Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Clin Ther ; 42(7): 1317-1329, 2020 07.
Article in English | MEDLINE | ID: mdl-32622784

ABSTRACT

PURPOSE: Fixed-dose combination glecaprevir (GLE) 300 mg + pibrentasvir (PIB) 120 mg is an orally administered once daily antiviral regimen approved for the treatment of hepatitis C virus (HCV) infection. The objective of this study was to evaluate the potential for cardiac repolarization following GLE + PIB administration in healthy adults. METHODS: This placebo- and active-controlled, randomized, single-dose, 4-period, 4-sequence crossover study enrolled 48 healthy subjects. The doses of GLE 400 mg + PIB 120 mg were selected to provide exposures comparable to those with the doses that are therapeutic in the HCV-infected population, GLE 300 mg + PIB 120 mg. The doses of GLE 600 mg + PIB 240 mg were selected to provide supratherapeutic exposures without exceeding the exposures of the GLE + PIB maximal tolerated doses. Moxifloxacin 400 mg (active control/open label) was used for confirming the sensitivity of the ECG assay in detecting QTc prolongation. Time-matched plasma concentrations and triplicate ECGs were obtained on treatment days -1 and 1. The primary end point was time-matched, placebo-corrected, baseline-adjusted Fridericia-corrected QT interval (ΔΔQTcF). Pharmacokinetic-pharmacodynamic analyses characterized the relationship between GLE and PIB plasma concentrations and ΔΔQTcF using a linear regression model and linear mixed-effects model. Findings from categorical analyses of ECG-interval data were also summarized. Tolerability was evaluated through adverse-events monitoring, physical examination including vital sign measurements, ECGs, and laboratory tests. FINDINGS: A total of 48 subjects (22 women [46%], 26 men [54%]), were enrolled in the study, and 47 subjects completed all 4 periods. None of the subjects had a change from baseline in QTcF interval of >30 msec or an absolute QTcF interval of >450 msec. Peak ΔΔQTcF values observed at 5 h postdose (Tmax) were 2.9 msec (upper 95% confidence limit, 4.9 msec) with the therapeutic dose and 3.1 msec (upper 95% confidence limit, 5.1 msec) with the supratherapeutic dose, with both upper 95% confidence limits well below the 10-msec threshold. Assay sensitivity was confirmed by peak ΔΔQTcF in the positive control (12.8 ms at 2 h postdose). No statistically significant GLE or PIB concentration-dependent effects on ΔΔQTcF were observed. Headache and skin irritation from ECG electrodes were the most commonly reported AEs. No clinically significant vital sign measurements, ECG findings, or laboratory measurements were observed. There were no patterns of T- and U-wave morphologic abnormalities. IMPLICATIONS: The fixed-dose combination regimen of GLE/PIB does not prolong the QTc interval. ClinicalTrials.gov identifier.


Subject(s)
Aminoisobutyric Acids/administration & dosage , Benzimidazoles/administration & dosage , Cyclopropanes/administration & dosage , Heart Rate/drug effects , Heart/drug effects , Lactams, Macrocyclic/administration & dosage , Leucine/analogs & derivatives , Proline/analogs & derivatives , Quinoxalines/administration & dosage , Sulfonamides/administration & dosage , Adult , Aminoisobutyric Acids/blood , Aminoisobutyric Acids/pharmacokinetics , Benzimidazoles/blood , Benzimidazoles/pharmacokinetics , Cross-Over Studies , Cyclopropanes/blood , Cyclopropanes/pharmacokinetics , Double-Blind Method , Drug Combinations , Electrocardiography/drug effects , Female , Healthy Volunteers , Heart/physiology , Humans , Lactams, Macrocyclic/blood , Lactams, Macrocyclic/pharmacokinetics , Leucine/administration & dosage , Leucine/blood , Leucine/pharmacokinetics , Long QT Syndrome , Male , Middle Aged , Proline/administration & dosage , Proline/blood , Proline/pharmacokinetics , Pyrrolidines , Quinoxalines/blood , Quinoxalines/pharmacokinetics , Single-Blind Method , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Young Adult
2.
J Clin Pharmacol ; 60(9): 1254-1267, 2020 09.
Article in English | MEDLINE | ID: mdl-32441835

ABSTRACT

While an initial clinical absorption, distribution, metabolism, and excretion (ADME) study (Study 1; N = 6) with 100 mg/100 µCi [14 C]lorlatinib, radiolabeled on the carbonyl carbon, confirmed that the primary metabolic pathways for lorlatinib are oxidation (N-demethylation, N-oxidation) and N-glucuronidation, it also revealed an unanticipated, intramolecular cleavage metabolic pathway of lorlatinib, yielding a major circulating benzoic acid metabolite (M8), and an unlabeled pyrido-pyrazole substructure. Concerns regarding the fate of unknown metabolites associated with this intramolecular cleavage pathway led to conduct of a second ADME study (Study 2; N = 6) of identical design but with the radiolabel positioned on the pyrazole ring. Results were similar with respect to the overall mass balance, lorlatinib plasma exposures, and metabolic profiles in excreta for the metabolites that retained the radiolabel in both studies. Differences were observed in plasma total radioactivity exposures (2-fold area under the plasma concentration-time curve from time 0 to infinity difference) and relative ratios of the percentage of dose recovered in urine vs feces (48% vs 41% in Study 1; 28% vs 64% in Study 2). In addition, an approximately 3-fold difference in the mean molar exposure ratio of M8 to lorlatinib was observed for values derived from metabolic profiling data relative to those derived from specific bioanalytical methods (0.5 vs 1.4 for Studies 1 and 2, respectively). These interstudy differences were attributed to a combination of factors, including alteration of radiolabel position, orthogonal analytical methodologies, and intersubject variability, and illustrate that results from clinical ADME studies are not unambiguous and should be interpreted within the context of the specific study design considerations.


Subject(s)
Carbon Radioisotopes/metabolism , Carbon Radioisotopes/pharmacokinetics , Lactams, Macrocyclic/metabolism , Lactams, Macrocyclic/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aminopyridines , Biotransformation , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/chemistry , Drug Elimination Routes , Feces/chemistry , Healthy Volunteers , Humans , Lactams , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/chemistry , Male , Middle Aged , Models, Biological , Pyrazoles , Solutions , Young Adult
3.
Cancer Med ; 9(12): 4350-4359, 2020 06.
Article in English | MEDLINE | ID: mdl-32347012

ABSTRACT

OBJECTIVE: To clarify the distribution of lorlatinib in the brain and elucidate the molecular mechanisms of lorlatinib penetration across the blood-brain barrier (BBB). METHODS: Cytological experiments were performed to investigate the growth inhibitory effect of lorlatinib on different cells (endothelial cells HUVEC, HMEC-1, and HCMEC/D3) and to investigate the protective effect of lorlatinib on neuronal cells after SH-SY5Y hypoxia/reoxygenation injury. Furthermore, rat brain tissue was sequenced, and the differentially expressed genes (secreted phosphoprotein 1 (SPP1), vascular endothelial growth factor (VEGF), transforming growth factor beta (TGF-ß), Claudin, ZO-1 and P-gp) in several different drug treatment groups were verified by Real-Time PCR. Lorlatinib brain distribution was predicted by physiologically based pharmacokinetics (PBPK). RESULTS: Lorlatinib and crizotinib both had inhibitory effects on endothelial cells, however lorlatinib inhibited the growth of HCMEC/D3 more efficaciously than crizotinib. In the SH-SY5Y hypoxia model, lorlatinib had a greater protective effect on nerve cell damage caused by hypoxia and reoxygenation than crizotinib. The expression of SPP1, VEGF, TGF-ß, and Claudin in brain tissue was significantly downregulated after lorlatinib administration, and the expression level of early growth transcription factor 1 (Egr-1) was significantly increased. The PBPK model successfully described lorlatinib concentrations in blood and brain tissue in the mouse model and gave a brain tissue partition coefficient of 0.7. CONCLUSION: Lorlatinib can increase the permeability of the blood-brain barrier whereby we suggest its underlying working mechanism is related to downregulating SPP1, inhibiting VEGF, TGF-ß, and Claudin subsequently reducing the number of tight junctions between BBB cells. Lorlatinib plays a protective role on injured nerve cells and does not change the amount of P-gp expression in brain tissue, which may be important for its ability to be efficacious across the BBB with a low incidence of resistance.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Membrane Permeability/drug effects , Hypoxia/complications , Ischemia/drug therapy , Lactams, Macrocyclic/pharmacology , Neuroblastoma/drug therapy , Reperfusion Injury/drug therapy , Aminopyridines , Animals , Biological Transport , Blood-Brain Barrier/drug effects , Humans , Ischemia/etiology , Ischemia/metabolism , Ischemia/pathology , Lactams , Lactams, Macrocyclic/pharmacokinetics , Male , Neuroblastoma/metabolism , Neuroblastoma/pathology , Pyrazoles , Rats , Rats, Sprague-Dawley , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tissue Distribution , Tumor Cells, Cultured
4.
J Clin Pharmacol ; 60(3): 331-339, 2020 03.
Article in English | MEDLINE | ID: mdl-31515816

ABSTRACT

Glecaprevir (GLE)/pibrentasvir (PIB) 300 mg/120 mg once daily (Mavyret/Maviret) is an all-oral, pangenotypic, interferon- and ribavirin-free combination regimen approved for the treatment of chronic hepatitis C virus (HCV) infection. The objective of the current analyses was to characterize the pharmacokinetics (PK) of GLE/PIB in HCV-infected Japanese patients. Data from 332 subjects enrolled in 2 Japan phase 3 trials, CERTAIN-1 and CERTAIN-2, were used in the analyses. Pharmacokinetics of GLE/PIB were characterized using a nonlinear mixed-effects modeling. The analyses evaluated the impact of covariates (concomitant medications and demographic and clinical covariates such as renal impairment, effect of cirrhotic status) on GLE/PIB PK. GLE and PIB PK were described by 1- and 2-compartment models, respectively. Presence of cirrhosis, age, and body weight were identified as significant covariates on GLE/PIB PK. A trend toward higher GLE and PIB exposures in older patients and higher PIB exposures in heavier patients was observed; however, these increases were not considered clinically meaningful. GLE and PIB exposures were higher in HCV-infected subjects with cirrhosis (Child-Pugh A; GLE area under the plasma concentration-time curve was 160% higher, and PIB area under the plasma concentration-time curve was 21% higher) compared to subjects without cirrhosis. Renal function (including subjects with end-stage renal disease with dialysis) had no impact on GLE or PIB exposures. The GLE/PIB dose was well tolerated in the Japanese population, and no dose adjustment is needed for the evaluated intrinsic and extrinsic factors.


Subject(s)
Aminoisobutyric Acids/pharmacokinetics , Antiviral Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Cyclopropanes/pharmacokinetics , Hepatitis C, Chronic/drug therapy , Lactams, Macrocyclic/pharmacokinetics , Leucine/analogs & derivatives , Proline/analogs & derivatives , Quinoxalines/pharmacokinetics , Sulfonamides/pharmacokinetics , Administration, Oral , Adult , Age Factors , Aged , Aged, 80 and over , Aminoisobutyric Acids/administration & dosage , Aminoisobutyric Acids/adverse effects , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Area Under Curve , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Biological Availability , Body Weight , Cyclopropanes/administration & dosage , Cyclopropanes/adverse effects , Drug Administration Schedule , Drug Combinations , Female , Hepatitis C, Chronic/blood , Humans , Japan , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/adverse effects , Leucine/administration & dosage , Leucine/adverse effects , Leucine/pharmacokinetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Middle Aged , Models, Biological , Proline/administration & dosage , Proline/adverse effects , Proline/pharmacokinetics , Pyrrolidines , Quinoxalines/administration & dosage , Quinoxalines/adverse effects , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Young Adult
5.
Adv Healthc Mater ; 9(2): e1901307, 2020 01.
Article in English | MEDLINE | ID: mdl-31814332

ABSTRACT

A nanoplatform that integrates diagnostic and therapeutic functions with intrinsic tumor microenvironment-responsive biodegradability is highly desired. Herein, a biodegradable nanotheranostic agent based on hollow mesoporous organosilica nanoparticles (HMONs), followed by encapsulating of heat shock protein 90 (Hsp 90) inhibitor is described. Then, the pore-engineering including gating with bovine serum albumin-iridium oxide nanoparticles (BSA-IrO2 ) and conjugation of polyethylene glycol (PEG) is conducted to yield 17AAG@HMONs-BSA-IrO2 -PEG (AHBIP) nanotheranostics for multimode computed tomography (CT)/photoacoustic (PA) imaging-guided photodynamic therapy (PDT) and low-temperature photothermal therapy (PTT). Such nanoplatforms show extraordinary photothermal conversion efficiency, high cargo loading (35.4% for 17AAG), and stimuli-responsive release of 17AAG for inhibition of Hsp90, which induces cell apoptosis at low-temperatures (≈41 °C). Also, the IrO2 simultaneously endows the nanotheranostics with catalytic activity in triggering the decomposition of H2 O2 into O2 and thus reducing the tumor hypoxia, as well as protecting normal tissues against H2 O2 -induced inflammation. AHBIP shows good photocatalysis activity for PDT as a result of the generation of superoxide anion by laser irradiation. The resulting AHBIP-mediated synergistic PTT/PDT offers an outstanding therapeutic outcome both in vitro and in vivo. Overall, the incorporation of the BSA-IrO2 and biodegradable HMONs into one nanoplatform has great potential for clinical applications.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoquinones/administration & dosage , Drug Delivery Systems/methods , Lactams, Macrocyclic/administration & dosage , Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzoquinones/pharmacokinetics , Biocompatible Materials/chemistry , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Hydrogen Peroxide/chemistry , Iridium/chemistry , Lactams, Macrocyclic/pharmacokinetics , Mice, Inbred C57BL , Mice, Nude , Oxygen/pharmacokinetics , Photoacoustic Techniques , Photochemotherapy/methods , Polyethylene Glycols/chemistry , Serum Albumin, Bovine/chemistry , Superoxides/metabolism , Theranostic Nanomedicine/instrumentation , Tomography, X-Ray Computed , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Adv Ther ; 37(2): 745-758, 2020 02.
Article in English | MEDLINE | ID: mdl-31863284

ABSTRACT

INTRODUCTION: Lorlatinib is a third-generation tyrosine kinase inhibitor approved for the treatment of anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer; cytochrome P450 (CYP) 3A plays an important role in the metabolism of lorlatinib. METHODS: This phase 1, open-label, two-period, crossover study estimated the effect of oral rifampin (a strong CYP3A inducer) on the pharmacokinetics and safety of oral lorlatinib (NCT02804399). Healthy participants received single-dose lorlatinib 100 mg in period 1 followed by rifampin 600 mg/day (days 1-12) and single-dose lorlatinib 100 mg (day 8) in period 2. Blood samples were collected for 120 h after each dose of lorlatinib. RESULTS: When a single dose of lorlatinib was administered during daily dosing with rifampin (period 2), the area under the plasma concentration-time profile extrapolated to infinity (AUCinf) and maximum plasma concentration (Cmax) of lorlatinib were 14.74% [90% confidence interval (CI) 12.78%, 17.01%] and 23.88% (90% CI 21.58%, 26.43%), respectively, of those in period 1 (lorlatinib alone). A single dose of lorlatinib was well tolerated in period 1, but elevations in transaminase values were observed in all participants (grade 2-4 in 11 participants) within 1-3 days after a single dose of lorlatinib was administered with ongoing rifampin in period 2. Rifampin dosing was therefore halted. Transaminase levels subsequently returned to normal (median time to recovery: 15 days). No elevations in bilirubin were observed. CONCLUSIONS: The addition of a single dose of lorlatinib to daily dosing with rifampin significantly reduced lorlatinib plasma exposure relative to a single dose of lorlatinib administered alone and was associated with severe but self-limiting transaminase elevations in all healthy participants. These observations support the contraindication in the product label against concomitant use of lorlatinib with all strong CYP3A inducers. TRIAL REGISTRATION: ClinicalTrials.gov identifier, NCT02804399.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lactams, Macrocyclic/pharmacokinetics , Lactams, Macrocyclic/therapeutic use , Lung Neoplasms/drug therapy , Rifampin/pharmacokinetics , Rifampin/therapeutic use , Adult , Aminopyridines , Area Under Curve , Cross-Over Studies , Drug Interactions , Female , Healthy Volunteers , Humans , Lactams , Male , Middle Aged , Pyrazoles , Young Adult
7.
Invest New Drugs ; 38(1): 131-139, 2020 02.
Article in English | MEDLINE | ID: mdl-31728714

ABSTRACT

Background The third-generation tyrosine kinase inhibitor lorlatinib is approved for the treatment of ALK-positive metastatic NSCLC. CYP3A plays a major role in lorlatinib metabolism; therefore, a drug-drug interaction study was warranted to evaluate the impact of the strong CYP3A inhibitor, itraconazole, on lorlatinib plasma exposure. Methods This phase 1, open-label, 2-period, crossover study estimated the effects of itraconazole on the plasma pharmacokinetics and safety of lorlatinib in healthy participants (NCT02838264). Single-dose lorlatinib 50 mg (n = 2), 75 mg (n = 2) and 100 mg (n = 12) was administered in Period 1. In Period 2, itraconazole oral solution 200 mg/day was administered on Days 1-11, and single-dose lorlatinib on Day 5. Blood samples were collected up to 168 h after lorlatinib dosing. Results During daily dosing with itraconazole (Period 2), the ratios of the adjusted geometric means for area under the plasma concentration-time profile extrapolated to infinity (AUCinf) and maximum plasma concentration (Cmax) of single-dose lorlatinib 100 mg were 141.79% (90% confidence interval, 128.71%, 156.21%) and 124.39% (110.20%, 140.41%), respectively, compared with Period 1 (lorlatinib alone). Lorlatinib was well tolerated alone and with itraconazole. No serious adverse events or withdrawals were reported. Conclusions Co-administration of itraconazole and lorlatinib increased the plasma exposure of lorlatinib relative to lorlatinib alone in healthy participants. Therefore, concomitant use of lorlatinib with strong CYP3A inhibitors should be avoided. If this combination is unavoidable, the starting dose of lorlatinib should be reduced from 100 mg to 75 mg.


Subject(s)
Itraconazole/pharmacology , Lactams, Macrocyclic/pharmacokinetics , Adult , Aminopyridines , Antifungal Agents/pharmacology , Area Under Curve , Cross-Over Studies , Drug Interactions , Female , Healthy Volunteers , Humans , Lactams , Lactams, Macrocyclic/pharmacology , Male , Pyrazoles , Tissue Distribution
8.
Clin Pharmacol Ther ; 107(6): 1325-1333, 2020 06.
Article in English | MEDLINE | ID: mdl-31868916

ABSTRACT

Fine-needle aspiration (FNA) for serial hepatic sampling may be an efficient and less invasive alternative to core needle biopsy (CNB), the current standard for liver tissue sampling. In this randomized, open-label trial in 31 participants with hepatitis C virus genotype 1 infection (NCT01678131/Merck protocol PN048), we evaluated the feasibility of using FNA to obtain human liver tissue samples appropriate for measuring hepatic pharmacokinetics (PK), using vaniprevir as a tool compound. The primary end point was successful retrieval of liver tissue specimens with measurable vaniprevir concentrations at two of three specified FNA time points. Twenty-nine patients met the primary end point and, therefore, were included in the PK analyses. Hepatic vaniprevir concentrations obtained with FNA were consistent with known vaniprevir PK properties. The shape of liver FNA and CNB concentration-time profiles were comparable. In conclusion, FNA may be effective for serial tissue sampling to assess hepatic drug exposure in patients with liver disease.


Subject(s)
Antiviral Agents/pharmacokinetics , Biopsy, Fine-Needle/methods , Cyclopropanes/pharmacokinetics , Hepatitis C, Chronic/drug therapy , Isoindoles/pharmacokinetics , Lactams, Macrocyclic/pharmacokinetics , Leucine/analogs & derivatives , Liver/metabolism , Proline/analogs & derivatives , Sulfonamides/pharmacokinetics , Adult , Antiviral Agents/administration & dosage , Cyclopropanes/administration & dosage , Female , Genotype , Hepacivirus/genetics , Hepacivirus/isolation & purification , Hepatitis C, Chronic/virology , Humans , Isoindoles/administration & dosage , Lactams, Macrocyclic/administration & dosage , Leucine/administration & dosage , Leucine/pharmacokinetics , Liver/virology , Male , Middle Aged , Proline/administration & dosage , Proline/pharmacokinetics , Sulfonamides/administration & dosage , Tissue Distribution , Young Adult
9.
Biol Pharm Bull ; 43(3): 384-392, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31685755

ABSTRACT

Transporter gene knockout models are a practical and widely used tool for pharmacokinetic studies in drug discovery. P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp) are major efflux transporters that control absorption and bioavailability, and are important when determining oral drug disposition. To the best of our knowledge, beyond the rule of five (bRo5) molecules launched on the market to date tend to be substrates for efflux transporters. The purpose of this study is to evaluate in vivo the impact of efflux transporters on the oral absorption process and systemic clearance using rats which lack P-gp and/or Bcrp expression. We administered five bRo5 substrates (asunaprevir, cyclosporine, danoprevir, ledipasvir, and simeprevir) intravenously or orally to wild-type and Mdr1a, Bcrp, and Mdr1a/Bcrp knockout rats, calculated the clearance, oral bioavailability, and absorption rate profile of each substrate, and compared the results. Systemic clearance of the substrates in knockout rats changed within approximately ±40% compared to wild-types, suggesting the efflux transporters do not have a significant influence on clearance in rats. On the other hand, the oral absorption of substrates in the knockout rats, especially those lacking Mdr1a, increased greatly-between 2- and 5-fold more than in wild-types. This suggests that rat efflux transporters, especially P-gp, greatly reduce the oral exposure of these substrates. Moreover, results on the absorption rate-time profile suggest that efflux transporters are constantly active during the absorption period in rats. Transporter knockout rats are a useful in vivo tool for estimating the transporter-mediated disposition of bRo5 molecules in drug discovery.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Benzimidazoles/pharmacokinetics , Cyclopropanes/pharmacokinetics , Cyclosporine/pharmacokinetics , Fluorenes/pharmacokinetics , Isoindoles/pharmacokinetics , Isoquinolines/pharmacokinetics , Lactams, Macrocyclic/pharmacokinetics , Proline/analogs & derivatives , Simeprevir/pharmacokinetics , Sulfonamides/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/blood , Biological Availability , Cyclopropanes/administration & dosage , Cyclopropanes/blood , Cyclosporine/administration & dosage , Cyclosporine/blood , Fluorenes/administration & dosage , Fluorenes/blood , Gene Knockout Techniques , Isoindoles/administration & dosage , Isoindoles/blood , Isoquinolines/administration & dosage , Isoquinolines/blood , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/blood , Male , Metabolic Clearance Rate/genetics , Oral Mucosal Absorption/genetics , Proline/administration & dosage , Proline/blood , Proline/pharmacokinetics , Rats , Rats, Sprague-Dawley , Simeprevir/administration & dosage , Simeprevir/blood , Sulfonamides/administration & dosage , Sulfonamides/blood
10.
Drugs ; 79(1): 93-98, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30604291

ABSTRACT

Lorlatinib is an oral small molecule inhibitor of anaplastic lymphoma kinase (ALK) and C-ros oncogene 1 (ROS1) kinase developed by Pfizer for the treatment of ALK-positive non-small cell lung cancer (NSCLC). Based on results from a phase I/II trial, lorlatinib received approval in Japan in September 2018 and in the USA in November 2018 for the treatment of ALK-positive NSCLC. This article summarizes the milestones in the development of lorlatinib leading to the first global approval for this indication.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Lactams, Macrocyclic , Protein Kinase Inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Aminopyridines , Child , Drug Approval , Humans , Japan , Lactams , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/adverse effects , Lactams, Macrocyclic/pharmacokinetics , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles , United States , United States Food and Drug Administration
11.
J Med Chem ; 61(19): 8774-8796, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30180567

ABSTRACT

Bifunctional derivatives of bis(phosphinate)-bearing cyclam (BPC) chelators bearing a carboxylate, amine, isothiocyanate, azide, or cyclooctyne in the BP side chain were synthesized. Conjugations required no protection of phosphinate or ring secondary amine groups. The ring amines were not reactive (proton protected) at pH < ∼8. For isothiocyanate coupling, oligopeptide N-terminal α-amines were more suitable than alkyl amines, e.g., Lys ω-amine (p Ka ∼7.5-8.5 and ∼10-11, respectively) due to lower basicity. The Cu-64 labeling was efficient at room temperature (specific activity ∼100 GBq/µmol; 25 °C, pH 6.2, ∼100 ligand equiv, 10 min). A representative Cu-64-BPC was tested in vivo showing fast clearance and no nonspecific radioactivity deposition. The monoclonal anti-PSCA antibody 7F5 conjugates with thiocyanate BPC derivative or NODAGA were radiolabeled and studied in PC3-PSCA tumor bearing mice by PET. The radiolabeled BPC conjugate was accumulated in the prostate tumor with a low off-target uptake, unlike Cu-64-labeled NODAGA-antibody conjugate. The BPC chelators have a great potential for theranostic applications of the Cu-64/Cu-67 matched pair.


Subject(s)
Antibodies, Monoclonal/chemistry , Chelating Agents/chemistry , Copper Radioisotopes/chemistry , Immunoconjugates/chemistry , Lactams, Macrocyclic/chemistry , Phosphinic Acids/chemistry , Radiopharmaceuticals/chemistry , Animals , Antibodies, Monoclonal/pharmacokinetics , Chelating Agents/pharmacokinetics , Copper Radioisotopes/pharmacokinetics , Drug Stability , Immunoconjugates/pharmacokinetics , Isotope Labeling , Lactams, Macrocyclic/pharmacokinetics , Ligands , Male , Mice , Positron-Emission Tomography , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Rats, Nude , Rats, Wistar , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
J Pharm Biomed Anal ; 161: 136-143, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30149189

ABSTRACT

Several second and third generation ALK inhibitors have been introduced in recent years. A bioanalytical assay for simultaneous quantification of alectinib, brigatinib, and lorlatinib was developed and validated for human plasma. The method was also partially validated for diluted mouse plasma and tissue homogenates of brain, liver, kidney, and spleen. Samples (40 µl) were pretreated in a 96-well plate by protein precipitation with acetonitrile containing the internal standard [2H8]-alectinib. After chromatographic separation on an ethylene bridged octadecyl silica column by gradient elution at 600 µl/min using 1% (v/v) formic acid (in water) and acetonitrile, compounds were ionized by a turbo electrospray and monitored by selected reaction monitoring on a triple quadrupole mass spectrometer. Validation was performed in a 2-2000 ng/ml concentration range for alectinib and lorlatinib and a 4-4000 ng/ml range for brigatinib. Precisions (within-day and between-day) were in the range 2.2-15.0% and accuracies were in between 87.2 and 110.2% for all matrices and levels. Compounds were sufficiently stable under most investigated conditions. Results of a pilot pharmacokinetic and tissue distribution study for brigatinib in mice are reported. Finally, successful incurred samples reanalysis of tissue homogenate samples containing brigatinib and lorlatinib is presented. Lorlatinib homogenate samples were also successfully reanalyzed using a second independent assay (cross-validation).


Subject(s)
Carbazoles/blood , Carbazoles/pharmacokinetics , Chromatography, Liquid/methods , Lactams, Macrocyclic/blood , Lactams, Macrocyclic/pharmacokinetics , Organophosphorus Compounds/blood , Organophosphorus Compounds/pharmacokinetics , Piperidines/blood , Piperidines/pharmacokinetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics , Tandem Mass Spectrometry/methods , Aminopyridines , Animals , Drug Stability , Humans , Lactams , Male , Mice , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles , Tissue Distribution
13.
Article in English | MEDLINE | ID: mdl-29866861

ABSTRACT

Hsp90 inhibitors, well studied in the laboratory and clinic for antitumor indications, have promising activity against protozoan pathogens, including Trypanosoma brucei which causes African sleeping sickness, and the malaria parasite, Plasmodium falciparum To progress these experimental drugs toward clinical use, we adapted an in vitro dynamic hollow-fiber system and deployed artificial pharmacokinetics to discover the driver of their activity: either concentration or time. The activities of compounds from three major classes of Hsp90 inhibitors in development were evaluated against trypanosomes. In all circumstances, the activities of the tested Hsp90 inhibitors were concentration driven. By optimally deploying the drug to match its kinetic driver, the efficacy of a given dose was improved up to 5-fold, and maximal efficacy was achieved with a significantly lower drug exposure. The superiority of concentration-driven regimens was evident in vitro over several logs of drug exposure and was predictive of efficacy in a mouse model of African trypanosomiasis. In studies with P. falciparum, antimalarial activity was similarly concentration driven. This experimental strategy offers an expedient and versatile translational tool to assess the impact of pharmacokinetics on antiprotozoal activity. Knowing kinetic governance early in drug development provides an additional metric for judging lead compounds and allows the incisive design of animal efficacy studies.


Subject(s)
Antiprotozoal Agents/pharmacokinetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antiprotozoal Agents/blood , Antiprotozoal Agents/pharmacology , Area Under Curve , Benzodioxoles/blood , Benzodioxoles/pharmacokinetics , Benzodioxoles/pharmacology , Benzoquinones/blood , Benzoquinones/pharmacokinetics , Benzoquinones/pharmacology , Biological Assay , Disease Models, Animal , Drug Repositioning , Female , Gene Expression , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Imidazoles/blood , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Isoxazoles/blood , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Lactams, Macrocyclic/blood , Lactams, Macrocyclic/pharmacokinetics , Lactams, Macrocyclic/pharmacology , Malaria, Falciparum/parasitology , Mice , Models, Biological , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Resorcinols/blood , Resorcinols/pharmacokinetics , Resorcinols/pharmacology , Trypanosoma brucei brucei/growth & development , Trypanosomiasis, African/parasitology
14.
Anticancer Drugs ; 29(8): 774-785, 2018 09.
Article in English | MEDLINE | ID: mdl-29878901

ABSTRACT

Triple-negative breast cancer (TNBC) is typically aggressive, difficult to treat, and commonly metastasizes to the visceral organs and soft tissues, including the lungs and the brain. Taxanes represent the most effective and widely used therapeutic class in metastatic TNBC but possess limiting adverse effects that often result in a delay, reduction, or cessation of their use. DZ-2384 is a candidate microtubule-targeting agent with a distinct mechanism of action and strong activity in several preclinical cancer models, with reduced toxicities. DZ-2384 is highly effective in patient-derived taxane-sensitive and taxane-resistant xenograft models of TNBC at lower doses and over a wider range relative to paclitaxel. When comparing compound exposure at minimum effective doses relative to safe exposure levels, the therapeutic window for DZ-2384 is 14-32 compared with 2.0 and less than 2.8 for paclitaxel and docetaxel, respectively. DZ-2384 is effective at reducing brain metastatic lesions when used at maximum tolerated doses and is equivalent to paclitaxel. Drug distribution experiments indicate that DZ-2384 is taken up more efficiently by tumor tissue but at equivalent levels in the brain compared with paclitaxel. Selective DZ-2384 uptake by tumor tissue may in part account for its wider therapeutic window compared with taxanes. In view of the current clinical efforts to combine chemotherapy with immune checkpoint inhibitors, we demonstrate that DZ-2384 acts synergistically with anti-CTLA-4 immunotherapy in a syngeneic murine model. These results demonstrate that DZ-2384 has a superior pharmacologic profile over currently used taxanes and is a promising therapeutic agent for the treatment of metastatic TNBC.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Oxazoles/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/administration & dosage , Brain/metabolism , CTLA-4 Antigen/immunology , Cell Line, Tumor , Drug Synergism , Female , Humans , Lactams, Macrocyclic/administration & dosage , Lactams, Macrocyclic/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Oxazoles/administration & dosage , Oxazoles/pharmacokinetics , Random Allocation , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
15.
J Med Chem ; 61(8): 3350-3369, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29590750

ABSTRACT

Rhodesain (RD) is a parasitic, human cathepsin L (hCatL) like cysteine protease produced by Trypanosoma brucei ( T. b.) species and a potential drug target for the treatment of human African trypanosomiasis (HAT). A library of hCatL inhibitors was screened, and macrocyclic lactams were identified as potent RD inhibitors ( Ki < 10 nM), preventing the cell-growth of Trypanosoma brucei rhodesiense (IC50 < 400 nM). SARs addressing the S2 and S3 pockets of RD were established. Three cocrystal structures with RD revealed a noncovalent binding mode of this ligand class due to oxidation of the catalytic Cys25 to a sulfenic acid (Cys-SOH) during crystallization. The P-glycoprotein efflux ratio was measured and the in vivo brain penetration in rats determined. When tested in vivo in acute HAT model, the compounds permitted up to 16.25 (vs 13.0 for untreated controls) mean days of survival.


Subject(s)
Cathepsin L/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Lactams, Macrocyclic/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei rhodesiense/drug effects , Animals , Binding Sites , Blood-Brain Barrier/metabolism , Cell Line , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacokinetics , Drug Repositioning , Humans , Lactams, Macrocyclic/chemical synthesis , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacokinetics , Ligands , Male , Mice, Inbred C57BL , Molecular Structure , Rats , Structure-Activity Relationship , Swine , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacokinetics
16.
Nat Commun ; 8: 15761, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28594000

ABSTRACT

Lorlatinib (PF-06463922) is a next-generation small-molecule inhibitor of the orphan receptor tyrosine kinase c-ros oncogene 1 (ROS1), which has a kinase domain that is physiologically related to anaplastic lymphoma kinase (ALK), and is undergoing Phase I/II clinical trial investigations for non-small cell lung cancers. An early goal is to measure the concentrations of this drug in brain tumour lesions of lung cancer patients, as penetration of the blood-brain barrier is important for optimal therapeutic outcomes. Here we prepare both 11C- and 18F-isotopologues of lorlatinib to determine the biodistribution and whole-body dosimetry assessments by positron emission tomography (PET). Non-traditional radiolabelling strategies are employed to enable an automated multistep 11C-labelling process and an iodonium ylide-based radiofluorination. Carbon-11-labelled lorlatinib is routinely prepared with good radiochemical yields and shows reasonable tumour uptake in rodents. PET imaging in non-human primates confirms that this radiotracer has high brain permeability.


Subject(s)
Carbon Radioisotopes/pharmacokinetics , Fluorine Radioisotopes/pharmacology , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacology , Positron-Emission Tomography/methods , Aminopyridines , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Animals , Carbon Radioisotopes/chemistry , Chemistry Techniques, Synthetic , Contrast Media/chemical synthesis , Contrast Media/pharmacokinetics , Fluorine Radioisotopes/chemistry , Humans , Isotope Labeling/methods , Lactams , Lactams, Macrocyclic/pharmacokinetics , Macaca mulatta , Male , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles , Tissue Distribution , Xenograft Model Antitumor Assays
17.
Colloids Surf B Biointerfaces ; 155: 83-92, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28410515

ABSTRACT

When exposed to cancer cells, cytotoxic drugs such as doxorubicin (DOX) can lead to the induction of heat shock protein 90 (Hsp90), a molecular chaperone associated with a number of cancer-related client proteins, and result in cell survival. Co-administration of DOX with tanespimycin (TNP), an Hsp90 inhibitor, can sensitize the cancer cells to the cytotoxic effects of DOX. The effect of such a combination has been found to depend on the schedule of administration. Sequential administration of DOX and TNP has been linked to highly synergistic combination effects. Therefore, we aimed to develop folate-receptor targeted hybrid lipid-core nanocapsules comprising a hybrid lipid core lodging TNP and a polymeric corona lodging DOX (F-DTN). These nanocarriers were capable of delivering DOX and TNP sequentially, which was well demonstrated by an in vitro release study. The in vitro release profiles displayed pH-dependent and sustained release features. F-DTN exhibited excellent morphological characteristics with highly monodispersed particles. In vitro tests with F-DTN in MCF-7 cell line demonstrated exceptional cytotoxicity, with high cellular uptake and apoptosis. These findings were appreciably more assertive than tests with free individual drugs (DOX, TNP), free drug combination (DOX/TNP), or non-folate receptor-targeted hybrid lipid-core nanocapsules (DTN). In vivo pharmacokinetic study revealed noticeable enhancement of bioavailability and plasma circulation time of the drugs when encapsulated in the carrier system. Therefore, hybrid lipid-core nanocapsules have the potential to be utilized for application in folate receptor-targeted combination chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Folate Receptor 1/metabolism , Lactams, Macrocyclic/pharmacology , Nanocapsules/chemistry , Neoplasm Proteins/metabolism , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Benzoquinones/chemistry , Benzoquinones/pharmacokinetics , Biomarkers/metabolism , Dimethyl Sulfoxide/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Fatty Acids, Monounsaturated/chemistry , Folate Receptor 1/genetics , Folic Acid/chemistry , Folic Acid/metabolism , Gene Expression , Humans , Injections, Intravenous , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacokinetics , MCF-7 Cells , Male , Micelles , Nanocapsules/administration & dosage , Nanocapsules/ultrastructure , Neoplasm Proteins/genetics , Phosphatidylethanolamines/chemistry , Quaternary Ammonium Compounds/chemistry , Rats , Rats, Sprague-Dawley
19.
Drug Dev Ind Pharm ; 42(1): 91-98, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25853294

ABSTRACT

In this study, the PEGylated nanostructured lipid carriers (PEG-NLC) were constructed for the intravenous delivery of 17-allylamino-17-demethoxygeldanamycin (17AAG). 17AAG-PEG-NLC was successfully prepared by the method of emulsion evaporation at a high temperature and solidification at a low temperature using a mixture of glycerol monostearate and PEG2000-stearate as solid lipids, and medium-chain triglyceride as the liquid lipid. The results revealed that the morphology of the NLC was spheroidal. The particle size, zeta potential and entrapment efficiency for 17AAG-PEG-NLC were observed as 189.4 nm, -20.2 mV and 83.42%, respectively. X-ray diffraction analysis revealed that 17AAG existed as amorphous structures in the nanoparticles. In the in vitro release study, the 17AAG from 17AAG-PEG-NLC exhibited a biphasic release pattern with burst release initially and sustained release afterwards. In addition, 17AAG-PEG-NLC showed a significantly higher in vitro antitumor efficacy and longer retention time in vivo than 17AAG solution. These results indicated that 17AAG-PEG-NLC may offer a promising alternative to the current 17AAG formulations for the treatment of solid tumors.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Benzoquinones/administration & dosage , Drug Carriers/chemistry , Drug Compounding/methods , Drug Liberation , Lactams, Macrocyclic/administration & dosage , Administration, Intravenous , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Benzoquinones/pharmacokinetics , Calorimetry, Differential Scanning , Drug Evaluation, Preclinical , Emulsions , Glycerol/chemistry , Humans , Lactams, Macrocyclic/pharmacokinetics , Lipids/chemistry , MCF-7 Cells , Nanoparticles/chemistry , Particle Size , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Solubility , Stearates/chemistry , Triglycerides/chemistry , X-Ray Diffraction
20.
Bioorg Med Chem Lett ; 25(17): 3744-7, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26159484

ABSTRACT

Biodegradable vehicles that degrade specifically at tumor sites are highly desirable since they can cause selective exposure of highly toxic drugs at tumor sites whereas keep the conjugates stable during blood circulation. Here, we evaluate the utility of a dendritic hexadecapeptide comprised of four arms, each having a tetrapeptide sequence recognized by an enzyme cathepsin B as a carrier system for heat shock protein 90 (HSP90) inhibitor geldanamycin (GDM). We report the synthesis of a carrier having GDM conjugated to the terminal end of each arm (>55% wt/wt drug). We further report the stability of the GDM containing peptidic dendrimer in various buffers and in the presence of serum along with its ability to release free drug in the presence of cathepsin B, the enzyme overexpressed in a variety of tumors. Using androgen-independent prostate cancer cell line (DU-145) we further demonstrate that the geldanamycin containing peptidic dendrimer has antiproliferative property similar to the free drug derivative.


Subject(s)
Benzoquinones/administration & dosage , Cathepsin B/metabolism , Dendrimers/administration & dosage , Drug Carriers/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/administration & dosage , Peptides/administration & dosage , Peptides/chemistry , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Benzoquinones/pharmacokinetics , Cathepsin B/administration & dosage , Cathepsin B/chemistry , Cell Line, Tumor/drug effects , Dendrimers/chemistry , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Stability , Humans , Inhibitory Concentration 50 , Lactams, Macrocyclic/pharmacokinetics , Male , Peptides/pharmacokinetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...