Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.009
Filter
1.
Expert Opin Drug Metab Toxicol ; 20(4): 197-205, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38497279

ABSTRACT

INTRODUCTION: Colorectal cancer (CRC) is the third most diagnosed cancer globally and despite therapeutic strides, the prognosis for patients with metastatic disease (mCRC) remains poor. Fruquintinib is an oral vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor (TKI) targeting VEGFR -1, -2, and -3, and has recently received approval by the U.S. Food and Drug Administration for treatment of mCRC refractory to standard chemotherapy, anti-VEGF therapy, and anti-epidermal growth factor receptor (EGFR) therapy. AREAS COVERED: This article provides an overview of the pre-clinical data, pharmacokinetics, clinical efficacy, and safety profile of fruquintinib, as well as the management of clinical toxicities associated with fruquintinib. EXPERT OPINION: Fruquintinib is a valuable additional treatment option for patients with refractory mCRC. The pivotal role of vigilant toxicity management cannot be understated. While fruquintinib offers a convenient and overall, well-tolerated treatment option, ongoing research is essential to determine its efficacy in different patient subsets, evaluate it in combination with chemotherapy and immunotherapy, and determine its role in earlier lines of therapy.


Subject(s)
Antineoplastic Agents , Benzofurans , Colorectal Neoplasms , Neoplasm Metastasis , Protein Kinase Inhibitors , Quinazolines , Receptors, Vascular Endothelial Growth Factor , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Benzofurans/administration & dosage , Benzofurans/adverse effects , Benzofurans/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Animals , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Prognosis
2.
Pak J Pharm Sci ; 36(5): 1527-1542, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37869929

ABSTRACT

S1 and S2, two structurally similar quinazoline derivatives, are novel anticancer drugs targeting the PI3K/AKT/mTOR signaling pathway channel. However, their pharmacokinetic and tissue distribution characteristics are unknown, which has hindered further development and in-depth studies. In this study, a simple, rapid and sensitive method using high performance liquid chromatography was established and validated to quantitatively study the pharmacokinetics and tissue distribution profiles of S1 and S2 in rats following intravenous injection. The results indicated that after intravenous injection, the elimination of S1 and S2 fit the two-compartment model and linear pharmacokinetics characteristics were observed. Furthermore, S1 and S2 were widely distributed and found in high concentrations in liver and kidney tissues and a small proportion of S1 and S2 could cross the blood-brain barrier and be distributed in the brain. The current findings will contribute to interpretation and understanding the relationship between dosage and pharmacodynamic effects of S1 and S2.


Subject(s)
Antineoplastic Agents , Quinazolines , Animals , Rats , Antineoplastic Agents/pharmacokinetics , MTOR Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Tissue Distribution , TOR Serine-Threonine Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics
3.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36902093

ABSTRACT

We report synthesis of a novel 1,2,3,4-tetrahydroquinazoline derivative, named 2-(6,8-dibromo-3-(4-hydroxycyclohexyl)-1,2,3,4-tetrahydroquinazolin-2-yl)phenol (1), which was obtained from the hydrochloride of 4-((2-amino-3,5-dibromobenzyl)amino)cyclohexan-1-ol (ambroxol hydrochloride) and salicylaldehyde in EtOH. The resulting compound was produced in the form of colorless crystals of the composition 1∙0.5EtOH. The formation of the single product was confirmed by the IR and 1H spectroscopy, single-crystal and powder X-ray diffraction, and elemental analysis. The molecule of 1 contains a chiral tertiary carbon of the 1,2,3,4-tetrahydropyrimidine fragment and the crystal structure of 1∙0.5EtOH is a racemate. Optical properties of 1∙0.5EtOH were revealed by UV-vis spectroscopy in MeOH and it was established that the compound absorbs exclusively in the UV region up to about 350 nm. 1∙0.5EtOH in MeOH exhibits dual emission and the emission spectra contains bands at about 340 and 446 nm upon excitation at 300 and 360 nm, respectively. The DFT calculations were performed to verify the structure as well as electronic and optical properties of 1. ADMET properties of the R-isomer of 1 were evaluated using the SwissADME, BOILED-Egg, and ProTox-II tools. As evidenced from the blue dot position in the BOILED-Egg plot, both human blood-brain barrier penetration and gastrointestinal absorption properties are positive with the positive PGP effect on the molecule. Molecular docking was applied to examine the influence of the structures of both R-isomer and S-isomer of 1 on a series of the SARS-CoV-2 proteins. According to the docking analysis results, both isomers of 1 were found to be active against all the applied SARS-CoV-2 proteins with the best binding affinities with Papain-like protease (PLpro) and nonstructural protein 3 (Nsp3_range 207-379-AMP). Ligand efficiency scores for both isomers of 1 inside the binding sites of the applied proteins were also revealed and compared with the initial ligands. Molecular dynamics simulations were also applied to evaluate the stability of complexes of both isomers with Papain-like protease (PLpro) and nonstructural protein 3 (Nsp3_range 207-379-AMP). The complex of the S-isomer with Papain-like protease (PLpro) was found to be highly unstable, while the other complexes are stable.


Subject(s)
Ambroxol , COVID-19 , Coronavirus Papain-Like Proteases , Quinazolines , SARS-CoV-2 , Humans , Ambroxol/analogs & derivatives , Ambroxol/pharmacokinetics , Ambroxol/pharmacology , Molecular Docking Simulation , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Coronavirus Papain-Like Proteases/chemistry
4.
Pharm Biol ; 60(1): 621-626, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35289238

ABSTRACT

CONTEXT: Tucatinib (CYP2C8 substrate) and quercetin (CYP2C8 inhibitor) are two common drugs for the treatment of cancer. However, the effect of quercetin on the metabolism of tucatinib remains unknown. OBJECTIVE: We validated a sensitive method to quantify tucatinib levels in rat plasma based on ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), which was successfully employed to explore the effect of quercetin on tucatinib pharmacokinetics in rats. MATERIALS AND METHODS: An Acquity UPLC BEH C18 column was applied to achieve the separation of tucatinib and internal standard (IS) talazoparib after protein precipitation with acetonitrile. Then, we used this assay to investigate the effect of different doses of quercetin (25, 50 and 100 mg/kg) on the exposure of orally administered tucatinib (30 mg/kg) in 24 Sprague-Dawley (SD) rats, which were randomly divided into three quercetin pre-treated groups and one control group (n = 6). RESULTS: Our developed assay was verified in all aspects of bioanalytical method validation, involving lower limit of quantification (LLOQ), selectivity, accuracy and precision, calibration curve, extraction recovery, matrix effect and stability. After pre-treatment with 100 mg/kg quercetin, AUC0→t, AUC0→∞ and Cmax of tucatinib were remarkably increased by 75.4%, 75.8% and 59.1% (p < 0.05), respectively, while CLz/F was decreased significantly by 47.3% (p < 0.05) when compared with oral administration of 30 mg/kg tucatinib alone. This change is dose-dependent. CONCLUSIONS: This study will help better understand the pharmacokinetic properties of tucatinib with concurrent use with quercetin, and more clinical verifications were inspired to confirm whether this interaction has clinical significance in humans.


Subject(s)
Chromatography, High Pressure Liquid/methods , Oxazoles/pharmacokinetics , Pyridines/pharmacokinetics , Quercetin/pharmacology , Quinazolines/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Drug Interactions , Limit of Detection , Male , Oxazoles/administration & dosage , Oxazoles/analysis , Pyridines/administration & dosage , Pyridines/analysis , Quercetin/administration & dosage , Quinazolines/administration & dosage , Quinazolines/analysis , Rats , Rats, Sprague-Dawley
5.
Clin Pharmacol Drug Dev ; 11(8): 938-948, 2022 08.
Article in English | MEDLINE | ID: mdl-35238179

ABSTRACT

Letermovir is a human cytomegalovirus terminase inhibitor for the prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant recipients. The pharmacokinetics, safety, and tolerability of letermovir were assessed in healthy Japanese subjects in 2 phase 1 trials: trial 1-single ascending oral doses (240, 480, and 720 mg) and intravenous (IV) doses (240, 480, and 960 mg), and trial 2-multiple oral doses (240 and 480 mg once daily for 7 days). Following administration of oral single and multiple doses, letermovir was absorbed with a median time to maximum plasma concentration of 2 to 4 hours, and concentrations declined in a biphasic manner with a terminal half-life of ≈10 to 13 hours. The post absorption plasma concentration-time profile of letermovir following oral administration was similar to the profile observed with IV dosing. There was minimal accumulation with multiple-dose administration. Letermovir exposure in healthy Japanese subjects was ≈1.5- to 2.5-fold higher than that observed in non-Japanese subjects. Based on the population pharmacokinetic analysis, weight differences primarily accounted for the higher exposures observed in Asians. Letermovir was generally well tolerated following oral and IV administration to healthy Japanese subjects.


Subject(s)
Acetates , Quinazolines , Acetates/adverse effects , Acetates/pharmacokinetics , Area Under Curve , Humans , Metabolic Clearance Rate , Quinazolines/adverse effects , Quinazolines/pharmacokinetics
6.
J Med Chem ; 65(1): 633-664, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34928601

ABSTRACT

The Janus family of tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) play an essential role in the receptor signaling of cytokines that have been implicated in the pathogenesis of severe asthma, and there is emerging interest in the development of small-molecule-inhaled JAK inhibitors as treatments. Here, we describe the optimization of a quinazoline series of JAK inhibitors and the results of mouse lung pharmacokinetic (PK) studies where only low concentrations of parent compound were observed. Subsequent investigations revealed that the low exposure was due to metabolism by aldehyde oxidase (AO), so we sought to identify quinazolines that were not metabolized by AO. We found that specific substituents at the quinazoline 2-position prevented AO metabolism and this was rationalized through computational docking studies in the AO binding site, but they compromised kinome selectivity. Results presented here highlight that AO metabolism is a potential issue in the lung.


Subject(s)
Aldehyde Oxidase/metabolism , Janus Kinase Inhibitors/pharmacokinetics , Lung/metabolism , Administration, Intranasal , Administration, Intravenous , Animals , Binding Sites , Drug Delivery Systems , Female , Humans , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/chemical synthesis , Liver/metabolism , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Docking Simulation , Quinazolines/chemical synthesis , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Structure-Activity Relationship
7.
J Med Chem ; 65(1): 838-856, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34967623

ABSTRACT

The leucine-rich repeat kinase 2 (LRRK2) protein has been genetically and functionally linked to Parkinson's disease (PD), a disabling and progressive neurodegenerative disorder whose current therapies are limited in scope and efficacy. In this report, we describe a rigorous hit-to-lead optimization campaign supported by structural enablement, which culminated in the discovery of brain-penetrant, candidate-quality molecules as represented by compounds 22 and 24. These compounds exhibit remarkable selectivity against the kinome and offer good oral bioavailability and low projected human doses. Furthermore, they showcase the implementation of stereochemical design elements that serve to enable a potency- and selectivity-enhancing increase in polarity and hydrogen bond donor (HBD) count while maintaining a central nervous system-friendly profile typified by low levels of transporter-mediated efflux and encouraging brain penetration in preclinical models.


Subject(s)
Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/pharmacology , Brain/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Antiparkinson Agents/pharmacokinetics , Biological Availability , Drug Design , Humans , Models, Molecular , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacokinetics , Structure-Activity Relationship
8.
Clin Pharmacol Ther ; 111(3): 664-675, 2022 03.
Article in English | MEDLINE | ID: mdl-34888851

ABSTRACT

Rifampin has acute inhibitory and chronic inductive effects that can cause complex drug-drug interactions. Rifampin inhibits transporters including organic-anion-transporting polypeptide (OATP)1B and P-glycoprotein (P-gp), and induces enzymes and transporters including cytochrome P450 3A, UDP-glucuronosyltransferase (UGT)1A, and P-gp. This study aimed to separate inhibitory and inductive effects of rifampin on letermovir disposition and elimination (indicated for cytomegalovirus prophylaxis in hematopoietic stem cell transplant recipients). Letermovir is a substrate of UGT1A1/3, P-gp, and OATP1B, with its clearance primarily mediated by OATP1B. Letermovir (single-dose) administered with rifampin (single-dose) resulted in increased letermovir exposure through transporter inhibition. Chronic coadministration with rifampin (inhibition plus potential OATP1B induction) resulted in modestly decreased letermovir exposure vs. letermovir alone. Letermovir administered 24 hours after the last rifampin dose (potential OATP1B induction) resulted in markedly decreased letermovir exposure. These data suggest rifampin may induce transporters that clear letermovir; the modestly reduced letermovir exposure with chronic rifampin coadministration likely reflects the net effect of inhibition and induction. OATP1B endogenous biomarkers coproporphyrin (CP) I and glycochenodeoxycholic acid-sulfate (GCDCA-S) were also analyzed; their exposures increased after single-dose rifampin plus letermovir, consistent with OATP1B inhibition and prior reports of inhibition by rifampin alone. CP I and GCDCA-S exposures were substantially reduced with letermovir administered 24 hours after the last dose of rifampin vs. letermovir plus chronic rifampin coadministration. This study suggests that OATP1B induction may contribute to reduced letermovir exposure after chronic rifampin administration, although given the complexity of letermovir disposition alternative mechanisms are not fully excluded.


Subject(s)
Acetates/pharmacokinetics , Drug Interactions/physiology , Organic Anion Transporters/metabolism , Quinazolines/pharmacokinetics , Rifampin/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Area Under Curve , Biomarkers/metabolism , Coproporphyrins/metabolism , Cytochrome P-450 CYP3A/metabolism , Female , Hepatocytes/metabolism , Humans , Liver-Specific Organic Anion Transporter 1/metabolism , Middle Aged , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Young Adult
9.
Clin Pharmacol Ther ; 111(2): 485-495, 2022 02.
Article in English | MEDLINE | ID: mdl-34674258

ABSTRACT

The cytomegalovirus (CMV) viral terminase inhibitor letermovir is approved for prophylaxis of CMV infection and disease in adult CMV-seropositive allogeneic hematopoietic stem cell transplantation recipients. In a phase III trial (NCT02137772), letermovir significantly reduced clinically significant CMV infection (CS-CMVi) rate vs. placebo through Week 24 (primary end point) and Week 14 (secondary end point) post transplantation. Here, exposure-response relationships were investigated using efficacy and selected safety end points from the phase III trial to inform the proposed clinical dose. Post hoc exposure estimates were derived from a population pharmacokinetic model. No significant exposure dependencies were found for CS-CMVi through Week 24 or Week 14 among letermovir-treated participants. Evaluated covariates had no impact on exposure-efficacy relationships and letermovir plasma exposure did not affect time of CS-CMVi onset. There was no dependence between adverse event incidence and letermovir exposure. These results support current dosing recommendations in several countries and regions, including the United States and European Union.


Subject(s)
Acetates/administration & dosage , Antiviral Agents/administration & dosage , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation , Quinazolines/administration & dosage , Acetates/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/pharmacokinetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Dosage Calculations , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Models, Biological , Quinazolines/pharmacokinetics , Transplantation, Homologous , Treatment Outcome , Young Adult
10.
Chem Biol Interact ; 350: 109700, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34648813

ABSTRACT

AIM: To investigate the enzymatic properties of cytochrome P450 3A4 (CYP3A4) variants and their ability to metabolize vandetanib (VNT) in vitro, and to study potential drug interactions in combination with VNT. METHOD: Recombinant CYP3A4 cell microsomes were prepared using a Bac-to-Bac baculovirus expression system. Enzymatic reactions were carried out, and the metabolites were determined by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS: The activities of 27 CYP3A4 variants were determined to assess the degree of VNT metabolism that occurred. Analysis indicated that there was enhanced intrinsic clearance (Vmax/Km, CLint) for eight variants (CYP3A4.2, 3, 9, 15, 16, 29, 32, and 33), while there was a significant decrease in CYP3A4.5, 7, 8, 10-14, 17-20, 23, 24, 28, 31, and 34. Compared with CYP3A4.1, no significant differences were found for CYP3A4.6 and 30. Furthermore, the relative clearances were compared between VNT and cabozantinib, which were all metabolized by CYP3A4 with the same indications. When combined with ketoconazole, which is a CYP inhibitor, obvious differences were observed in the potency of VNT between different variants, including CYP3A4.2, 15, and 18. CONCLUSION: This comprehensive assessment of CYP3A4 variants provides significant insights into the allele-specific metabolism of VNT and drug interactions in vitro. We hope that these comprehensive data will provide references and predictions for the clinical application of VNT.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Piperidines/metabolism , Protein Kinase Inhibitors/metabolism , Quinazolines/metabolism , Alleles , Biotransformation , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Interactions , Genetic Association Studies , Genetic Variation , Humans , In Vitro Techniques , Ketoconazole/administration & dosage , Kinetics , Metabolic Clearance Rate , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Mol Pharm ; 18(9): 3509-3518, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34410132

ABSTRACT

Although KRAS has been an important target for many cancers, direct inhibition of oncogenic RAS remains challenging. Until recently, covalent KRAS G12C-specific inhibitors have been developed and progressed to the clinics. Nevertheless, not all patients benefit from these covalent inhibitors. At present, identification of candidates for this treatment requires tissue biopsies and gene sequencing, which are invasive, time-consuming, and could be of insufficient quality and limited predictive value owing to tumor heterogeneity. The use of noninvasive molecular imaging techniques such as PET and SPECT for spying KRAS G12C mutation in tumors provide a promising strategy for circumventing these hurdles. In the present study, based on the covalent G12C-specific inhibitor ARS-1620, we sought to develop radiolabeled small molecules for direct imaging of the KRAS mutation status in tumors. [131I]I-ARS-1620 and [18F]F-ARS-1620 were successfully prepared with high radiochemical yield, radiochemical purity, and molar activity. In vitro and in vivo studies have demonstrated the affinity, specificity, and capacity of [131I]I-ARS-1620 for direct imaging of the oncogenic KRAS G12C mutant. This initial attempt allows us to directly screen the KRAS G12C mutant for the first time in vivo.


Subject(s)
Neoplasms/diagnosis , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Quinazolines/administration & dosage , Radiopharmaceuticals/administration & dosage , Animals , Cell Line, Tumor , Female , Fluorine Radioisotopes , Humans , Iodine Radioisotopes , Mice , Molecular Docking Simulation , Molecular Imaging/methods , Mutation , Neoplasms/genetics , Piperazines/pharmacokinetics , Quinazolines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Single Photon Emission Computed Tomography Computed Tomography/methods , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Chem Pharm Bull (Tokyo) ; 69(7): 646-651, 2021.
Article in English | MEDLINE | ID: mdl-34193713

ABSTRACT

With the aim of studying the pharmacokinetics of letermovir, which is a newly developed antiviral agent for human cytomegalovirus, a rapid and simple ultra-performance liquid chromatography coupled with mass spectrometry (UPLC/MS) method was developed and validated for the quantification of letermovir in human plasma. Separation was performed in reverse phase mode using an ACQUITY UPLC BEH C18 column (130 Å, 1.7 µm, 2.1 × 50 mm) at a flow rate of 0.3 mL/min, 10 mM ammonium acetate-0.1% formic acid solution as mobile phase A, and acetonitrile as mobile phase B with a gradient elution. The method was validated over a linear range of 10-1000 ng/mL with a coefficient of determination (R2) >0.99 using weighted linear regression analysis. The intra- and inter-assay accuracy (nominal%) and precision (relative standard deviation%) were within ±15 and ≤15%, respectively. The specificity, recovery, matrix effect, stability, and dilution integrity of this method were also within acceptable limits. This method could be useful in studying the pharmacokinetics and pharmacodynamics, as well as performing the therapeutic drug monitoring of letermovir.


Subject(s)
Acetates/blood , Chromatography, High Pressure Liquid/methods , Quinazolines/blood , Tandem Mass Spectrometry/methods , Acetates/pharmacokinetics , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Half-Life , Humans , Limit of Detection , Quinazolines/pharmacokinetics , Reproducibility of Results
13.
Invest New Drugs ; 39(6): 1641-1648, 2021 12.
Article in English | MEDLINE | ID: mdl-34322775

ABSTRACT

Background The phosphatidylinositol-3 kinase pathway is often altered in head and neck squamous cell carcinoma (HNSCC), and is involved in the resistance to EGFR inhibitors. Objective We investigated the dose-limiting toxicities (DLTs), maximum tolerated dose, pharmacokinetics, and preliminary efficacy of the combination of copanlisib, an intravenous, pan-class I PI3K inhibitor, with the anti-EGFR monoclonal antibody cetuximab in recurrent and/or metastatic HNSCC patients in a phase I dose-escalation trial. Patients and methods Copanlisib was given intravenously on days 1, 8, and 15 of 28-day cycles at the dose of 45 mg and 30 mg, in combination with standard doses of weekly cetuximab (400 mg/m2 loading dose followed by 250 mg/m2 on days 8, 15, and 22, and weekly thereafter). Results Three patients received copanlisib 45 mg, of whom two experienced grade 3 hyperglycemia during Cycle 1 that met the DLT criteria. Eight patients were then treated with copanlisib at the dose of 30 mg. Because of the occurrence of hyperglycemia, a premedication with metformine was introduced on the day of the injections. No DLTs were reported at this dose level. The trial was stopped early because of the unfavourable toxicity profile of the combination. Among eight evaluable patients for response, four patients (50%) had disease stabilization according to RECIST1.1 as best response. Conclusion Copanlisib combined with cetuximab demonstrated unfavorable toxicity and limited efficacy in heavily pretreated recurrent and/or metastatic HNSCC patients.Trial registration NCT02822482, Date of registration: June 2016.


Subject(s)
Antineoplastic Agents/therapeutic use , Head and Neck Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Cetuximab/therapeutic use , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Middle Aged , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Phosphoinositide-3 Kinase Inhibitors/adverse effects , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/adverse effects , Quinazolines/pharmacokinetics
14.
Drug Metab Dispos ; 49(10): 892-901, 2021 10.
Article in English | MEDLINE | ID: mdl-34312304

ABSTRACT

Icotinib (ICT) is an antitumor drug approved by China National Medical Products Administration and is found to be effective against non-small cell lung cancer. The present study aimed at the interaction of ICT with CYP3A. ICT exhibited time-, concentration-, and NADPH-dependent inhibitory effect on recombinant human CYP3A4/5. About 60% of CYP3A activity was suppressed by ICT at 50 µM after 30 minutes. The observed enzyme inhibition could not be recovered by dialysis. Nifedipine protected CYP3A from the inactivation by ICT. The inhibitory effects of ICT on CYP3A were influenced neither by glutathione/N-acetyl lysine nor by superoxide dismutase/catalase. Incubation of ICT with human hepatic microsomes produced a ketene reactive intermediate trapped by 4-bromobenzylamine. CYP3A4 dominated the metabolic activation of ICT to the ketene intermediate. Ethyl and vinyl analogs of ICT did not induce inactivation of recombinant human CYP3A4/5, which indicates that acetylenic bioactivation of ICT contributed to the enzyme inactivation. Moreover, the metabolic activation of ICT resulted in heme destruction. In conclusion, this study demonstrated that ICT was a mechanism-based inactivator of recombinant human CYP3A4/5, and heme destruction by the ketene metabolite may be responsible for the observed CYP3A inactivation. SIGNIFICANCE STATEMENT: Cytochrome P450 enzymes play an important role in drug-drug interactions. The present study demonstrated that icotinib, an inhibitor of epidermal growth factor receptor used to treat non-small cell lung cancer, is a mechanism-based inactivator of recombinant human CYP3A4/5. The study provided solid evidence for the involvement of acetylene moiety in the metabolic activation as well as the inactivation of the enzyme. Furthermore, the resulting ketene intermediate was found to destroy heme, which is possibly responsible for the observed enzyme inactivation.


Subject(s)
Activation, Metabolic/drug effects , Carcinoma, Non-Small-Cell Lung , Crown Ethers/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Quinazolines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Enzyme Activation/drug effects , ErbB Receptors/antagonists & inhibitors , Ethylenes/metabolism , Heme/metabolism , Humans , Ketones/metabolism , Microsomes, Liver/metabolism , Recombinant Proteins/metabolism
15.
Int J Mol Sci ; 22(8)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33923880

ABSTRACT

A tyrosine kinase inhibitor, vandetanib (Van), is an anticancer drug affecting the signaling of VEGFR, EGFR and RET protooncogenes. Van is primarily used for the treatment of advanced or metastatic medullary thyroid cancer; however, its usage is significantly limited by side effects, particularly cardiotoxicity. One approach to minimize them is the encapsulation or binding of Van in- or onto a suitable carrier, allowing targeted delivery to tumor tissue. Herein, we constructed a nanocarrier based on apoferritin associated with Van (ApoVan). Based on the characteristics obtained by analyzing the average size, the surface ζ-potential and the polydispersive index, ApoVan nanoparticles exhibit long-term stability and maintain their morphology. Experiments have shown that ApoVan complex is relatively stable during storage. It was found that Van is gradually released from its ApoVan form into the neutral environment (pH 7.4) as well as into the acidic environment (pH 6.5). The effect of free Van and ApoVan on neuroblastoma and medullary thyroid carcinoma cell lines revealed that both forms were toxic in both used cell lines, and minimal differences between ApoVan and Van were observed. Thus, we assume that Van might not be encapsulated into the cavity of apoferritin, but instead only binds to its surface.


Subject(s)
Apoferritins/chemistry , Apoferritins/pharmacokinetics , Piperidines/chemistry , Piperidines/pharmacokinetics , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Cell Line, Tumor , Drug Stability , Humans , Hydrogen-Ion Concentration , Nanoparticles/chemistry
16.
Pharm Biol ; 59(1): 457-464, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33899675

ABSTRACT

CONTEXT: Dacomitinib and poziotinib, irreversible ErbB family blockers, are often used for treatment of non-small cell lung cancer (NSCLC) in the clinic. OBJECTIVE: This study investigates the effect of dacomitinib on the pharmacokinetics of poziotinib in rats. MATERIALS AND METHODS: Twelve Sprague-Dawley rats were randomly divided into two groups: the test group (20 mg/kg dacomitinib for 14 consecutive days) and the control group (equal amounts of vehicle). Each group was given an oral dose of 10 mg/kg poziotinib 30 min after administration of dacomitinib or vehicle at the end of the 14 day administration. The concentration of poziotinib in plasma was quantified by UPLC-MS/MS. Both in vitro effects of dacomitinib on poziotinib and the mechanism of the observed inhibition were studied in rat liver microsomes and human liver microsomes. RESULTS: When orally administered, dacomitinib increased the AUC, Tmax and decreased CL of poziotinib (p < 0.05). The IC50 values of M1 in RLM, HLM and CYP3A4 were 11.36, 30.49 and 19.57 µM, respectively. The IC50 values of M2 in RLM, HLM and CYP2D6 were 43.69, 0.34 and 0.11 µM, respectively, and dacomitinib inhibited poziotinib by a mixed way in CYP3A4 and CYP2D6. The results of the in vivo experiments were consistent with those of the in vitro experiments. CONCLUSIONS: This research demonstrates that a drug-drug interaction between poziotinib and dacomitinib possibly exists when readministered with poziotinib; thus, clinicians should pay attention to the resulting changes in pharmacokinetic parameters and accordingly, adjust the dose of poziotinib in clinical settings.


Subject(s)
Microsomes, Liver/metabolism , Quinazolines/pharmacokinetics , Quinazolinones/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Area Under Curve , Chromatography, High Pressure Liquid , Drug Interactions , Humans , Inhibitory Concentration 50 , Quinazolines/administration & dosage , Quinazolinones/administration & dosage , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
17.
Curr Drug Metab ; 22(10): 784-794, 2021.
Article in English | MEDLINE | ID: mdl-33622223

ABSTRACT

BACKGROUND: Letermovir is approved for prophylaxis of cytomegalovirus infection and disease in cytomegalovirus-seropositive hematopoietic stem-cell transplant (HSCT) recipients. OBJECTIVE: HSCT recipients are required to take many drugs concomitantly. The pharmacokinetics, absorption, distribution, metabolism, and excretion of letermovir and its potential to inhibit metabolizing enzymes and transporters in vitro were investigated to inform on the potential for drug-drug interactions (DDIs). METHODS: A combination of in vitro and in vivo studies described the absorption, distribution, metabolism, and routes of elimination of letermovir, as well as the enzymes and transporters involved in these processes. The effect of letermovir to inhibit and induce metabolizing enzymes and transporters was evaluated in vitro and its victim and perpetrator DDI potentials were predicted by applying the regulatory guidance for DDI assessment. RESULTS: Letermovir was a substrate of CYP3A4/5 and UGT1A1/3 in vitro. Letermovir showed concentration- dependent uptake into organic anionic transporting polypeptide (OATP)1B1/3-transfected cells and was a substrate of P-glycoprotein (P-gp). In a human ADME study, letermovir was primarily recovered as unchanged drug and minor amounts of a direct glucuronide in feces. Based on the metabolic pathway profiling of letermovir, there were few oxidative metabolites in human matrix. Letermovir inhibited CYP2B6, CYP2C8, CYP3A, and UGT1A1 in vitro, and induced CYP3A4 and CYP2B6 in hepatocytes. Letermovir also inhibited OATP1B1/3, OATP2B1, OAT3, OCT2, BCRP, BSEP, and P-gp. CONCLUSION: The body of work presented in this manuscript informed on the potential for DDIs when letermovir is administered both intravenously and orally in HSCT recipients.


Subject(s)
Acetates , Biotransformation , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/immunology , Drug Elimination Routes/physiology , Drug Interactions , Quinazolines , Tissue Distribution/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Acetates/metabolism , Acetates/pharmacokinetics , Adult , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Glucuronosyltransferase/metabolism , Healthy Volunteers , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Medication Therapy Management/standards , Neoplasm Proteins/metabolism , Organic Anion Transporters/metabolism , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Rats
18.
CPT Pharmacometrics Syst Pharmacol ; 10(3): 255-267, 2021 03.
Article in English | MEDLINE | ID: mdl-33440077

ABSTRACT

Letermovir is indicated for prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Two-stage population pharmacokinetic (PK) modeling of letermovir was conducted to support dose rationale and evaluate the impact of intrinsic/extrinsic factors. Data from healthy phase I study participants over a wide dose range were modeled to evaluate the effects of selected intrinsic factors, including pharmacogenomics; next, phase III HSCT-recipient data at steady-state following clinical doses were modeled. The model in HSCT recipients adequately described letermovir PK following both oral or i.v. administration, and was consistent with the healthy participant model at steady-state clinical doses. Intrinsic factor effects were not clinically meaningful. These staged analyses indicate that letermovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate. The HSCT recipient model was suitable for predicting exposure for exposure-response analysis supporting final dose selection.


Subject(s)
Acetates/pharmacokinetics , Antiviral Agents/pharmacokinetics , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Quinazolines/pharmacokinetics , Acetates/administration & dosage , Acetates/therapeutic use , Administration, Intravenous , Administration, Oral , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Biological Availability , Case-Control Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers/statistics & numerical data , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Pharmacogenetics , Placebos/administration & dosage , Quinazolines/administration & dosage , Quinazolines/therapeutic use , Transplant Recipients/statistics & numerical data
19.
Clin Pharmacol Drug Dev ; 10(2): 198-206, 2021 02.
Article in English | MEDLINE | ID: mdl-32700459

ABSTRACT

Letermovir is a prophylactic agent for cytomegalovirus infection and disease in adult cytomegalovirus-seropositive recipients of allogeneic hematopoietic stem cell transplant. As the antifungal agent fluconazole is administered frequently in transplant recipients, a drug-drug interaction study was conducted between oral letermovir and oral fluconazole. A phase 1 open-label, fixed-sequence study was performed in healthy females (N = 14, 19-55 years). In Period 1, a single dose of fluconazole 400 mg was administered. Following a 14-day washout, a single dose of letermovir 480 mg was administered (Period 2), and after a 7-day washout, single doses of fluconazole 400 mg and letermovir 480 mg were coadministered in Period 3. Pharmacokinetics and safety were evaluated. The pharmacokinetics of fluconazole and letermovir were not meaningfully changed following coadministration. Fluconazole geometric mean ratio (GMR; 90% confidence interval [CI]) with letermovir for area under the concentration-versus-time curve from time 0 to infinity (AUC0-∞ ) was 1.03 (0.99-1.08); maximum concentration (Cmax ) was 0.95 (0.92-0.99). Letermovir AUC0-∞ GMR (90%CI) was 1.11 (1.01-1.23), and Cmax was 1.06 (0.93-1.21) following coadministration with fluconazole. Coadministration of fluconazole and letermovir was generally well tolerated.


Subject(s)
Acetates/administration & dosage , Antifungal Agents/administration & dosage , Antiviral Agents/administration & dosage , Fluconazole/administration & dosage , Quinazolines/administration & dosage , Acetates/adverse effects , Acetates/pharmacokinetics , Adult , Antifungal Agents/adverse effects , Antifungal Agents/pharmacokinetics , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Area Under Curve , Drug Interactions , Female , Fluconazole/adverse effects , Fluconazole/pharmacokinetics , Humans , Middle Aged , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Young Adult
20.
Biomed Chromatogr ; 35(4): e5015, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33125719

ABSTRACT

Phosphatidylinositol 3-kinase (PI3K) inhibitors are a novel class of anticancer drugs that are approved to treat various malignancies. We report the development and validation of a HPLC method for the simultaneous quantitation of three PI3K inhibitors, namely copanlisib, duvelisib and idelalisib, in rat plasma as per the regulatory guidelines of the United States Food and Drug Administration. The method involves extraction of copanlisib, duvelisib and idelalisib along with an internal standard (IS; filgotinib) from rat plasma (100 µL) using a liquid-liquid extraction process. The chromatographic separation of the analytes was achieved using step-wise gradient elution on a Hypersil Gold C18 column. The UV detection wavelength was set at λmax = 280 nm. Copanlisib, duvelisib, idelalisib and the IS eluted at 7.16, 12.6, 11.9 and 9.86 min, respectively, with a total run time of 15 min. The calibration curve ranged from 50 to 5000 ng/mL for all the analytes. Inter- and intra-day precision and accuracy, stability studies, dilution integrity and incurred sample reanalysis were investigated for all three analytes, and the results met the acceptance criteria. The validated HPLC method was successfully applied to a pharmacokinetic study in rats.


Subject(s)
Chromatography, High Pressure Liquid/methods , Liquid-Liquid Extraction/methods , Phosphoinositide-3 Kinase Inhibitors/blood , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Isoquinolines/blood , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Linear Models , Male , Phosphoinositide-3 Kinase Inhibitors/chemistry , Purines/blood , Purines/chemistry , Purines/pharmacokinetics , Pyrimidines/blood , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Quinazolines/blood , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolinones/blood , Quinazolinones/chemistry , Quinazolinones/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...