to predict the magnitude of drug-drug interaction (DDI) potential of elagolix as an inhibitor of P-glycoprotein (P-gp) and inducer of cytochrome P450 (CYP) 3A4. METHODS: A PBPK model (SimCYP® version 15.0.86.0) was developed using elagolix data from in vitro, clinical PK and DDI studies. Data from DDI studies were used to quantify contributions of the uptake transporter organic anion transporting polypeptide (OATP) 1B1 and CYP3A4 in the disposition of elagolix, and to quantitatively assess the perpetrator potential of elagolix as a CYP3A4 inducer and P-gp inhibitor. RESULTS: After accounting for the interplay between elagolix metabolism by CYP3A4 and uptake by OATP1B1, the model-predicted PK parameters of elagolix along with the DDI AUC∞ and Cmax ratios, were within 1.5-fold of the observed data. Based on model simulations, elagolix 200 mg administered twice daily is a moderate inducer of CYP3A4 (approximately 56% reduction in midazolam AUC∞). Simulations of elagolix 150 mg administered once daily with digoxin predicted an increase in digoxin Cmax and AUC∞ by 68% and 19%, respectively. CONCLUSIONS: A PBPK model of elagolix was developed, verified, and applied to characterize the disposition interplay between CYP3A4 and OATP1B1, and to predict the DDI potential of elagolix as a perpetrator under dosing conditions that were not tested clinically. PBPK model-based predictions were used to support labeling language for DDI recommendations of elagolix.


Subject(s)
Hydrocarbons, Fluorinated/pharmacokinetics , Models, Biological , Pyrimidines/pharmacokinetics , Cytochrome P-450 CYP3A , Drug Interactions , Endometriosis/drug therapy , Female , Humans , Liver-Specific Organic Anion Transporter 1
13.
Ecotoxicol Environ Saf ; 181: 138-145, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31176248

ABSTRACT

While N-ethyl perfluorooctane sulfonamidoethanol (EtFOSE) is a precursor of perfluorooctane sulfonate (PFOS), its bioaccumulation, transformation and toxicological effects in earthworms (Eisenia fetida) exposed to quartz sands are poorly understood. The present study showed that except for parent EtFOSE, N-ethylperfluorooctane sulfonamide acetate (EtFOSAA), N-ethyl perfluorooctane sulfonamide (EtFOSA), perfluorooctane sulfonamide acetate (FOSAA), perfluorooctane sulfonamide (FOSA) and PFOS were detected in earthworms, with EtFOSAA as the primary biotransformation product. The biota-to-sand accumulation factor (BSAF) and uptake rate coefficient (ku) of EtFOSE were 5.7 and 0.542/d, respectively. The elimination rate constants (ke) decreased in the order EtFOSA (0.167/d) ∼ FOSAA (0.147/d) > FOSA (0.119/d) ∼ EtFOSAA (0.117/d) > EtFOSE (0.095/d) > PFOS (0.069/d). No significant effects were observed in malondialdehyde (MDA) contents and acetylcholinesterase (AChE) activities between EtFOSE treatments and controls. EtFOSE could cause significant accumulation of reactive oxygen species (ROS) in earthworms. Peroxidase (POD), superoxide dismutase (SOD) and catalase (CAT) were significantly activated by 41.4-74.3%, 37.2-44.4% and 32.4-52.3% from day 4-10, respectively, while 8-Hydroxy-2-deoxyguanosine (8-OHdG) levels were elevated by 47.7-70.3% from day 8-10, demonstrating that EtFOSE induced oxidative stress and oxidative DNA damage in earthworms. Significant increase of glutathione-S-transferase (GST) with 41.6-62.8% activation (8-10 d) gave indirect evidence on the conjugation of EtFOSE or its corresponding metabolites during phase II of detoxication. This study provides important information on the fate and potential risks of EtFOSE to terrestrial invertebrates.


Subject(s)
Hydrocarbons, Fluorinated/toxicity , Oligochaeta/metabolism , Quartz , Sulfonamides/toxicity , Animals , Biodegradation, Environmental , Biotransformation , DNA Damage , Fluorocarbons/metabolism , Hydrocarbons, Fluorinated/pharmacokinetics , Oxidative Stress , Silicon Dioxide , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics
14.
J Med Chem ; 62(5): 2651-2665, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30776234

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, incurable cancer with a 20% 1 year survival rate. While standard-of-care therapy can prolong life in a small fraction of cases, PDAC is inherently resistant to current treatments, and novel therapies are urgently required. Histone deacetylase (HDAC) inhibitors are effective in killing pancreatic cancer cells in in vitro PDAC studies, and although there are a few clinical studies investigating combination therapy including HDAC inhibitors, no HDAC drug or combination therapy with an HDAC drug has been approved for the treatment of PDAC. We developed an inhibitor of HDACs, AES-135, that exhibits nanomolar inhibitory activity against HDAC3, HDAC6, and HDAC11 in biochemical assays. In a three-dimensional coculture model, AES-135 kills low-passage patient-derived tumor spheroids selectively over surrounding cancer-associated fibroblasts and has excellent pharmacokinetic properties in vivo. In an orthotopic murine model of pancreatic cancer, AES-135 prolongs survival significantly, therefore representing a candidate for further preclinical testing.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Hydroxamic Acids/chemistry , Pancreatic Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Disease Models, Animal , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/therapeutic use , Mice , Pancreatic Neoplasms/pathology , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
15.
Bioorg Med Chem ; 27(1): 161-166, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30503782

ABSTRACT

Inspite of progress made for the discovery of novel antiepileptic drugs, epilepsy remains an unmet medical need. We synthesized nine trifluoromethylated enaminone derivatives and tested them for their anticonvulsant activity using maximal electroshock seizure (MES) test, subcutaneous pentylenetetrazole (scPTZ) test, and rotorod test for neurotoxicity. Among the compounds tested 3-(4-fluoro-3-(trifluomethyl)benzylamino)-5-(trifluoromethyl)cyclohex-2-enone (4f) showed ED50 of 23.47 mg/kg, when given orally to rats, 3-(4-chlorophenylamino)-5-(trifluoromethyl)cyclohex-2-enone (5a), which was previously reported by us but for which no quantitative data was available at the time, exhibited an ED50 of 62.39 mg/kg. Under the same conditions commercially available carbamazepine showed an ED50 of 28.20 mg/kg. There were no neurotoxicity observed upto a dose of 300 mg/kg for all the tested compounds. Compounds 4f and 5a represent good lead compounds for further development.


Subject(s)
Anticonvulsants/pharmacology , Benzylamines/pharmacology , Cyclohexanones/pharmacology , Cyclohexylamines/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Seizures/prevention & control , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacokinetics , Anticonvulsants/toxicity , Benzylamines/chemical synthesis , Benzylamines/pharmacokinetics , Benzylamines/toxicity , Computer Simulation , Cyclohexanones/chemical synthesis , Cyclohexanones/pharmacokinetics , Cyclohexanones/toxicity , Cyclohexylamines/chemical synthesis , Cyclohexylamines/pharmacokinetics , Cyclohexylamines/toxicity , Drug Design , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/toxicity , Male , Mice , Molecular Structure , Rats , Structure-Activity Relationship
16.
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
17.
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
18.
Int J Pharm ; 549(1-2): 388-396, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30107219

ABSTRACT

CKD-519, a potent cholesteryl ester transfer protein (CETP) inhibitor, is a clinical candidate being developed for the treatment of dyslipidemia. It is considered a Biopharmaceutical Classification System II compound with low solubility and high permeability. The objective of this study was to develop early formulations focusing on the dissolution rate of the compound to achieve dose-dependent exposure. High performance formulation strategies including solid dispersion (SD) and a self-microemulsifying drug delivery system (SMEDDS) were investigated and their in vivo and in vitro correlations were also evaluated in monkeys along with dose optimization in human volunteers. The SD granules were prepared in a fluid bed granulator using microcrystalline cellulose and mannitol as carriers. Poloxamer 407 and Eudragit E PO were each found to be a suitable solubilizing agent and polymer for the improvement of the CKD-519 dissolution rate. Pharmacokinetic studies in monkeys showed that the SD tablets exhibited better absorption than the SMEDDS in a dose-dependent manner from 1.5 mg to 100 mg. The mannitol-based SD tablet formulations were bioequivalent. However, pharmacokinetics studies in humans showed that the dose was saturable above 100 mg of CKD-519. This study was performed to determine how to develop early formulations for clinical studies and to identify rational formulation development strategies for CKD-519 to establish the pharmaceutical proof-of-concept in humans.


Subject(s)
Anticholesteremic Agents/administration & dosage , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Drug Development/methods , Hydrocarbons, Fluorinated/administration & dosage , Oxazoles/administration & dosage , Administration, Oral , Adult , Animals , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacokinetics , Biological Availability , Cellulose/chemistry , Cholesterol Ester Transfer Proteins/metabolism , Double-Blind Method , Drug Carriers , Drug Compounding , Excipients/chemistry , Female , Humans , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/pharmacokinetics , Macaca mulatta , Male , Mannitol/chemistry , Middle Aged , Models, Biological , Oxazoles/chemistry , Oxazoles/pharmacokinetics , Permeability , Poloxamer/chemistry , Polymethacrylic Acids/chemistry , Proof of Concept Study , Solubility , Tablets , Young Adult
19.
Clin Pharmacokinet ; 57(10): 1295-1306, 2018 10.
Article in English | MEDLINE | ID: mdl-29476499

ABSTRACT

INTRODUCTION: Elagolix is a novel, orally active, non-peptide, competitive gonadotropin-releasing hormone (GnRH) receptor antagonist in development for the management of endometriosis with associated pain and heavy menstrual bleeding due to uterine fibroids. The pharmacokinetics of elagolix have been well-characterized in phase I studies; however, elagolix population pharmacokinetics have not been previously reported. Therefore, a robust model was developed to describe elagolix population pharmacokinetics and to evaluate factors affecting elagolix pharmacokinetic parameters. METHODS: The data from nine clinical studies (a total of 1624 women) were included in the analysis: five phase I studies in healthy, premenopausal women and four phase III studies in premenopausal women with endometriosis. RESULTS: Elagolix population pharmacokinetics were best described by a two-compartment model with a lag time in absorption. Of the 15 covariates tested for effect on elagolix apparent clearance (CL/F) and/or volume of distribution only one covariate, organic anion transporting polypeptide (OATP) 1B1 genotype status, had a statistically significant, but not clinically meaningful, effect on elagolix CL/F. CONCLUSION: Elagolix pharmacokinetics were not affected by patient demographics and were similar between healthy women and women with endometriosis. Clinical Trial Registration Numbers NCT01403038, NCT01620528, NCT01760954, NCT01931670, NCT02143713.


Subject(s)
Endometriosis/metabolism , Hydrocarbons, Fluorinated/pharmacokinetics , Models, Biological , Premenopause/metabolism , Pyrimidines/pharmacokinetics , Adult , Clinical Trials, Phase I as Topic , Clinical Trials, Phase III as Topic , Endometriosis/drug therapy , Female , Genotype , Humans , Inactivation, Metabolic , Liver-Specific Organic Anion Transporter 1/genetics , Receptors, LHRH/antagonists & inhibitors
20.
Adv Healthc Mater ; 7(5)2018 03.
Article in English | MEDLINE | ID: mdl-29280311

ABSTRACT

The use of small interfering RNA (siRNA) in cancer treatment has been limited by the lack of effective systemic delivery methods. Although synthetic polycations have been widely explored in siRNA delivery, polycation/siRNA polyplexes often suffer from insufficient stability in vivo. Here, rationally designed siRNA delivery systems that meet the requirements for systemic siRNA delivery to distant tumors are reported. The hypothesis that modular design of delivery systems based on poly(amido amine)s that combine fluorination for systemic stability with bioreducibility for easy intracellular siRNA release, and PEGylation for improved safety and colloidal stability will overcome problems with contradicting siRNA delivery demands is tested. PEGylated, fluorinated, and bioreducible copolymers (PEG-PCD-F) with different degree of fluorination are thus synthesized. The fluorinated copolymers readily formed polyplexes with siRNA and achieved greatly improved gene silencing efficacy in multiple cell lines in vitro when compared with nonfluorinated controls. The results show fluorination-induced enhancement of stability, cellular uptake, and endosomal escape of the polyplexes, while exhibiting efficient siRNA release in reducing intracellular environment. PEG-PCD-F polyplexes with siRNA against Bcl2 inhibit breast tumor growth following systemic intravenous administration. The results provide strong evidence of successful combination of bioreducibility with fluorination and PEGylation to achieve systemic siRNA polyplex delivery.


Subject(s)
Hydrocarbons, Fluorinated , Mammary Neoplasms, Experimental/drug therapy , Polyamines , RNA, Small Interfering , Animals , CHO Cells , Cricetulus , Female , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Polyamines/chemistry , Polyamines/pharmacokinetics , Polyamines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...