Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 84
1.
Cancer Chemother Pharmacol ; 93(5): 439-453, 2024 May.
Article En | MEDLINE | ID: mdl-38270613

PURPOSE: Midostaurin, approved for treating FLT-3-mutated acute myeloid leukemia and advanced systemic mastocytosis, is metabolized by cytochrome P450 (CYP) 3A4 to two major metabolites, and may inhibit and/or induce CYP3A, CYP2B6, and CYP2C8. Two studies investigated the impact of midostaurin on CYP substrate drugs and oral contraceptives in healthy participants. METHODS: Using sentinel dosing for participants' safety, the effects of midostaurin at steady state following 25-day (Study 1) or 24-day (Study 2) dosing with 50 mg twice daily were evaluated on CYP substrates, midazolam (CYP3A4), bupropion (CYP2B6), and pioglitazone (CYP2C8) in Study 1; and monophasic oral contraceptives (containing ethinylestradiol [EES] and levonorgestrel [LVG]) in Study 2. RESULTS: In Study 1, midostaurin resulted in a 10% increase in midazolam peak plasma concentrations (Cmax), and 3-4% decrease in total exposures (AUC). Bupropion showed a 55% decrease in Cmax and 48-49% decrease in AUCs. Pioglitazone showed a 10% decrease in Cmax and 6% decrease in AUC. In Study 2, midostaurin resulted in a 26% increase in Cmax and 7-10% increase in AUC of EES; and a 19% increase in Cmax and 29-42% increase in AUC of LVG. Midostaurin 50 mg twice daily for 28 days ensured that steady-state concentrations of midostaurin and the active metabolites were achieved by the time of CYP substrate drugs or oral contraceptive dosing. No safety concerns were reported. CONCLUSION: Midostaurin neither inhibits nor induces CYP3A4 and CYP2C8, and weakly induces CYP2B6. Midostaurin at steady state has no clinically relevant PK interaction on hormonal contraceptives. All treatments were well tolerated.


Bupropion , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP3A , Drug Interactions , Midazolam , Staurosporine , Humans , Area Under Curve , Bupropion/pharmacokinetics , Bupropion/administration & dosage , Contraceptives, Oral/administration & dosage , Contraceptives, Oral/pharmacology , Contraceptives, Oral/pharmacokinetics , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2B6/genetics , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Combinations , Ethinyl Estradiol/pharmacokinetics , Ethinyl Estradiol/administration & dosage , Ethinyl Estradiol/pharmacology , Healthy Volunteers , Levonorgestrel/pharmacokinetics , Levonorgestrel/administration & dosage , Levonorgestrel/pharmacology , Midazolam/pharmacokinetics , Midazolam/administration & dosage , Pioglitazone/pharmacology , Pioglitazone/administration & dosage , Pioglitazone/pharmacokinetics , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Staurosporine/pharmacokinetics , Staurosporine/administration & dosage , Male , Female , Adolescent , Young Adult , Adult , Middle Aged
2.
Neuropharmacology ; 205: 108907, 2022 03 01.
Article En | MEDLINE | ID: mdl-34856203

Pioglitazone, an agonist at peroxisome proliferator-activated receptor gamma, is FDA-approved for the treatment of insulin resistance in type 2 diabetes. Numerous studies in male rodents suggest that pioglitazone inhibits inflammatory and neuropathic pain, but few included female subjects. To address this gap, we compared the effects of pioglitazone in both sexes in the intraplantar methylglyoxal model (MG) model of chemical pain and painful diabetic neuropathy (PDN), the plantar incision model (PIM) of postoperative pain, the spared nerve injury (SNI) model of traumatic nerve injury, and the ZDF rat and db/db mouse models of PDN. We administered pioglitazone by one-time intrathecal or intraperitoneal injection or by adding it to chow for 6 weeks, followed by measurement of hypersensitivity to non-noxious mechanical, noxious mechanical, heat, and/or cold stimuli. In all mouse models, injection of pioglitazone decreased pain-like behaviors with greater potency and/or efficacy in females as compared to males: heat and mechanical hypersensitivity in the MG model (0.1-10 mg/kg); mechanical hypersensitivity in the PIM model (10 µg); mechanical and cold hypersensitivity in the SNI model (100 mg/kg); and heat hypersensitivity in the db/db model (100 mg/kg). Furthermore, co-administration of low doses of morphine (1 mg/kg) and pioglitazone (10 mg/kg) decreased SNI-induced mechanical and cold hypersensitivity in female but not male mice. In the ZDF rat, pioglitazone (100 mg/kg) decreased heat and mechanical hypersensitivity with no sex difference. In the db/db model, pioglitazone had no effect when given into chow for 6 weeks at 0.3, 3 or 30 mg/kg doses. We conclude that females exhibit greater anti-hyperalgesic responses to pioglitazone in mouse models of chemical-induced nociception, postsurgical pain, neuropathic pain, and PDN. These findings set the stage for clinical trials to determine whether pioglitazone has analgesic properties across a broad spectrum of chronic pain conditions, particularly in women.


Analgesics/pharmacology , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Nociceptive Pain/drug therapy , PPAR gamma/agonists , Pain, Postoperative/drug therapy , Pioglitazone/pharmacology , Analgesics/administration & dosage , Animals , Diabetic Neuropathies/complications , Disease Models, Animal , Female , Male , Mice , Morphine/pharmacology , Neuralgia/etiology , Nociceptive Pain/chemically induced , Pain, Postoperative/etiology , Peripheral Nerve Injuries/complications , Pioglitazone/administration & dosage , Pyruvaldehyde/pharmacology , Sex Characteristics
3.
Respir Res ; 22(1): 230, 2021 Aug 19.
Article En | MEDLINE | ID: mdl-34412637

Agonists of peroxisome proliferator-activated receptor (PPAR)-γ have been suggested as potential adjuvant therapy in bacterial pneumonia because of their capacity to inhibit inflammation and enhance bacterial clearance. Previous studies only assessed the effects of pretreatment with these compounds, thereby bearing less relevance for the clinical scenario. Moreover, PPAR-γ agonists have not been studied in pneumonia caused by Klebsiella pneumoniae, a common human respiratory pathogen of which antibiotic treatment is hampered by increasing antimicrobial resistance. Here we show that administration of the PPAR-γ agonist pioglitazone 6 or 8 h after infection of mice with a highly virulent strain of Klebsiella pneumoniae via the airways results in reduced cytokine and myeloperoxidase levels in the lungs at 24 h after infection, as well as reduced bacterial growth in the lungs and decreased dissemination to distant organs at 42 h post-infection. These results suggest that pioglitazone may be an interesting agent in the treatment of Klebsiella pneumonia.


Klebsiella Infections/drug therapy , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , PPAR gamma/agonists , Pioglitazone/administration & dosage , Animals , Female , Inflammation/drug therapy , Inflammation/physiopathology , Injections, Intraperitoneal , Klebsiella Infections/physiopathology , Mice , Mice, Inbred C57BL
4.
Med Oncol ; 38(9): 100, 2021 Jul 24.
Article En | MEDLINE | ID: mdl-34302533

The Estudo de Descontinuação de Imatinibe após Pioglitazona (EDI-PIO) is a single-center, longitudinal, prospective, phase 2, non-randomized, open, clinical trial (NCT02852486, August 2, 2016 retrospectively registered) for the discontinuation of imatinib after concomitant use of pioglitazone, being the first of its kind in a Brazilian population with chronic myeloid leukemia. Due to remaining of leukemic quiescent cells that are not affected by tyrosine kinase inhibitors, it has been suggested the use of pioglitazone, a PPARγ agonist, together with imatinib as a strategy for the maintenance of deep molecular response. The clinical benefit to this association is still controversial, and the metabolic alteration along this process remains unclear. Therefore, we applied a metabolomic protocol using high-resolution mass spectrometry to profile plasmatic metabolic response of a prospective cohort of ten individuals under discontinuation of imatinib and pioglitazone protocol. By comparing patients under pioglitazone and imatinib treatment with imatinib monotherapy and discontinuation phase, we were able to annotate 41 and 36 metabolites, respectively. The metabolic alterations observed during imatinib-pioglitazone combined therapy are associated with an extensive lipid remodeling, with activation of ß-oxidation pathway, in addition to the presence of markers that suggest mitochondrial dysfunction.


Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Metabolic Diseases/pathology , Metabolome , Withholding Treatment , Adult , Aged , Female , Follow-Up Studies , Humans , Imatinib Mesylate/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Longitudinal Studies , Male , Metabolic Diseases/chemically induced , Metabolic Diseases/metabolism , Middle Aged , Non-Randomized Controlled Trials as Topic , Pioglitazone/administration & dosage , Prognosis , Prospective Studies , Retrospective Studies , Young Adult
5.
J Diabetes Investig ; 12(11): 2002-2009, 2021 Nov.
Article En | MEDLINE | ID: mdl-34002953

AIMS/INTRODUCTION: This study aimed to investigate whether insulin resistance (IR) in individuals with type 2 diabetes undergoing intensive glycemic control determines the extent of improvement in neuropathy. MATERIALS AND METHODS: This was an exploratory substudy of an open-label, randomized controlled trial of individuals with poorly controlled type 2 diabetes treated with exenatide and pioglitazone or insulin to achieve a glycated hemoglobin <7.0% (<53 mmol/mol). Baseline IR was defined using homeostasis model assessment of IR, and change in neuropathy was assessed using corneal confocal microscopy. RESULTS: A total of 38 individuals with type 2 diabetes aged 50.2 ± 8.5 years with (n = 25, 66%) and without (n = 13, 34%) IR were studied. There was a significant decrease in glycated hemoglobin (P < 0.0001), diastolic blood pressure (P < 0.0001), total cholesterol (P < 0.01) and low-density lipoprotein (P = 0.05), and an increase in bodyweight (P < 0.0001) with treatment. Individuals with homeostasis model assessment of IR <1.9 showed a significant increase in corneal nerve fiber density (P ≤ 0.01), length (P ≤ 0.01) and branch density (P ≤ 0.01), whereas individuals with homeostasis model assessment of IR ≥1.9 showed no change. IR was negatively associated with change in corneal nerve fiber density after adjusting for change in bodyweight (P < 0.05). CONCLUSIONS: Nerve regeneration might be limited in individuals with type 2 diabetes and IR undergoing treatment with pioglitazone plus exenatide or insulin to improve glycemic control.


Cornea/innervation , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/physiopathology , Insulin Resistance/physiology , Nerve Regeneration , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/blood , Diabetic Neuropathies/etiology , Exenatide/administration & dosage , Female , Glycated Hemoglobin/drug effects , Glycemic Control/methods , Humans , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Male , Microscopy, Confocal , Middle Aged , Nerve Fibers/pathology , Pioglitazone/administration & dosage , Treatment Outcome
6.
Drug Metab Dispos ; 49(7): 540-547, 2021 07.
Article En | MEDLINE | ID: mdl-33863817

Clinical induction liability is assessed with human hepatocytes. However, underpredictions in the magnitude of clinical induction have been reported. Unfortunately, in vivo studies in animals do not provide additional insight because of species differences in drug metabolizing enzymes and their regulatory pathways. To circumvent this limitation, transgenic animals expressing human orthologs were developed. The aim of this work was to investigate the utility of mouse models expressing human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 (Tg-Composite) in evaluating clinical induction. Rifampin, efavirenz, and pioglitazone, which were employed to represent strong, moderate, and weak inducers, were administered at multiple doses to Tg-Composite animals. In vivo CYP3A activity was monitored by measuring changes in the exposure of the CYP3A probe substrate triazolam. After the in vivo studies, microsomes were prepared from their livers to measure changes of in vitro CYP3A4 activity. In both in vivo and in vitro, distinction of clinic induction was recapitulated as rifampin yielded the greatest inductive effect followed by efavirenz and pioglitazone. Interestingly, with rifampin, in vivo CYP3A activity was approximately 4-fold higher than in vitro activity. Conversely, there was no difference between in vivo and in vitro CYP3A activity with efavirenz. These findings are consistent with the report that, although rifampin exhibits differential inductive effects between the intestines and liver, efavirenz does not. These data highlight the promise of transgenic models, such as Tg-Composite, to complement human hepatocytes to enhance the translatability of clinical induction as well as become a powerful tool to further study mechanisms of drug disposition. SIGNIFICANCE STATEMENT: Underprediction of the magnitude of clinical induction when using human hepatocytes has been reported, and transgenic models may improve clinical translatability. The work presented here showcases the human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 model, which was able to recapitulate the magnitude of clinical induction and to differentiate tissue-dependent induction observed with rifampin but not with efavirenz. These results not only foreshadow the potential application of such transgenic models in assessing clinical induction but also in further investigation of the mechanism of drug disposition.


Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Alkynes/administration & dosage , Alkynes/pharmacokinetics , Animals , Benzoxazines/administration & dosage , Benzoxazines/pharmacokinetics , Constitutive Androstane Receptor/genetics , Constitutive Androstane Receptor/metabolism , Cyclopropanes/administration & dosage , Cyclopropanes/pharmacokinetics , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inducers/administration & dosage , Drug Evaluation, Preclinical/methods , Drug Interactions , Feasibility Studies , Female , Humans , Mice , Mice, Transgenic , Microsomes, Liver , Pioglitazone/administration & dosage , Pioglitazone/pharmacokinetics , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism , Rifampin/administration & dosage , Rifampin/pharmacokinetics , Species Specificity , Triazolam/administration & dosage , Triazolam/pharmacokinetics
7.
Sci Rep ; 11(1): 8129, 2021 04 14.
Article En | MEDLINE | ID: mdl-33854134

Exposed rats to normal saline and paraquat (PQ) aerosol as control and PQ group, rats exposed to PQ and treated with 20 and 80 mg/kg/day carvacrol, 5 and 10 mg/kg/day pioglitazone, low dose of pioglitazone + carvacrol and 0.03 mg/kg/day dexamethasone (Dexa) for 16 days after the end of PQ exposure were studied (n = 6 in each group). Lung pathological changes, tracheal responsiveness to methacholine and ovalbumin (OVA) as well as transforming growth factor beta (TGF-ß) and interleukin (IL)-6 level in the lung tissue homogenize as well as TGF-ß, IL-6, oxidant and antioxidant levels oxidant and antioxidants were increased in PQ group (p < 0.01 to p < 0.001). Lung pathological changes, tracheal responsiveness to methacholine and OVA as well as TGF-ß, IL-6 oxidant and antioxidant levels were improved in all treated groups except lung pathological changes in treated group with low dose of pioglitazone (p < 0.05 to p < 0.001). The effects of low dose of pioglitazone and carvacrol alone were significantly lower than in the combination group of low dose of pioglitazone + carvacrol (p < 0.05 to p < 0.001). Carvacrol treatment improved inhaled PQ-induced lug injury similar to the effects of dexamethasone. The synergic effect of carvacrol and pioglitazone suggests PPAR-γ receptor mediated effects of carvacrol on inhaled PQ-induced lung injury.


Cymenes/administration & dosage , Dexamethasone/administration & dosage , Lung Injury/drug therapy , Paraquat/adverse effects , Pioglitazone/administration & dosage , Animals , Case-Control Studies , Cymenes/pharmacology , Dexamethasone/pharmacology , Disease Models, Animal , Drug Synergism , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Lung Injury/chemically induced , Lung Injury/immunology , Male , Oxidative Stress/drug effects , Pioglitazone/pharmacology , Rats , Rats, Wistar , Transforming Growth Factor beta/metabolism , Treatment Outcome
8.
Mol Pharm ; 18(4): 1711-1719, 2021 04 05.
Article En | MEDLINE | ID: mdl-33629861

An in vitro methodology for simulating the change in the pH and composition of gastrointestinal fluid associated with the transition of orally administered drugs from the stomach to the small intestine was developed (the stomach-to-intestine fluid changing system (the SIFC system)). This system was applied to in vitro sensitivity analysis on the dissolution of weakly basic drugs, and the obtained results were discussed in relation to the intrasubject variability in the plasma exposure in human bioequivalence (BE) study. Three types of protocols were employed (steep pH change: pH 1.6 FaSSGF → pH 6.5 FaSSIF, gradual pH change: pH 1.6 FaSSGF → pH 6.5 FaSSIF, and high gastric pH: pH 4.0 FaSSGF → pH 6.5 FaSSIF). Regardless of the protocols and the forms of drug applied in active pharmaceutical ingredient powder or formulation, dissolution profiles of pioglitazone after fluid shift were similar and the final concentrations in FaSSIF were approximately equal to the saturation solubility in FaSSIF, supporting its small intrasubject variance in human BE study. In contrast, dissolved concentration of terbinafine in the SIFC system became less than half in the high gastric pH protocol than that in other protocols, suggesting the fluctuation of gastric pH as one of the factors of high intrasubject variance of terbinafine in human. Plasma exposure of telmisartan was highly variable especially at the high dose. Although the dissolution of telmisartan in the SIFC system was greatly improved by formulation, it considerably fluctuated during fluid shift especially at the high dose, which corresponds well to in vivo results.


Body Fluids/chemistry , Gastric Mucosa/metabolism , Gastrointestinal Absorption/physiology , Intestinal Mucosa/metabolism , Administration, Oral , Biological Variation, Population , Chemistry, Pharmaceutical , Computer Simulation , Humans , Hydrogen-Ion Concentration , Permeability , Pioglitazone/administration & dosage , Pioglitazone/chemistry , Pioglitazone/pharmacokinetics , Solubility , Tablets , Taurocholic Acid/administration & dosage , Taurocholic Acid/pharmacokinetics , Telmisartan/administration & dosage , Telmisartan/pharmacokinetics , Terbinafine/administration & dosage , Terbinafine/chemistry , Terbinafine/pharmacokinetics
9.
JCI Insight ; 6(6)2021 03 22.
Article En | MEDLINE | ID: mdl-33571166

BACKGROUNDBeige and brown adipose tissue (BAT) are associated with improved metabolic homeostasis. We recently reported that the ß3-adrenergic receptor agonist mirabegron induced beige adipose tissue in obese insulin-resistant subjects, and this was accompanied by improved glucose metabolism. Here we evaluated pioglitazone treatment with a combination pioglitazone and mirabegron treatment and compared these with previously published data evaluating mirabegron treatment alone. Both drugs were used at FDA-approved dosages.METHODSWe measured BAT by PET CT scans, measured beige adipose tissue by immunohistochemistry, and comprehensively characterized glucose and lipid homeostasis and insulin sensitivity by euglycemic clamp and oral glucose tolerance tests. Subcutaneous white adipose tissue, muscle fiber type composition and capillary density, lipotoxicity, and systemic inflammation were evaluated by immunohistochemistry, gene expression profiling, mass spectroscopy, and ELISAs.RESULTSTreatment with pioglitazone or the combination of pioglitazone and mirabegron increased beige adipose tissue protein marker expression and improved insulin sensitivity and glucose homeostasis, but neither treatment induced BAT in these obese subjects. When the magnitude of the responses to the treatments was evaluated, mirabegron was found to be the most effective at inducing beige adipose tissue. Although monotherapy with either mirabegron or pioglitazone induced adipose beiging, combination treatment resulted in less beiging than either alone. The 3 treatments also had different effects on muscle fiber type switching and capillary density.CONCLUSIONThe addition of pioglitazone to mirabegron treatment does not enhance beiging or increase BAT in obese insulin-resistant research participants.TRIAL REGISTRATIONClinicalTrials.gov NCT02919176.FUNDINGNIH DK112282 and P20GM103527 and Clinical and Translational Science Awards grant UL1TR001998.


Acetanilides/pharmacology , Adipose Tissue, Beige/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Pioglitazone/pharmacology , Thiazoles/pharmacology , Acetanilides/administration & dosage , Drug Synergism , Female , Glucose Tolerance Test , Humans , Hypoglycemic Agents/administration & dosage , Insulin Resistance , Male , Middle Aged , Obesity/metabolism , Pioglitazone/administration & dosage , Thiazoles/administration & dosage
10.
Diabetologia ; 64(1): 159-167, 2021 01.
Article En | MEDLINE | ID: mdl-33001232

AIMS/HYPOTHESIS: In vitro and rodent studies suggest that pioglitazone, a thiazolidinedione, can promote adipogenesis in adipose tissue (AT); however, there is a lack of in vivo studies in humans to support these findings. The objectives of this randomised, placebo-controlled, parallel-arm trial were to test if pioglitazone stimulates in vivo adipogenesis in the subcutaneous adipose tissue depots and if these measures were related to metabolic health outcomes in women with obesity. METHODS: Forty-one healthy women with obesity (20 black; 21 white; 29 ± 6 years; BMI 32.0 ± 1.7 kg/m2; 44.0 ± 3.6% body fat) were randomised to consume 30 mg/day of pioglitazone (n = 21) or placebo (n = 20) for 16 weeks. SAS v9.4 was used to generate the block randomisation code sequence (stored in password-protected files) with a 1:1 allocation ratio. The participants and study staff involved in assessing and analysing data outcomes were blinded to the group assignments. The trial was conducted at Pennington Biomedical Research Center and ended in 2016. At baseline and post-intervention, subcutaneous abdominal (scABD) and femoral (scFEM) AT biopsies were collected, and in vivo cellular kinetics (primary endpoint of the trial) were assessed by an 8 week labelling protocol of deuterium (2H) into the DNA of adipose cells. Body composition was measured by dual-energy x-ray absorptiometry (DXA), scABD and visceral AT (VAT) by MRI, ectopic fat by 1H-MRS, and insulin sensitivity by an OGTT. RESULTS: After the 16 week intervention, there was a significant decrease in visceral fat (VAT:total abdominal AT [as a %]; p = 0.002) and an increase in the Matsuda index (i.e. improved insulin sensitivity; p = 0.04) in the pioglitazone group relative to the placebo group. A significant increase in the formation of new adipocytes was observed in the scFEM (Δ = 3.3 ± 1.6%; p = 0.04) but not the scABD depot (Δ = 2.0 ± 2.1%; p = 0.32) in the pioglitazone group relative to the placebo group. No serious adverse events were reported. CONCLUSIONS/INTERPRETATION: Pioglitazone may elicit distinct differences in in vivo adipogenesis in subcutaneous adipose depots in women with obesity, with increased rates in the protective scFEM. Trial registration ClinicalTrials.gov NCT01748994 Funding This study was funded by R01DK090607, P30DK072476, and R03DK112006 from the National Institute of Diabetes and Digestive and Kidney Diseases of the National Institutes of Health. U54 GM104940 from the National Institute of General Medical Sciences of the National Institutes of Health. The Robert C. and Veronica Atkins Foundation. Graphical abstract.


Adipogenesis/drug effects , Obesity/pathology , Pioglitazone/administration & dosage , Abdominal Fat/drug effects , Abdominal Fat/pathology , Adipocytes/pathology , Adult , Biopsy , Black People , Body Composition , Double-Blind Method , Female , Humans , Hypoglycemic Agents , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Obesity/metabolism , Placebos , Subcutaneous Fat/drug effects , Subcutaneous Fat/pathology , Waist-Hip Ratio , White People
11.
Mol Med ; 26(1): 128, 2020 12 11.
Article En | MEDLINE | ID: mdl-33308138

Autosomal dominant polycystic kidney disease (ADPKD) is an inherited chronic kidney disorder (CKD) that is characterized by the development of numerous fluid-filled cysts in kidneys. It is caused either due to the mutations in the PKD1 or PKD2 gene that encodes polycystin-1 and polycystin-2, respectively. This condition progresses into end-stage renal disorder if the renal or extra-renal clinical manifestations remain untreated. Several clinical trials with a variety of drugs have failed, and the only Food and Drugs Administration (FDA) approved drug to treat ADPKD to date is tolvaptan that works by antagonizing the vasopressin-2 receptor (V2R). The pathology of ADPKD is complex and involves the malfunction of different signaling pathways like cAMP, Hedgehog, and MAPK/ERK pathway owing to the mutated product that is polycystin-1 or 2. A measured yet substantial number of preclinical studies have found pioglitazone to decrease the cystic burden and improve the renal function in ADPKD. The peroxisome proliferator-activated receptor-gamma is found on the epithelial cells of renal collecting tubule and when it gets agonized by pioglitazone, confers efficacy in ADPKD treatment through multiple mechanisms. There is only one clinical trial (ongoing) wherein it is being assessed for its benefits and risk in patients with ADPKD, and is expected to get approval from the regulatory body owing to its promising therapeutic effects. This article would encompass the updated information on the epidemiology, pathophysiology of ADPKD, different mechanisms of action of pioglitazone in the treatment of ADPKD with preclinical and clinical shreds of evidence, and related safety updates.


Genetic Predisposition to Disease , Pioglitazone/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Animals , Biomarkers , Disease Management , Gene Expression Regulation , Genetic Association Studies , Humans , Mutation , Phenotype , Pioglitazone/administration & dosage , Pioglitazone/adverse effects , Polycystic Kidney, Autosomal Dominant/diagnosis , Polycystic Kidney, Autosomal Dominant/epidemiology , Signal Transduction , Treatment Outcome
12.
Drug Des Devel Ther ; 14: 4493-4502, 2020.
Article En | MEDLINE | ID: mdl-33122892

AIM: Evogliptin is a newly developed oral glucose-lowering medication of the dipeptidyl peptidase 4 (DPP-4) inhibitor class for type 2 diabetes mellitus. The combination of a DPP-4 inhibitor with pioglitazone is a promising therapeutic option. The aim of the present study was to evaluate the pharmacokinetic and pharmacodynamic interaction between evogliptin and pioglitazone. MATERIALS AND METHODS: A randomized, open-label, multiple-dose, three-treatment, three-period, six-sequence crossover study was conducted in healthy Korean male subjects. All subjects received evogliptin 5 mg once daily for 7 days (EVO), pioglitazone 30 mg once daily for 7 days (PIO) and co-administration of evogliptin 5 mg and pioglitazone 30 mg once daily for 7 days (EVO+PIO) according to the assigned sequence and period. Serial blood samples were collected for 24 hours for pharmacokinetic analysis and 3 hours after the oral glucose tolerance test for the pharmacodynamic analysis. RESULTS: Thirty-four subjects completed the study. EVO+PIO and EVO showed a similar maximum plasma concentration at steady state (Cmax,ss) and area under the concentration-time curve during the dosing interval at the steady state (AUCτ,ss) of evogliptin, with geometric mean ratios (GMRs) (90% confidence interval (CI)) of 1.01 (0.97-1.05) and 1.01 (0.98-1.04), respectively. EVO+PIO and PIO showed a similar Cmax,ss and AUCτ,ss of pioglitazone, with GMRs (90% CI) of 1.07 (0.99-1.17) and 1.08 (0.99-1.17), respectively. Reduction of the glucose level after EVO+PIO was larger compared to PIO and similar with EVO. CONCLUSION: Concomitant administration of evogliptin and pioglitazone showed similar glucose-lowering effects with those of evogliptin alone without pharmacokinetic interactions when compared to the intake of each drug alone.


Pioglitazone/pharmacokinetics , Piperazines/pharmacokinetics , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Male , Pioglitazone/administration & dosage , Pioglitazone/metabolism , Piperazines/administration & dosage , Piperazines/metabolism
13.
Biomed Pharmacother ; 132: 110741, 2020 Dec.
Article En | MEDLINE | ID: mdl-33038582

Capsaicin (N-vanillyl-8-methyl-alpha-nonenamide), a spicy, neurotoxic component of hot pepper is a ligand of vanilloid type-I (TRPV1) receptor of anti-cancer potential. However, molecular mechanism of its action is not fully understood. We found that capsaicin stimulated intrinsic and extrinsic pathway of apoptosis in human glioblastoma LN-18 cell line and this phenomenon was not dependent on TRPV1. Activation of peroxisome proliferator-activated receptor gamma (PPARγ), a ligand-dependent transcription factor, also induced apoptosis in glioblastoma cells. Although PPARγ ligands (thiazolidinediones - rosiglitazone, pioglitazone) promoted apoptosis in LN-18 cells, capsaicin augmented this effect. We found that capsaicin in a dose dependent manner induced expression of PPARγ in glioblastoma LN-18 cells. These findings suggest that capsaicin-dependent up-regulation of PPARγ represent the mechanism for augmentation of cell death by thiazolidinediones.


Apoptosis/drug effects , Capsaicin/pharmacology , Glioblastoma/drug therapy , Pioglitazone/pharmacology , Rosiglitazone/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Capsaicin/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Glioblastoma/pathology , Humans , Ligands , PPAR gamma/metabolism , Pioglitazone/administration & dosage , Rosiglitazone/administration & dosage , TRPV Cation Channels/metabolism , Up-Regulation/drug effects
14.
J Stroke Cerebrovasc Dis ; 29(10): 105048, 2020 Oct.
Article En | MEDLINE | ID: mdl-32912514

OBJECTIVE: Standards for reporting and analyzing adherence to medical therapy have recently improved due to international consensus efforts. If applied to clinical trial research in patients with stroke, these improvements have the potential to identify when in the sequence of trial operations participants are at risk for non-adherence and opportunities to safeguard adherence. METHODS: We analyzed three phases of adherence according to the European Society for Patient Adherence, COMpliance, and Persistence (ESPACOMP) Medication Adherence Reporting Guideline (EMERGE) taxonomy in the Insulin Resistance Intervention after Stroke (IRIS) trial: initiation (did patient start drug), implementation (did patient take a drug holiday, defined as temporary cessation lasting ≥14 days), and persistence (did patient prematurely and permanently discontinue drug). IRIS was a randomized, placebo controlled, double-blind trial testing pioglitazone to prevent stroke or myocardial infarction in patients with a recent ischemic stroke or transient ischemic attack. Adherence was classified by self-report. Researchers used coaching algorithms to seek adherence recovery if participants went off drug. RESULTS: During 2005-2013, 3876 participants were enrolled from 179 sites in seven countries and followed for a mean of 4.8 years. Less than 1% of participants in each group did not initiate study drug. 20% of patients assigned to pioglitazone and 17% assigned to placebo took at least one drug holiday. 36% and 30%, respectively, discontinued the study drug prematurely with or without a prior holiday. The risk for stopping the study drug (temporarily or permanently) in the first year after randomization was twice the risk in each of the subsequent four years. This was true both for patients assigned to active therapy and placebo. More participants assigned to pioglitazone, compared to placebo, took a drug holiday or permanently stopped study drug, but the difference in rates of discontinuation was only evident in year one. In years two through five, rates of discontinuation were similar in the two treatment groups. The difference in rates during year one was the result of adverse effects related to the active study drug, pioglitazone. During the remainder of the trial, the attribution of discontinuations to adverse effects potentially related to pioglitazone was reduced but still higher in those assigned to active drug. Other reasons for discontinuation were similar between treatment groups and were largely unrelated to pharmacodynamic effects of the study drug. Rates of discontinuation varied widely among research sites. CONCLUSION: Patients in a drug trial for stroke prevention are at greatest risk for premature drug discontinuation early after randomization. Reasons for discontinuation change over time. Variable discontinuation rates among sites suggests that adherence can be improved by using best practices from high-performing sites.


Hypoglycemic Agents/administration & dosage , Insulin Resistance , Ischemic Attack, Transient/prevention & control , Medication Adherence , Myocardial Infarction/prevention & control , Pioglitazone/administration & dosage , Secondary Prevention , Stroke/prevention & control , Double-Blind Method , Drug Administration Schedule , Female , Humans , Hypoglycemic Agents/adverse effects , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/physiopathology , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology , Pioglitazone/adverse effects , Recurrence , Risk Factors , Stroke/etiology , Stroke/physiopathology , Time Factors , Treatment Outcome
15.
Biomed Pharmacother ; 131: 110692, 2020 Nov.
Article En | MEDLINE | ID: mdl-32942156

BACKGROUND AND OBJECTIVES: Bone cancer pain (BCP) remains a difficult clinical problem. This study examined whether pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, is effective for attenuating BCP, and investigated the interaction between activation of PPARγ and phosphatase and tensin homolog deleted from chromosome 10 (PTEN) / mammalian target of rapamycin (mTOR) signal in the spinal dorsal horn (SDH) of BCP rats. METHODS: We tested the effects of intrathecal (i.t.) injection of adenovirus-mediated PTEN (Ad-PTEN), PTEN antisense oligonucleotide (Ad-antisense PTEN), mTOR inhibitor rapamycin, pioglitazone and PPARγ antagonist GW9662 on bone cancer-induced mechanical allodynia by measuring the paw withdrawal threshold (PWT). Western blot or immunofluorescence examined the expression of spinal PPARγ, PTEN, mTOR, p-mTOR and p-S6K1. RESULTS: Bone cancer did not alter total mTOR expression but caused significant downregulation of PTEN and upregulation of p-mTOR and p-S6K1 in spinal neurons. Rapamycin markedly reduced BCP. Upregulation of spinal PTEN by i.t. Ad-PTEN significantly relieved BCP and downregulated p-mTOR and p-S6K1; while i.t. Ad-antisense PTEN led to the opposite effects of Ad-PTEN. Spinal PPARγ expression increased in BCP rats, co-localizing mainly with neurons and a few astrocytes, but not in microglia. Pioglitazone (500 µg/day i.t. for one week, from 7 days after surgery) attenuated BCP, further increased expression of PPARγ, and inhibited downregulation of PTEN and upregulation of p-mTOR and p-S6K1 in the SDH. Pioglitazone's analgesic effect was enhanced by Ad-PTEN and attenuated by Ad-antisense PTEN. Blockade of PPARγ with GW9662 (300 µg i.t. 15 min prior to pioglitazone) reversed the effects of pioglitazone on BCP and regulations of PPARγ/PTEN/mTOR signal. CONCLUSIONS: Intrathecal pioglitazone administration alleviates BCP by regulating the PPARγ/PTEN/mTOR signal in the SDH. Our data provided new insight in the therapeutic strategy in BCP management.


Bone Neoplasms/metabolism , Cancer Pain/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Pioglitazone/administration & dosage , Spinal Cord Dorsal Horn/metabolism , TOR Serine-Threonine Kinases/metabolism , Analgesics/administration & dosage , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cancer Pain/drug therapy , Cancer Pain/pathology , Female , Injections, Spinal , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology
16.
Am J Physiol Endocrinol Metab ; 319(5): E835-E851, 2020 11 01.
Article En | MEDLINE | ID: mdl-32865011

Cardiac autonomic neuropathy (CAN) is an early cardiovascular manifestation of type 2 diabetes (T2D) that constitutes an independent risk factor for cardiovascular mortality and morbidity. Nevertheless, its underlying pathophysiology remains poorly understood. We recently showed that localized perivascular adipose tissue (PVAT) inflammation underlies the incidence of parasympathetic CAN in prediabetes. Here, we extend our investigation to provide a mechanistic framework for the evolution of autonomic impairment as the metabolic insult worsens. Early metabolic dysfunction was induced in rats fed a mild hypercaloric diet. Two low-dose streptozotocin injections were used to evoke a state of late decompensated T2D. Cardiac autonomic function was assessed by invasive measurement of baroreflex sensitivity using the vasoactive method. Progression into T2D was associated with aggravation of CAN to include both sympathetic and parasympathetic arms. Unlike prediabetic rats, T2D rats showed markers of brainstem neuronal injury and inflammation as well as increased serum levels of IL-1ß. Experiments on PC12 cells differentiated into sympathetic-like neurons demonstrated that brainstem injury observed in T2D rats resulted from exposure to possible proinflammatory mediators in rat serum rather than a direct effect of the altered metabolic profile. CAN and the associated cardiovascular damage in T2D only responded to combined treatment with insulin to manage hyperglycemia in addition to a nonhypoglycemic dose of metformin or pioglitazone providing an anti-inflammatory effect, coincident with the effect of these combinations on serum IL-1ß. Our present results indicate that CAN worsening upon progression to T2D involves brainstem inflammatory changes likely triggered by systemic inflammation.


Baroreflex/physiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Neuropathies/physiopathology , Hypoglycemic Agents/therapeutic use , Inflammation/physiopathology , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/blood , Diabetic Neuropathies/drug therapy , Disease Progression , Hemodynamics/drug effects , Hemodynamics/physiology , Hypoglycemic Agents/administration & dosage , Inflammation/blood , Inflammation/drug therapy , Insulin/administration & dosage , Insulin/therapeutic use , Interleukin-1beta/blood , Male , Pioglitazone/administration & dosage , Pioglitazone/therapeutic use , Rats , Rats, Sprague-Dawley
18.
Nat Commun ; 11(1): 4448, 2020 09 07.
Article En | MEDLINE | ID: mdl-32895370

Substance abuse disorders are linked to alteration of circadian rhythms, although the molecular and neuronal pathways implicated have not been fully elucidated. Addictive drugs, such as cocaine, induce a rapid increase of dopamine levels in the brain. Here, we show that acute administration of cocaine triggers reprogramming in circadian gene expression in the striatum, an area involved in psychomotor and rewarding effects of drugs. This process involves the activation of peroxisome protein activator receptor gamma (PPARγ), a nuclear receptor involved in inflammatory responses. PPARγ reprogramming is altered in mice with cell-specific ablation of the dopamine D2 receptor (D2R) in the striatal medium spiny neurons (MSNs) (iMSN-D2RKO). Administration of a specific PPARγ agonist in iMSN-D2RKO mice elicits substantial rescue of cocaine-dependent control of circadian genes. These findings have potential implications for development of strategies to treat substance abuse disorders.


Circadian Clocks/drug effects , Cocaine-Related Disorders/physiopathology , Cocaine/adverse effects , Nucleus Accumbens/drug effects , PPAR gamma/metabolism , Receptors, Dopamine D2/metabolism , Administration, Oral , Animals , Circadian Clocks/physiology , Cocaine/administration & dosage , Cocaine-Related Disorders/drug therapy , Dopamine/metabolism , Injections, Intraperitoneal , Locomotion/physiology , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/physiopathology , PPAR gamma/agonists , Pioglitazone/administration & dosage , Receptors, Dopamine D2/genetics , Reward , Signal Transduction
19.
Physiol Rep ; 8(15): e14515, 2020 08.
Article En | MEDLINE | ID: mdl-32776495

BACKGROUND: Thrombosis is a potentially life-threatening nephrotic syndrome (NS) complication. We have previously demonstrated that hypercoagulopathy is proportional to NS severity in rat models and that pioglitazone (Pio) reduces proteinuria both independently and in combination with methylprednisolone (MP), a glucocorticoid (GC). However, the effect of these treatments on NS-associated hypercoagulopathy remains unknown. We thus sought to determine the ability of Pio and GC to alleviate NS-associated hypercoagulopathy. METHODS: Puromycin aminonucleoside-induced rat NS was treated with sham, Low- or High-dose MP, Pio, or combination (Pio + Low-MP) and plasma was collected at day 11. Plasma samples were collected from children with steroid-sensitive NS (SSNS) and steroid-resistant NS (SRNS) upon presentation and after 7 weeks of GC therapy. Plasma endogenous thrombin potential (ETP), antithrombin (AT) activity, and albumin (Alb) were measured using thrombin generation, amidolytic, and colorimetric assays, respectively. RESULTS: In a rat model of NS, both High-MP and Pio improved proteinuria and corrected hypoalbuminemia, ETP and AT activity (p < .05). Proteinuria (p = .005) and hypoalbuminemia (p < .001) were correlated with ETP. In childhood NS, while ETP was not different at presentation, GC therapy improved proteinuria, hypoalbuminemia, and ETP in children with SSNS (p < .001) but not SRNS (p = .330). CONCLUSIONS: Both Pio and GC diminish proteinuria and significantly alleviate hypercoagulopathy. Both Pio and MP improved hypercoagulopathy in rats, and successful GC therapy (SSNS) also improved hypercoagulopathy in childhood NS. These data suggest that even a partial reduction in proteinuria may reduce NS-associated thrombotic risk.


Blood Coagulation/drug effects , Glucocorticoids/therapeutic use , Nephrotic Syndrome/drug therapy , Pioglitazone/therapeutic use , Thrombosis/drug therapy , Animals , Child , Female , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacology , Humans , Male , Nephrotic Syndrome/complications , PPAR gamma/agonists , Pioglitazone/administration & dosage , Pioglitazone/pharmacology , Rats , Rats, Wistar , Receptors, Glucocorticoid/agonists , Thrombosis/etiology
20.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article En | MEDLINE | ID: mdl-32707656

Peroxisome proliferator-activated receptor alpha (PPARα) and gamma (PPARγ) agonists have anti-inflammatory and anti-neovascularization effects, but few reports have tested the combination of PPARα and PPARγ agonists. In this study, we investigated the therapeutic effects of ophthalmic solutions of agonists of PPARα, PPARγ, and the combination in a rat corneal alkali burn model. After alkali injury, an ophthalmic solution of 0.05% fenofibrate (PPARα group), 0.1% pioglitazone (PPARγ group), 0.05% fenofibrate + 0.1% pioglitazone (PPARα+γ group), or vehicle (vehicle group) was topically instilled onto the rat's cornea twice a day. After instillation, upregulation was seen of PPAR mRNA corresponding to each agonist group. Administration of agonists for PPARα, PPARγ, and PPARα+γ suppressed inflammatory cells, neovascularization, and fibrotic changes. In addition, the PPARγ agonist upregulated M2 macrophages, which contributed to wound healing, whereas the PPARα agonist suppressed immature blood vessels in the early phase. Administration of PPARα+γ agonists showed therapeutic effects in corneal wound healing, combining the characteristics of both PPARα and PPARγ agonists. The results indicate that the combination of PPARα and γ agonists may be a new therapeutic strategy.


Burns, Chemical/drug therapy , Corneal Injuries/drug therapy , Eye Burns/drug therapy , PPAR alpha/agonists , PPAR gamma/agonists , Animals , Burns, Chemical/metabolism , Burns, Chemical/pathology , Corneal Injuries/metabolism , Corneal Injuries/pathology , Corneal Neovascularization/prevention & control , Cytokines/genetics , Disease Models, Animal , Drug Therapy, Combination , Eye Burns/metabolism , Eye Burns/pathology , Fenofibrate/administration & dosage , Fibrosis , Keratitis/prevention & control , Male , Ophthalmic Solutions , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Pioglitazone/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
...