Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 499
Filter
1.
Cancer Chemother Pharmacol ; 93(5): 509-517, 2024 May.
Article in English | MEDLINE | ID: mdl-38520556

ABSTRACT

Tazemetostat, a novel oral selective inhibitor of enhancer of zeste homolog 2 (EZH2), was approved by the Food and Drug Administration (FDA) in 2020 for use in patients with advanced epithelioid sarcoma or relapsed/refractory (R/R) EZH2-mutated follicular lymphoma. These indications were approved by the FDA trough accelerated approval based on objective response rate and duration of response that resulted from phase 2 clinical trials. Tazemetostat competes with S-adenosylmethionine (SAM) cofactor to inhibit EZH2, reducing the levels of trimethylated lysine 27 of histone 3 (H3K27me3), considered as pharmacodynamic marker. Tazemetostat is orally bioavailable, characterized by rapid absorption and dose-proportional exposure, which is not influenced by coadministration with food or gastric acid reducing agents. It highly distributes in tissues, but with limited access to central nervous system. Tazemetostat is metabolized by CYP3A in the liver to 3 major inactive metabolites (M1, M3, and M5), has a short half-life and is mainly excreted in feces. Drug-drug interactions were shown with moderate CYP3A inhibitors as fluconazole, leading the FDA to recommend a 50% dose reduction, while studies investigating coadministration of tazemetostat with strong inhibitors/inducers are ongoing. No dosage modifications are recommended based on renal or hepatic dysfunctions. Overall, tazemetostat is the first-in-class EZH2 inhibitor approved by the FDA for cancer treatment. Current clinical studies are evaluating combination therapies in patients with several malignancies.


Subject(s)
Benzamides , Biphenyl Compounds , Drug Interactions , Morpholines , Humans , Morpholines/pharmacokinetics , Morpholines/pharmacology , Morpholines/administration & dosage , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Biphenyl Compounds/administration & dosage , Pyridones/pharmacokinetics , Pyridones/pharmacology , Pyridones/administration & dosage , Pyridones/therapeutic use , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Sulfonamides/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Animals , Organophosphates/pharmacokinetics , Organophosphates/pharmacology
2.
Eur J Drug Metab Pharmacokinet ; 47(1): 57-67, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34635990

ABSTRACT

BACKGROUND AND OBJECTIVES: Huanglian-Houpo decoction (HH), which is recorded in the famous traditional Chinese medicine monograph "Puji Fang," contains two individual herbs, Huanglian (Rhizoma coptidis) and Houpo (Magnoliae officinalis cortex). It was regularly used to treat seasonal epidemic colds and influenzas in ancient China. Our laboratory discovered that HH has a significant anti-H1N1 influenza virus effect. However, no pharmacokinetic and pharmacodynamic data concerning the anti-H1N1 influenza virus activity of HH are available to date. In the current study, the concentration-time profiles of two major components of HH, berberine and magnolol, in rat plasma were investigated. METHODS: An integrate pharmacokinetic approach was developed for evaluating the holistic pharmacokinetic characteristics of berberine and magnolol from HH. Additionally, the inhibition rate and levels of IFN-ß in MDCK cells infected by influenza virus were analyzed. Data were calculated using 3p97 with pharmacokinetic analysis. RESULTS: The estimated pharmacokinetic parameters were maximum plasma concentration (Cmax) 0.9086 µg/ml, area under the concentration-time curve (AUC) 347.74 µg·min/ml, and time to reach Cmax (Tmax) 64.69 min for berberine and Cmax = 0.9843 µg/ml, AUC= 450.64 µg·min/ml, Tmax = 56.86 min for magnolol, respectively. Furthermore, integrated pharmacokinetic and pharmacodynamic analysis showed that the highest plasma concentration, inhibition rate and interferon-ß (IFN-ß) secretion of HH first increased and then weakened over time, reaching their peaks at 60 min. The plasma concentration of HH is directly related to the anti-influenza virus effect. CONCLUSION: The results indicated that berberine and magnolol are the main active ingredients of HH related to its anti-influenza virus effect, which is related to the improvement of IFN-ß secretion.


Subject(s)
Antiviral Agents/pharmacology , Berberine/pharmacology , Biphenyl Compounds/pharmacology , Drugs, Chinese Herbal/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Lignans/pharmacology , Animals , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Area Under Curve , Berberine/blood , Berberine/pharmacokinetics , Biphenyl Compounds/blood , Biphenyl Compounds/pharmacokinetics , China , Drugs, Chinese Herbal/pharmacokinetics , Humans , Influenza, Human/drug therapy , Lignans/blood , Lignans/pharmacokinetics , Male , Models, Animal , Phytotherapy , Rats , Rats, Inbred Strains
3.
Bioanalysis ; 14(1): 29-45, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34786955

ABSTRACT

Aim: To develop a new sensitive RP-HPLC method for simultaneous estimation of 5-fluorouracil (5-FU) and sonidegib (SDG). Materials & methods: Analytical and bioanalytical methods for simultaneous quantification of 5-FU and SDG in bulk, nanoformulations and in rat plasma were developed and validated using a gradient elution technique. Results: Separation of the analytes was effected on a Luna® C18 LC column using a mobile mixture comprising acetonitrile and acidified water. 5-FU and SDG were extracted from plasma matrix using liquid-liquid extraction. The applicability of the method was verified through single-dose oral pharmacokinetic study in Wistar rats. Conclusion: The developed methods allow a specific, sensitive and steady analytical procedure for the simultaneous estimation of 5-FU and SDG in nanoformulations and biological matrix.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy , Pyridines/pharmacokinetics , Animals , Chromatography, High Pressure Liquid/methods , Male , Rats , Rats, Wistar
4.
Drug Deliv ; 28(1): 2574-2581, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34866522

ABSTRACT

In the present study, the porous starch (PS) was used as an efficient carrier of honokiol (HK), and the HK-loaded PS (HPS) delivery system was prepared by melting method without using organic solvents. Its physical-chemical properties, solubility and oral bioavailability were also investigated. The obtained results proved that the HK in the HPS was mostly amorphous when it was loaded into the PSs with 87.54 ± 1.52% of encapsulation efficiency (EE) and 12.51 ± 0.22% of drug loading (DL) capacity. The water-solubility of the HPS was increased to 115.27 ± 2.92 µg/mL (pH = 1.2, artificial gastric juice (AGJ)), 161.58 ± 3.42 (pH = 6.8, artificial intestinal juice (AIJ)) and 148.5 ± 1.89 µg/mL (pH = 5.5, simulated tumor microenvironment), being 6.07, 4.38 and 4.87-folds higher than free HK. In vitro dissolution tests showed the HK was significantly higher from HPS than from free HK. Furthermore, compared with free HK, the release rate and the bioavailability was also substantially improved for HK from the HPS. Meanwhile, the HPS generated a higher inhibition to HepG2 cells than free HK.


Subject(s)
Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacokinetics , Drug Carriers/chemistry , Lignans/administration & dosage , Lignans/pharmacokinetics , Starch/chemistry , Animals , Area Under Curve , Drug Liberation , Half-Life , Hep G2 Cells , Humans , Porosity , Random Allocation , Rats , Rats, Sprague-Dawley , Solubility , Surface Properties , Technology, Pharmaceutical , Tumor Microenvironment
5.
Arch Pharm Res ; 44(12): 1109-1119, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34817825

ABSTRACT

Candesartan cilexetil is an angiotensin II receptor blocker and it is widely used to treat hypertension and heart failure. This drug is a prodrug that rapidly converts to candesartan after oral administration. Candesartan is metabolized by cytochrome P450 2C9 (CYP2C9) enzyme or uridine diphosphate glucurinosyltransferase 1A3, or excreted in an unchanged form through urine, biliary tract and feces. We investigated the effect of genetic polymorphism of CYP2C9 enzyme on drug pharmacokinetics using physiologically based pharmacokinetic (PBPK) modeling. In addition, by introducing the age and ethnicity into the model, we developed a model that can propose an appropriate dosage regimen taking into account the individual characteristics of each patient. To evaluate the suitability of the model, the results of a clinical trial on twenty-two healthy Korean subjects and their CYP2C9 genetic polymorphism data was applied. In this study, PK-Sim® was used to develop the PBPK model of candesartan.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Benzimidazoles/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Cytochrome P-450 CYP2C9/genetics , Models, Biological , Tetrazoles/pharmacokinetics , Adult , Age Factors , Asian People/genetics , Child , Child, Preschool , Female , Humans , Infant , Male , Polymorphism, Genetic , Young Adult
6.
JAMA Cardiol ; 6(12): 1415-1423, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34524394

ABSTRACT

Importance: The US Food and Drug Administration (FDA) expanded labeling for sacubitril/valsartan for use in individuals with chronic heart failure (HF) with left ventricular ejection fraction (LVEF) lower than normal. The population-level implications of implementation of sacubitril/valsartan at higher LVEF ranges is unknown. While the Prospective Comparison of ARNI With ARB Global Outcomes in HF With Preserved Ejection Fraction (PARAGON-HF) trial did not meet its primary end point, the trial may provide useful information in projecting expected clinical events among treated individuals. Objective: To quantify newly eligible treatment candidates for sacubitril/valsartan under the expanded FDA labeling and to apply treatment effects and the number needed to treat (NNT) to prevent 1 worsening HF event derived from subgroups of the PARAGON-HF trial who fall under the revised FDA label. Design, Setting, and Participants: Newly eligible treatment candidates were estimated by mapping the LVEF distribution from 559 520 adult patients hospitalized between 2014 and 2019 in the Get With The Guidelines-Heart Failure registry to adults self-identifying with HF in the National Health and Nutrition Examination Survey (2015 to 2018). The NNT with 3 years of treatment for 3 end points of interest (total HF hospitalizations, total HF hospitalizations and cardiovascular death, and total HF hospitalizations and urgent HF visits and cardiovascular death) were estimated from the PARAGON-HF trial. Data were analyzed from February to June 2021. Main Outcomes and Measures: Number of worsening HF events prevented or postponed if eligible patients were treated with sacubitril/valsartan for 3 years. Results: Of an estimated 4 682 098 adults, the mean (SE) age was 66.3 (0.8) years, 1 995 037 (42.6%) were women, and 748 045 (16.0%) were Black. The potential number of adults projected to be newly eligible varied by the definition of FDA labeling of lower than normal LVEF from 643 161 (95% CI, 534 433-751 888; LVEF of 41% to 50%) to 1 838 756 (95% CI, 1 527 911-2 149 601; LVEF of 41% to 60%). In the PARAGON-HF trial, the NNT to prevent a worsening HF event (range, 7 to 12 patients) was consistent irrespective of specific LVEF range selected. Comprehensive implementation of sacubitril/valsartan among newly eligible patients was empirically estimated to prevent up to 69 268 (95% CI, 57 558-80 978) worsening HF events (LVEF of 41% to 50%) to 182 592 (95% CI, 151 725-213 460) worsening HF events (LVEF of 41% to 60%). Conclusions and Relevance: The expanded FDA labeling is positioned to substantially increase the potential HF population eligible for sacubitril/valsartan by up to 1.8 million individuals and has the potential to prevent or postpone as many as 180 000 worsening HF events, depending on the definition of normal LVEF.


Subject(s)
Aminobutyrates/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Drug Labeling/methods , Heart Failure/drug therapy , United States Food and Drug Administration/statistics & numerical data , Valsartan/pharmacokinetics , Ventricular Function, Left/physiology , Aged , Angiotensin Receptor Antagonists/pharmacology , Disease Progression , Drug Combinations , Female , Follow-Up Studies , Heart Failure/epidemiology , Humans , Incidence , Male , Neprilysin , Retrospective Studies , Time Factors , United States/epidemiology , Ventricular Function, Left/drug effects
7.
Drug Des Devel Ther ; 15: 3675-3683, 2021.
Article in English | MEDLINE | ID: mdl-34465979

ABSTRACT

BACKGROUND AND OBJECTIVE: Bioequivalence tests are fundamental step in assessing the equivalence in bioavailability between a test and reference product. In practice, two separate linear mixed models (LMMs) with random subject effects, which have an area under the concentration-time curve (AUC) and the peak concentration (Cmax) as the responses, have become the gold standard for evaluating bioequivalence. Recently, Lee et al developed a multivariate hierarchical generalized linear model (HGLM) for several responses that modeled correlations among multivariate responses via correlated random effects. The objective of this study was to apply this multivariate analysis to the bioequivalence test in practice and to compare the performance of multivariate HGLM and separate LMMs. METHODS: Three pharmacokinetic datasets, fixed-dose combination (naproxen and esomeprazole), tramadol and fimasartan data were analyzed. We compared the 90% confidence interval (CI) for the geometric mean ratio (GMR) of a test product to a reference product using the multivariate HGLM and two conventional separate LMMs. RESULTS: We found that the 90% CIs for the GMRs of both AUC and Cmax from the multivariate HGLM were narrower than those from the separate LMMs: (0.843, 1.152) vs (0.825, 1.177) for Cmax of esomeprazole in fixed-dose combination data; (0.805, 0.931) vs (0.797, 0.941) for Cmax in tramadol data; (0.801, 1.501) vs (0.762, 1.578) for Cmax and (1.163, 1.332) vs (1.009, 1.341) for AUC in fimasartan data, consistent with the random subject effects from two separate LMMs being highly correlated in the three datasets (correlation coefficient r = 0.883; r = 0.966; r = 0.832). CONCLUSION: This multivariate HGLM had good performance in the bioequivalence test with multiple endpoints. This method would provide a more reasonable option to reduce the 90% CI by adding correlation parameters and thus an advantage especially in evaluating the bioequivalence of highly variable drugs with broad 90% CIs.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Esomeprazole/pharmacokinetics , Models, Statistical , Pyrimidines/pharmacokinetics , Tetrazoles/pharmacokinetics , Tramadol/pharmacokinetics , Area Under Curve , Biological Availability , Biphenyl Compounds/administration & dosage , Datasets as Topic , Drug Combinations , Esomeprazole/administration & dosage , Humans , Linear Models , Male , Multivariate Analysis , Naproxen/administration & dosage , Naproxen/pharmacokinetics , Pyrimidines/administration & dosage , Randomized Controlled Trials as Topic , Tetrazoles/administration & dosage , Therapeutic Equivalency , Tramadol/administration & dosage
8.
Expert Opin Drug Saf ; 20(8): 877-882, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33888008

ABSTRACT

Introduction: Hedgehog inhibitors are an alternative treatment option for patients with advanced BCCs not eligible for standard therapies due to lack of efficacy, high recurrence risk, and high-rate morbidity. Sonidegib, an oral smoothened antagonist, has been approved for the treatment of adult patients with locally advanced basal cell carcinoma. Several studies and randomized controlled trials have been conducted in order to evaluate the efficacy, safety, and tolerability of this new molecule.Areas covered: The aim of this article is to provide a complete overview on the use of sonidegib for the treatment of advanced BCCs describing the efficacy, safety, and drug tolerability of this drug.Expert opinion: Sonidegib, with a different pharmacokinetics profile from that of the other SMO-inhibitor vismodegib, demonstrated to be an efficacious and well-tolerated treatment in patients with locally advanced BCC. Although several drug-related adverse events have already been described, different strategies should be taken into account to better manage this small molecule while avoiding treatment discontinuation. The use of sonidegib as neoadjuvant therapy or combined with other hedgehog pathway inhibitors targeting different sites and to date, only available for pre-clinical studies, should also be considered.


Subject(s)
Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Pyridines/administration & dosage , Skin Neoplasms/drug therapy , Anilides/administration & dosage , Anilides/adverse effects , Anilides/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Carcinoma, Basal Cell/pathology , Hedgehog Proteins/antagonists & inhibitors , Humans , Pyridines/adverse effects , Pyridines/pharmacokinetics , Randomized Controlled Trials as Topic , Skin Neoplasms/pathology , Smoothened Receptor/antagonists & inhibitors
9.
Cancer Rep (Hoboken) ; 4(5): e1394, 2021 10.
Article in English | MEDLINE | ID: mdl-33811471

ABSTRACT

BACKGROUND: Chimeric antigen receptor (CAR) T-cell therapy of pediatric sarcomas is challenged by the paucity of targetable cell surface antigens. A candidate target in osteosarcoma (OS) is the ganglioside GD2 , but heterogeneous expression of GD2 limits its value. AIM: We aimed to identify mechanisms that upregulate GD2 target expression in OS. METHODS AND RESULTS: GD2 surface expression in OS cells, studied by flow cytometry, was found to vary both among and within individual OS cell lines. Pharmacological approaches, including inhibition of the histone methyltransferase Enhancer of Zeste Homolog 2 (EZH2) and modulation of the protein kinase C, failed to increase GD2 expression. Instead, cell confluency was found to be associated with higher GD2 expression levels both in monolayer cultures and in tumor spheroids. The sensitivity of OS cells to targeting by GD2 -specific CAR T cells was compared in an in vitro cytotoxicity assay. Higher cell confluencies enhanced the sensitivity of OS cells to GD2 -antigen specific, CAR T-cell-mediated in vitro cytolysis. Mechanistic studies revealed that confluency-dependent upregulation of GD2 expression in OS cells is mediated by increased de novo biosynthesis, through a yet unknown mechanism. CONCLUSION: Expression of GD2 in OS cell lines is highly variable and associated with increasing cell confluency in vitro. Strategies for selective upregulation of GD2 are needed to enable effective therapeutic targeting of this antigen in OS.


Subject(s)
Bone Neoplasms/metabolism , Cell Culture Techniques/standards , Gangliosides/metabolism , Osteosarcoma/metabolism , T-Lymphocytes/immunology , Benzamides/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Brefeldin A/pharmacology , Cytotoxicity, Immunologic/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Morpholines/pharmacokinetics , Osteosarcoma/immunology , Osteosarcoma/pathology , Protein Synthesis Inhibitors/pharmacology , Pyridones/pharmacokinetics , Surface Properties , Tumor Cells, Cultured
10.
Int J Nanomedicine ; 16: 651-665, 2021.
Article in English | MEDLINE | ID: mdl-33536753

ABSTRACT

OBJECTIVE: This study was intended to utilize lecithin-based mixed polymeric micelles (lbMPMs) for enhancing the solubility and bioavailability of honokiol and magnolol to resolve the hindrance of their extreme hydrophobicity on the clinical applications. METHODS: Lecithin was selected to increase the volume of the core of lbMPMs, thereby providing a greater solubilization capacity. A series of amphiphilic polymers (sodium deoxycholate [NaDOC], Cremophor®, and Pluronic® series) were included with lecithin for screening and optimization. RESULTS: After preliminary evaluation and subsequentially optimization, two lbMPMs formulations composed of honokiol/magnolol:lecithin:NaDOC (lbMPMs[NaDOC]) and honokiol/magnolol:lecithin:PP123 (lbMPMs[PP123]) in respective ratios of 6:2:5 and 1:1:10 were optimally obtained with the mean particle sizes of 80-150 nm, encapsulation efficacy (EEs) of >90%, and drug loading (DL) of >9.0%. These lbMPMs efficiently stabilized honokiol/magnolol in phosphate-buffered saline (PBS) at room temperature or 4 °C and in fetal bovine serum or PBS at 37 °C. PK study demonstrated that lbMPMs[NaDOC] showed much improvement in enhancing bioavailability than that by lbMPMs[PP123] for both honokiol and magnolol. The absolute bioavailability for honokiol and magnolol after intravenous administration of lbMPMs[NaDOC] exhibited 0.93- and 3.4-fold increases, respectively, compared to that of free honokiol and magnolol. For oral administration with lbMPMs[NaDOC], the absolute bioavailability of honokiol was 4.8%, and the absolute and relative bioavailability of magnolol were 20.1% and 2.9-fold increase, respectively. CONCLUSION: Overall, honokiol/magnolol loaded in lbMPMs[NaDOC] showed an improvement of solubility with suitable physical characteristics leading to enhance honokiol and magnolol bioavailability and facilitating their wider application as therapeutic agents for treating human disorders.


Subject(s)
Biphenyl Compounds/pharmacology , Lecithins/chemistry , Lignans/pharmacology , Micelles , Polymers/chemistry , Administration, Oral , Animals , Biological Availability , Biphenyl Compounds/blood , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Drug Liberation , Humans , Lignans/blood , Lignans/chemistry , Lignans/pharmacokinetics , Male , Particle Size , Rats, Sprague-Dawley , Solubility
11.
J Drugs Dermatol ; 20(2): 156-165, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33538567

ABSTRACT

Basal cell carcinoma (BCC) is the most common malignancy in fair-skinned populations. Most cases are successfully treated with surgery, but in advanced BCC—including locally advanced BCC and metastatic BCC—surgery is likely to result in substantial morbidity or unlikely to be effective. In those patients, the systemic Hedgehog inhibitors (HHIs) sonidegib and vismodegib are the only approved pharmacologic treatment option. Although a number of clinical studies highlight the similarities and differences between the two HHIs, no head-to-head clinical comparison is available. Results from the pivotal BOLT and ERIVANCE clinical studies for sonidegib and vismodegib, respectively, demonstrate similar efficacy measured by objective response rate, complete response rate, and histologic tumor subtype. Safety results for both studies are comparable with similar common adverse events reported for muscle spasms, alopecia, and dysgeusia. A notable difference between sonidegib and vismodegib is their respective pharmacokinetic profiles with sonidegib reaching peak concentration in plasma within 2–4 hours of dosing and steady state in plasma achieved by week 17 of treatment, while vismodegib reaches peak plasma concentration approximately 2 days after a single dose and steady state within 21 days of repeated dosing. This review compares efficacy, safety, and pharmacokinetics of sonidegib and vismodegib based on published literature to date. J Drugs Dermatol. 2021;20(2):156-165. doi:10.36849/JDD.5657 THIS ARTICLE HAD BEEN MADE AVAILABLE FREE OF CHARGE. PLEASE SCROLL DOWN TO ACCESS THE FULL TEXT OF THIS ARTICLE WITHOUT LOGGING IN. NO PURCHASE NECESSARY. PLEASE CONTACT THE PUBLISHER WITH ANY QUESTIONS.


Subject(s)
Anilides/administration & dosage , Biphenyl Compounds/administration & dosage , Carcinoma, Basal Cell/drug therapy , Hedgehog Proteins/antagonists & inhibitors , Pyridines/administration & dosage , Skin Neoplasms/drug therapy , Alopecia/chemically induced , Alopecia/epidemiology , Anilides/adverse effects , Anilides/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Carcinoma, Basal Cell/blood , Carcinoma, Basal Cell/mortality , Carcinoma, Basal Cell/pathology , Clinical Trials, Phase II as Topic , Dysgeusia/chemically induced , Dysgeusia/epidemiology , Hedgehog Proteins/metabolism , Humans , Multicenter Studies as Topic , Progression-Free Survival , Pyridines/adverse effects , Pyridines/pharmacokinetics , Randomized Controlled Trials as Topic , Signal Transduction/drug effects , Skin Neoplasms/blood , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Spasm/chemically induced , Spasm/epidemiology
12.
Cancer Sci ; 112(3): 1123-1131, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33492746

ABSTRACT

BACKGROUND: Tazemetostat is a selective and orally available inhibitor of enhancer of zeste homolog 2 (EZH2), a histone methyltransferase and epigenetic regulator of cellular differentiation programs. We carried out a phase I study of tazemetostat in Japanese patients with relapsed or refractory B-cell non-Hodgkin-type lymphoma (B-NHL) to evaluate its tolerability, safety, pharmacokinetics, and preliminary antitumor activity. METHODS: Tazemetostat was given orally at a single dose of 800 mg on the first day and 800 mg twice daily (BID: total 1600 mg/d) on following days in a 28-day/cycle manner. Tazemetostat dose-limiting toxicity (DLT) was evaluated up to the end of the first treatment cycle. Archival tumor tissues were analyzed for hotspot EZH2 mutations. RESULTS: As of 15 January 2018, seven patients (four follicular lymphoma [FL] and three diffuse large B-cell lymphoma [DLBCL]) were enrolled. The median age was 73 (range, 59-85) years, and the median number of prior chemotherapy regimens was three (range, one to five). No DLT was observed (one patient was not evaluable due to early disease progression). The common treatment-related adverse events (AEs) were thrombocytopenia and dysgeusia (three patients each; 42.9%). No treatment-related serious AEs were observed. The objective response rate was 57% (4/7 patients), including responses in three of four patients with FL and one of three patients with DLBCL. An EZH2 mutation was detected in one patient with FL responding to treatment. CONCLUSIONS: Tazemetostat at 800 mg BID showed an acceptable safety profile and promising antitumor activity in Japanese patients with relapsed or refractory B-NHL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/adverse effects , Biphenyl Compounds/adverse effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma, B-Cell/drug therapy , Morpholines/adverse effects , Neoplasm Recurrence, Local/drug therapy , Pyridones/adverse effects , Administration, Oral , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacokinetics , Drug Administration Schedule , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Humans , Japan , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Middle Aged , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Treatment Outcome
13.
Clin Transl Sci ; 14(1): 194-203, 2021 01.
Article in English | MEDLINE | ID: mdl-32702160

ABSTRACT

Heart failure (HF) causes pathological changes in multiple organs, thus affecting the pharmacokinetics (PK) of drugs. The aim of this study was to investigate the PK of candesartan in patients with HF while examining significant covariates and their related impact on estimated clearance using a population PK (Pop-PK) modeling approach. Data from a prospective, multicenter study were used. Modeling and simulations were conducted using Nonlinear Mixed-Effects Modeling (NONMEM) and R software. A total of 281 white patients were included to develop the Pop-PK model. The final model developed for apparent oral clearance (CL/F) included weight, estimated glomerular filtration rate (eGFR), and diabetes, which partly explained its interindividual variability. The mean CL/F value estimated was 7.6 L/h (1.7-22.6 L/h). Simulations revealed that an important decrease in CL/F (> 25%) is obtained with the combination of the factors retained in the final model. Considering these factors, a more individualized approach of candesartan dosing should be investigated in patients with HF.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Benzimidazoles/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Heart Failure/drug therapy , Models, Biological , Tetrazoles/pharmacokinetics , Administration, Oral , Aged , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Benzimidazoles/administration & dosage , Biological Variation, Population , Biphenyl Compounds/administration & dosage , Canada , Chronic Disease/drug therapy , Female , Heart Failure/blood , Humans , Male , Middle Aged , Prospective Studies , Tetrazoles/administration & dosage
14.
Biomed Chromatogr ; 35(2): e4981, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32895916

ABSTRACT

An LC-tandem mass spectrometry method was developed and validated for the simultaneous quantitation of fimasartan and sacubitrilat using positive ion mode. The protein precipitation method was employed for the extraction of fimasartan, sacubitrilat and alprazolam (internal standard) from rat heparinized plasma. Baseline separation of the analytes was accomplished using an ACE-5, C18 (4.6 × 50 mm) column and gradient elution of mobile phase A (5 mm ammonium formate and 0.1% formic acid in purified water) and B (acetonitrile:methanol, 80:20; v/v). All peaks of interest were eluted within a 5-min runtime. The quantitation was achieved in the selected reaction monitoring mode. The developed method was validated as per US Food and Drug Administration guidelines and met the pre-defined acceptance criteria. The method showed linearity from 5 to 10,000 ng/mL. The accuracy/precision of intra- and inter-batch assays was 96.64%/2.05% to 109.17%/13.70% and 100.74%/3.76% to 106.39%/9.75% for fimasartan and 100.02%/1.49% to 113.80%/9.38% and 100.75%/2.31% to 108.40%/7.74% for sacubitrilat, respectively, in rat plasma. Fimasartan and sacubitrilat remained stable in rat plasma at different experimental conditions up to 21 days. The developed method was sensitive, selective and applied successfully to monitor plasma concentrations of fimasartan and sacubitrilat in an oral rat pharmacokinetic study.


Subject(s)
Aminobutyrates/blood , Biphenyl Compounds/blood , Chromatography, Liquid/methods , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Tetrazoles/blood , Aminobutyrates/chemistry , Aminobutyrates/pharmacokinetics , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Linear Models , Male , Prodrugs , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics
15.
Clin Pharmacol Drug Dev ; 10(2): 190-197, 2021 02.
Article in English | MEDLINE | ID: mdl-32388918

ABSTRACT

For efficient cardiovascular risk protection antihypertensive treatment is often combined with cholesterol-lowering treatment, although solid data of interaction and side effects are missing. This is a prospective, single-center interaction study conducted in a fixed sequence design at steady state of candesartan, amlodipine, and atorvastatin. Five-day monotherapy of candesartan 8 mg was followed by 5-day atorvastatin 40 mg monotherapy and subsequently 9-day amlodipine 5 mg monotherapy; each treatment separated by washout phases. Immediately after amlodipine monotherapy, all 3 drugs were administered concomitantly for 5 days. Pharmacokinetic parameters as well as safety were assessed. Eighteen healthy subjects enrolled and completed the study. No significant difference in the maximum concentration (Cmax ) and the area the under plasma concentration-time curve (AUC) for amlodipine and AUC of atorvastatin was detected following combination versus monotherapy. Cmax of atorvastatin decreased slightly but clinically not relevantly when given in combination. A statistically significant but not below 0.80-fold decrease between candesartan following combination vs monotherapy was detected for Cmax and AUC. In general, all treatments were well tolerated. Concluding, systemic exposure of candesartan, amlodipine, and atorvastatin is not clinically significantly changed upon coadministration. These data support a fixed-dose combination of the 3 components for dual cardiovascular risk prevention.


Subject(s)
Amlodipine/administration & dosage , Atorvastatin/administration & dosage , Benzimidazoles/administration & dosage , Biphenyl Compounds/administration & dosage , Tetrazoles/administration & dosage , Adolescent , Adult , Amlodipine/adverse effects , Amlodipine/pharmacokinetics , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacokinetics , Area Under Curve , Atorvastatin/adverse effects , Atorvastatin/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Drug Interactions , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Male , Middle Aged , Prospective Studies , Tetrazoles/adverse effects , Tetrazoles/pharmacokinetics , Young Adult
16.
Int Immunopharmacol ; 91: 107308, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33383448

ABSTRACT

Bicyclol, an innovative chemical drug with proprietary intellectual property rights in China, is based on derivative of traditional Chinese medicine (TCM) Schisandra chinensis (Wuweizi) of North. Mounting data has proved that bicyclol has therapeutic potential in various pathological conditions in liver. In this narrative review, we provide the first summary of pharmacological activities, pharmacokinetic characteristics and toxicity of bicyclol, and discuss future research perspectives. Our results imply that bicyclol has a wide spectrum of pharmacological properties, including anti-viral, anti-inflammatory, immuno-regulatory, anti-oxidative, antisteatotic, anti-fibrotic, antitumor, cell death regulatory effects and modulation of heat shock proteins. Pharmacokinetic studies have indicated that bicyclol is the main substrate of CYP3A/2E1. Additionally, no obvious drug interactions have been found when bicyclol is administered simultaneously with other prescriptions. Furthermore, the results of chronic toxicity have strongly addressed that bicyclol has no noticeable toxic effects on all biochemical indices and pathological examinations of the main organs. In view of good pharmacological actions and safety, bicyclol is anticipated to be a potential candidate for various liver diseases, including acute liver injury, fulminant hepatitis, non-alcoholic fatty liver disease, fibrosis and hepatocellular carcinoma. Further studies are therefore required to delineate its molecular mechanisms and targets to confer this well-designed drug a far greater potency. We hope that bicyclol-based therapeutics for liver diseases might be broadly used in clinical practice worldwide.


Subject(s)
Biphenyl Compounds/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Liver Diseases/drug therapy , Liver/drug effects , Medicine, Chinese Traditional , Animals , Biphenyl Compounds/adverse effects , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacokinetics , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/pharmacokinetics , Humans , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Diseases/diagnosis , Liver Diseases/immunology , Liver Diseases/metabolism , Treatment Outcome
17.
Diabetes ; 69(12): 2619-2629, 2020 12.
Article in English | MEDLINE | ID: mdl-33004472

ABSTRACT

Sodium-glucose cotransporter 2 inhibitors (SGLT2i) effectively lower plasma glucose (PG) concentration in patients with type 2 diabetes, but studies have suggested that circulating glucagon concentrations and endogenous glucose production (EGP) are increased by SGLT2i, possibly compromising their glucose-lowering ability. To tease out whether and how glucagon may influence the glucose-lowering effect of SGLT2 inhibition, we subjected 12 patients with type 2 diabetes to a randomized, placebo-controlled, double-blinded, crossover, double-dummy study comprising, on 4 separate days, a liquid mixed-meal test preceded by single-dose administration of either 1) placebo, 2) the SGLT2i empagliflozin (25 mg), 3) the glucagon receptor antagonist LY2409021 (300 mg), or 4) the combination empagliflozin + LY2409021. Empagliflozin and LY2409021 individually lowered fasting PG compared with placebo, and the combination further decreased fasting PG. Previous findings of increased glucagon concentrations and EGP during acute administration of SGLT2i were not replicated in this study. Empagliflozin reduced postprandial PG through increased urinary glucose excretion. LY2409021 reduced EGP significantly but gave rise to a paradoxical increase in postprandial PG excursion, which was annulled by empagliflozin during their combination (empagliflozin + LY2409021). In conclusion, our findings do not support that an SGLT2i-induced glucagonotropic effect is of importance for the glucose-lowering property of SGLT2 inhibition.


Subject(s)
Benzhydryl Compounds/therapeutic use , Biphenyl Compounds/therapeutic use , Diabetes Mellitus, Type 2/therapy , Glucagon/metabolism , Glucosides/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/blood , Benzhydryl Compounds/pharmacokinetics , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/blood , Biphenyl Compounds/pharmacokinetics , Blood Glucose/drug effects , C-Peptide/metabolism , Drug Therapy, Combination , Energy Metabolism/drug effects , Gastric Emptying/drug effects , Glucosides/administration & dosage , Glucosides/blood , Glucosides/pharmacokinetics , Glycerol/blood , Glycerol/metabolism , Half-Life , Humans , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/blood , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics
18.
Eur J Pharm Sci ; 154: 105518, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32818657

ABSTRACT

Bicyclol, a novel hepatoprotective agent, has been widely used to treat chronic viral hepatitis and drug-induced liver injury (DILI). However, its metabolic characteristics remains to be explored, especially in humans. The current study aimed to identify major metabolites and specific metabolizing enzymes involved in bicyclol metabolism in vitro and in vivo using high performance liquid chromatography coupled with Q-Exactive orbitrap mass spectrometry (HPLC-Q-Exactive Orbitrap/MS). After incubation with liver microsomes and oral administration to rats, dogs and humans, a total of nine metabolites of bicyclol were identified including M1 (methyl ester hydrolysate product), M2-M3 (demethylated bicyclol), M4-M5 (demethoxy or dehydroxymethyl bicyclol), M6 (glucuronidated bicyclol) and M7-M9 (glucuronide conjugates of metabolites). Among these metabolites, M2 and M3 were the major phase I metabolites mainly mediated by CYP2C19 and CYP3A4, while M6 was the dominant phase II metabolite primarily catalyzed by UGT2B4. In this study, species-related metabolic difference among rats, dogs and humans were observed. In humans and dogs, M6 (glucuronidated bicyclol) was the most abundant circulating metabolite (higher than the parent drug) in the blood after oral administration, while the parent drug was the highest in rats. M4 and M5 were rats-specific metabolites whereas M1 and M9 were absent in dogs in vivo. The metabolism of bicyclol was demonstrated as demethylation and glucuronidation mediated by multiple drug metabolizing enzymes in different species. Our findings systematically elucidated the metabolic sites and routes of bicyclol in human for the first time, which may be helpful for rational combined application in clinic and further study of metabolites-related efficacy or toxicity.


Subject(s)
Biphenyl Compounds , Microsomes, Liver , Animals , Biphenyl Compounds/pharmacokinetics , Chromatography, High Pressure Liquid , Dogs , Humans , Rats , Rats, Sprague-Dawley
19.
Int J Nanomedicine ; 15: 5239-5252, 2020.
Article in English | MEDLINE | ID: mdl-32801689

ABSTRACT

INTRODUCTION: The main pathological mechanism of restenosis after percutaneous coronary intervention (PCI) is intimal hyperplasia, which is mainly caused by proliferation and migration of vascular smooth muscle cells (VSMCs). Our previous study found that honokiol (HNK), a small-molecule polyphenol, can inhibit neointimal hyperplasia after balloon injury, but its specific mechanism is still unclear. Moreover, poor water solubility as well as low bioavailability of honokiol has limited its practical use. METHODS: We used mesoporous silica nanoparticles (MSNPs) as a standard substance to encapsulate HNK and then assemble into honokiol-mesoporous silica nanoparticles, and we investigated the effect of these nanoparticles on the process of restenosis after common carotid artery injury in rats. RESULTS: We report a promising delivery system that loads HNK into MSNPs and finally assembles it into a nanocomposite particle. These HNK-MSNPs not merely inhibited proliferation and migration of VSMCs by reducing phosphorylation of Smad3, but also showed a higher suppression of intimal thickening than the free-honokiol-treated group in a rat model of balloon injury. CONCLUSION: To sum up, this drug delivery system supplies a potent nano-platform for improving the biological effects of HNK and provides a promising strategy for preventing vascular restenosis.


Subject(s)
Biphenyl Compounds/pharmacology , Coronary Restenosis/drug therapy , Drug Delivery Systems/methods , Lignans/pharmacology , Nanoparticles/chemistry , Percutaneous Coronary Intervention/adverse effects , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacokinetics , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Line , Cell Proliferation/drug effects , Coronary Restenosis/metabolism , Disease Models, Animal , Humans , Lignans/administration & dosage , Lignans/pharmacokinetics , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nanoparticles/administration & dosage , Poloxamer/chemistry , Rats, Sprague-Dawley , Silicon Dioxide/chemistry
20.
J Nanobiotechnology ; 18(1): 114, 2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32811502

ABSTRACT

Diacid metabolite as the stable form of norcantharidin (DM-NCTD) derived from Chinese blister beetle (Mylabris spp.). The previous studies reported that DM-NCTD could enhance ABT-737-triggered cell viability inhibition and apoptosis in hepatocellular carcinoma (HCC) cell lines. To translate this synergistic therapy into in vivo anticancer treatment, a folate receptor-targeted lipid bilayer-supported chlorodimethyloctadecylsilane-modified mesoporous silica nanoparticle (FA-LB-CHMSN) with DM-NCTD loaded in CHMSN and ABT-737 in lipid bilayer was prepared, which could promote the cancer cell uptake of the drugs through folate receptor-mediated endocytosis. The structure and the properties of the nanoparticle were evaluated. FA-LB-CHMSN with DM-NCTD/ABT-737 loaded induced apparent tumor cell apoptosis and showed remarkably tumor inhibition in H22 tumor-bearing mice model, with significant cellular apoptosis in the tumor and no obvious toxicity to the tissues. We expect that this nanoparticle could be of interest in both biomaterial investigations for HCC treatment and the combination of chemotherapeutic drugs for synergistic therapies.


Subject(s)
Antineoplastic Agents , Biphenyl Compounds , Bridged Bicyclo Compounds, Heterocyclic , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nitrophenols , Sulfonamides , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Carcinoma, Hepatocellular/pathology , Folic Acid/chemistry , Lipid Bilayers/chemistry , Liver/chemistry , Liver/pathology , Liver Neoplasms/pathology , Mice , Nanoparticles/chemistry , Nitrophenols/chemistry , Nitrophenols/pharmacokinetics , Piperazines/chemistry , Piperazines/pharmacokinetics , Silicon Dioxide/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...