Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 402
Filter
1.
J Med Chem ; 67(8): 6268-6291, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38619191

ABSTRACT

Overactivation of cyclic GMP-AMP synthase (cGAS) is implicated in the occurrence of many inflammatory and autoimmune diseases, and inhibition of cGAS with a specific inhibitor has been proposed as a potential therapeutic strategy. However, only a few low-potency cGAS inhibitors have been reported, and few are suitable for clinical investigation. As a continuation of our structural optimization on the reported cGAS inhibitor 6 (G140), we developed a series of spiro[carbazole-3,3'-pyrrolidine] derivatives bearing a unique 2-azaspiro[4.5]decane structural motif, among which compound 30d-S was identified with high cellular effects against cGAS. This compound showed improved plasma exposure, lower clearance, and an oral bioavailability of 35% in rats. Moreover, in the LPS-induced acute lung injury (ALI) mice model, oral administration of compound 30d-S at 30 mg/kg markedly reduced lung inflammation and alleviated histopathological changes. These results confirm that 30d-S is a new efficacious cGAS inhibitor and is worthy of further investigation.


Subject(s)
Acute Lung Injury , Carbazoles , Drug Design , Nucleotidyltransferases , Pyrrolidines , Acute Lung Injury/drug therapy , Animals , Mice , Male , Humans , Rats , Carbazoles/chemical synthesis , Carbazoles/pharmacology , Carbazoles/chemistry , Carbazoles/therapeutic use , Carbazoles/pharmacokinetics , Pyrrolidines/pharmacology , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Pyrrolidines/therapeutic use , Pyrrolidines/pharmacokinetics , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/metabolism , Lipopolysaccharides , Rats, Sprague-Dawley , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/therapeutic use , Spiro Compounds/pharmacokinetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/chemistry , Structure-Activity Relationship , Molecular Docking Simulation
2.
Clin Transl Sci ; 16(12): 2557-2564, 2023 12.
Article in English | MEDLINE | ID: mdl-37828717

ABSTRACT

The traditional design of food-effect studies has a high patient burden for toxic drugs with long half-lives (e.g., anticancer agents). Microtracers could be used to assess food-effect in patients without influencing their ongoing treatment. The feasibility of a microtracer food-effect study during steady-state of the therapeutic drug was investigated in an in silico simulation study with alectinib as an example for a relative toxic drug with a long half-life. Microtracer pharmacokinetics were simulated based on a previously published population pharmacokinetic model and used for estimation of a model with and a model without food as a covariate on oral bioavailability of alectinib (assuming a 40% food-effect). Power was defined as the fraction of clinical trials where a significant (p < 0.01) food-effect was identified. The proposed study design of 10 patients on steady-state treatment, 10 blood samples collected within 24 h after administration and an assumed food-effect of 40% had a power of 99.9%. The mean estimated food-effect was 39.8% (80% confidence interval: 31.0%-48.6%). The feasibility of microtracer food-effect studies was demonstrated. The design of the microtracer food-effect study allowed estimation of the food-effect with minimal influence on therapeutic treatment and reducing patient burden compared to the traditional study design for toxic drugs with long half-lives.


Subject(s)
Carbazoles , Piperidines , Humans , Pharmaceutical Preparations , Half-Life , Carbazoles/adverse effects , Carbazoles/pharmacokinetics , Piperidines/adverse effects , Piperidines/pharmacokinetics , Administration, Oral
3.
J Korean Med Sci ; 38(22): e173, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37272562

ABSTRACT

BACKGROUND: Carvedilol is a beta-adrenergic receptor antagonist primarily metabolized by cytochromes P450 (CYP) 2D6. This study established a carvedilol population pharmacokinetic (PK)-pharmacodynamic (PD) model to describe the effects of CYP2D6 genetic polymorphisms on the inter-individual variability of PK and PD. METHODS: The PK-PD model was developed from a clinical study conducted on 21 healthy subjects divided into three CYP2D6 phenotype groups, with six subjects in the extensive metabolizer (EM, *1/*1, *1/*2), seven in the intermediate metabolizer-1 (IM-1, *1/*10, *2/*10), and eight in the intermediate metabolizer-2 (IM-2, *10/*10) groups. The PK-PD model was sequentially developed, and the isoproterenol-induced heart rate changes were used to establish the PD model. A direct effect response and inhibitory Emax model were used to develop a carvedilol PK-PD model. RESULTS: The carvedilol PK was well described by a two-compartment model with zero-order absorption, lag time, and first-order elimination. The carvedilol clearance in the CYP2D6*10/*10 group decreased by 32.8% compared with the other groups. The inhibitory concentration of carvedilol estimated from the final PK-PD model was 16.5 ng/mL regardless of the CYP2D6 phenotype. CONCLUSION: The PK-PD model revealed that the CYP2D6 genetic polymorphisms were contributed to the inter-individual variability of carvedilol PK, but not PD.


Subject(s)
Cytochrome P-450 CYP2D6 , Propanolamines , Carvedilol/pharmacology , Cytochrome P-450 CYP2D6/genetics , Heart Rate , Propanolamines/pharmacokinetics , Carbazoles/pharmacokinetics , Genotype
4.
F1000Res ; 12: 1438, 2023.
Article in English | MEDLINE | ID: mdl-38778814

ABSTRACT

Background: In the current work, co-rotating twin-screw processor (TSP) was utilized to formulate solid crystal suspension (SCS) of carvedilol (CAR) for enhancing its solubility, dissolution rate, permeation and bioavailability using mannitol as a hydrophilic carrier. Methods: In-silico molecular dynamics (MD) studies were done to simulate the interaction of CAR with mannitol at different kneading zone temperatures (KZT). Based on these studies, the optimal CAR: mannitol ratios and the kneading zone temperatures for CAR solubility enhancement were assessed. The CAR-SCS was optimized utilizing Design-of-Experiments (DoE) methodology using the Box-Behnken design. Saturation solubility studies and in vitro dissolution studies were performed for all the formulations. Physicochemical characterization was performed using differential scanning calorimetry , Fourier transform infrared spectroscopy, X-ray diffraction studies, and Raman spectroscopy analysis. Ex vivo permeation studies and in vivo pharmacokinetic studies for the CAR-SCS were performed. Stability studies were performed for the DoE-optimized CAR-SCS at accelerated stability conditions at 40 ºC/ 75% RH for three months. Results: Experimentally, the formulation with CAR: mannitol ratio of 20:80, prepared using a KZT of 120 ºC at 100 rpm screw speed showed the highest solubility enhancement accounting for 50-fold compared to the plain CAR. Physicochemical characterization confirmed the crystalline state of DoE-optimized CAR-SCS. In-vitro dissolution studies indicated a 6.03-fold and 3.40-fold enhancement in the dissolution rate of optimized CAR-SCS in pH 1.2 HCl solution and phosphate buffer pH 6.8, respectively, as compared to the pure CAR. The enhanced efficacy of the optimized CAR-SCS was indicated in the ex vivo and in vivo pharmacokinetic studies wherein the apparent permeability was enhanced 1.84-fold and bioavailability enhanced 1.50-folds compared to the plain CAR. The stability studies showed good stability concerning the drug content. Conclusions: TSP technology could be utilized to enhance the solubility, bioavailability and permeation of poor soluble CAR by preparing the SCS.


Subject(s)
Biological Availability , Carvedilol , Solubility , Carvedilol/pharmacokinetics , Carvedilol/chemistry , Carvedilol/administration & dosage , Animals , Administration, Oral , Carbazoles/pharmacokinetics , Carbazoles/chemistry , Carbazoles/administration & dosage , Propanolamines/pharmacokinetics , Propanolamines/chemistry , Propanolamines/administration & dosage , Permeability , Male , Mannitol/chemistry , Mannitol/pharmacokinetics , Suspensions , Molecular Dynamics Simulation , Rats
5.
Drug Des Devel Ther ; 15: 2961-2968, 2021.
Article in English | MEDLINE | ID: mdl-34262261

ABSTRACT

PURPOSE: The present report describes findings from a Phase I clinical study that evaluated the single- and multiple-dose pharmacokinetics of frovatriptan succinate tablet in Chinese healthy subjects. METHODS: A total of 24 healthy subjects were enrolled. In single-dose study, 2.5, 5, and 10 mg oral doses of frovatriptan succinate tablet were administrated. A 2.5 mg frovatriptan succinate tablet was administrated 12 times in 7 days in the multiple-dose study. Blood samples were collected at scheduled time points. RESULTS: The results in single-dose study indicated that the blood levels were proportional to the administered dose, with the mean Cmax and AUClast ranging from approximately 6.27 ng/mL-17.35 ng/mL and 92.52 h⋅ng/mL - 287.40 h⋅ng/mL over the dose range. In the multiple-dose study, moderate drug accumulation was noted, which was attributable to forvatriptan's long t1/2 of about 26.47 to 30.63 h. Gender differences were noticed in both single- and multiple-dose study; exposure PK parameters were consistently higher in female than in male. CONCLUSION: These pharmacokinetic evaluations in healthy Chinese subjects found that frovatriptan succinate tablet has an acceptable pharmacokinetic profile in Chinese subjects.


Subject(s)
Asian People , Carbazoles/administration & dosage , Serotonin Receptor Agonists/administration & dosage , Tryptamines/administration & dosage , Administration, Oral , Adult , Area Under Curve , Carbazoles/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Serotonin Receptor Agonists/pharmacokinetics , Sex Factors , Tablets , Time Factors , Tryptamines/pharmacokinetics , Young Adult
6.
Res Vet Sci ; 137: 235-242, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34049110

ABSTRACT

Osteoarthritis is treated with COX or fosfolipase A2 inhibitors such as carprofen, a propionic acid NSAID. The enhancement of its action over the articular cartilage is mandatory to facilitate its therapeutic application. Drug uptake into the cartilage requires high synovial fluid concentrations, anticipating its rapid distribution towards bloodstream. Thus, intraarticular administration improves local targeting of the drug, lining with the site of action. A pharmacokinetic study in rabbits has been performed to evaluate carprofen nanoparticles after intraarticular administration. Pharmacokinetic analysis of plasma profiles through a modelling approach, has demonstrated the rapid distribution of drug outside of synovial chamber but mainly remaining in plasma. The data modelling has demonstrated the existence of two release-absorption processes when the nanoparticles are administered in the synovial space. Additionally, results are predictive of the PK profile of some other species such as cat, dogs or humans.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Carbazoles/pharmacokinetics , Drug Delivery Systems/veterinary , Injections, Intra-Articular/veterinary , Nanoparticles , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Carbazoles/administration & dosage , Cartilage, Articular , Male , Rabbits
7.
Biopharm Drug Dispos ; 42(6): 263-284, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33904202

ABSTRACT

Renal (RIP) and hepatic (HIP) impairments are prevalent conditions in cancer patients. They can cause changes in gastric emptying time, albumin levels, hematocrit, glomerular filtration rate, hepatic functional volume, blood flow rates, and metabolic activity that can modify drug pharmacokinetics. Performing clinical studies in such populations has ethical and practical issues. Using predictive physiologically-based pharmacokinetic (PBPK) models in the evaluation of the PK of alectinib, ruxolitinib, and panobinostat exposures in the presence of cancer, RIP, and HIP can help in using optimal doses with lower toxicity in these populations. Verified PBPK models were customized under scrutiny to account for the pathophysiological changes induced in these diseases. The PBPK model-predicted plasma exposures in patients with different health conditions within average 2-fold error. The PBPK model predicted an area under the curve ratio (AUCR) of 1, and 1.8, for ruxolitinib and panobinostat, respectively, in the presence of severe RIP. On the other hand, the severe HIP was associated with AUCR of 1.4, 2.9, and 1.8 for alectinib, ruxolitinib, and panobinostat, respectively, in agreement with the observed AUCR. Moreover, the PBPK model predicted that alectinib therapeutic cerebrospinal fluid levels are achieved in patients with non-small cell lung cancer, moderate HIP, and severe HIP at 1-, 1.5-, and 1.8-fold that of healthy subjects. The customized PBPK models showed promising ethical alternatives for simulating clinical studies in patients with cancer, RIP, and HIP. More work is needed to quantify other pathophysiological changes induced by simultaneous affliction by cancer and RIP or HIP.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carbazoles/pharmacokinetics , Liver Diseases/blood , Models, Biological , Neoplasms/blood , Nitriles/pharmacokinetics , Panobinostat/pharmacokinetics , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Renal Insufficiency/blood , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Area Under Curve , Carbazoles/blood , Fasting/metabolism , Female , Humans , Liver Diseases/metabolism , Male , Middle Aged , Neoplasms/metabolism , Nitriles/blood , Panobinostat/blood , Piperidines/blood , Protein Kinase Inhibitors/blood , Pyrazoles/blood , Pyrimidines/blood , Renal Insufficiency/metabolism
8.
Clin Pharmacol Drug Dev ; 10(7): 789-796, 2021 07.
Article in English | MEDLINE | ID: mdl-33345463

ABSTRACT

The effect of age on the pharmacokinetics and safety of chiglitazar was evaluated in patients < 65 and ≥ 65 years with type 2 diabetes mellitus (T2DM). A total of 20 T2DM patients (<65 vs ≥65 years 1:1) completed the study. Patients received multiple doses of 48 mg chiglitazar once daily for 7 days consecutively. After the first dosing, chiglitazar maximum plasma concentration (Cmax ) and area under the plasma concentration-time curve (AUC) in patients ≥ 65 years were similar to those observed in patients < 65 years, with the geometric mean ratio (GMR) for Cmax and AUC being 97.22% and 96.83%, respectively. No significant difference was observed in Cmax (GMR, 97.23%) in the steady state. Compared with the patients < 65 years, a slight increase (8%-13%) of AUC was observed in the patients ≥ 65 years after multiple doses. Chiglitazar was generally well tolerated following multiple doses in both age groups. In conclusion, there were no significant clinical influences on the pharmacokinetic properties and safety profiles of chiglitazar between patients with T2DM < 65 and ≥ 65 years, indicating that in the future it is not required to adjust the dosing regimen by age for T2DM patients ≥ 65 years.


Subject(s)
Carbazoles/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Propionates/administration & dosage , Age Factors , Aged , Area Under Curve , Carbazoles/adverse effects , Carbazoles/pharmacokinetics , Female , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Male , Middle Aged , Peroxisome Proliferator-Activated Receptors/agonists , Propionates/adverse effects , Propionates/pharmacokinetics
9.
Vet Anaesth Analg ; 48(1): 35-41, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33303396

ABSTRACT

OBJECTIVE: To investigate the pharmacokinetics of carprofen after a single intravenous (IV) dose and multiple oral doses administered to pigs undergoing electroporation of the pancreas. STUDY DESIGN: Prospective experimental study. ANIMALS: A group of eight female pigs weighing 31.74 ± 2.24 kg (mean ± standard deviation). METHODS: Carprofen 4 mg kg-1 was administered IV after placement of a central venous catheter during general anaesthesia with isoflurane. Blood samples were collected 30 seconds before and 5, 10, 20, 30 and 60 minutes and 2, 4, 6, 8, 12 and 24 hours after carprofen administration. Subsequently, the same dose of carprofen was administered orally, daily, for 6 consecutive days and blood collected at 36, 48, 60, 72, 96, 120, 144 and 168 hours after initial carprofen administration. Plasma was analysed using liquid chromatography with mass spectrometry. Standard pharmacokinetic parameters were calculated by compartmental analysis of plasma concentration-time curves. Data are presented as mean ± standard error. RESULTS: The initial plasma concentration of IV carprofen was estimated at 54.57 ± 3.92 µg mL-1 and decreased to 8.26 ± 1.07 µg mL-1 24 hours later. The plasma elimination curve showed a bi-exponential decline: a rapid distribution phase with a distribution half-life of 0.21 ± 0.03 hours and a slower elimination phase with an elimination half-life of 17.31 ± 3.78 hours. The calculated pharmacokinetic parameters were as follows: the area under the plasma concentration-time curve was 357.3 ± 16.73 µg mL-1 hour, volume of distribution was 0.28 ± 0.07 L kg-1 and plasma clearance rate was 0.19 ± 0.009 mL minute-1 kg-1. The plasma concentration of carprofen, administered orally from days 2 to 7, varied from 9.03 ± 1.87 to 11.49 ± 2.15 µg mL-1. CONCLUSIONS AND CLINICAL RELEVANCE: Carprofen can be regarded as a long-acting non-steroidal anti-inflammatory drug in pigs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Carbazoles/pharmacokinetics , Administration, Intravenous/veterinary , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Area Under Curve , Female , Half-Life , Prospective Studies , Swine
10.
Cancer Sci ; 111(12): 4540-4547, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33010107

ABSTRACT

Anaplastic lymphoma kinase (ALK) inhibition is expected to be a promising therapeutic strategy for ALK-positive malignancies. We aimed to examine the efficacy and safety of alectinib, a second-generation ALK inhibitor, in patients with relapsed or refractory ALK-positive anaplastic large cell lymphoma (ALCL). This open-label, phase II trial included patients (aged 6 years or older) with relapsed or refractory ALK-positive ALCL. Alectinib 300 mg was given orally twice a day (600 mg/d) for 16 cycles, and the duration of each cycle was 21 days. Patients who weighed less than 35 kg were given a reduced dose of alectinib of 150 mg twice a day (300 mg/d). Ten patients were enrolled, and the median age was 19.5 years (range, 6-70 years). Objective responses were documented in eight of 10 patients (80%; 90% confidence interval, 56.2-95.9), with six complete responses. The 1-year progression-free survival, event-free survival, and overall survival rates were 58.3%, 70.0%, and 70.0%, respectively. The median duration of therapy was 340 days. No unexpected adverse events occurred. The most common grade 3 and higher adverse event was a decrease in neutrophil count in two patients. Alectinib showed favorable clinical activity and was well tolerated in patients with ALK-positive ALCL who had progressed on standard chemotherapy. Based on the results of the current study, the Ministry of Health, Labour and Welfare of Japan approved alectinib for the treatment of recurrent or refractory ALK-positive ALCL in February 2020.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Carbazoles/administration & dosage , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/enzymology , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Administration, Oral , Adult , Aged , Anaplastic Lymphoma Kinase/blood , Carbazoles/adverse effects , Carbazoles/pharmacokinetics , Child , Confidence Intervals , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Japan , Lymphoma, Large-Cell, Anaplastic/mortality , Male , Piperidines/adverse effects , Piperidines/pharmacokinetics , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Recurrence , Survival Rate , Treatment Outcome , Young Adult
11.
Pharmacol Biochem Behav ; 193: 172918, 2020 06.
Article in English | MEDLINE | ID: mdl-32247816

ABSTRACT

Synthetic cannabinoid receptor agonists (SCRAs) possess high abuse liability and complex toxicological profiles, making them serious threats to public health. EG-018 is a SCRA that has been detected in both illicit products and human samples, but it has received little attention to date. The current studies investigated EG-018 at human CB1 and CB2 receptors expressed in HEK293 cells in [3H]CP55,940 competition binding, [35S]GTPγS binding and forskolin-stimulated cAMP production. EG-018 was also tested in vivo for its ability to produce cannabimimetic and abuse-related effects in the cannabinoid tetrad and THC drug discrimination, respectively. EG-018 exhibited high affinity at CB1 (21 nM) and at CB2 (7 nM), but in contrast to typical SCRAs, behaved as a weak partial agonist in [35S]GTPγS binding, exhibiting lower efficacy but greater potency, than that of THC at CB1 and similar potency and efficacy at CB2. EG-018 inhibited forskolin-stimulated cAMP with similar efficacy but lower potency, compared to THC, which was likely due to high receptor density facilitating saturation of this signaling pathway. In mice, EG-018 (100 mg/kg, 30 min) administered intraperitoneally (i.p.) did not produce effects in the tetrad or drug discrimination nor did it shift THC's ED50 value in drug discrimination when administered before THC, suggesting EG-018 has negligible occupancy of brain CB1 receptors following i.p. administration. Following intravenous (i.v.) administration, EG-018 (56 mg/kg) produced hypomotility, catalepsy, and hypothermia, but only catalepsy was blocked by the selective CB1 antagonist rimonabant (3 mg/kg, i.v.). Additional studies of EG-018 and its structural analogues could provide further insight into how cannabinoids exert efficacy through the cannabinoid receptors.


Subject(s)
Behavior, Animal/drug effects , Body Temperature/drug effects , Cannabinoid Receptor Agonists/pharmacokinetics , Carbazoles/pharmacokinetics , Locomotion/drug effects , Microsomes/drug effects , Naphthalenes/pharmacokinetics , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Signal Transduction/drug effects , Synthetic Drugs/pharmacokinetics , Animals , Cannabinoid Receptor Agonists/pharmacology , Carbazoles/pharmacology , Cyclic AMP/metabolism , Dronabinol/pharmacology , HEK293 Cells , Humans , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Naphthalenes/pharmacology , Rats , Rats, Long-Evans , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Synthetic Drugs/metabolism
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 226: 117652, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31654902

ABSTRACT

Transient absorption spectroscopy in combination with in silico methods has been employed to study the interactions between human serum albumin (HSA) and the anti-psychotic agent chlorpromazine (CPZ) as well as its two demethylated metabolites (MCPZ and DCPZ). Thus, solutions containing CPZ, MCPZ or DCPZ and HSA (molar ligand:protein ratios between 1:0 and 1:3) were submitted to laser flash photolysis and the ΔAmax value at λ = 470 nm, corresponding to the triplet excited state, was monitored. In all cases, the protein-bound ligand exhibited higher ΔAmax values measured after the laser pulse and were also considerably longer-lived than the non-complexed forms. This is in agreement with an enhanced hydrophilicity of the metabolites, due to the replacement of methyl groups with H that led to a lower extent of protein binding. For the three compounds, laser flash photolysis displacement experiments using warfarin or ibuprofen indicated Sudlow site I as the main binding site. Docking and molecular dynamics simulation studies revealed that the binding mode of the two demethylated ligands with HSA would be remarkable different from CPZ, specially for DCPZ, which appears to come from the different ability of their terminal ammonium groups to stablish hydrogen bonding interactions with the negatively charged residues within the protein pocket (Glu153, Glu292) as well as to allocate the methyl groups in an apolar environment. DCPZ would be rotated 180° in relation to CPZ locating the aromatic ring away from the Sudlow site I of HSA.


Subject(s)
Chlorpromazine/chemistry , Chlorpromazine/pharmacokinetics , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Binding Sites , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Chlorpromazine/analogs & derivatives , Chlorpromazine/pharmacology , Drug Interactions , Humans , Hydrogen Bonding , Inactivation, Metabolic , Methylation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding/drug effects , Serum Albumin, Human/drug effects , Spectrophotometry, Ultraviolet , Stereoisomerism
13.
Nucleic Acids Res ; 47(22): 11931-11942, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31740959

ABSTRACT

BMVC is the first fluorescent probe designed to detect G-quadruplexes (G4s) in vivo. The MYC oncogene promoter forms a G4 (MycG4) which acts as a transcription silencer. Here, we report the high-affinity and specific binding of BMVC to MycG4 with unusual slow-exchange rates on the NMR timescale. We also show that BMVC represses MYC in cancer cells. We determined the solution structures of the 1:1 and 2:1 BMVC-MycG4 complexes. BMVC first binds the 5'-end of MycG4 to form a 1:1 complex with a well-defined structure. At higher ratio, BMVC also binds the 3'-end to form a second complex. In both complexes, the crescent-shaped BMVC recruits a flanking DNA residue to form a BMVC-base plane stacking over the external G-tetrad. Remarkably, BMVC adjusts its conformation to a contracted form to match the G-tetrad for an optimal stacking interaction. This is the first structural example showing the importance of ligand conformational adjustment in G4 recognition. BMVC binds the more accessible 5'-end with higher affinity, whereas sequence specificity is present at the weaker-binding 3'-site. Our structures provide insights into specific recognition of MycG4 by BMVC and useful information for design of G4-targeted anticancer drugs and fluorescent probes.


Subject(s)
Carbazoles/chemistry , Carbazoles/pharmacokinetics , G-Quadruplexes/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacokinetics , Binding Sites/drug effects , Circular Dichroism , DNA/chemistry , DNA/drug effects , DNA/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Humans , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Nucleic Acid Conformation/drug effects , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/drug effects , Substrate Specificity
14.
J Exp Med ; 216(11): 2492-2502, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31455602

ABSTRACT

The pathways of circulation and clearance of cerebrospinal fluid (CSF) in the spine have yet to be elucidated. We have recently shown with dynamic in vivo imaging that routes of outflow of CSF in mice occur along cranial nerves to extracranial lymphatic vessels. Here, we use near-infrared and magnetic resonance imaging to demonstrate the flow of CSF tracers within the spinal column and reveal the major spinal pathways for outflow to lymphatic vessels in mice. We found that after intraventricular injection, a spread of CSF tracers occurs within both the central canal and the spinal subarachnoid space toward the caudal end of the spine. Outflow of CSF tracers from the spinal subarachnoid space occurred predominantly from intravertebral regions of the sacral spine to lymphatic vessels, leading to sacral and iliac LNs. Clearance of CSF from the spine to lymphatic vessels may have significance for many conditions, including multiple sclerosis and spinal cord injury.


Subject(s)
Carbazoles/cerebrospinal fluid , Lymphatic Vessels/metabolism , Magnetic Resonance Imaging/methods , Sacrum/metabolism , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/cerebrospinal fluid , Analgesics, Opioid/pharmacokinetics , Animals , Buprenorphine/administration & dosage , Buprenorphine/cerebrospinal fluid , Buprenorphine/pharmacokinetics , Carbazoles/administration & dosage , Carbazoles/pharmacokinetics , Cerebrospinal Fluid/metabolism , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Lumbosacral Region/diagnostic imaging , Lymphatic System/metabolism , Lymphatic Vessels/diagnostic imaging , Mice, Inbred C57BL , Mice, Transgenic , Sacrum/diagnostic imaging , Subarachnoid Space
15.
Cancer Radiother ; 23(5): 432-438, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31331844

ABSTRACT

Anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (3 to 5% of all non-small cell lung cancers) carries a particularly high risk of central nervous system dissemination (60% to 90%). As the use of ALK inhibitors improves treatment outcomes over chemotherapy, the determent of central nervous system metastases has become an increasingly relevant therapeutic dilemma considering young age and possible extended overall survival. The goal of brain metastases management is to optimize both overall survival and quality of life, with the high priority of neurocognitive function preservation. Unfortunately in the first year on crizotinib, the pioneering ALK inhibitors, approximately one third of these patients fail in the central nervous system, which is explained by an inadequate central nervous system drug penetration through the blood-brain barrier. Central nervous system-directed radiotherapy represents the most important strategy to control intracranial disease burden and extend the survival benefit with crizotinib. The role of whole brain irradiation in the treatment of brain metastases diminishes, as this technique is associated with the risk of neurocognitive decline. Stereotactic radiotherapy represents an alternative technique that delivers ablative doses of ionizing radiation to the limited volume of oligometastatic brain disease, offering sparing of the adjacent brain parenchyma and reduced neurotoxicity. The next generation ALK inhibitors were designed to cross the blood-brain barrier more efficiently than crizotinib and achieve higher concentration in the cerebrospinal fluid, offering prominent ability to control central nervous system spread. In the phase III ALEX trial the intracranial control was significantly better with alectinib as compared to crizotinib and it translated into survival benefit. Other next generation ALK inhibitors (i.e. ceritinib, brigatinib, lorlatinib) also demonstrated promising activity in the central nervous system.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms , Meningeal Neoplasms/secondary , Neoplasm Proteins/antagonists & inhibitors , Anaplastic Lymphoma Kinase/analysis , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/enzymology , Brain Neoplasms/radiotherapy , Carbazoles/pharmacokinetics , Carbazoles/therapeutic use , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cisplatin/administration & dosage , Clinical Trials as Topic , Combined Modality Therapy , Cranial Irradiation/adverse effects , Cranial Irradiation/methods , Crizotinib/pharmacokinetics , Crizotinib/therapeutic use , Disease Management , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/enzymology , Meningeal Neoplasms/enzymology , Meningeal Neoplasms/radiotherapy , Mice , Molecular Targeted Therapy , Neoplasm Proteins/analysis , Neurocognitive Disorders/etiology , Neurocognitive Disorders/prevention & control , Observational Studies as Topic , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/antagonists & inhibitors , Pemetrexed/administration & dosage , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Radiosurgery/adverse effects , Radiosurgery/methods
16.
Drug Metab Dispos ; 47(7): 699-709, 2019 07.
Article in English | MEDLINE | ID: mdl-31068367

ABSTRACT

Alectinib is a tyrosine kinase inhibitor currently used as a first-line treatment of anaplastic lymphoma kinase-positive metastatic nonsmall cell lung cancer (NSCLC). In the present work, we investigated possible interactions of this novel drug with ATP-binding cassette (ABC) drug efflux transporters and cytochrome P450 (P450) biotransformation enzymes that play significant roles in the phenomenon of multidrug resistance (MDR) of cancer cells as well as in pharmacokinetic drug-drug interactions. Using accumulation studies in Madin-Darby canine kidney subtype 2 (MDCKII) cells alectinib was identified as an inhibitor of ABCB1 and ABCG2 but not of ABCC1. In subsequent drug combination studies, we demonstrated the ability for alectinib to effectively overcome MDR in ABCB1- and ABCG2-overexpressing MDCKII and A431 cells. To describe the pharmacokinetic interaction profile of alectinib in a complete fashion, its possible inhibitory properties toward clinically relevant P450 enzymes (i.e., CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, or CYP3A5) were evaluated using human P450-expressing insect microsomes, revealing alectinib as a poor interactor. Advantageously for its use in pharmacotherapy, alectinib further exhibited negligible potential to cause any changes in expression of ABCB1, ABCG2, ABCC1, CYP1A2, CYP3A4, and CYP2B6 in intestine, liver, and NSCLC models. Our in vitro observations might serve as a valuable foundation for future in vivo studies that could support the rationale for our conclusions and possibly enable providing more efficient and safer therapy to many oncological patients.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Carbazoles/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Biotransformation , Carbazoles/pharmacokinetics , Dogs , Humans , Madin Darby Canine Kidney Cells , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics
17.
Clin Drug Investig ; 39(6): 553-563, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31037611

ABSTRACT

BACKGROUND AND OBJECTIVES: Chiglitazar is a novel configuration-restricted non-thiazolidinedione peroxisome proliferator-activated receptor pan-agonist currently in the Phase III clinical development stage for type 2 diabetes mellitus patients. The objective of this Phase I study was to evaluate the pharmacokinetics, safety and tolerability of single and multiple doses of chiglitazar tablets taken orally and the effect of food on its pharmacokinetics in healthy Chinese subjects. METHODS: A single-centre, open-label, randomised, two-stage Phase I study was carried out. In the first-stage study, we evaluated a single dose of 8, 16, or 32 mg, and multiple doses of 16 mg, taken once daily for 9 days. The effect of food consumption was also studied in this stage. In the second-stage study, a greater range of single doses (24, 48 or 72 mg) were further evaluated. Pharmacokinetics, safety and tolerability profiles were assessed at each study stage. RESULTS: After a single oral dose of chiglitazar, at doses ranging from 8 to 72 mg, the maximum plasma concentration (Cmax) and area under the concentration-time curve (AUC) were proportionally increased (165-1599 ng/mL for the mean Cmax and 1356-12,584 ng·h/mL for the mean AUC0-t), with low inter-subject variability. There were no significant changes in the mean terminal phase half-life (t1/2), which ranged from 9.0 to 11.9 h, and the clearance and volume of distribution were similar for all evaluated doses. The results from the examination of multiple dose of 16 mg once daily for nine consecutive days showed that a steady-state condition was achieved by Day 6. There was no apparent accumulation of chiglitazar observed at Day 9, as compared with the first administration. While food increased the AUC0-t of chiglitazar by about 13%, there were no effects on other parameters, including Cmax, Tmax and t1/2. There were no serious or severe adverse events observed in the single- or multiple-dose studies. CONCLUSIONS: Chiglitazar tablets showed a good dose-dependent linear pharmacokinetic profile in the dose range of 8-72 mg. There was no accumulation after multiple daily administration of chiglitazar at a dose of 16 mg.  High-fat/calorie food increased the absorption of the drug, but there were no significant changes in exposure and other pharmacokinetic parameters. Chiglitazar was safe and well tolerated in healthy Chinese subjects at the dose levels and administration regimens evaluated.


Subject(s)
Carbazoles/administration & dosage , Peroxisome Proliferator-Activated Receptors/agonists , Propionates/administration & dosage , Administration, Oral , Adult , Area Under Curve , Asian People , Carbazoles/adverse effects , Carbazoles/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Propionates/adverse effects , Propionates/pharmacokinetics , Tablets , Young Adult
18.
Clin Pharmacol Drug Dev ; 8(7): 934-941, 2019 10.
Article in English | MEDLINE | ID: mdl-30809967

ABSTRACT

Chiglitazar (CHI) is a potent and selective peroxisome proliferator-activated receptor potentially for the treatment of patients with type 2 diabetes mellitus (T2DM). An open-label, randomized, 3-period crossover and self-controlled study was conducted to investigate drug-drug interaction potential between CHI and metformin hydrochloride (MET). Eligible subjects received a single oral dose of CHI (48 mg), MET (1000 mg), or a combination in each period, followed by serial blood sampling collected for up to 48 hours postdose, and safety was assessed throughout the trial. The area under the plasma concentration-time curves from time 0 to 48 hours (AUC0-48 h ) of CHI was similar following administration alone or with MET (AUC0-48h , 12 540 ng·h/mL [9811-15 269 ng·h/mL] vs 12 130 ng·h/mL [9304-14 956 ng·h/mL]; 90% confidence interval [CI] of its geometric mean ratio [GMR], 89.7%-103.8%), whereas the maximum concentration (Cmax ) of CHI was reduced during coadministration, as its 90%CI of the GMR was slightly outside the acceptance range for bioequivalence (Cmax , 1620 ng/mL [1418-1822 ng/mL] vs 1420 ng/mL [1049-1791 ng/mL], 90%CI GMR, 77.%-94.1%). However, it was not considered clinically meaningful. The MET exposures remained consistent in the absence or presence of CHI (AUC0-48 h , 12 570 ng·h/mL [10681-14 459 ng·h/mL] vs 13 190 [10973-15 407 ng·h/mL); 90%CI of GMR: 99.1%-110.5%; Cmax , 1790 ng/mL [1448-2132 ng/mL] vs 1820 ng/mL [1510-2130 ng/mL]; 90%CI of GMR, 94.2%-110.9%). No moderate to severe adverse events were reported. Our study indicated no clinically significant pharmacokinetic drug-drug interaction between CHI and MET and demonstrated good tolerance in subjects. These results support future application of CHI in combination with MET for treatment of T2DM.


Subject(s)
Carbazoles/administration & dosage , Carbazoles/pharmacokinetics , Metformin/administration & dosage , Metformin/pharmacokinetics , Propionates/administration & dosage , Propionates/pharmacokinetics , Administration, Oral , Area Under Curve , China , Cross-Over Studies , Drug Combinations , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Therapeutic Equivalency
19.
Pharmazie ; 74(2): 79-82, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30782255

ABSTRACT

IG-105, N-(2, 6-dimethoxypyridine-3-yl)-9-methylcarbazole-3-sulfonamide, a novel carbazole sulfonamide, shows a potent anticancer activity in a variety of human tumor cells in vitro and in vivo. In the present study, a rapid and convenient liquid chromatography/tandem mass spectrometry (LC-MS/MS) method was developed and applied to the pharmacokinetic study of IG-105 in rats. Chromatographic separation was accomplished on a C18 column using an isocratic mobile phase of acetonitrile-water-acetic acid (56:44:0.2, v/v/v). The ion transitions of IG-105 and combretastatin A4 (internal standard) in selected reaction monitoring mode were m/z 398→154 and m/z 317→286, respectively. The assay exhibited good linearity over the range of 2-512 ng/mL. Intra- and inter-day precisions were within 8.2 %, and the accuracies ranged from -6.0 to 3.7 %. The extraction recoveries were higher than 90 %, and the matrix effects were negligible. All quality control samples were stable at different storage conditions. The validated LC-MS/MS method was successfully applied to a preclinical pharmacokinetic study of IG-105 in rats after a single oral dose of 100, 250, or 1000 mg/kg which showed tumor growth inhibition activity. The absorption of IG-105 was proved to be rapid but saturated to a certain extent into the blood circulation, from where it was distributed and eliminated gradually.


Subject(s)
Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Carbazoles/blood , Carbazoles/pharmacokinetics , Chromatography, Liquid/methods , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Tandem Mass Spectrometry/methods , Tubulin Modulators , Animals , Dose-Response Relationship, Drug , Female , Male , Rats , Rats, Sprague-Dawley , Tubulin Modulators/blood , Tubulin Modulators/pharmacokinetics
20.
Clin Pharmacokinet ; 58(4): 403-420, 2019 04.
Article in English | MEDLINE | ID: mdl-29915924

ABSTRACT

The identification of anaplastic lymphoma kinase rearrangements in 2-5% of patients with non-small-cell lung cancer led to rapid advances in the clinical development of oral tyrosine kinase inhibitors. Anaplastic lymphoma kinase inhibitors are an effective treatment in preclinical models and patients with anaplastic lymphoma kinase-translocated cancers. Four anaplastic lymphoma kinase inhibitors (crizotinib, ceritinib, alectinib, and brigatinib) have recently been approved. Post-marketing studies provided additional pharmacokinetic information on their pharmacokinetic parameters. The pharmacokinetic properties of approved anaplastic lymphoma kinase inhibitors have been reviewed herein. Findings from additional studies on the effects of drug-metabolizing enzymes, drug transporters, and drug-drug interactions have been incorporated. Crizotinib, ceritinib, and alectinib reach their maximum plasma concentrations after approximately 6 h and brigatinib after 1-4 h. These drugs are primarily metabolized by cytochrome P450 3A with other cytochrome P450 enzymes. They are mainly excreted in the feces, with only a minor fraction being eliminated in urine. Crizotinib, ceritinib, and brigatinib are substrates for the adenosine triphosphate binding-cassette transporter B1, whereas alectinib is not. The different substrate specificities of the transporters play a key role in superior blood-brain barrier penetration by alectinib than by crizotinib and ceritinib. Although the absorption, distribution, and excretion of anaplastic lymphoma kinase inhibitors are regulated by drug transporters, their transporter-mediated pharmacokinetics have not yet been elucidated in detail in patients with non-small-cell lung cancer. Further research to analyze the contribution of drug transporters to the pharmacokinetics of anaplastic lymphoma kinase inhibitors in patients with non-small-cell lung cancer will be helpful for understanding the mechanisms of the inter-individual differences in the pharmacokinetics of anaplastic lymphoma kinase inhibitors.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Carbazoles/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Crizotinib/pharmacokinetics , Lung Neoplasms/metabolism , Organophosphorus Compounds/pharmacokinetics , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Sulfones/pharmacokinetics , Animals , Drug Interactions , Humans , Membrane Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...