Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Mol Cancer Ther ; 21(7): 1115-1124, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35499386

ABSTRACT

Patients with prostate cancer whose tumors bear deleterious mutations in DNA-repair pathways often respond to PARP inhibitors. Studies were conducted to compare the activity of several PARP inhibitors in vitro and their tissue exposure and in vivo efficacy in mice bearing PC-3M-luc-C6 prostate tumors grown subcutaneously or in bone. Niraparib, olaparib, rucaparib, and talazoparib were compared in proliferation assays, using several prostate tumor cell lines and in a cell-free PARP-trapping assay. PC-3M-luc-C6 cells were approximately 12- to 20-fold more sensitive to PARP inhibition than other prostate tumor lines, suggesting that these cells bear a DNA damage repair defect. The tissue exposure and efficacy of these PARP inhibitors were evaluated in vivo in PC-3M-luc-C6 subcutaneous and bone metastasis tumor models. A steady-state pharmacokinetic study in PC-3M-luc-C6 tumor-bearing mice showed that all of the PARP inhibitors had favorable subcutaneous tumor exposure, but niraparib was differentiated by superior bone marrow exposure compared with the other drugs. In a PC-3M-luc-C6 subcutaneous tumor efficacy study, niraparib, olaparib, and talazoparib inhibited tumor growth and increased survival to a similar degree. In contrast, in the PC-3M-luc-C6 bone metastasis model, niraparib showed the most potent inhibition of bone tumor growth compared with the other therapies (67% vs. 40%-45% on day 17), and the best survival improvement over vehicle control [hazard ratio (HR), 0.28 vs. HR, 0.46-0.59] and over other therapies (HR, 1.68-2.16). These results show that niraparib has superior bone marrow exposure and greater inhibition of tumor growth in bone, compared with olaparib, rucaparib, and talazoparib.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Humans , Indazoles , Male , Mice , Piperidines , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Prostate/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Tissue Distribution
2.
CPT Pharmacometrics Syst Pharmacol ; 10(9): 1107-1118, 2021 09.
Article in English | MEDLINE | ID: mdl-34273250

ABSTRACT

Erdafitinib is a potent oral pan-fibroblast growth factor receptor inhibitor being developed as oncology drug for patients with alterations in the fibroblast growth factor receptor pathway. Erdafitinib binds preferentially to α1-acid glycoprotein (AGP) and is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. This article describes a physiologically based pharmacokinetic (PBPK) model for erdafitinib to assess the drug-drug interaction (DDI) potential of CYP3A4 and CYP2C9 inhibitors and CYP3A4/CYP2C9 inducers on erdafitinib pharmacokinetics (PK) in patients with cancer exhibiting higher AGP levels and in populations with different CYP2C9 genotypes. Erdafitinib's DDI potential as a perpetrator for transporter inhibition and for time-dependent inhibition and/or induction of CYP3A was also evaluated. The PBPK model incorporated input parameters from various in vitro and clinical PK studies, and the model was verified using a clinical DDI study with itraconazole and fluconazole. Erdafitinib clearance in the PBPK model consisted of multiple pathways (CYP2C9/3A4, renal, intestinal; additional hepatic clearance), making the compound less susceptible to DDIs. In poor-metabolizing CYP2C9 populations carrying the CYP2C9*3/*3 genotype, simulations shown clinically relevant increase in erdafitinib plasma concentrations. Simulated luminal and enterocyte concentration showed potential risk of P-glycoprotein inhibition with erdafitinib in the first 5 h after dosing, and simulations showed this interaction can be avoided by staggering erdafitinib and digoxin dosing. Other than a simulated ~ 60% exposure reduction with strong CYP3A/2C inducers such as rifampicin, other DDI liabilities were minimal and considered not clinically relevant.


Subject(s)
Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Models, Biological , Pyrazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP2C9/drug effects , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drug Interactions , Genotype , Humans , Orosomucoid/metabolism
3.
Cancer Chemother Pharmacol ; 88(1): 25-37, 2021 07.
Article in English | MEDLINE | ID: mdl-33754187

ABSTRACT

PURPOSE: To assess the safety and pharmacokinetics and determine the recommended phase 2 dose (RP2D) of niraparib with apalutamide or abiraterone acetate plus prednisone (AAP) in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS: BEDIVERE was a multicenter, open-label, phase 1b study of niraparib 200 or 300 mg/day with apalutamide 240 mg or AAP (abiraterone acetate 1000 mg; prednisone 10 mg). Patients with mCRPC were previously treated with ≥ 2 lines of systemic therapy, including ≥ 1 androgen receptor-axis-targeted therapy for prostate cancer. RESULTS: Thirty-three patients were enrolled (niraparib-apalutamide, 6; niraparib-AAP, 27). No dose-limiting toxicities (DLTs) were reported when combinations included niraparib 200 mg; five patients receiving niraparib 300 mg experienced DLTs [niraparib-apalutamide, 2/3 patients (66.7%); niraparib-AAP, 3/8 patients (37.5%)]. Although data are limited, niraparib exposures were lower when given with apalutamide compared with historical niraparib monotherapy exposures in patients with solid tumors. Because of the higher incidence of DLTs, the niraparib-apalutamide combination and niraparib 300 mg combination with AAP were not further evaluated. Niraparib 200 mg was selected as the RP2D with AAP. Of 19 patients receiving niraparib 200 mg with AAP, 12 (63.2%) had grade 3/4 treatment-emergent adverse events, the most common being thrombocytopenia (26.3%) and hypertension (21.1%). Five patients (26.3%) had adverse events leading to treatment discontinuation. CONCLUSIONS: These results support the choice of niraparib 200 mg as the RP2D with AAP. The niraparib-AAP combination was tolerable in patients with mCRPC, with no new safety signals. An ongoing phase 3 study is further assessing this combination in patients with mCRPC. TRIAL REGISTRATION NO: NCT02924766 (ClinicalTrials.gov).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Indazoles/adverse effects , Indazoles/pharmacokinetics , Piperidines/adverse effects , Piperidines/pharmacokinetics , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Aged , Aged, 80 and over , Androstenes/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Humans , Indazoles/therapeutic use , Male , Middle Aged , Piperidines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prednisone/therapeutic use , Prostatic Neoplasms, Castration-Resistant/metabolism , Thiohydantoins/therapeutic use
5.
Xenobiotica ; 51(2): 177-193, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32902324

ABSTRACT

This article describes in vivo biotransformation and disposition of erdafitinib following single oral dose of 3H-erdafitinib and 14C-erdafitinib to intact and bile duct-cannulated (BC) rats (4 mg/kg), 3H-erdafitinib to intact dogs (0.25 mg/kg), and 14C-erdafitinib to humans (12 mg; NCT02692677). Peak plasma concentrations of total radioactivity were achieved rapidly (Tmax: animals, 1 h; humans, 2-3 h). Recovery of drug-derived radioactivity was significantly slower in humans (87%, 384 h) versus animals (rats: 91-98%, 48 h; dogs: 81%, 72 h). Faeces was the primary route of elimination in intact rats (95%), dogs (76%), and humans (69%); and bile in BC rats (48%). Renal elimination of radioactivity was relatively low in animals (2-12%) versus humans (19%). Unchanged erdafitinib was major component in human excreta (faeces, 17%; urine, 11%) relative to animals. M6 (O-desmethyl) was the major faecal metabolite in humans (24%) and rats (intact, 46%; BC, 11%), and M2 (O-glucuronide of M6) was the prevalent biliary metabolite in rats (14%). In dogs, besides M6, majority of radioactive dose in faeces was composed of multiple minor metabolites. In humans, unchanged erdafitinib was the major circulating entity. O-demethylation of erdafitinib was the major metabolic pathway in humans and animals.


Subject(s)
Protein Kinase Inhibitors/metabolism , Pyrazoles/metabolism , Quinoxalines/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Administration, Oral , Animals , Area Under Curve , Bile/metabolism , Biotransformation , Dogs , Feces , Glucuronides/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Species Specificity
6.
Health Policy Open ; 1: 100004, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33392500

ABSTRACT

India's rapid economic growth has been accompanied by slower improvements in population health. Given the need to reconcile the ambitious goal of achieving Universal Coverage with limited resources, a robust priority-setting mechanism is required to ensure that the right trade-offs are made and the impact on health is maximised. Health Technology Assessment (HTA) is endorsed by the World Health Assembly as the gold standard approach to synthesizing evidence systematically for evidence-informed priority setting (EIPS). India is formally committed to institutionalising HTA as an integral component of the EIPS process. The effective conduct and uptake of HTA depends on a well-functioning ecosystem of stakeholders adept at commissioning and generating policy-relevant HTA research, developing and utilising rigorous technical, transparent, and inclusive methods and processes, and a strong multisectoral and transnational appetite for the use of evidence to inform policy. These all require myriad complex and complementary capacities to be built at each level of the health system . In this paper we describe how a framework for targeted and locally-tailored capacity building for EIPS, and specifically HTA, was collaboratively developed and implemented by an international network of priority-setting expertise, and the Government of India.

7.
Eur J Drug Metab Pharmacokinet ; 45(1): 101-111, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31673875

ABSTRACT

BACKGROUND AND OBJECTIVES: Erdafitinib, an oral selective pan-fibroblast growth factor receptor (FGFR) kinase inhibitor, is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. The aim of this phase 1 study was to assess the pharmacokinetics and safety of erdafitinib in healthy participants when coadministered with fluconazole (moderate CYP2C9 and CYP3A inhibitor), and itraconazole (a strong CYP3A4 and P-glycoprotein inhibitor). The effect of CYP2C9 genotype variants (*1/*1, *1/*2, *1/*3) on the pharmacokinetics of erdafitinib was also investigated. METHODS: In this open-label, parallel-group, single-center study, eligible healthy adults were randomized by CYP2C9 genotype to receive Treatment A (single oral dose of erdafitinib 4 mg) on day 1, Treatment B (fluconazole 400 mg/day orally) on days 1-11, or Treatment C (itraconazole 200 mg/day orally) on days 1-11. Healthy adults randomized to Treatment B and C received a single oral 4-mg dose of erdafitinib on day 5. The pharmacokinetic parameters, including mean maximum plasma concentration (Cmax), area under the curve (AUC) from time 0 to 168 h (AUC168h), AUC from time 0 to the last quantifiable concentration (AUClast), and AUC from time 0 to infinity (AUC∞) were calculated from individual plasma concentration-time data using standard non-compartmental methods. RESULTS: Coadministration of erdafitinib with fluconazole increased Cmax of erdafitinib by approximately 21%, AUC168h by 38%, AUClast by 49%, and AUC∞ by 48% while coadministration with itraconazole resulted in no change in erdafitinib Cmax and increased AUC168h by 20%, AUClast by 33% and AUC∞ by 34%. Erdafitinib exposure was comparable between participants with CYP2C9 *1/*2 or *1/*3 and with wild-type CYP2C9 genotype. The ratio of total amount of erdafitinib excreted in the urine (inhibited to non-inhibited) was 1.09, the ratio of total amount of excreted metabolite M6 was 1.21, and the ratio of the metabolite to parent ratio in the urine was 1.11, when coadministration of erdafitinib with itraconazole was compared with single-dose erdafitinib. Treatment-emergent adverse events (TEAEs) were generally Grade 1 or 2 in severity; the most commonly reported TEAE was headache. No safety concerns were identified with single-dose erdafitinib when administered alone and in combination with fluconazole or itraconazole in healthy adults. CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. TRIAL REGISTRATION: ClinicalTrials.gov identifier number: NCT03135106.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drug Interactions , Fluconazole/pharmacology , Itraconazole/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Adult , Area Under Curve , Cytochrome P-450 CYP2C9/genetics , Drug Combinations , Female , Healthy Volunteers , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/urine , Pyrazoles/adverse effects , Pyrazoles/blood , Pyrazoles/urine , Quinoxalines/adverse effects , Quinoxalines/blood , Quinoxalines/urine , Receptors, Fibroblast Growth Factor/antagonists & inhibitors
8.
RSC Adv ; 8(21): 11509-11516, 2018 Mar 21.
Article in English | MEDLINE | ID: mdl-35542808

ABSTRACT

Here we report the influence of terminal -F, -Cl and -NO2 substitution on the elastic, dielectric and polar switching behavior of four-ring bent-core liquid crystals (LCs). Elastic constants of nematic liquid crystals are the key parameters in determining the threshold voltage and sensitivity to electro-optical response in a device. The elastic properties of bent-core liquid crystal systems show atypical temperature dependence and there is no hard-core theory to explain the behavior. However based on molecular simulation and atomistic calculations it is found in earlier studies that the bend angle dominates the behavior of elastic constants and the terminal or lateral substitutions have very little effect. Here we have studied three bent-core compounds which are differentiated only by their terminal polar substitution. The bend angle is identical (∼146°) for all the three compounds yet they show dramatically different elastic properties. In the fluoro-substituted compound K 11 > K 33, while for the other two compounds K 33 > K 11. Thus it is evident that the terminal polar substitution plays vital role in determining the elastic properties of bent-core systems. Correlating the mesophase ranges with the respective dipole moments of the samples it is observed that the fluoro-substituted compound (11-2M-F) with lowest dipole moment favours only nematic phase with smallest mesophase range (46.1 °C), compound 11-2M-Cl with moderate dipole moment favours short range nematic, broad range smectic with moderate mesophase range (53.1 °C), whereas the compound 11-2M-NO2 possesses the widest mesophase range (99.8 °C) with a very narrow nematic and a broad smectic phase amongst the three studied compounds.

9.
Chem Biol Interact ; 277: 85-90, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28916336

ABSTRACT

During preclinical development of canagliflozin, an SGLT2 inhibitor, treatment-related pheochromocytomas, renal tubular tumors (RTT), and testicular Leydig cell tumors were reported in the 2-year rat toxicology study. In a previous 6-month rat mechanistic study, feeding a glucose free diet prevented canagliflozin effects on carbohydrate malabsorption as well as the increase in cell proliferation in adrenal medulla and kidneys, implicating carbohydrate malabsorption as the mechanism for tumor formation. In this chronic study male Sprague-Dawley rats were dosed orally with canagliflozin at high dose-levels (65 or 100 mg/kg/day) for 15 months and received either a standard diet or a glucose-free diet. Canagliflozin-dosed rats on standard diet showed presence of basophilic renal tubular tumors (6/90) and an increased incidence of adrenal medullary hyperplasia (35/90), which was fully prevented by feeding a glucose-free diet (no RTT's; adrenal medullary hyperplasia in ≤5/90). These data further confirm that kidney and adrenal medullary tumors in the 2-year rat study were secondary to carbohydrate (glucose) malabsorption and were not due to a direct effect of canagliflozin on these target tissues.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Canagliflozin/therapeutic use , Glucose/metabolism , Hypoglycemic Agents/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Tubules/drug effects , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Animals , Dietary Sucrose/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Rats , Rats, Sprague-Dawley
10.
Spectrochim Acta A Mol Biomol Spectrosc ; 178: 142-150, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28182984

ABSTRACT

A newly designed asymmetrical four-ring bent-core compound (4'-n-decyloxyphenylazo)-phenyl-4-yl-3-[N-(4'-n-octadecyloxy-2-hydroxybenzylidene) amino]-2-methyl benzoate exhibiting liquid crystalline behavior was synthesized and characterized. The thermal and textural morphology were studied using differential scanning calorimetry and polarizing optical microscopy, respectively. The study of hydrogen bonding and dynamics of the phase transition has been performed at the molecular level using temperature dependent Fourier transform infrared (FTIR) spectroscopy. The spectral analysis of OH, CH2/CH3, CO, and CN stretching vibrational bands revealed clear signatures of Cr→SmA phase transition at 125°C. Density functional theory has been adopted for the geometry optimization and conformational study of the monomer using the B3LYP/6-31G(d) method. The conformational analysis has been performed to predict the most stable conformer along with the possible conformers using one-dimensional potential energy scan employing the same level of theory. The combination of experimental findings and theoretical analysis helped to understand the mechanism of phase transitions at the molecular level.

11.
Br J Clin Pharmacol ; 83(5): 1082-1096, 2017 05.
Article in English | MEDLINE | ID: mdl-27862160

ABSTRACT

AIMS: Canagliflozin is a recently approved drug for use in the treatment of type 2 diabetes. The potential for canagliflozin to cause clinical drug-drug interactions (DDIs) was assessed. METHODS: DDI potential of canagliflozin was investigated using in vitro test systems containing drug metabolizing enzymes or transporters. Basic predictive approaches were applied to determine potential interactions in vivo. A physiologically-based pharmacokinetic (PBPK) model was developed and clinical DDI simulations were performed to determine the likelihood of cytochrome P450 (CYP) inhibition by canagliflozin. RESULTS: Canagliflozin was primarily metabolized by uridine 5'-diphospho-glucuronosyltransferase 1A9 and 2B4 enzymes. Canagliflozin was a substrate of efflux transporters (P-glycoprotein, breast cancer resistance protein and multidrug resistance-associated protein-2) but was not a substrate of uptake transporters (organic anion transporter polypeptide isoforms OATP1B1, OATP1B3, organic anion transporters OAT1 and OAT3, and organic cationic transporters OCT1, and OCT2). In inhibition assays, canagliflozin was shown to be a weak in vitro inhibitor (IC50 ) of CYP3A4 (27 µmol l -1 , standard error [SE] 4.9), CYP2C9 (80 µmol l -1 , SE 8.1), CYP2B6 (16 µmol l-1 , SE 2.1), CYP2C8 (75 µmol l -1 , SE 6.4), P-glycoprotein (19.3 µmol l -1 , SE 7.2), and multidrug resistance-associated protein-2 (21.5 µmol l -1 , SE 3.1). Basic models recommended in DDI guidelines (US Food & Drug Administration and European Medicines Agency) predicted moderate to low likelihood of interaction for these CYPs and efflux transporters. PBPK DDI simulations of canagliflozin with CYP probe substrates (simvastatin, S-warfarin, bupropion, repaglinide) did not show relevant interaction in humans since mean areas under the concentration-time curve and maximum plasma concentration ratios for probe substrates with and without canagliflozin and its 95% CIs were within 0.80-1.25. CONCLUSIONS: In vitro DDI followed by a predictive or PBPK approach was applied to determine DDI potential of canagliflozin. Overall, canagliflozin is neither a perpetrator nor a victim of clinically important interactions.


Subject(s)
Canagliflozin/administration & dosage , Hypoglycemic Agents/administration & dosage , Models, Biological , Animals , Area Under Curve , Canagliflozin/pharmacokinetics , Canagliflozin/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Xenopus laevis
12.
J Clin Pharmacol ; 55(9): 1061-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25827774

ABSTRACT

O-glucuronidation is the major metabolic elimination pathway for canagliflozin. The objective was to identify enzymes and tissues involved in the formation of 2 major glucuronidated metabolites (M7 and M5) of canagliflozin and subsequently to assess the impact of genetic variations in these uridine diphosphate glucuronosyltransferases (UGTs) on in vivo pharmacokinetics in humans. In vitro incubations with recombinant UGTs revealed involvement of UGT1A9 and UGT2B4 in the formation of M7 and M5, respectively. Although M7 and M5 were formed in liver microsomes, only M7 was formed in kidney microsomes. Participants from 7 phase 1 studies were pooled for pharmacogenomic analyses. A total of 134 participants (mean age, 41 years; men, 63%; white, 84%) were included in the analysis. In UGT1A9*3 carriers, exposure of plasma canagliflozin (Cmax,ss , 11%; AUCτ,ss , 45%) increased relative to the wild type. An increase in exposure of plasma canagliflozin (Cmax,ss , 21%; AUCt,ss , 18%) was observed in participants with UGT2B4*2 genotype compared with UGT2B4*2 noncarriers. Metabolites further delineate the role of both enzymes. The pharmacokinetic findings in participants carrying the UGT1A9*3 and UGT2B4*2 allele implicate that UGT1A9 and UGT2B4 are involved in the metabolism of canagliflozin to M7 and M5, respectively.


Subject(s)
Canagliflozin/pharmacokinetics , Gene Expression Regulation, Enzymologic/physiology , Genetic Variation , Glucuronosyltransferase/metabolism , Microsomes/metabolism , Adult , Canagliflozin/administration & dosage , Canagliflozin/blood , Canagliflozin/metabolism , Female , Genotype , Glucuronosyltransferase/genetics , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Intestinal Mucosa/metabolism , Kidney , Liver/metabolism , Male , Middle Aged , Recombinant Proteins
13.
Clin Pharmacol Drug Dev ; 4(4): 295-304, 2015 07.
Article in English | MEDLINE | ID: mdl-27136910

ABSTRACT

Absolute oral bioavailability of canagliflozin was assessed by simultaneous oral administration with intravenous [(14) C]-canagliflozin microdose infusion in nine healthy men. Pharmacokinetics of canagliflozin, [(14) C]-canagliflozin, and total radioactivity, and safety and tolerability were assessed at prespecified timepoints. On day 1, single-dose oral canagliflozin (300 mg) followed 105 minutes later by intravenous [(14) C]-canagliflozin (10 µg, 200 nCi) was administered. After oral administration, the mean (SD) Cmax of canagliflozin was 2504 (482) ng/mL at 1.5 hours, AUC∞ 17,375 (3555) ng.h/mL, and t1/2 11.6 (0.70) hours. After intravenous administration, the mean (SD) Cmax of unchanged [(14) C]-canagliflozin was 17,605 (6901) ng/mL, AUC∞ 27,100 (10,778) ng.h/mL, Vdss 83.5 (29.2) L, Vdz 119 (41.6) L, and CL 12.2 (3.79) L/h. Unchanged [(14) C]-canagliflozin and metabolites accounted for about 57% and 43% of the plasma total [(14) C] radioactivity AUC∞ , respectively. For total [(14) C] radioactivity, the mean (SD) Cmax was 15,981 (2721) ng-eq/mL, and AUC∞ 53,755 (15,587) ng-eq.h/mL. Renal (34.5% in urine) and biliary (34.1% in feces) excretions were the major elimination pathways for total [(14) C] radioactivity. The absolute oral bioavailability of canagliflozin was 65% (90% confidence interval: 55.41; 76.07). Overall, oral canagliflozin 300 mg coadministered with intravenous [(14) C]-canagliflozin (10 µg) was generally well-tolerated in healthy men, with no treatment-emergent adverse events.


Subject(s)
Canagliflozin/administration & dosage , Canagliflozin/pharmacokinetics , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Biological Availability , Canagliflozin/adverse effects , Canagliflozin/blood , Feces/chemistry , Healthy Volunteers , Hepatobiliary Elimination , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/blood , Infusions, Intravenous , Male , Middle Aged , Models, Biological , Young Adult
14.
Clin Pharmacol Drug Dev ; 4(3): 226-36, 2015.
Article in English | MEDLINE | ID: mdl-27140803

ABSTRACT

Drug-drug interactions between canagliflozin, a sodium glucose co-transporter 2 inhibitor, and glyburide, metformin, and simvastatin were evaluated in three phase-1 studies in healthy participants. In these open-label, fixed sequence studies, participants received: Study 1-glyburide 1.25 mg/day (Day 1), canagliflozin 200 mg/day (Days 4-8), canagliflozin with glyburide (Day 9); Study 2-metformin 2,000 mg/day (Day 1), canagliflozin 300 mg/day (Days 4-7), metformin with canagliflozin (Day 8); Study 3-simvastatin 40 mg/day (Day 1), canagliflozin 300 mg/day (Days 2-6), simvastatin with canagliflozin (Day 7). Pharmacokinetic parameters were assessed at prespecified intervals. Co-administration of canagliflozin and glyburide did not affect the overall exposure (maximum plasma concentration [Cmax ] and area under the plasma concentration-time curve [AUC]) of glyburide and its metabolites (4-trans-hydroxy-glyburide and 3-cis-hydroxy-glyburide). Canagliflozin did not affect the peak concentration of metformin; however, AUC increased by 20%. Though Cmax and AUC were slightly increased for simvastatin (9% and 12%) and simvastatin acid (26% and 18%) following coadministration with canagliflozin, compared with simvastatin administration alone; however, no effect on active 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase inhibitory activity was observed. There were no serious adverse events or hypoglycemic episodes. No drug-drug interactions were observed between canagliflozin and glyburide, metformin, or simvastatin. All treatments were well-tolerated in healthy participants.


Subject(s)
Canagliflozin/administration & dosage , Glyburide/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Simvastatin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Argentina , Biological Availability , Biotransformation , Canagliflozin/adverse effects , Drug Administration Schedule , Drug Interactions , Female , Glyburide/administration & dosage , Glyburide/adverse effects , Glyburide/blood , Half-Life , Healthy Volunteers , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/blood , Male , Metabolic Clearance Rate , Metformin/administration & dosage , Metformin/adverse effects , Metformin/blood , Middle Aged , Models, Biological , Simvastatin/administration & dosage , Simvastatin/adverse effects , Simvastatin/blood , United States , Young Adult
15.
Int J Clin Pharmacol Ther ; 53(2): 115-28, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25407255

ABSTRACT

OBJECTIVE: Canagliflozin, a sodium-glucose co-transporter 2 inhibitor, approved for the treatment of type-2 diabetes mellitus (T2DM), is metabolized by uridine diphosphate-glucuronosyltransferases (UGT) 1A9 and UGT2B4, and is a substrate of P-glycoprotein (P-gp). Canagliflozin exposures may be affected by coadministration of drugs that induce (e.g., rifampin for UGT) or inhibit (e.g. probenecid for UGT; cyclosporine A for P-gp) these pathways. The primary objective of these three independent studies (single-center, open-label, fixed-sequence) was to evaluate the effects of rifampin (study 1), probenecid (study 2), and cyclosporine A (study 3) on the pharmacokinetics of canagliflozin in healthy participants. METHODS: Participants received; in study 1: canagliflozin 300 mg (days 1 and 10), rifampin 600 mg (days 4-12); study 2: canagliflozin 300 mg (days 1-17), probenecid 500 mg twice daily (days 15-17); and study 3: canagliflozin 300 mg (days 1-8), cyclosporine A 400 mg (day 8). Pharmacokinetics were assessed at prespecified intervals on days 1 and 10 (study 1); on days 14 and 17 (study 2), and on days 2-8 (study 3). RESULTS: Rifampin decreased the maximum plasma canagliflozin concentration (Cmax) by 28% and its area under the curve (AUC) by 51%. Probenecid increased the Cmax by 13% and the AUC by 21%. Cyclosporine A increased the AUC by 23% but did not affect the Cmax. CONCLUSION: Coadministration of canagliflozin with rifampin, probenecid, and cyclosporine A was well-tolerated. No clinically meaningful interactions were observed for probenecid or cyclosporine A, while rifampin coadministration modestly reduced canagliflozin plasma concentrations and could necessitate an appropriate monitoring of glycemic control.


Subject(s)
Cyclosporine/pharmacology , Glucosides/pharmacokinetics , Probenecid/pharmacology , Rifampin/pharmacology , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/pharmacokinetics , Adult , Canagliflozin , Cyclosporine/adverse effects , Drug Interactions , Female , Glucosides/adverse effects , Humans , Male , Probenecid/adverse effects , Rifampin/adverse effects , Thiophenes/adverse effects
16.
Toxicol Pathol ; 43(1): 48-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25398756

ABSTRACT

Canagliflozin, a sodium glucose co-transporter 2 (SGLT2) inhibitor, has been developed for the treatment of adults with type 2 diabetes mellitus (T2DM). During the phase 3 program, treatment-related pheochromocytomas, renal tubular tumors, and testicular Leydig cell tumors were reported in the 2-year rat toxicology study. Treatment-related tumors were not seen in the 2-year mouse study. A cross-functional, mechanism-based approach was undertaken to determine whether the mechanisms responsible for tumorigenesis in the rat were of relevance to humans. Based on findings from nonclinical and clinical studies, the treatment-related tumors observed in rats were not deemed to be of clinical relevance. Here, we describe the scientific and regulatory journey from learning of the 2-year rat study findings to the approval of canagliflozin for the treatment of T2DM.


Subject(s)
Canagliflozin/toxicity , Hypoglycemic Agents/toxicity , Neoplasms, Experimental/chemically induced , Animals , Canagliflozin/administration & dosage , Carcinogenicity Tests , Clinical Trials as Topic , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/administration & dosage , Male , Mice , Rats , Sodium-Glucose Transporter 2 Inhibitors , Testosterone
17.
Int J Clin Pharmacol Ther ; 53(1): 41-53, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25345427

ABSTRACT

OBJECTIVE: Drug-drug interactions between canagliflozin, a sodium glucose co-transporter 2 inhibitor approved for the management of type-2 diabetes mellitus, and an oral contraceptive (OC), warfarin, and digoxin were evaluated in three phase 1 studies in healthy participants. METHODS: All studies were open-label; study 1 included a fixed-sequence design, and studies 2 and 3 used a crossover design. Regimens were: study 1: OC (levonorgestrel (150 µg) + ethinyl estradiol (30 µg))/day (day 1), canagliflozin 200 mg/day (days 4 - 8), and canagliflozin with OC (day 9); study 2: canagliflozin 300 mg/day (days 1 - 12) with warfarin 30 mg/day (day 6) in period 1, and only warfarin 30 mg/day (day 1) in period 2, or vice versa; study 3: digoxin alone (0.5 mg/day (day 1) + 0.25 mg/day (days 2 - 7)) in period 1, and with canagliflozin 300 mg/day (days 1 - 7) in period 2, or vice versa. Pharmacokinetics (PK) were assessed at prespecified intervals; OC: days 1 and 9, canagliflozin: days 8 - 9 (study 1); warfarin: days 6 (period 1) and 1 (period 2) (study 2); and digoxin: days 5 - 7 (periods 1 and 2) (study 3). Warfarin's pharmacodynamics (PD; International Normalized Ratio (INR)) was assessed on days 6 (period 1) and 1 (period 2). RESULTS: Canagliflozin increased the plasma exposure of OC (maximum plasma concentration (Cmax): 22%, area under the curve (AUC): 6%) and digoxin (Cmax: 36%, AUC: 20%); but did not alter warfarin'€™s PK and PD. No clinically relevant safety findings (including hypoglycemia) were noted. CONCLUSION: Canagliflozin can be coadministered with OC, warfarin, or digoxin without dose adjustments. All treatments were well-tolerated.


Subject(s)
Anticoagulants/pharmacokinetics , Cardiotonic Agents/pharmacokinetics , Contraceptives, Oral, Combined/pharmacokinetics , Digoxin/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Glucosides/administration & dosage , Hypoglycemic Agents/administration & dosage , Levonorgestrel/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/administration & dosage , Warfarin/pharmacokinetics , Adult , Anticoagulants/administration & dosage , Anticoagulants/blood , Area Under Curve , Blood Coagulation/drug effects , Canagliflozin , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/blood , Contraceptives, Oral, Combined/administration & dosage , Contraceptives, Oral, Combined/blood , Cross-Over Studies , Digoxin/administration & dosage , Digoxin/blood , Drug Administration Schedule , Drug Combinations , Drug Dosage Calculations , Drug Interactions , Ethinyl Estradiol/administration & dosage , Ethinyl Estradiol/blood , Female , Glucosides/adverse effects , Half-Life , Healthy Volunteers , Humans , Hypoglycemic Agents/adverse effects , International Normalized Ratio , Levonorgestrel/administration & dosage , Levonorgestrel/blood , Male , Metabolic Clearance Rate , Middle Aged , Polypharmacy , Risk Assessment , Sodium-Glucose Transporter 2/metabolism , Thiophenes/adverse effects , Warfarin/administration & dosage , Warfarin/blood , Young Adult
18.
Chem Biol Interact ; 224: 1-12, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25289773

ABSTRACT

The carcinogenicity potential of canagliflozin, an inhibitor of SGLT2, was evaluated in a 2-year rat study (10, 30, and 100 mg/kg). Rats showed an increase in pheochromocytomas, renal tubular tumors, and testicular Leydig cell tumors. Systemic exposure multiples at the highest dose relative to the maximum clinical dose were 12- to 21-fold. Pheochromocytomas and renal tubular tumors were noted in both sexes at 100 mg/kg. Leydig cell tumors were observed in males in all dose groups and were associated with increased luteinizing hormone levels. Hyperplasia was increased in the adrenal medulla at 100 mg/kg, but only a limited increase in simple tubular hyperplasia was observed in the kidney of males at 100 mg/kg. Hyperostosis occurred and was accompanied by substantial effects on calcium metabolism, including increased urinary calcium excretion and decreased levels of calcium regulating hormones (1,25-dihydroxyvitamin D and parathyroid hormone). A separate study with radiolabeled calcium confirmed that increased urinary calcium excretion was mediated via increased calcium absorption from the gastrointestinal tract. It was hypothesized that, at high doses, canagliflozin might have inhibited glucose absorption in the intestine via SGLT1 inhibition that resulted in glucose malabsorption, which increased calcium absorption by stimulating colonic glucose fermentation and reducing intestinal pH. Pheochromocytomas and adrenal medullary hyperplasia were attributed to altered calcium homeostasis, which have a known relationship in the rat. In conclusion, Leydig cell tumors were associated with increased luteinizing hormone levels and pheochromocytomas were most likely related to glucose malabsorption and altered calcium homeostasis. Renal tubular tumors may also have been linked to glucose malabsorption.


Subject(s)
Adrenal Gland Neoplasms/chemically induced , Carcinogenesis/chemically induced , Glucosides/toxicity , Kidney Neoplasms/chemically induced , Leydig Cell Tumor/chemically induced , Pheochromocytoma/chemically induced , Sodium-Glucose Transporter 2 Inhibitors , Testicular Neoplasms/chemically induced , Thiophenes/toxicity , Adrenal Gland Neoplasms/pathology , Animals , Canagliflozin , Carcinogenicity Tests , Dose-Response Relationship, Drug , Glucosides/chemistry , Kidney Neoplasms/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Leydig Cell Tumor/pathology , Male , Pheochromocytoma/pathology , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 2 , Structure-Activity Relationship , Testicular Neoplasms/pathology , Thiophenes/chemistry
19.
Chem Biol Interact ; 221: 109-18, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25130857

ABSTRACT

Canagliflozin is an SGLT2 inhibitor used for the treatment of type 2 diabetes mellitus. Studies were conducted to investigate the mechanism responsible for renal tubular tumors and pheochromocytomas observed at the high dose in a 2-year carcinogenicity study in rats. At the high dose (100mg/kg) in rats, canagliflozin caused carbohydrate malabsorption evidenced by inhibition of intestinal glucose uptake, decreased intestinal pH and increased urinary calcium excretion. In a 6-month mechanistic study utilization of a glucose-free diet prevented carbohydrate malabsorption and its sequelae, including increased calcium absorption and urinary calcium excretion, and hyperostosis. Cell proliferation in the kidney and adrenal medulla was increased in rats maintained on standard diet and administered canagliflozin (100mg/kg), and in addition an increase in the renal injury biomarker KIM-1 was observed. Increased cell proliferation is considered as a proximal event in carcinogenesis. Effects on cell proliferation, KIM-1 and calcium excretion were inhibited in rats maintained on the glucose-free diet, indicating they are secondary to carbohydrate malabsorption and are not direct effects of canagliflozin.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Carcinogenesis , Glucosides/pharmacology , Malabsorption Syndromes , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/pharmacology , Animals , Canagliflozin , Cell Adhesion Molecules/metabolism , Cell Proliferation , Immunohistochemistry , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley
20.
Drug Metab Dispos ; 42(5): 903-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24568888

ABSTRACT

Canagliflozin is an oral antihyperglycemic agent used for the treatment of type 2 diabetes mellitus. It blocks the reabsorption of glucose in the proximal renal tubule by inhibiting the sodium-glucose cotransporter 2. This article describes the in vivo biotransformation and disposition of canagliflozin after a single oral dose of [(14)C]canagliflozin to intact and bile duct-cannulated (BDC) mice and rats and to intact dogs and humans. Fecal excretion was the primary route of elimination of drug-derived radioactivity in both animals and humans. In BDC mice and rats, most radioactivity was excreted in bile. The extent of radioactivity excreted in urine as a percentage of the administered [(14)C]canagliflozin dose was 1.2%-7.6% in animals and approximately 33% in humans. The primary pathways contributing to the metabolic clearance of canagliflozin were oxidation in animals and direct glucuronidation of canagliflozin in humans. Unchanged canagliflozin was the major component in systemic circulation in all species. In human plasma, two pharmacologically inactive O-glucuronide conjugates of canagliflozin, M5 and M7, represented 19% and 14% of total drug-related exposure and were considered major human metabolites. Plasma concentrations of M5 and M7 in mice and rats from repeated dose safety studies were lower than those in humans given canagliflozin at the maximum recommended dose of 300 mg. However, biliary metabolite profiling in rodents indicated that mouse and rat livers had significant exposure to M5 and M7. Pharmacologic inactivity and high water solubility of M5 and M7 support glucuronidation of canagliflozin as a safe detoxification pathway.


Subject(s)
Glucosides/metabolism , Hypoglycemic Agents/metabolism , Thiophenes/metabolism , Administration, Oral , Adult , Animals , Bile/metabolism , Canagliflozin , Carbon Radioisotopes , Dogs , Feces/chemistry , Female , Glucosides/blood , Glucosides/pharmacokinetics , Glucosides/urine , Glucuronides/metabolism , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/urine , Magnetic Resonance Spectroscopy , Male , Mice , Middle Aged , Rats , Rats, Sprague-Dawley , Species Specificity , Thiophenes/blood , Thiophenes/pharmacokinetics , Thiophenes/urine , Tissue Distribution , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...