Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Diabetes Obes Metab ; 26(7): 2860-2868, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38646838

ABSTRACT

AIM: To investigate the metabolism and disposition characteristics of HSK7653 in healthy male Chinese participants. METHODS: A single oral dose of 80 µCi (25 mg) [14C]HSK7653 capsules was administered to six healthy participants, and blood, plasma, urine and faeces were collected. Quantitative and qualitative analysis was conducted to investigate the pharmacokinetics, blood-to-plasma ratio, mass balance and metabolism of HSK7653. RESULTS: The drug was well absorbed and reached a maximum concentration at 1.25 h. The drug-related components (HSK7653 and its metabolites) were eliminated slowly, with a half-life (t1/2) of 111 h. Unchanged HSK7653 contributed to more than 97% of the total radioactivity in all plasma samples. The blood-to-plasma ratio (0.573-0.845) indicated that HSK7653 did not tend to distribute into blood cells. At 504 h postdose, up to 95.9% of the dose was excreted, including 79.8% in urine and 16.1% in faeces. Most of the radioactivity (75.5% dose) in excreta was unchanged HSK7653. In addition, nine metabolites were detected in urine and faeces. The most abundant metabolite was M6-2, a dioxidation product of HSK7653, which accounted for 4.73% and 2.63% of the dose in urine and faeces, respectively. The main metabolic pathways of HSK7653 in vivo included oxidation, pyrrole ring opening and sulphonamide hydrolysation. CONCLUSION: HSK7653 was well absorbed, slightly metabolized and slowly excreted in humans. The high plasma exposure and long t1/2 of HSK7653 may contribute to its long-lasting efficacy as a long-acting dipeptidyl peptidase-4 inhibitor.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Humans , Male , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Adult , Biotransformation , Half-Life , Feces/chemistry , Young Adult , Healthy Volunteers , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Administration, Oral
2.
Eur J Clin Pharmacol ; 80(1): 1-10, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37906300

ABSTRACT

PURPOSE: This study intends to assess the reference range of lamotrigine concentration for treating childhood epilepsy. METHODS: PubMed, Ovid-Embase, The Cochrane Library, CNKI, WanFang data and VIP databases were searched from database inception to January 2022. RCT, cohort study, case-control study, cross-sectional study that estimated the reference range of lamotrigine for children epilepsy treatment were included. The data extracted included basic information, statistical methods, data type, and results of reference range. Descriptive analysis was performed for them. RESULTS: 8 studies were included and estimated the reference range, and all of them were calculated based on efficacy data and/or concentration data. Statistical methods including ROC curve, concentration-effect curve, mean ± standard deviation, 95% confidence interval and percentile interval were utilized. For lamotrigine monotherapy, the lower limits ranged from 2.06 mg/L to 3.99 mg/L, and the upper limits ranged from 8.43 mg/L to 9.08 mg/L, showing basic consistency. However, for lamotrigine concomitant with valproate, the lower limits ranged from 2.00 mg/L to 8.00 mg/L, and the upper limit was 11.50 mg/L, for lamotrigine concomitant with other antiepileptics, the lower limits ranged from 1.00 mg/L to 3.09 mg/L, and the upper limits varied from 5.90 mg/L to 16.24 mg/L, indicating inconsistency. CONCLUSION: Several studies have estimated the reference range of lamotrigine for childhood epilepsy, while controversy exist and no studies have determined the upper limit of the range based on safety data. To establish the optimal reference range, further high-quality studies are necessary that consider both efficacy and safety data.


Subject(s)
Anticonvulsants , Epilepsy , Child , Humans , Anticonvulsants/therapeutic use , Lamotrigine/therapeutic use , Case-Control Studies , Cohort Studies , Cross-Sectional Studies , Reference Values , Triazines/therapeutic use , Epilepsy/drug therapy , Valproic Acid/therapeutic use
3.
Arch Gynecol Obstet ; 309(4): 1387-1393, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37004537

ABSTRACT

PURPOSE: To investigate the effect of intraperitoneal infusion of ropivacaine combined with dexmedetomidine and ropivacaine alone on the quality of postoperative recovery of patients undergoing total laparoscopic hysterectomy (TLH). METHODS: Female patients scheduled to undergo a TLH under general anesthesia at Fujian Maternity and Child Health Hospital were included. Before the end of pneumoperitoneum, patients were laparoscopically administered an intraperitoneal infusion of 0.25% ropivacaine 40 ml (R group) or 0.25% ropivacaine combined with 1 µg/kg dexmedetomidine 40 ml (RD group). The primary outcome was QoR-40, which was assessed before surgery and 24 h after surgery. Secondary outcomes included postoperative NRS scores, postoperative anesthetic dosage, the time to ambulation, urinary catheter removal, and anal exhaust. The incidence of dizziness, nausea, and vomiting was also analyzed. RESULTS: A total of 109 women were recruited. The RD group had higher QoR scores than the R group at 24 h after surgery (p < 0.05). Compared with the R group, NRS scores in the RD group decreased at 2, 6, 12, and 24 h after surgery (all p < 0.05). In the RD group, the time to the first dosage of postoperative opioid was longer and the cumulative and effective times of PCA compression were less than those in the R group (all p < 0.05). Simultaneously, the time to ambulation (p = 0.033), anal exhaust (p = 0.002), and urethral catheter removal (p = 0.018) was shortened in the RD group. The RD group had a lower incidence of dizziness, nausea, and vomiting (p < 0.05). CONCLUSION: Intraperitoneal infusion of ropivacaine combined with dexmedetomidine improved the quality of recovery in patients undergoing TLH. TRIAL REGISTRATION: ChiCTR2000033209, Registration Date: May 24, 2020.


Subject(s)
Dexmedetomidine , Laparoscopy , Child , Female , Humans , Pregnancy , Ropivacaine , Dexmedetomidine/therapeutic use , Anesthetics, Local , Dizziness/complications , Dizziness/drug therapy , Pain, Postoperative/drug therapy , Pain, Postoperative/prevention & control , Amides/therapeutic use , Hysterectomy/adverse effects , Double-Blind Method , Infusions, Parenteral/adverse effects , Laparoscopy/adverse effects , Nausea , Vomiting
4.
Stroke ; 54(9): 2420-2433, 2023 09.
Article in English | MEDLINE | ID: mdl-37465997

ABSTRACT

BACKGROUND: Hematoma clearance has been a proposed therapeutic strategy for hemorrhagic stroke. This study investigated the impact of CX3CR1 (CX3C chemokine receptor 1) activation mediated by r-FKN (recombinant fractalkine) on hematoma resolution, neuroinflammation, and the underlying mechanisms involving AMPK (AMP-activated protein kinase)/PPARγ (peroxisome proliferator-activated receptor gamma) pathway after experimental germinal matrix hemorrhage (GMH). METHODS: A total of 313 postnatal day 7 Sprague Dawley rat pups were used. GMH was induced using bacterial collagenase by a stereotactically guided infusion. r-FKN was administered intranasally at 1, 25, and 49 hours after GMH for short-term neurological evaluation. Long-term neurobehavioral tests (water maze, rotarod, and foot-fault test) were performed 24 to 28 days after GMH with the treatment of r-FKN once daily for 7 days. To elucidate the underlying mechanism, CX3CR1 CRISPR, or selective CX3CR1 inhibitor AZD8797, was administered intracerebroventricularly 24 hours preinduction of GMH. Selective inhibition of AMPK/PPARγ signaling in microglia via intracerebroventricularly delivery of liposome-encapsulated specific AMPK (Lipo-Dorsomorphin), PPARγ (Lipo-GW9662) inhibitor. Western blot, Immunofluorescence staining, Nissl staining, Hemoglobin assay, and ELISA assay were performed. RESULTS: The brain expression of FKN and CX3CR1 were elevated after GMH. FKN was expressed on both neurons and microglia, whereas CX3CR1 was mainly expressed on microglia after GMH. Intranasal administration of r-FKN improved the short- and long-term neurobehavioral deficits and promoted M2 microglia polarization, thereby attenuating neuroinflammation and enhancing hematoma clearance, which was accompanied by an increased ratio of p-AMPK (phosphorylation of AMPK)/AMPK, Nrf2 (nuclear factor erythroid 2-related factor 2), PPARγ, CD36 (cluster of differentiation 36), CD163 (hemoglobin scavenger receptor), CD206 (the mannose receptor), and IL (interleukin)-10 expression, and decreased CD68 (cluster of differentiation 68), IL-1ß, and TNF (tumor necrosis factor) α expression. The administration of CX3CR1 CRISPR or CX3CR1 inhibitor (AZD8797) abolished the protective effect of FKN. Furthermore, selective inhibition of microglial AMPK/PPARγ signaling abrogated the anti-inflammation effects of r-FKN after GMH. CONCLUSIONS: CX3CR1 activation by r-FKN promoted hematoma resolution, attenuated neuroinflammation, and neurological deficits partially through the AMPK/PPARγ signaling pathway, which promoted M1/M2 microglial polarization. Activating CX3CR1 by r-FKN may provide a promising therapeutic approach for treating patients with GMH.


Subject(s)
Chemokine CX3CL1 , Infant, Newborn, Diseases , Rats , Animals , Humans , Infant, Newborn , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/pharmacology , PPAR gamma/metabolism , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Rats, Sprague-Dawley , Neuroinflammatory Diseases , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Microglia/metabolism , Hematoma/metabolism , CX3C Chemokine Receptor 1/metabolism
5.
Xenobiotica ; 53(2): 69-83, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36745485

ABSTRACT

SHR0302, a selective JAK1 inhibitor developed by Jiangsu Hengrui Pharmaceutical Co., was intended for the treatment of rheumatoid arthritis. In this study, we evaluated the pharmacokinetics, mass balance, and metabolism of SHR0302 in six healthy Chinese male subjects after a single 8 mg (80 µCi) oral dose of [14C]SHR0302.SHR0302 was absorbed rapidly (Tmax = 0.505 h), and the average t1/2 of the SHR0302-related components in plasma was approximately 9.18 h. After an oral dose was administered, the average cumulative excretion of the radioactive components was 100.56% ± 1.51%, including 60.95% ± 11.62% in urine and 39.61% ± 10.52% in faeces.A total of 16 metabolites were identified. In plasma, the parent drug SHR0302 accounted for 90.42% of the total plasma radioactivity. In urine, SHR161279 was the main metabolite, accounting for 33.61% of the dose, whereas the parent drug SHR0302 only accounted for 5.1% of the dose. In faeces, the parent drug SHR0302 accounted for 23.73% of the dose, and SHR161279 was the significant metabolite, accounting for 5.67% of the dose. In conclusion, SHR0302-related radioactivity was mainly excreted through urine (60.95%) and secondarily through faeces (39.61%).The metabolic reaction of SHR0302 in the human body is mainly through mono-oxidation and glucuronidation. The main metabolic location of SHR0302 in the human body is the pyrrolopyrimidine ring.


Subject(s)
Body Fluids , Sulfuric Acids , Humans , Male , Feces , Administration, Oral , Carbon Radioisotopes , Janus Kinase 1
6.
Drug Metab Dispos ; 50(6): 798-808, 2022 06.
Article in English | MEDLINE | ID: mdl-34862252

ABSTRACT

HR011303, a promising selective urate transporter 1 inhibitor, is currently being studied in a phase III clinical trial in China for the treatment of hyperuricemia and gout. In the current study, the pharmacokinetics, mass balance, and metabolism of HR011303 were examined in six healthy Chinese male subjects who received a single oral dose of 10 mg of [14C]HR011303 (80 µCi). The results showed that HR011303 was rapidly absorbed with a median time to reach C max of 1.50 hours postdose, and the arithmetic mean half-life of total radioactivity was approximately 24.2 hours in plasma. The mean blood-to-plasma radioactivity concentration ratio was 0.66, suggesting the preferential distribution of drug-related components in plasma. At 216 hours postdose, the mean cumulative excreted radioactivity was 91.75% of the dose, including 81.50% in urine and 10.26% in feces. Six metabolites were identified, and the parent drug HR011303 was the most abundant component in plasma and feces, but a minor component in urine. Glucuronidation of the carboxylic acid moiety of HR011303 was the primary metabolic pathway in humans, amounting to 69.63% of the dose (M5, 51.57% of the dose; M5/2, 18.06% of the dose) in the urine; however, it was not detected in plasma. UDP-glucuronosyltransferase (UGT) 2B7 was responsible for the formation of M5. Overall, after a single oral dose of 10 mg of [14C]HR011303 (80 µCi), HR011303 and its main metabolites were eliminated via renal excretion. The major metabolic pathway was carboxylic acid glucuronidation, which was catalyzed predominantly by UGT2B7. SIGNIFICANCE STATEMENT: This study determined the absorption and disposition of HR011303, a selective urate transporter (URAT) 1 inhibitor currently in development for the treatment of hyperuricemia and gout. This work helps to characterize the major metabolic pathways of new URAT inhibitors and identify the absorption and clearance mechanism.


Subject(s)
Gout , Hyperuricemia , Administration, Oral , Carboxylic Acids , Feces , Glucuronosyltransferase/metabolism , Gout/drug therapy , Humans , Male , Organic Anion Transporters , Uricosuric Agents , Uridine Diphosphate
7.
Eur J Nucl Med Mol Imaging ; 49(13): 4419-4426, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35902411

ABSTRACT

BACKGROUND: A novel reporter system, streptavidin (SA)- [68 Ga]Ga-labeled biotin ([68 Ga]Ga-DOTA-biotin), was constructed and its ability for PET imaging the behaviors of CAR T cells were also evaluated in this study. METHODS: In vitro activity and cytotoxicity of the SA transduced anti-CD19-CAR T (denoted as SA-CD19-CAR T) cells were determined. The feasibility of monitoring proliferation profiles of SA-CD19-CAR T cells using [68 Ga]Ga-DOTA-biotin was firstly investigated in a solid tumor model. Also, the pharmacodynamics and pharmacokinetics of the CAR T cells in whole-body hematologic neoplasms were evaluated by bioluminescence imaging and [68 Ga]Ga-DOTA-biotin PET imaging simultaneously. RESULTS: After transduction with SA, the activity and cytotoxicity of the modified CAR T cells were not affected. PET images revealed that the uptakes of [68 Ga]Ga-DOTA-biotin in CD19+ K562 solid tumors were 0.67 ± 0.32 ID%/g and 1.26 ± 0.13 ID%/g at 30 min and 96 h p.i. after administration of SA-CD19-CAR T cells respectively. It confirmed that the SA-CD19-CAR T cells could effectively inhibit the growth of Raji hematologic tumors. However, low radioactivity related to the proliferation of CD19-CAR T cells was detected in the Raji model. CONCLUSION: SA-CD19-CAR T cells were constructed successfully without disturbing the antitumor functions of the cells. The proliferation of the CAR T cells in solid tumors could be early detected by [68 Ga]Ga-DOTA-biotin PET imaging.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Streptavidin , Biotin/pharmacokinetics , Feasibility Studies , Positron-Emission Tomography/methods , T-Lymphocytes , Cell Line, Tumor , Gallium Radioisotopes/pharmacokinetics
8.
Eur J Nucl Med Mol Imaging ; 49(13): 4394-4405, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35829748

ABSTRACT

PURPOSE: Positron emission tomography (PET) with specific diagnostic probes for quantifying CD8+ T cells has emerged as a powerful technique for monitoring the immune response. However, most CD8+ T cell radiotracers are based on antibodies or antibody fragments, which are slowly cleared from circulation. Herein, we aimed to develop and assess 68 Ga-NODAGA-SNA006 for instant PET (iPET) imaging of CD8+ T cells. METHODS: A novel nanobody without a hexahistidine (His6) tag, SNA006-GSC, was designed, site-specifically conjugated with NODAGA-maleimide and radiolabelled with 68 Ga. The PET imaging profiles of 68 Ga-NODAGA-SNA006 were evaluated in BALB/c MC38-CD8+/CD8- tumour models and cynomolgus monkeys. Three volunteers with lung cancer underwent whole-body PET/CT imaging after 68 Ga-NODAGA-SNA006 administration. The biodistribution, pharmacokinetics and dosimetry of patients were also investigated. In addition, combined with immunohistochemistry (IHC), the quantitative performance of the tracer for monitoring CD8 expression was evaluated in BALB/c MC38-CD8+/CD8- and human subjects. RESULTS: 68 Ga-NODAGA-SNA006 was prepared with RCP > 98% and SA > 100 GBq/µmol. 68 Ga-NODAGA-SNA006 exhibited specific uptake in MC38-CD8+ xenografts tumours, CD8-rich tissues (such as the spleen) in monkeys and CD8+ tumour lesions in patients within 1 h. Fast washout from circulation was observed in three volunteers (t1/2 < 20 min). A preliminary quantitative linear relationship (R2 = 0.9668, p < 0.0001 for xenografts and R2 = 0.7924, p = 0.0013 for lung patients) appeared between 68 Ga-NODAGA-SNA006 uptake and CD8 expression. 68 Ga-NODAGA-SNA006 was well tolerated by all patients. CONCLUSION: 68 Ga-NODAGA-SNA006 PET imaging can instantly quantify CD8 expression with an ideal safety profile and is expected to be important for dynamically tracking CD8+ T cells and monitoring immune responses for individualised cancer immunotherapy. TRIAL REGISTRATION: NCT05126927 (19 November 2021, retrospectively registered).


Subject(s)
Neoplasms , Positron Emission Tomography Computed Tomography , Humans , Pilot Projects , Tissue Distribution , CD8-Positive T-Lymphocytes , Tomography, X-Ray Computed , Heterocyclic Compounds, 1-Ring , Positron-Emission Tomography/methods , Acetates , Maleimides , Immunoglobulin Fragments , Gallium Radioisotopes , Cell Line, Tumor
9.
Anal Biochem ; 647: 114664, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35300971

ABSTRACT

Warfarin is a common first line anticoagulant with a narrow therapeutic window. Because of the large blood volume needed, previous warfarin determination methods were not applicable to small animals, such as mice. To reduce the number of small animals used needed, we developed and validated a sensitive rapid assay for the simultaneous detection of warfarin enantiomers in mouse dried blood spot (DBS) samples. Analytes were extracted by tert-butyl methyl ether and then separated by a chiral Cellulose-1 column with a mobile phase of 75% acetonitrile (containing 0.1% formic acid). The total chromatographic run time was 3 min. Negative mode electrospray ionization was used for MS/MS detection, where the monitored ion transitions were m/z 307.1 â†’ 161.0 and 341.1 â†’ 284.0 for warfarin and coumachlor (internal standard) respectively. The calibration curves were linear with a correlation coefficient of ≥0.994 for both enantiomers over a concentration range of 10-1000 ng/mL. The satisfactory accuracy and adequate reproducibility of both warfarin enantiomers were validated in terms of intra- and interday precision with mouse DBS cards. The samples were stable at room temperature for at least 14 days. The validated method was applied to a pharmacokinetic study in mice.


Subject(s)
Tandem Mass Spectrometry , Warfarin , Animals , Calibration , Chromatography, High Pressure Liquid/methods , Dried Blood Spot Testing/methods , Mice , Reproducibility of Results , Tandem Mass Spectrometry/methods
10.
Br J Clin Pharmacol ; 88(7): 3307-3320, 2022 07.
Article in English | MEDLINE | ID: mdl-35112382

ABSTRACT

AIM: This trial (NCT04013048) investigated the metabolite profiles, mass balance and pharmacokinetics of fuzuloparib, a novel poly (ADP-ribose) polymerase inhibitor, in subjects with advanced solid cancers. METHODS: A single dose of 150 mg [14 C]fuzuloparib was administered to five subjects with advanced solid cancers. Blood, urine and faecal samples were collected, analysed for radioactivity and unchanged fuzuloparib, and profiled for metabolites. The safety of the medicine was assessed during the study. RESULTS: The maximum concentrations (Cmax ) of the total radioactivity (TRA) and unchanged fuzuloparib in plasma were 5.39 µg eq./mL and 4.19 µg/mL, respectively, at approximately 4 hours post dose. The exposure (AUC0-t ) of fuzuloparib accounted for 70.7% of the TRA in plasma, and no single metabolite was observed accounting for more than 10% of the plasma TRA. The recovery of TRA in excreta was 103.3 ± 3.8% in 288 hours, including 59.1 ± 9.9% in urine and 44.2 ± 10.8% in faeces. Sixteen metabolites of fuzuloparib were identified, including mono-oxidation (M1), hydrogenation (M2), di-oxidation (M3), trioxidation (M4), glucuronidation (M5, M7, M8) and de-ethylation (M6) products, and there was no specific binding between these metabolites and blood cells. Aliphatic hydroxylated fuzuloparib (M1-1) was the primary metabolite in the excreta, accounting for more than 40% of the dose for subjects. There were no serious adverse events observed in the study. CONCLUSION: Fuzuloparib was widely metabolized and excreted completely through urine and faeces in subjects with advanced solid cancer. Unchanged fuzuloparib was indicated to be the primary drug-related compound in circulation. [14 C]fuzuloparib was well-tolerated at the study dose.


Subject(s)
Antineoplastic Agents , Neoplasms , Adenosine Diphosphate/analysis , Administration, Oral , Antineoplastic Agents/adverse effects , Feces/chemistry , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/analysis , Ribose/analysis
11.
Br J Clin Pharmacol ; 88(5): 2456-2472, 2022 05.
Article in English | MEDLINE | ID: mdl-34729817

ABSTRACT

AIMS: A lot of medication risks related to high-dose methotrexate (HDMTX) therapy still remain to be identified and standardized. This study aims to establish an evidence-based practice guideline for individualized medication of HDMTX. METHODS: The practice guideline was launched by the Division of Therapeutic Drug Monitoring, Chinese Pharmacological Society. The guideline was developed following the WHO handbook for guideline development and the methodology of evidence-based medicine (EBM). The guideline was initially registered in the International Practice Guidelines Registry Platform (IPGRP-2017CN021). Systematic reviews were conducted to synthesize available evidence. A multicentre cross-sectional study was conducted using questionnaires to evaluate patients' perception and willingness concerning individualized medication of HDMTX. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to rate the quality of evidence and to grade the strength of recommendations. RESULTS: Multidisciplinary working groups were included in this guideline, including clinicians, pharmacists, methodologists, pharmacologists and pharmacoeconomic specialists. A total of 124 patients were involved to integrate patient values and preferences. Finally, the guideline presents 28 recommendations, regarding evaluation prior to administration (renal function, liver function, pleural effusion, comedications, genetic testing), pre-treatment and routine dosing regimen, therapeutic drug monitoring (necessity, method, timing, target concentration), leucovorin rescue (initial timing, dosage regimen and optimization), and management of toxicities. Of these, 12 are strong recommendations. CONCLUSIONS: We developed an evidence-based practice guideline with respect to HDMTX medication using a rigorous and multidisciplinary approach. This guideline provides comprehensive and practical recommendations involving the whole process of HDMTX administration to health care providers.


Subject(s)
Drug Monitoring , Methotrexate , China , Cross-Sectional Studies , Evidence-Based Medicine/methods , Humans , Methotrexate/adverse effects
12.
Pharm Res ; 39(8): 1921-1933, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35725843

ABSTRACT

PURPOSE: Venetoclax (VEN), an anti-tumor drug that is a substrate of cytochrome P450 3A enzyme (CYP3A4), is used to treat leukemia. Voriconazole (VCZ) is an antifungal medication that inhibits CYP3A4. The goal of this study is to predict the effect of VCZ on VEN exposure. METHOD: Two physiological based pharmacokinetics (PBPK) models were developed for VCZ and VEN using the bottom-up and top-down method. VCZ model was also developed to describe the effect of CYP2C19 polymorphism on its pharmacokinetics (PK). The reversible inhibition constant (Ki) of VCZ for CYP3A4 was calibrated using drug-drug interaction (DDI) data of midazolam and VCZ. The clinical verified VCZ and VEN model were used to predict the DDI of VCZ and VEN at clinical dosing scenario. RESULT: VCZ model predicted VCZ exposure in the subjects of different CYP2C19 genotype and DDI related fold changes of sensitive CYP3A substrate with acceptable prediction error. VEN model can capture PK of VEN with acceptable prediction error. The DDI PBPK model predicted that VCZ increased the exposure of VEN by 4.5-9.6 fold. The increase in VEN exposure by VCZ was influenced by subject's CYP2C19 genotype. According to the therapeutic window, VEN dose should be reduced to 100 mg when co-administered with VCZ. CONCLUSION: The PBPK model developed here could support individual dose adjustment of VEN and DDI risk assessment. Predictions using the robust PBPK model confirmed that the 100 mg dose adjustment is still applicable in the presence of VCZ with high inter-individual viability.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Cytochrome P-450 CYP3A , Models, Biological , Sulfonamides , Voriconazole , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Humans , Sulfonamides/pharmacokinetics , Voriconazole/pharmacokinetics
13.
Acta Pharmacol Sin ; 43(2): 494-503, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33927359

ABSTRACT

Furmonertinib was designed for the treatment of non-small cell lung cancer (NSCLC) patients with EGFR T790M mutation. In this study, we investigated the metabolic disposition and mass balance in humans and tissue distribution in rats. After a single oral administration of 97.9 µCi/81.5 mg [14C]-furmonertinib mesylate to six healthy male volunteers, the absorption process of furmonertinib was fast with a tmax of total plasma radioactivity at 0.75 h. Afterward, furmonertinib was extensively metabolized, with the parent drug and active metabolite AST5902 accounting for 1.68% and 0.97% of total radioactivity in plasma. The terminal t1/2 of total radioactivity in plasma was as long as 333 h, suggesting that the covalent binding of drug-related substances to plasma proteins was irreversible to a great extent. The most abundant metabolites identified in feces were desmethyl metabolite (AST5902), cysteine conjugate (M19), and parent drug (M0), which accounted for 6.28%, 5.52%, and 1.38% of the dose, respectively. After intragastric administration of 124 µCi/9.93 mg/kg [14C]-furmonertinib to rats, drug-related substances were widely and rapidly distributed in tissues within 4 h. The concentration of total radioactivity in the lung was 100-fold higher than that in rat plasma, which could be beneficial to the treatment of lung cancer. Mass balance in humans was achieved with 77.8% of the administered dose recovered in excretions within 35 days after administration, including 6.63% and 71.2% in urine and feces, respectively. In conclusion, [14C]-furmonertinib is completely absorbed and rapidly distributed into lung tissue, extensively metabolized in humans, presented mostly as covalent conjugates in plasma, and slowly eliminated mostly via fecal route.


Subject(s)
Antineoplastic Agents , Adult , Animals , Female , Humans , Male , Rats , Administration, Oral , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Chromatography, High Pressure Liquid , ErbB Receptors/antagonists & inhibitors , Rats, Sprague-Dawley , Tissue Distribution
14.
Xenobiotica ; 52(3): 254-264, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35373704

ABSTRACT

YY-20394, a highly selective PI3Kδ inhibitor, is under NDA submission for treating follicular lymphoma in China. The absorption, metabolism, and excretion of YY-20394 were evaluated in healthy Chinese male subjects following a single oral dose of 80 mg [14C]YY-20394 (100 µCi).Within 264 h post-dose, 92.1% of the administered dose was recovered, with 58.1% from urine and 34.0% from faeces. YY-20394 was rapidly absorbed in humans, and the peak plasma concentrations occurred at 1.0 h. The absorbed drug fraction was at least 58.1% according to urine recovery.In addition to the parent drug, nine metabolites were identified in plasma, urine, and faeces. Unchanged YY-20394 was the predominant drug-related component in plasma (accounting for 68.4% of the total radioactivity), urine (accounting for 90.0% of the urinary radioactivity) and faeces (accounting for 41.7% of the faecal radioactivity). In humans, the major metabolic sites were the morphine ring and side chains of piperidine rings. The major metabolic pathways involved N-dealkylation, O-dealkylation, glucuronidation and acetylation.Overall, renal elimination played a significant role in the disposition of YY-20394, and the morphine ring and the side chain of the piperidine ring was the predominant metabolic sites.


Subject(s)
Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors , Administration, Oral , Angiogenesis Inhibitors , Carbon Radioisotopes/analysis , Feces/chemistry , Humans , Male , Morphine Derivatives/analysis , Phosphoinositide-3 Kinase Inhibitors , Piperidines
15.
Scand J Clin Lab Invest ; 82(7-8): 556-562, 2022.
Article in English | MEDLINE | ID: mdl-36300737

ABSTRACT

Accurate therapeutic drug monitoring (TDM) of vancomycin, meropenem, linezolid and teicoplanin are conducive to developing optimal therapeutic regimes for patients. However, the measurement status of those drugs in different laboratories has not been reported. In this study, four samples including two frozen plasma samples and two lyophilized plasma samples were measured by over 35 laboratories across China. The inter- and intra-laboratory %CV, biases (%) of laboratories and intra- and inter-measurement-system %CV were calculated and analyzed. The short-term stability and homogeneity of those drugs in samples were studied. The results of frozen and lyophilized samples were also compared to determine whether there were significant differences in their matrix effects on various measurement systems. Results showed most laboratories' intra-laboratory %CVs were less than 9% for all drugs, and the mean inter-laboratory %CVs were 18.4%, 86.4%, 19.1% and 37.1% for vancomycin, meropenem, linezolid and teicoplanin measurements, respectively. For vancomycin, the intra-measurement %CV of commercial measurement systems was found to be smaller than that of other measurement systems. For meropenem, linezolid and teicoplanin, the agreement among laboratories using self-developed methods (Liquid chromatography-mass spectrometry [LC-MS] or high-performance liquid chromatography [HPLC]) was not satisfactory as most intra-measurement system CVs% were over 20%. Drugs in lyophilized samples were found to be more stable than in frozen samples, and no obvious differences in matrix effects were found for those two kinds of processed samples on most measurement systems. In conclusion, this study depicted the measurement status of those drugs in clinical laboratories, and found the lyophilized samples were more suitable EQA material for those drugs.


Subject(s)
Anti-Bacterial Agents , Vancomycin , Humans , Anti-Bacterial Agents/therapeutic use , Linezolid , Meropenem , Teicoplanin , Drug Monitoring/methods
16.
BMC Anesthesiol ; 22(1): 357, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418946

ABSTRACT

BACKGROUND: To evaluate the safety and effectiveness of different dosages of intranasal Dexmedetomidine (DEX) in combination with oral midazolam for sedation of young children during brain MRI examination. METHODS: Included in this prospective single-blind randomized controlled trial were 156 children aged from 3 months to 6 years and weighing from 4 to 20 Kg with ASA I-II who underwent brain MRI examination between March 2021 and February 2022. Using the random number table method, they were divided into group A (using 3 ug/kg intranasal DEX plus 0.2 mg/Kg oral midazolam) and group B (using 2 ug/kg intranasal DEX plus 0.2 mg/Kg oral Midazolam). The one-time success rate of sedation, sedation onset time, recovery time, overall sedation time, and occurrence of adverse reactions during MRI examination were compared between the two groups. The heart rate (HR), mean arterial pressure (MAP), and percutaneous SpO2before and after drug administration were observed in both groups. Differences in sedation scores between the two groups were compared before intranasal drug administration (T0), 10 min after drug administration (T1), at the time of falling asleep (T2), at the end of examination (T3), and at the time of recovery (T4). RESULTS: The one-time success rate of sedation in group A and B was 88.31% and 79.75% respectively, showing no significant difference between the two groups (P>0.05). The sedation onset time in group A was 24.97±16.94 min versus 27.92±15.83 min in group B, and the recovery time was 61.88±22.18 min versus 61.16±28.16 min, both showing no significance difference between the two groups (P>0.05). Children in both groups exhibited good drug tolerance without presenting nausea and vomiting, hypoxia, or bradycardia and hypotension that needed clinical interventions. There was no significant difference in the occurrence of abnormal HR, MAP or other adverse reactions between the two groups (P>0.05). CONCLUSION: 3 ug/kg or 2 ug/kg intranasal DEX in combination with 0.2 mg/kg oral Midazolam both are safe and effective for sedation of children undergoing MRI examination with the advantages of fast-acting and easy application. TRIAL REGISTRATION: It was registered at the Chinese Clinical Trial Registry ( ChiCTR1800015038 ) on 02/03/2018.


Subject(s)
Dexmedetomidine , Midazolam , Child , Humans , Child, Preschool , Administration, Intranasal , Dexmedetomidine/adverse effects , Hypnotics and Sedatives , Single-Blind Method , Prospective Studies , Magnetic Resonance Imaging , Brain/diagnostic imaging
17.
Arch Gynecol Obstet ; 305(4): 1055-1067, 2022 04.
Article in English | MEDLINE | ID: mdl-34982175

ABSTRACT

OBJECTIVE: The purpose of this study was to evaluate the effect of propofol on lipopolysaccharide (LPS)-induced toxicity in HTR-8/SVneo cells. METHODS: In this study, HTR-8/SVneo cells were induced by LPS. The cells were treated with different concentrations of propofol. Cell proliferation, apoptosis, invasion, and wound healing rate were measured by MTT, flow cytometry, Transwell, and wound-healing assay. The relative mRNA expression levels of miR-216a-5p, TLR, MyD88, and NF-κB(p65) were measured by qRT-PCR. The protein expression levels of TLR, MyD88, and p-NF-κB(p65) were detected by western blot. The p-NF-κB(p65) nuclear volume was evaluated by cell immunofluorescence. RESULTS: Compared with control group, the cell proliferation, invasion, and wound healing rate were significantly decreased and the cell apoptosis rate was significantly increased in LPS- induced HTR-8/SVneo cells (P < 0.01). With propofol supplement, the cell proliferation, migration, and invasion abilities were significantly recovered and apoptosis rate was significantly inhibited (P < 0.05). The expression levels of miR-216a-5p, TLR4, MyD88, NF-κB(p65), and p-NF-κB(p65), and p-NF-κB(p65) nuclear volume were significantly different between propofol group and model group (P < 0.05). However, after knockdown of miR-216a-5p expression by si-miR-216a-5p transfection, the cell proliferation, migration, and invasion abilities were significantly inhibited and apoptosis rate was notably increased (P < 0.05). CONCLUSION: Propofol improves LPS-induced toxicity in HTR-8/SVneo cells via regulation miR-216a-5p/TLR4 axis.


Subject(s)
MicroRNAs , Propofol , Toll-Like Receptor 4 , Cell Proliferation , Humans , Lipopolysaccharides , MicroRNAs/metabolism , Propofol/pharmacology , Toll-Like Receptor 4/metabolism
18.
J Cell Mol Med ; 25(15): 7451-7461, 2021 08.
Article in English | MEDLINE | ID: mdl-34245101

ABSTRACT

In recent years, chimeric antigen receptor T (CAR T)-cell therapy has shown great potential in treating haematologic disease, but no breakthrough has been achieved in solid tumours. In order to clarify the antitumour mechanism of CAR T cell in solid tumours, the pharmacokinetic (PK) and pharmacodynamic (PD) investigations of CD19 CAR T cell were performed in human leukaemic xenograft mouse models. For PK investigation, we radiolabelled CD19 CAR T cell with 89 Zr and used PET imaging in the CD19-positive and the CD19-negative K562-luc animal models. For PD evaluation, optical imaging, tumour volume measurement and DNA copy-number detection were performed. Unfortunately, the qPCR results of the DNA copy number in the blood were below the detection limit. The tumour-specific uptake was higher in the CD19-positive model than in the CD19-negative model, and this was consistent with the PD results. The preliminary PK and PD studies of CD19 CAR T cell in solid tumours are instructive. Considering the less efficiency of CAR T-cell therapy of solid tumours with the limited number of CAR T cells entering the interior of solid tumours, this study is suggestive for the subsequent CAR T-cell design and evaluation of solid tumour therapy.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Leukemia, Experimental/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Female , Humans , K562 Cells , Leukemia, Experimental/diagnostic imaging , Mice , Mice, Inbred NOD , Multimodal Imaging/methods , Optical Imaging/methods , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Zirconium/chemistry
19.
Cancer Immunol Immunother ; 70(2): 365-376, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32761423

ABSTRACT

CD47, an immune checkpoint receptor frequently unregulated in various blood and solid tumors, interacts with ligand SIPRα on innate immune cells, and conveys a "do not eat me" signal to inhibit macrophage-mediated tumor phagocytosis. This makes CD47 a valuable target for cancer immunotherapy. However, the therapeutic utility of CD47-SIRPα blockade monoclonal antibodies is largely compromised due to significant red blood cell (RBCs) toxicities and fast target-mediated clearance as a result of extensive expression of CD47 on normal cells. To overcome these limitations and further improve therapeutic efficacy, we designed IBI322, a CD47/PD-L1 bispecific antibody which attenuated CD47 activity in monovalent binding and blocked PD-L1 activity in bivalent binding. IBI322 selectively bound to CD47+PD-L1+ tumor cells, effectively inhibited CD47-SIRPα signal and triggered strong tumor cell phagocytosis in vitro, but only with minimal impact on CD47 single positive cells such as human RBCs. In addition, as a dual blocker of innate and adaptive immune checkpoints, IBI322 effectively accumulated in PD-L1-positive tumors and demonstrated synergistic activity in inducing complete tumor regression in vivo. Furthermore, IBI322 showed only marginal RBCs depletion and was well tolerated in non-human primates (NHP) after repeated weekly injections, suggesting a sufficient therapeutic window in future clinical development of IBI322 for cancer treatment.


Subject(s)
Antibodies, Bispecific/therapeutic use , B7-H1 Antigen/therapeutic use , CD47 Antigen/antagonists & inhibitors , Immunotherapy/methods , Neoplasms/drug therapy , Animals , Antibodies, Bispecific/pharmacology , B7-H1 Antigen/pharmacology , Humans , Mice , Mice, Inbred NOD , Neoplasms/pathology
20.
Br J Clin Pharmacol ; 87(1): 93-105, 2021 01.
Article in English | MEDLINE | ID: mdl-32415708

ABSTRACT

AIMS: This trial (NCT03751956) investigated the mass balance, pharmacokinetics and pharmacodynamics of HSK3486, a novel anaesthetic, in healthy subjects. METHODS: A single dose of 0.4 mg/kg [14 C]HSK3486 was administered to six healthy subjects. Blood, urine and faecal samples were collected, analysed for radioactivity, unchanged HSK3486 and profiled for metabolites. The Modified Observer's Assessment of Alertness/Sedation (MOAA/S) scale and vital signs were closely monitored during the study. RESULTS: The mean recovery of total radioactivity in excreta was 87.3% in 240 h, including 84.6% in urine and 2.65% in faeces. The exposure (AUC0-t ) of total radioactivity was much higher than that of unchanged HSK3486 in plasma, indicating there were circulating metabolites in plasma. The glucuronide conjugate of HSK3486 (M4) was found as the only major circulating metabolite in plasma (79.3%), while unchanged HSK3486 accounted for only 3.97% of the total radiation exposure. M4 also resulted in a longer estimated elimination half-life (t1/2 ) of total radioactivity than that of unchanged HSK3486 in plasma. Fortunately, the metabolite was detected to be not specific to red blood cells and was suggested to be nonhypnotic and nontoxic. All the subjects were quickly anaesthetized (2 min) after drug administration and woke up smoothly after a short time (5.5-14.1 min) with few residual effects. The only adverse event in the study was mild (grade 1) and consisted of hypotension. CONCLUSION: HSK3486 is a promising anaesthetic candidate with rapid onset of action and clear absorption, distribution, metabolism, excretion (ADME) processes. HSK3486 showed favourable pharmacokinetic characteristics, pharmacodynamic responses and safety at the study dose.


Subject(s)
Anesthetics , Administration, Intravenous , Administration, Oral , Feces , Healthy Volunteers , Humans , Metabolic Clearance Rate
SELECTION OF CITATIONS
SEARCH DETAIL