Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
J Parkinsons Dis ; 14(2): 325-334, 2024.
Article En | MEDLINE | ID: mdl-38251063

Background: Pre-clinical studies suggest that c-Abl activation may play an important role in the etiology of Parkinson's disease, making c-Abl an important target to evaluate for potential disease-modification. Objective: To assess safety, tolerability, and pharmacokinetics of the c-Abl inhibitor risvodetinib (IkT-148009) in healthy subjects and participants with Parkinson's disease. Methods: Part 1 (single ascending dose (SAD)) and Part 2 (7-day multiple ascending dose (MAD)) studies were in healthy volunteers. Participants were randomized 3 : 1 across 9 SAD doses and 3 MAD doses of risvodetinib (IkT-148009) or placebo. Part 3 was a MAD study conducted at two doses in 14 participants with mild-to-moderate PD (MAD-PD). Primary outcome measures were safety, tolerability and pharmacokinetics. Exploratory outcomes in PD participants included clinical measures of PD state, GI function, and cerebrospinal fluid (CSF) concentration. Results: 108 patients were treated with no dropouts. The SAD tested doses ranging from 12.5 to 325 mg, while the MAD tested 25 to 200 mg and MAD-PD tested 50 to 100 mg in Parkinson's participants. All active doses had a favorable safety profile with no clinically meaningful adverse events. Single dose pharmacokinetics were approximately linear between 12.5 mg and 200 mg for both Cmax and AUC0 - inf without distinction between healthy volunteers and participants with PD. Exposures at each dose were high relative to other drugs in the same kinase inhibitor class. Conclusions: Risvodetinib (IkT-148009) was well tolerated, had a favorable safety and pharmacology profile over 7-day dosing, did not induce serious adverse events and did not appear to induce deleterious side-effects in participants administered anti-PD medications.


Parkinson Disease , Aged , Humans , Area Under Curve , Healthy Volunteers , Parkinson Disease/drug therapy
2.
Epilepsia ; 63(7): 1714-1723, 2022 07.
Article En | MEDLINE | ID: mdl-35377464

OBJECTIVE: Current diazepam nasal spray labeling requires waiting 4 h before administering a second dose. The objective of the current analyses was to examine safety and pharmacokinetic profiles of second doses of diazepam nasal spray given 0-4 h after the first dose. METHODS: Two datasets were analyzed. The first, a long-term, repeat-dose safety study of diazepam nasal spray, compared rates of treatment-emergent adverse events (TEAEs), serious TEAEs, and treatment-related TEAEs for patients receiving ≥1 second dose ≤4 h versus all second doses >4 h after the first. The second was a population pharmacokinetic analysis using data from three phase 1 studies to model drug exposure when a second dose of diazepam nasal spray was administered across multiple time points (1 min-4 h) following the first dose. RESULTS: In the repeat-dose safety study, a second dose of diazepam nasal spray was administered ≤24 h after the first to treat 485 seizure clusters in 79 patients. Rates of TEAEs were similar between patients receiving ≥1 second dose in ≤4 h (89.5%, n = 38) compared with >4-24 h only (80.5%, n = 41). The most common treatment-related TEAEs were associated with nasal discomfort, which was mild or moderate and transient. There were no reports of respiratory or cardiac depression. The pharmacokinetic simulations of second doses predicted comparable elevations of plasma diazepam concentrations with administrations across a range of intervals after the first dose (1 min-4 h). SIGNIFICANCE: These data indicate that the safety and pharmacokinetic profiles of a second dose of diazepam nasal spray administered within 4 h of the first dose are consistent with those associated with current labeling. This is potentially important for patients with seizure clusters who have a recurrent seizure within 4 h of first treatment and might benefit from immediate retreatment to reduce the risk of progression to status epilepticus.


Diazepam , Epilepsy, Generalized , Administration, Intranasal , Anticonvulsants/therapeutic use , Diazepam/therapeutic use , Epilepsy, Generalized/drug therapy , Humans , Nasal Sprays , Seizures/chemically induced , Seizures/drug therapy
3.
Cancer ; 127(8): 1246-1259, 2021 04 15.
Article En | MEDLINE | ID: mdl-33270904

BACKGROUND: CXCR4 mediates the retention and survival of acute myelogenous leukemia blasts in bone marrow and contributes to their resistance to chemotherapy. The authors evaluated a combination of the high-affinity CXCR4 antagonist BL-8040 with high-dose cytarabine (HiDAC) chemotherapy in a phase 2a study of patients with relapsed and refractory AML. METHODS: Forty-two patients received treatment with BL-8040 monotherapy for 2 days followed by a combination of BL-8040 with HiDAC for 5 days. Six escalating BL-8040 dose levels were investigated (0.5, 0.75, 1.0, 1.25, 1.5, and 2.0 mg/kg), and 1.5 mg/kg was selected as the dose for the expansion phase (n = 23). RESULTS: BL-8040 in combination with HiDAC was safe and well tolerated at all dose levels. Clinical response was observed with BL-8040 doses ≥1.0 mg/kg. The composite response rate (complete remissions plus complete remissions with incomplete hematologic recovery of platelets or neutrophils) was 29% (12 of 42) in all patients and 39% (9 of 23) in the 1.5-mg/kg phase. The median overall survival was 8.4 months for all patients, 10.8 months in the 1.5-mg/kg phase, and 21.8 months for responding patients in the 1.5-mg/kg cohort. Two days of BL-8040 monotherapy triggered the mobilization of blasts into peripheral blood, with significantly higher mean fold-changes in responders versus nonresponders. This was accompanied by a decrease in bone marrow blasts. CONCLUSIONS: The current results demonstrate the efficacy of CXCR4 targeting with BL-8040 and support continued clinical development in acute myelogenous leukemia.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cytarabine/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Peptides/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Marrow Cells/drug effects , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Hematopoietic Stem Cell Mobilization , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Peptides/adverse effects , Peptides/pharmacokinetics , Recurrence , Remission Induction
4.
Clin Pharmacol Drug Dev ; 8(1): 49-59, 2019 01.
Article En | MEDLINE | ID: mdl-29786964

In this randomized double-blind study, 4 groups of healthy subjects (50 per arm) participated to evaluate the effect of laquinimod, an oral treatment in development for multiple sclerosis and Huntington disease, on the QTc interval. Subjects received a dose of either 0.6 or 1.2 mg/day laquinimod for 14 days, placebo for 14 days, or 13 days of placebo followed by a dose of 400 mg moxifloxacin on day 14. Continuous 12-lead electrocardiograms were recorded on day -1 (baseline) and days 14 to 17,  and quadruplicate electrocardiograms were extracted at predefined time points. The primary measure was time-matched change from baseline in individual QTc (QTcI), and an analysis of variance was conducted on the placebo-corrected change from baseline data (ddQTcI). Pharmacokinetic-pharmacodynamic and safety assessments were included. Results showed that the upper limits of the 2-sided 90%CI for ddQTcI for both laquinimod doses were below 10 millisconds at all time points, whereas lower limits for moxifloxacin were above 5 milliseconds. No notable changes in ECG parameters were observed. Pharmacokinetic/pharmacodynamic analysis showed no positive correlation between laquinimod plasma levels and QTcI. In conclusion, laquinimod was not found to affect cardiac repolarization or to cause prolongation of QTcI at doses of 0.6 and 1.2 mg/day.


Electrocardiography/drug effects , Heart/drug effects , Immunologic Factors/pharmacology , Quinolones/pharmacology , Adolescent , Adult , Double-Blind Method , Female , Heart/physiology , Heart Rate/drug effects , Humans , Huntington Disease/drug therapy , Immunologic Factors/blood , Immunologic Factors/pharmacokinetics , Long QT Syndrome , Male , Middle Aged , Multiple Sclerosis/drug therapy , Quinolones/blood , Quinolones/pharmacokinetics , Young Adult
5.
J Neuroinflammation ; 14(1): 172, 2017 Aug 31.
Article En | MEDLINE | ID: mdl-28859672

BACKGROUND: Laquinimod is an oral immunomodulator in clinical development to treat relapsing-remitting multiple sclerosis (RRMS). Laquinimod is in clinical development for the treatment of multiple sclerosis and Huntington Disease (HD). The objective of this study is to assess the safety, tolerability, pharmacokinetics (PK) and cytoimmunologic effects following escalating doses of laquinimod in patients with RRMS. METHODS: One hundred twelve patients were randomly assigned to laquinimod/placebo in a series of separate dose-escalating cohorts starting from a daily oral dose of 0.9 mg/1.2 mg escalating to 2.7 mg, in 0.3 mg increments. RESULTS: Twenty-eight patients received placebo and 84 received laquinimod ranging from 0.9 to 2.7 mg. No deaths occurred. One serious adverse event (SAE) of perichondritis was reported, which was unrelated to laquinimod (0.9 mg). There was no increased incidence of adverse events (AEs) with escalating doses. Laquinimod-treated patients showed more abnormal laboratory levels in liver enzymes, P-amylase, C-reactive protein (CRP), and fibrinogen, but most shifts were clinically non-significant. The exposure of laquinimod was dose proportional and linear in the tested dose range. An immunological substudy showed significant dose-dependent decreases in 6-sulpho LacNAc + dendritic cell (slanDC) frequency following laquinimod compared to placebo. CONCLUSION: Laquinimod doses up to 2.7 mg were safely administered to patients with RRMS. An in vivo effect of laquinimod on the innate immune system was demonstrated. TRIAL REGISTRATION: EudraCT Number: 2009-011234-99 . Registered 23 June 2009.


Immunity, Innate/immunology , Immunologic Factors/administration & dosage , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Quinolones/administration & dosage , Administration, Oral , Adolescent , Adult , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Humans , Immunity, Innate/drug effects , Male , Middle Aged , Young Adult
6.
J Pain Res ; 9: 587-98, 2016.
Article En | MEDLINE | ID: mdl-27621666

A randomized, placebo-controlled crossover trial utilizing vaporized cannabis containing placebo and 6.7% and 2.9% delta-9-tetrahydrocannabinol (THC) was performed in 42 subjects with central neuropathic pain related to spinal cord injury and disease. Subjects received two administrations of the study medication in a 4-hour interval. Blood samples for pharmacokinetic evaluation were collected, and pain assessment tests were performed immediately after the second administration and 3 hours later. Pharmacokinetic data, although limited, were consistent with literature reports, namely dose-dependent increase in systemic exposure followed by rapid disappearance of THC. Dose-dependent improvement in pain score was evident across all pain scale elements. Using mixed model regression, an evaluation of the relationship between plasma concentrations of selected cannabinoids and percent change in items from the Neuropathic Pain Scale was conducted. Changes in the concentration of THC and its nonpsychotropic metabolite, 11-nor-9-carboxy-THC, were related to percent change from baseline of several descriptors (eg, itching, burning, and deep pain). However, given the large number of multiple comparisons, false-discovery-rate-adjusted P-values were not significant. Plans for future work are outlined to explore the relationship of plasma concentrations with the analgesic response to different cannabinoids. Such an appraisal of descriptors might contribute to the identification of distinct pathophysiologic mechanisms and, ultimately, the development of mechanism-based treatment approaches for neuropathic pain, a condition that remains difficult to treat.

7.
AAPS J ; 16(6): 1372-9, 2014 Nov.
Article En | MEDLINE | ID: mdl-25155824

The empirical scaling from adult to pediatric using allometric size adjustments based on body weight continued to be the mainstream method for pediatric dose selection. Due to the flexibility of a polynomial function to conform to the data trend, an empirical function for simulating age-matched weight and body mass index by gender in the pediatric population is developed by using a polynomial function and a constant coefficient to describe the interindividual variability in weight. A polynomial of up to fifth order sufficiently described the pediatric data from the Center for Disease Control (CDC) and the World Health Organization (WHO). The coefficients of variation to describe the variability were within 17%. The percentages of the CDC simulated weights for pediatrics between 0 and 5 years that fell outside the WHO 90% and 95% confidence boundaries were well within the expected percentage values, indicating that the CDC dataset can be used to substitute for the WHO dataset for the purpose of pediatric drug development. To illustrate the utility of this empirical function, the CDC-based age-matched weights were simulated and were used in the prediction of the concentration-time profiles of tenofovir in children based on a population pharmacokinetic model whose parameters were allometrically scaled. We have shown that the resulting 95% prediction interval of tenofovir in newborn to 5 years of age was almost identical whether the weights were simulated based on WHO or CDC dataset. The approach is simple and is broadly applicable in adjusting for pediatric dosages using allometry.


Aging/physiology , Body Mass Index , Body Weight/physiology , Models, Biological , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Adolescent , Child , Child, Preschool , Computer Simulation , Female , Humans , Infant , Infant, Newborn , Male , Models, Statistical , Organophosphonates/pharmacokinetics , Predictive Value of Tests , Software , Tenofovir
8.
J Clin Pharmacol ; 54(1): 53-60, 2014 Jan.
Article En | MEDLINE | ID: mdl-24150835

Dalfampridine extended release tablets (D-ER; prolonged-release fampridine in Europe) are available to improve walking in patients with multiple sclerosis (MS). D-ER is mainly renally eliminated; the approved 10-mg twice daily dose is contraindicated in the United States in patients with moderate or severe renal impairment. This study evaluated single-dose and steady-state pharmacokinetics of a 7.5-mg dose of D-ER in healthy subjects (n = 13) and subjects with mild (n = 17) and moderate (n = 12) renal impairment. D-ER plasma concentrations were consistently higher in subjects with renal impairment relative to healthy individuals with a significant (P < .0001) inverse linear relationship between creatinine clearance and drug exposure. Steady-state AUC0-12 among healthy subjects, 167.0 ± 55.3 ng h/mL, increased 74% and 151% with mild and moderate renal impairment, respectively. The overall incidence of adverse events was 61.5%, 47.1%, and 33.3% in healthy subjects, and subjects with mild and moderate renal impairment, respectively, and for treatment-related adverse events the rates were 0%, 17.6%, and 8.3%, respectively. The most common adverse events were headache, dizziness, and arthralgia. The pharmacokinetics of D-ER 7.5-mg twice daily in subjects with mild renal impairment was comparable to 10-mg twice daily in patients with MS who had normal renal function. Exposure was significantly higher in moderate renal impairment.


4-Aminopyridine/pharmacokinetics , Kidney Diseases/metabolism , Potassium Channel Blockers/pharmacokinetics , 4-Aminopyridine/adverse effects , Adolescent , Adult , Age Factors , Aged , Analysis of Variance , Area Under Curve , Body Weight , Chromatography, High Pressure Liquid , Delayed-Action Preparations , Ethnicity , Female , Half-Life , Humans , Male , Middle Aged , Potassium Channel Blockers/adverse effects , Spectrometry, Mass, Electrospray Ionization , Tablets , Tandem Mass Spectrometry , Young Adult
9.
Antimicrob Agents Chemother ; 56(4): 2067-73, 2012 Apr.
Article En | MEDLINE | ID: mdl-22252798

A population pharmacokinetic model of telavancin, a lipoglycopeptide antibiotic, was developed and used to identify sources of interindividual variability. Data were obtained from healthy subjects (seven phase 1 studies), patients with complicated skin and skin structure infections (cSSSI; two phase 2 and two phase 3 studies), and patients with hospital-acquired pneumonia (HAP; two phase 3 studies). A two-compartment open model with zero-order input best fit the telavancin data from healthy individuals and patients with cSSSI or HAP. Telavancin clearance was highly correlated with renal function and, to a lesser extent, with body weight. Other covariates were related to at least one parameter in cSSSI (gender, bacterial eradication, and surgery) or HAP (age of ≥ 75 years) but did not markedly affect exposure. These analyses support current dosing recommendations for telavancin based on patient weight and renal function.


Aminoglycosides/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Bacterial Infections/metabolism , Adult , Aged , Aged, 80 and over , Algorithms , Aminoglycosides/blood , Anti-Bacterial Agents/blood , Area Under Curve , Bayes Theorem , Body Weight/physiology , Calibration , Cross Infection/metabolism , Female , Humans , Lipoglycopeptides , Male , Middle Aged , Models, Statistical , Population , Sex Characteristics , Young Adult
10.
J Clin Pharmacol ; 51(9): 1310-8, 2011 Sep.
Article En | MEDLINE | ID: mdl-21209239

Fostamatinib (R788) is being investigated as an add-on therapy for the treatment of rheumatoid arthritis (RA) in patients with inadequate response to methotrexate (MTX). This study evaluated the potential pharmacokinetic interaction between R788 and MTX. Sixteen RA subjects on a stable weekly MTX regimen were enrolled and received MTX on days 1 and 8. Twelve subjects received 100 mg of R788 orally, and 4 subjects received a matching placebo twice daily from days 4 to 8 and once daily on days 3 and 9. Blood samples were collected on days 1 and 8 for MTX and 7-hydroxymethotrexate (7-OH-MTX), and days 3 and 9 for R788 and its active metabolite, R406. MTX and 7-OH-MTX pharmacokinetic parameters were similar on days 1 and 8. In the R788 group, the mean day 8 to day 1 ratios (90% confidence intervals) of maximum concentration and area under the plasma concentration-time curve estimates were 1.01 (0.85-1.20) and 1.12 (0.90-1.40) for MTX and 1.06 (0.82-1.35) and 1.06 (0.83-1.36) for 7-OH-MTX, respectively. Urinary excretion of MTX and 7-OH-MTX was also similar with or without R788, averaging 58% to 69% and 4% to 5% of the MTX dose, respectively. The data suggest that there is no clinically significant pharmacokinetic interaction of R788 and MTX in RA patients.


Arthritis, Rheumatoid/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Methotrexate/pharmacokinetics , Methotrexate/therapeutic use , Oxazines/pharmacokinetics , Oxazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Aminopyridines , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Interactions/physiology , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Morpholines , Protein-Tyrosine Kinases/metabolism , Pyrimidines , Syk Kinase
11.
J Pharm Sci ; 94(9): 1949-56, 2005 Sep.
Article En | MEDLINE | ID: mdl-16052544

Defining a quantitative and reliable relationship between in vitro drug release and in vivo absorption is highly desired for rational development, optimization, and evaluation of controlled-release dosage forms and manufacturing process. During the development of a once-daily extended-release (ER) tablet of divalproex sodium, a predictive in vitro drug release method was designed and statistically evaluated using three formulations with varying release rates. In order to establish an internally and externally validated Level A IVIVC, a total of five different ER formulations of divalproex sodium were used to evaluate a linear IVIVC model based on the in vitro test method. For internal validation, a single-dose four-way crossover study (N = 16) was performed using fast-, medium-, and slow-releasing ER formulations and a 12-h IV infusion of valproic acid as reference. To validate the IVIVC externally, a second three-way crossover study (N = 36) was performed using slightly-fast-, medium-, and slightly-slow-releasing ER formulations. The in vivo absorption-time profile was inferred by deconvolution of the observed plasma concentration-time profiles against the unit disposition function (UDF). A linear IVIVC model was established in which the in vivo absorption was expressed as a function of in vitro drug release. Plasma profiles of ER formulations were estimated via convolution of in vitro release profiles with the UDF. Successful internal and external validations of the model were demonstrated by individual and average absolute percent prediction errors of

Anticonvulsants/pharmacokinetics , Valproic Acid/pharmacokinetics , Adolescent , Adult , Anticonvulsants/administration & dosage , Anticonvulsants/blood , Area Under Curve , Cross-Over Studies , Delayed-Action Preparations , Female , Half-Life , Humans , In Vitro Techniques , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Reproducibility of Results , Solubility , Valproic Acid/administration & dosage , Valproic Acid/blood
12.
Clin Cancer Res ; 11(10): 3633-41, 2005 May 15.
Article En | MEDLINE | ID: mdl-15897558

PURPOSE: To evaluate the therapeutic and biological effects of CHIR-258, an orally bioavailable, potent inhibitor of class III-V receptor tyrosine kinases, in colon cancer models. EXPERIMENTAL DESIGN: The pharmacologic activity of CHIR-258 was characterized by monitoring target modulation as well as by evaluating the antitumor and antiangiogenic effects in human colon xenograft models. RESULTS: CHIR-258 inhibits vascular endothelial growth factor receptor 1/2, fibroblast growth factor receptor 1/3, and platelet-derived growth factor receptor beta (PDGFRbeta) and shows both antitumor and antiangiogenic activities in vivo. Treatment of KM12L4a human colon cancer cells with CHIR-258 resulted in a dose-dependent inhibition of vascular endothelial growth factor receptor 1 and PDGFRbeta phosphorylation and reduction of phosphorylated extracellular signal-regulated kinase (ERK) levels, indicating modulation of target receptors and downstream signaling. In vivo administration of CHIR-258 resulted in significant tumor growth inhibition and tumor regressions, including large, established tumors (500-1,000 mm(3)). Immunohistochemical analysis showed a reduction of phosphorylated PDGFRbeta and phosphorylated ERK in tumor cells after oral dosing with CHIR-258 compared with control tumors. These changes were accompanied by decreased tumor cell proliferation rate and reduced intratumoral microvessel density. CHIR-258 inhibited the phosphorylation of PDGFRbeta and ERK phosphorylation in tumors within 2 hours following dosing and the inhibitory activity was sustained for >24 hours. Significant antitumor activity was observed with intermittent dosing schedules, indicating a sustained biological activity. CONCLUSION: These studies provide evidence that biological activity of CHIR-258 in tumors correlates with efficacy and aids in the identification of potential biomarkers of this multitargeted receptor tyrosine kinase inhibitor. CHIR-258 exhibits properties that make it a promising candidate for clinical development in a variety of solid and hematologic malignancies.


Benzimidazoles/pharmacology , Biomarkers, Tumor/analysis , Colonic Neoplasms/drug therapy , Growth Substances/biosynthesis , Neovascularization, Pathologic , Quinolones/pharmacology , Animals , Benzimidazoles/pharmacokinetics , Cell Proliferation , Colonic Neoplasms/pathology , Colonic Neoplasms/veterinary , Female , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins c-sis/metabolism , Quinolones/pharmacokinetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/metabolism
...