Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kwang-Hee Shin is active.

Publication


Featured researches published by Kwang-Hee Shin.


Clinical Therapeutics | 2008

Pharmacokinetics, pharmacodynamics, and tolerability of the dipeptidyl peptidase IV inhibitor LC15-0444 in healthy Korean men: a dose-block-randomized, double-blind, placebo-controlled, ascending single-dose, Phase I study.

Kyoung Soo Lim; J. Kim; Yunjung Choi; Kwang-Hee Shin; K. H. Kim; Jang-Hee Hong; Joo-Youn Cho; Hyun-Suk Shin; Kyung-Sang Yu; Sang-Goo Shin; O Hwan Kwon; Dal-Mi Hwang; Jeong-Ae Kim; In-Jin Jang

BACKGROUND LC15-0444 is a selective inhibitor of dipeptidyl peptidase (DPP) IV under investigation in Korea for the treatment of type 2 diabetes. OBJECTIVE The aim of this study was to investigate the pharmacokinetic (PK), pharmacodynamic (PD), and tolerability profiles of a single dose of LC15-0444 in healthy male subjects. METHODS A dose-block-randomized, double-blind, placebo-controlled, ascending single-dose, Phase I study was performed in healthy Korean male subjects assigned to receive 25, 50, 100, 200, 400, or 600 mg of LC15-0444 capsules. Blood and urine samples were collected up to 72 hours after administration. Plasma and urine drug concentrations were determined by tandem mass spectrometry coupled with high-performance liquid chromatography. DPP IV activity was measured by continuous spectrophotometric assay. An additional food effect study was performed in the 100-mg dose group; changes in PK and PD parameters after high-fat diet were evaluated. Adverse events (AEs) were detected through investigator inquiries, spontaneous reports, and clinical evaluations such as physical examinations, vital sign measurements, 12-lead electrocardiography, clinical laboratory tests (eg, hematology, blood chemistry, coagulation, urinalysis), and computerized impedance cardiography. RESULTS Sixty Korean men (mean age, 25.3 years [range, 19-39 years]; weight, 68.3 kg [range, 53.6-84.9 kg]) were enrolled, providing 10 subjects for each dose group. After administration, LC15-0444 reached T(max) at 0.5 to 5.1 hours, and was eliminated with a t((1/2)) of 16.7 to 21.3 hours. The mean fraction of unchanged drug excreted in urine ranged from 0.21 to 0.34 and mean renal clearance was 15.5 to 23.6 L/h. The dose-normalized AUC exhibited dose-linearity over the range of 50 to 400 mg. All doses of LC15-0444 =200 mg were found to inhibit 80% of DPP IV activity for 24 hours. High-fat diet did not significantly influence the AUC of LC15-0444. LC15-0444 was generally well tolerated. None of the subjects developed any serious clinical or laboratory AEs or discontinued the study due to an AE. All AEs were mild or moderate, and no dose-related trends were observed. Forty-six AEs were reported in 18 subjects (30.0%). AEs considered to be related to the study drug were headache (6 cases), dizziness (2), nausea (1), epistaxis (1), and increased heart rate (1). All AEs resolved spontaneously. CONCLUSIONS A single dose of LC15-0444 exhibited linear PK properties over the range of 50 to 400 mg in these healthy Korean male subjects. PK characteristics were not significantly influenced by food. In addition, doses >or=200 mg of LC15-0444 inhibited plasma DPP IV activity by >80% over a 24-hour dosing interval, and a 600-mg dose increased active glucagon-like peptide-1 levels after a standardized meal. LC15-0444 was generally well tolerated.


Therapeutic Drug Monitoring | 2012

Reduced valproic acid serum concentrations due to drug interactions with carbapenem antibiotics: overview of 6 cases.

Min Kyu Park; Kyoung Soo Lim; Tae-Eun Kim; HyeKyung Han; SoJeong Yi; Kwang-Hee Shin; Joo-Youn Cho; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

Background: The plasma concentrations of valproic acid (VPA) are known to decrease during the concomitant administration of carbapenem antibiotics, such as meropenem, imipenem, and ertapenem. This study summarizes 6 cases of drug–drug interactions between VPA and carbapenem antibiotics. Methods: To investigate the onset and severity of the reductions in the concentration of VPA in patients with or without the coadministration of carbapenem antibiotics, the authors performed a retrospective evaluation of therapeutic drug monitoring (TDM) reports that described a decrease in the serum concentrations of VPA during the concomitant use of carbapenem antibiotics from January 2008 to December 2010 in the Seoul National University Hospital. The evaluated TDM reports included 6 cases. The decrement ratio of the VPA serum concentration was calculated from the TDM reports, and the change in the half-life of the VPA was also estimated. Results: Six cases presented with changes in the VPA serum concentration before and after the administration of carbapenem antibiotics. (Three cases were treated with meropenem, 2 were treated with ertapenem, and 1 was treated with imipenem.) The VPA concentrations reduced by (mean ± SD) 88.7 ± 5.3% (3 cases of meropenem), 74.0 ± 9.8% (2 cases of ertapenem), and 73.3% (1 case of imipenem), respectively, and the half-life of VPA reduced by 80.1 ± 9.0%, 64.4 ± 24.2%, and 50.6%, respectively. Conclusion: The interaction between VPA and carbapenem antibiotics caused decreases in the VPA serum concentrations; the extent of this decrease was greater in the meropenem-treated patients than in the imipenem-treated or ertapenem-treated cases. Because the therapeutic effect of VPA depends on its serum concentration, it should be recognized that there may be a loss of seizure control in patients using VPA with carbapenem antibiotics.


Clinical Therapeutics | 2011

Tolerability and Pharmacokinetics of Lobeglitazone (CKD-501), a Peroxisome Proliferator-Activated Receptor-γ Agonist: A Single- and Multiple-Dose, Double-Blind, Randomized Control Study in Healthy Male Korean Subjects

Jung Won Kim; J. Kim; SoJeong Yi; Kwang-Hee Shin; Hyun-Suk Shin; Seo Hyun Yoon; Joo-Youn Cho; Dal-Hyun Kim; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

BACKGROUND Lobeglitazone, a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist with partial PPAR-α affinity, was developed to treat type 2 diabetes mellitus. OBJECTIVE This studys aim was to evaluate the tolerability and pharmacokinetic (PK) properties of lobeglitazone to satisfy regulatory requirements for marketing approval in Korea. METHODS A block-randomized, double-blind, placebo-controlled, single- and multiple-dose study was conducted in healthy subjects. In the pilot study, 4 subjects were administered 0.5 mg, including 1 receiving a placebo. Then, the single-dose study was conducted with 1, 2, 4, and 8 mg doses (8 subjects in each group, including 2 receiving placebos), followed by the multiple-dose study with 1, 2, and 4 mg doses (once daily for 7 days; 8 subjects in each group, including 2 receiving placebos). Serial samples of blood and urine were collected and drug concentrations were determined by high turbulence liquid chromatography-LC/MS/MS. Tolerability assessments were performed throughout the study. Adverse events (AEs) were determined from general health-related questions and self-reports. RESULTS Thirty-six (mean [SD]; age, 23.6 [2.7] years; weight, 70.0 [6.9] kg) and 25 Korean male subjects (age, 23.5 [3.1] years; weight 69.4 [9.4] kg) were enrolled in the single- and multiple-dose studies, respectively. The data from subjects administered lobeglitazone who completed the study (27, single; 18, multiple) was included in the PK analyses. In the single-dose study, the AUC and C(max) of lobeglitazone increased with the dose. After repeated dosing for 7 days, the accumulation ratio ranged from 1.1 to 1.4. A total of 25 AEs were reported by 11 (30.6%) and 8 subjects (33.3%) in the single- and multiple-dose studies, respectively. All AEs were mild in intensity and not serious. CONCLUSIONS Lobeglitazone was well tolerated in this small, selected group of healthy male Korean volunteers. The AUC and C(max) of lobeglitazone increased in a dose-proportional manner from 1 to 4 mg.


Journal of Cardiovascular Pharmacology | 2011

The effect of the newly developed angiotensin receptor II antagonist fimasartan on the pharmacokinetics of atorvastatin in relation to OATP1B1 in healthy male volunteers

Kwang-Hee Shin; Tae-Eun Kim; Sung Eun Kim; Min Goo Lee; Im-Sook Song; Seo Hyun Yoon; Joo-Youn Cho; In-Jin Jang; Sang-Goo Shin; Kyung-Sang Yu

Objective Interactions between coadministered drugs may unfavorably affect pharmacokinetics. This study evaluated whether fimasartan, an angiotensin receptor II antagonist, affected the pharmacokinetics of atorvastatin. Methods A randomized, open-label, 2-period, 2-sequence, crossover, multiple-dosing study was conducted with 24 healthy male volunteers. Twelve subjects received 80-mg atorvastatin once daily for 7 days; later, they received 80-mg atorvastatin with 240-mg fimasartan for 7 days. Twelve other subjects received the same drugs in the opposite sequence. Blood samples were collected scheduled intervals for 24 hours after the last dosing to determine plasma concentrations of atorvastatin acid, atorvastatin lactone, 2-hydroxy atorvastatin acid, and 2-hydroxy atorvastatin lactone. Results Compared with atorvastatin alone, coadministration of fimasartan and atorvastatin increased the atorvastatin acid mean (95% confidence interval) maximum concentration (Cmax,ss) by 1.89-fold (1.49–2.39) and the area under the concentration curve (AUC&tgr;,ss) by 1.19-fold (0.96–1.48). Fimasartan also increased the mean 2-hydroxy atorvastatin acid Cmax,ss and AUC&tgr;,ss by 2.45-fold (1.80–3.35) and 1.42-fold (1.09–1.85), respectively. The Cmax,ss and AUC&tgr;,ss of the lactone forms of atorvastatin showed smaller changes than those observed for the acidic forms. Conclusion We showed that fimasartan raised plasma atorvastatin concentrations. In vitro tests suggested that this effect may have been mediated by fimasartan inhibition of organic anion-transporting polypeptide 1B1.


Journal of Cardiovascular Pharmacology | 2012

Assessment of the drug-drug interactions between fimasartan and hydrochlorothiazide in healthy volunteers.

Hyewon Jeon; Kyoung Soo Lim; Kwang-Hee Shin; JaeWoo Kim; Seo Hyun Yoon; Joo-Youn Cho; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

Aim Fimasartan is a selective angiotensin II receptor blocker. Hydrochlorothiazide (HCTZ), which is used to treat hypertension and edematous conditions, is coadministered with many antihypertensive agents. Methods An open-label, randomized, multiple-dosing, 2-arm, 1-sequence, 2-period study was conducted to assess the effects of fimasartan (240 mg) on HCTZ (25 mg) or vice versa in 18 and 14 healthy male volunteers, respectively. During each drug administration period, drugs were given once daily for 7 days, with a 7-day washout period between the 2 administration periods. Results The respective geometric mean ratios of fimasartan for AUC&tgr;,ss and Cmax,ss with HCTZ were 1.30 [90% confidence interval (CI), 0.84–2.01] and 1.17 (90% CI, 0.93–1.47) compared with fimasartan alone. The respective geometric mean ratios of HCTZ for AUC&tgr;,ss and Cmax,ss with fimasartan were 0.94 (90% CI, 0.84–1.04) and 0.88 (90% CI, 0.80–0.97) compared with HCTZ alone. Plasma renin activity indicated no significant differences between fimasartan monotherapy and coadministered treatment. Conclusions Fimasartan administered alone or in combination with HCTZ was well tolerated at the described dosages. Coadministration of fimasartan increased the urinary excretion of HCTZ and urine volume, but these observations are unlikely to have any clinical relevance.


Clinical Therapeutics | 2009

The Effects of Ketoconazole and Rifampicin on the Pharmacokinetics of Mirodenafil in Healthy Korean Male Volunteers: An Open-Label, One-Sequence, Three-Period, Three-Treatment Crossover Study

Kwang-Hee Shin; Bo-Hyung Kim; Tae-Eun Kim; JaeWoo Kim; SoJeong Yi; Seo-Hyun Yoon; Joo-Youn Cho; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

BACKGROUND Mirodenafil, a phosphodiesterase 5 inhibitor reported to be effective in the treatment of erectile dysfunction, is metabolized by cytochrome P450 (CYP) 3A4 to the active metabolite N-dehydroxyethyl mirodenafil. Mirodenafil may have drug-drug interactions with ketoconazole and/or rifampicin. OBJECTIVE The aim of this study was to investigate the effects of a potent inhibitor (ketoconazole) and inducer (rifampicin) of the CYP3A4 isozyme on the pharmacokinetics of mirodenafil to meet the regulatory requirements for the marketing of mirodenafil in Korea. METHODS An open-label, 1-sequence, 3-period, 3-treatment crossover study was conducted over 22 days in healthy Korean male volunteers. Each subject received 100 mg of mirodenafil in each of 3 study periods: mirodenafil alone (period 1); mirodenafil after pretreatment with ketoconazole 400 mg once daily for 3 days (period 2); and mirodenafil after pretreatment with rifampicin 600 mg once daily for 10 days (period 3). Serial blood samples were collected for pharmacokinetic analysis after the administration of mirodenafil in each study period. Plasma concentration-time data for mirodenafil and its major metabolite, N-dehydroxyethyl mirodenafil, were determined using LC-MS/MS and analyzed by a noncompartmental method. The results for mirodenafil coadministration with either ketoconazole or rifampicin were compared with those for mirodenafil alone. Adverse events (AEs) were identified by asking general health-related questions of the subjects, by physical examination, and by subject self-report throughout the study period. RESULTS Nineteen subjects were enrolled (mean [SD] age, 23.2 [2.76] years [range, 19-29 years]; weight, 69.3 [6.50] kg [range, 61.0-84.0 kg]; body mass index, 22.4 [1.77] kg/m(2) [range, 20.0-26.0 kg/m(2)]) and 18 subjects completed the study. One subject discontinued the study due to protocol violation and was replaced. The AUC(0-infinity) of mirodenafil increased 5.04-fold (90% CI, 3.78-6.72) and the metabolic ratio decreased 0.21-fold after pretreatment with ketoconazole compared with mirodenafil alone. After pretreatment with rifampicin, the AUC(0-infinity) of mirodenafil decreased 0.03-fold (90% CI, 0.02-0.05) and the metabolic ratio increased 2.9-fold. Twelve cases of headache, 6 of nasal congestion, 2 of feeling hot, 2 of epistaxis, and 1 each of dizziness, nausea, and somnolence were considered to be related to administration of mirodenafil. Twenty-eight AEs were reported in period 2 (in 68.4% of subjects), during which systemic exposure to mirodenafil was highest, whereas 7 AEs were reported in period 1 (in 31.6% of subjects) and 5 AEs in period 3 (in 16.7% of subjects). CONCLUSION In these healthy Korean male volunteers, the coadministration of ketoconazole and rifampicin resulted in significant changes in systemic exposure to mirodenafil.


Clinical Therapeutics | 2009

Pharmacokinetic Comparison of a New Glimepiride 1-mg + Metformin 500-mg Combination Tablet Formulation and a Glimepiride 2-mg + Metformin 500-mg Combination Tablet Formulation: A Single-Dose, Randomized, Open-Label, Two-Period, Two-Way Crossover Study in Healthy, Fasting Korean Male Volunteers

Bo-Hyung Kim; Kwang-Hee Shin; JaeWoo Kim; Kyoung Soo Lim; K. H. Kim; J. Kim; Joo-Youn Cho; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

BACKGROUND Coadministration of glimepiride and metformin has been used to achieve glucose control. Because compliance with a multiple medication regimen can be difficult for some patients, combination tablets of glimepiride + metformin might be a suitable alternative for these patients. OBJECTIVE This study was conducted to compare the pharmacokinetics of test and reference formulations of glimepiride + metformin fixed-dose combination tablets under fasting conditions to meet the regulatory requirements for marketing approval of a new drug in Korea. METHODS This was a single-dose, randomized, open-label, 2-period, 2-way crossover study conducted between March 2007 and May 2007. Healthy fasting Korean men were randomized to 1 of 2 dosing sequences: a single oral administration of a fixed-dose glimepiride 1-mg + metformin 500-mg combination tablet (test) followed by single oral administration of a fixed-dose glimepiride 2 mg + metformin 500 mg combination tablet (reference), separated by a 1-week washout period between doses; or a single oral administration of a fixed-dose glimepiride 2-mg + metformin 500-mg combination tablet followed by single oral administration of a fixed-dose glimepiride 1 mg + metformin 500-mg combination tablet, separated by a 1-week washout period between doses. Serial samples of blood were collected up to 24 hours after oral administration, and drug concentrations in plasma were determined by HPLC-MS/MS. Tolerability was assessed based on adverse events and changes in clinical parameters. Serious adverse events included those that resulted in death, a life-threatening condition, congenital anomaly or birth defect, or required hospitalization or prolongation of existing hospitalization. RESULTS A total of 30 healthy male subjects (mean age, 25.6 years [range, 20-36 years]; weight, 69.5 kg [range, 58.2-90.7 kg]) participated in the study. After administration of the test and reference formulations, glimepiride was rapidly absorbed, reaching C(max) with a median T(max) of 1.75 and 2.0 hours, respectively, and then declined exponentially with an average t(1/2) of 8.2 and 8.5 hours. The individual C(max) and AUC(last) of glimepiride were observed to be proportionally increased according to the administered glimepiride dose. The mean (SD) dose-normalized Cmax values of glimepiride 1 and 2 mg were 168.2 (54.9) and 149.9 (47.4) ng/mL/mg, respectively; the mean dose-normalized AUC(last) values of glimepiride 1 and 2 mg were 681.5 (190.3) and 635.8 (194.1) ng x h/mL/mg. Individual plots of dose-normalized C(max) and AUC(last) values identified a similarity between the 2 groups but no significant between-group differences. A total of 25 adverse events (12 after the test dose and 13 after the reference dose) were reported by 13 of the 30 subjects. All adverse events were considered mild. Twenty-one adverse events were considered related to the study drug (8 after the test dose and 13 after the reference dose). Adverse events believed to be related to the test formulation were diarrhea (4 cases), dizziness (1), headache (1), tingling sensation (1), and weakness (1). Adverse events believed to be related to the reference formulation were diarrhea (6 cases), headache (3), cold sweats (1), dyspepsia (1), epigastric discomfort (1), and lethargy (1). There were no clinically significant findings in the laboratory test results or vital sign monitoring during the study. There were no serious adverse events reported. CONCLUSIONS The C(max) and AUC(last) of glimepiride increased proportionally according to the administered glimepiride dose in this study of healthy, fasting Korean men. The safety profiles of the 2 combination tablets were comparable.


Current Medical Research and Opinion | 2013

Comparison of pharmacokinetics between new quinolone antibiotics: the zabofloxacin hydrochloride capsule and the zabofloxacin aspartate tablet

HyeKyung Han; Sung Eun Kim; Kwang-Hee Shin; Cheolhee Lim; Kyoung Soo Lim; Kyung-Sang Yu; Joo-Youn Cho

Abstract Objectives: Zabofloxacin is being developed as a new fluoroquinolone antibiotic that is a potent and selective inhibitor of the essential bacterial type II topoisomerases and topoisomerase IV. Zabofloxacin is indicated for community-acquired respiratory infections due to Gram-positive bacteria. The aim of this study was to compare the pharmacokinetics (PK) of the zabofloxacin hydrochloride 400 mg capsule (DW224a, 366.7 mg as zabofloxacin) with the PK of the zabofloxacin aspartate 488 mg tablet (DW224aa, 366.5 mg as zabofloxacin) in healthy Korean male volunteers to assess the bioequivalence between the two drug formulations. Methods: A randomized, open-label, single-dose, two-way crossover study was performed. The subjects received either DW224a or DW224aa according to their sequence group. Plasma concentrations of zabofloxacin were determined by liquid chromatography–tandem mass spectrometry. The maximum plasma concentrations (Cmax), the area under the plasma concentration versus time curve (AUC) from the time of dosing to 48 hours post-dosing (AUClast), and the AUC extrapolated to infinity (AUCinf) were determined from the plasma concentration–time profile. (ClinicalTrials.gov identifier: NCT01341249). Results: Twenty-nine of the 32 subjects enrolled completed the study. The Cmax. AUClast, and AUCinf (mean ± SD) values of DW224a were 1889.7 ± 493.4 ng/mL, 11,110 ± 2005.0 ng*h/mL, and 11,287 ± 2012.6 ng*h/mL, respectively, and those of DW224aa were 2005.0 ± 341.3 ng/mL, 11,719 ± 2507.5 ng*h/mL, and 11,913 ± 2544.8 ng*h/mL, respectively. The geometric mean ratios (90% confidence intervals) of the Cmax. AUClast, and AUCinf were 1.08 (1.00–1.17), 1.05 (1.00–1.10), and 1.05 (1.00–1.10), respectively, and were within the bioequivalence acceptance range of 0.8–1.25. Both drugs were well tolerated with no serious adverse events. Conclusion: A single oral dose of DW224a or DW224aa to healthy volunteers appeared to be well tolerated. Both DW224a and DW224aa exhibited comparable PK profiles and were bioequivalent in terms of PK parameters. Further studies in patients are needed to corroborate the result of this study. Trial registration: ClinicalTrials.gov identifier: NCT01341249.


Therapeutic Drug Monitoring | 2014

Trough concentration over 12.1 mg/L is a major risk factor of vancomycin-related nephrotoxicity in patients with therapeutic drug monitoring.

Hye Kyung Han; Hyungmi An; Kwang-Hee Shin; Dong Hoon Shin; Sue Hyun Lee; Ju Han Kim; Sang-Heon Cho; Hye-Ryun Kang; In-Jin Jang; Kyung-Sang Yu; Kyoung Soo Lim

Background: High doses of vancomycin increase the risk of nephrotoxicity, but the quantitative relationship between vancomycin exposure and nephrotoxicity is still controversial. This study evaluated the relationship between vancomycin trough concentration and nephrotoxicity, and risk factors for nephrotoxicity in patients undergoing therapeutic drug monitoring. Methods: A total of 1269 cases from patients who underwent therapeutic drug monitoring were collected from 2006 to 2010. Receiver operating characteristic curve analysis was used to evaluate the relationship between trough concentration and the incidence of nephrotoxicity. Logistic regression using the generalized Least Absolute Shrinkage and Selection Operator (lasso) method was used to evaluate possible risk factors for nephrotoxicity. The data were divided into high/low-concentration groups by the cutoff value obtained from the receiver operating characteristic curve, and additional logistic regression using the generalized lasso method was performed for each group. Results: The cutoff value of the vancomycin trough concentration was 12.1 mg/L. Patients with high concentrations (>12.1 mg/L) were more likely to develop nephrotoxicity (odds ratio = 16.0, 95% confidence interval, 8.2–31.1). The vancomycin trough concentration was the only significant risk factor for nephrotoxicity identified using the generalized lasso (P < 0.001). In contrast, no factor was associated with nephrotoxicity in the low-concentration group. Conclusions: Vancomycin trough concentrations over 12.1 mg/L were associated with an increased risk of nephrotoxicity. This is lower than the known threshold. Trough vancomycin concentration over the threshold was the only risk factor of nephrotoxicity among demographic factors, dosing regimen, and other clinical conditions in this study. It is suggested that vancomycin trough concentrations greater than 12.1 mg/L require close monitoring for nephrotoxicity.


principles and practice of constraint programming | 2012

Evaluation of the pharmacokinetic interaction between the dipeptidyl peptidase IV inhibitor LC15-0444 and pioglitazone in healthy volunteers.

Sung Eun Kim; SoJeong Yi; Kwang-Hee Shin; Tae-Eun Kim; Min-Jeong Kim; Youn Hoa Kim; Seo Hyun Yoon; Joo-Youn Cho; Sang-Goo Shin; In-Jin Jang; Kyung-Sang Yu

OBJECTIVE LC15-0444, a newly developed selective dipeptidyl peptidase IV inhibitor, has the potential to be administered with other antihyperglycemic agents. The aim of this study was to investigate the interaction between LC15-0444 and pioglitazone by comparing the pharmacokinetics of both compounds and their metabolites. METHODS A randomized, open-label, multiple dosing, three-sequence, three-period, three-treatment crossover study was performed in healthy volunteers. The three treatment groups were comprised of LC15-0444 200 mg, pioglitazone 30 mg, or coadministration of both drugs once daily for 12 days. Blood samples were collected up to 48 hours after the last dosing. Safety and tolerability were assessed throughout the study. RESULTS The geometric mean ratios (GMRs; (LC15-0444+Pioglitazone coadministered)/(LC15-0444 or Pioglitazone alone)) (90% confidence intervals (CIs)) for Cmax,ss and AUCt,ss of LC15-0444 were 1.06 (0.96-1.16) and 0.98 (0.93-1.03), respectively. In the case of pioglitazone, the GMRs (90% CIs) for Cmax,ss and AUCt,ss were 0.84 (0.73-0.96) and 0.85 (0.76-0.96), respectively. All reported adverse events were mild in intensity. CONCLUSIONS The pharmacokinetics of LC15-0444 and its metabolites were not altered by pioglitazone. The systemic exposure of pioglitazone was decreased by 15% after coadministration of LC15-0444 with pioglitazone, but this was not judged to be clinically relevant, considering the total active moiety of pioglitazone.

Collaboration


Dive into the Kwang-Hee Shin's collaboration.

Top Co-Authors

Avatar

Kyung-Sang Yu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

In-Jin Jang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Joo-Youn Cho

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Goo Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tae-Eun Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seo Hyun Yoon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jae Sung Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji Who Kim

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge