Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
ACS Chem Neurosci ; 13(23): 3342-3351, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36417797

ABSTRACT

Demyelination, the loss of the insulating sheath of neurons, causes failed or slowed neuronal conduction and contributes to the neurological symptoms in multiple sclerosis, traumatic brain and spinal cord injuries, stroke, and dementia. In demyelinated neurons, the axonal potassium channels Kv1.1 and Kv1.2, generally under the myelin sheath, become exposed and upregulated. Therefore, imaging these channels using positron emission tomography can provide valuable information for disease diagnosis and monitoring. Here, we describe a novel tracer for Kv1 channels, [11C]3-methyl-4-aminopyridine ([11C]3Me4AP). [11C]3Me4AP was efficiently synthesized via Pd(0)-Cu(I) comediated Stille cross-coupling of a stannyl precursor containing a free amino group. Evaluation of its imaging properties in rats and nonhuman primates showed that [11C]3Me4AP has a moderate brain permeability and slow kinetics. Additional evaluation in monkeys showed that the tracer is metabolically stable and that a one-tissue compartment model can accurately model the regional brain time-activity curves. Compared to the related tracers [18F]3-fluoro-4-aminopyridine ([18F]3F4AP) and [11C]3-methoxy-4-aminopyridine ([11C]3MeO4AP), [11C]3Me4AP shows lower initial brain uptake, which indicates reduced permeability to the blood-brain barrier and slower kinetics, suggesting higher binding affinity consistent with in vitro studies. While the slow kinetics and strong binding affinity resulted in a tracer with less favorable properties for imaging the brain than its predecessors, these properties may make 3Me4AP useful as a therapeutic.


Subject(s)
4-Aminopyridine , Brain , Demyelinating Diseases , Kv1.1 Potassium Channel , Kv1.2 Potassium Channel , Molecular Imaging , Radioactive Tracers , Animals , Rats , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemical synthesis , 4-Aminopyridine/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Permeability , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/metabolism , Demyelinating Diseases/diagnostic imaging , Molecular Imaging/methods , Primates , Blood-Brain Barrier/metabolism
2.
Expert Opin Pharmacother ; 20(11): 1309-1320, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31237780

ABSTRACT

Introduction: Multiple sclerosis (MS) is a demyelinating disease, causing axonal damage and disability. Dalfampridine (DAL) is an extended-release formulation of 4-aminopyridine (4AP) and broad-spectrum voltage-dependent potassium channel blocker that is reported to improve motor, visual and cognitive functions. Furthermore, it is presently the only approved drug for walking impairment in MS. Areas covered: Herein, the authors evaluate DAL as a relapsing-remitting MS treatment, reporting and commenting on all aspects of the drug including its chemistry, safety, pharmacokinetics, and cost-effectiveness. A bibliographic search was performed on PubMed using the terms 'dalfampridine OR fampridine OR 4-aminopyridine'. Expert opinion: Evidence from post-marketing studies suggests that DAL, consistent with the effects of 4AP, may not only improve walking speed, but also arm function, fatigue, mood and cognition through restored nerve conduction in central nervous system demyelinated areas. Long-term safety data confirm that the approved dose of 10 mg twice daily is generally well tolerated. However, despite the reported efficacy, the extent of the benefits is limited in real life activities, although significant improvements have been demonstrated in the clinical setting. Patients often complain of side effects (such as cramps and painful paraesthesia) or lack of efficacy. Also, its considerably higher pricing in comparison to 4AP represents an important limitation.


Subject(s)
4-Aminopyridine/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Potassium Channel Blockers/therapeutic use , 4-Aminopyridine/adverse effects , 4-Aminopyridine/chemistry , 4-Aminopyridine/pharmacokinetics , Clinical Trials as Topic , Cognitive Dysfunction/complications , Cost-Benefit Analysis , Databases, Factual , Fatigue/complications , Half-Life , Humans , Multiple Sclerosis, Relapsing-Remitting/economics , Potassium Channel Blockers/adverse effects , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacokinetics , Seizures/etiology , Treatment Outcome
3.
CNS Neurosci Ther ; 25(6): 697-703, 2019 06.
Article in English | MEDLINE | ID: mdl-30756475

ABSTRACT

AIM: To examine whether the velocity of saccadic eye movements in internuclear ophthalmoparesis (INO) improves with fampridine treatment in patients with multiple sclerosis (MS). METHODS: Randomized, double-blind, placebo-controlled, cross-over trial with fampridine in patients with MS and INO. Horizontal saccades were recorded at baseline and at multiple time points post-dose. Main outcome measures were the change of peak velocity versional dysconjugacy index (PV-VDI) and first-pass amplitude VDI (FPA-VDI). Both parameters were compared between fampridine and placebo using a mixed model analysis of variance taking patients as their own control. Pharmacokinetics was determined by serial blood sampling. RESULTS: Thirteen patients had a bilateral and 10 had a unilateral INO. One patient had an INO of abduction (posterior INO of Lutz) and was excluded. Fampridine significantly reduced both PV-VDI (-17.4%, 95% CI: -22.4%, -12.1%; P < 0.0001) and FPA-VDI (-12.5%, 95% CI: -18.9%, -5.5%; P < 0.01). Pharmacokinetics demonstrated that testing coincided with the average tmax at 2.08 hours (SD 45 minutes). The main adverse event reported after administration of fampridine was dizziness (61%). CONCLUSION: Fampridine improves saccadic eye movements due to INO in MS. Treatment response to fampridine may gauge patient selection for inclusion to remyelination strategies in MS using saccadic eye movements as primary outcome measure.


Subject(s)
4-Aminopyridine/therapeutic use , Multiple Sclerosis/complications , Ophthalmoplegia/drug therapy , Potassium Channel Blockers/therapeutic use , 4-Aminopyridine/blood , 4-Aminopyridine/pharmacokinetics , Adult , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Ophthalmoplegia/blood , Ophthalmoplegia/etiology , Potassium Channel Blockers/blood , Potassium Channel Blockers/pharmacokinetics , Saccades/drug effects , Treatment Outcome
4.
Clin Pharmacol Drug Dev ; 8(3): 355-360, 2019 04.
Article in English | MEDLINE | ID: mdl-29749716

ABSTRACT

Dalfampridine is a medication that is approved by the US Food and Drug Administration to improve walking impairments in patients with multiple sclerosis (MS). The branded dalfampridine is enormously expensive; hence, the availability of generic dalfampridine will provide better access to the medication, especially for uninsured patients with MS. Bioequivalence studies are demanded by the regulatory authorities to allow the marketing of new generics of dalfampridine. The aim of this study was to assess the bioavailability of the generic (test) and branded (reference) formulations of 10 mg dalfampridine of extended-release tablets after oral administration to healthy adults under fed conditions. The current report methodology was based on a comparative, randomized, single-dose, 2-way crossover open-label study design. Twenty-seven subjects were given a single dose of the test dalfampridine tablet and completed the clinical study. The pharmacokinetic parameters Cmax and AUC0→t, Kel , AUC0→∞ , tmax , and t1/2el were estimated to prove bioequivalence. The confidence intervals for the log-transformed test/reference ratios for dalfampridine 100.96% (97.09%-104.97%) and 99.77% (95.81%-103.87%) for Cmax and AUC0→∞ , respectively, were within the allowed limit specified by the regulatory authorities (80%-125%). Hence, clinically, the test tablet can be prescribed as an alternative to the reference for the indication of improving walking impairments in patients with MS.


Subject(s)
4-Aminopyridine/pharmacokinetics , Drugs, Generic/pharmacokinetics , Mobility Limitation , Potassium Channel Blockers/pharmacokinetics , 4-Aminopyridine/blood , 4-Aminopyridine/chemistry , Administration, Oral , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Delayed-Action Preparations , Drug Compounding , Drugs, Generic/chemistry , Healthy Volunteers , Humans , Male , Middle Aged , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/blood , Potassium Channel Blockers/chemistry , Tablets , Therapeutic Equivalency , Young Adult
5.
Pharm Res ; 35(8): 159, 2018 Jun 18.
Article in English | MEDLINE | ID: mdl-29915999

ABSTRACT

PURPOSE: The renal clearance of fampridine (Fampyra®, or Ampyra®) significantly exceeds the glomerular filtration rate, suggesting active renal secretion is likely the major elimination pathway. The goal of this study was to identify the renal transporters that are involved in the renal active secretion, and elucidate the active renal secretion mechanism of fampridine. METHODS: The uptake of fampridine to HEK-293 cells overexpressing human OCT2, MATE1 or MATE2K was determined in the absence and presence of Cimetidine, the prototypical inhibitor of the transporters. The inhibition potential of fampridine on the renal transporters was evaluated by determining the uptake of TEA and Metformin, the probe substrates of the transporters of OCT2 and MATEs, respectively, in the absence or presence of fampridine. RESULTS: Significant time- and concentration-dependent uptake of fampridine by human OCT2 was observed. The Km and Vmax were determined as 51.0 ± 17.1 µM and 1107 ± 136 pmole/min/106 cells, respectively. Fampridine also inhibited OCT2 mediated uptake of Metformin with estimated IC50 of 66.8 µM. In contrast, there was not significant uptake of fampridine by human MATE1 or MATE2K, and fampridine did not inhibit MATE1 or MATE2K mediated uptake of TEA. CONCLUSION: The studies indicated fampridine is a substrate and inhibitor of OCT2, but not MATE1 or MATE2K. Results from the study suggested the active renal secretion of fampridine is mediated by human OCT2 but not MATE1 or MATE2K. To our knowledge, fampridine is the first reported substrate specific to OCT2 but not to MATE1 or MATE2K.


Subject(s)
4-Aminopyridine/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Potassium Channel Blockers/pharmacokinetics , 4-Aminopyridine/metabolism , 4-Aminopyridine/pharmacology , Biological Transport/drug effects , HEK293 Cells , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Metformin/metabolism , Metformin/pharmacokinetics , Organic Cation Transporter 2/antagonists & inhibitors , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology
6.
CPT Pharmacometrics Syst Pharmacol ; 6(9): 625-634, 2017 09.
Article in English | MEDLINE | ID: mdl-28623849

ABSTRACT

Lambert-Eaton myasthenia (LEM) is a rare autoimmune disorder associated with debilitating muscle weakness. There are limited treatment options and 3,4-diaminopyridine (3,4-DAP) free base is an investigational orphan drug used to treat LEM-related weakness. We performed a population pharmacokinetic/pharmacodynamic (PK/PD) analysis using 3,4-DAP and metabolite concentrations collected from a phase II study in patients with LEM. The Triple Timed Up & Go (3TUG) assessment, which measures lower extremity weakness, was the primary outcome measure. A total of 1,270 PK samples (49 patients) and 1,091 3TUG data points (32 randomized patients) were included in the PK/PD analysis. A two-compartment and one-compartment model for parent and metabolite, respectively, described the PK data well. Body weight and serum creatinine partially explained the variability in clearance for the final PK model. A fractional inhibitory maximum effect (Emax ) model characterized the exposure-response relationship well. The PK/PD model was applied to identify a suggested dosing approach for 3,4-DAP free base.


Subject(s)
4-Aminopyridine/analogs & derivatives , Lambert-Eaton Myasthenic Syndrome/drug therapy , Models, Biological , Muscle Weakness/drug therapy , Potassium Channel Blockers , 4-Aminopyridine/blood , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , 4-Aminopyridine/therapeutic use , Adult , Aged , Aged, 80 and over , Amifampridine , Arylamine N-Acetyltransferase/genetics , Female , Humans , Lambert-Eaton Myasthenic Syndrome/blood , Lambert-Eaton Myasthenic Syndrome/physiopathology , Lower Extremity/physiopathology , Male , Middle Aged , Muscle Weakness/blood , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Polymorphism, Single Nucleotide , Potassium Channel Blockers/blood , Potassium Channel Blockers/pharmacokinetics , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Treatment Outcome , Young Adult
7.
Clin Ther ; 39(7): 1360-1370, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28641995

ABSTRACT

PURPOSE: The purpose of this study is to evaluate safety, tolerability, and pharmacokinetic (PK) properties of amifampridine phosphate (Firdapse™) and its major inactive 3-N-acetyl metabolite in renally impaired and healthy individuals with slow acetylator (SA) and rapid acetylator (RA) phenotypes. METHODS: This was a Phase I, multicenter, open-label study of the PK properties and safety profile of amifampridine phosphate in individuals with normal, mild, moderate, or severely impaired renal function. Amifampridine phosphate was given as a single 10 mg (base equivalent) dose, and the plasma and urine PK properties of amifampridine and its 3-N-acetyl metabolite were determined. The safety profile was evaluated by monitoring adverse events (AEs), clinical laboratory tests, and physical examinations. FINDINGS: Amifampridine clearance was predominantly metabolic through N-acetylation, regardless of renal function in both acetylator phenotypes. In individuals with normal renal function, mean renal clearance represented approximately 3% and 18% of the total clearance of amifampridine in RA and SA, respectively. Large differences in amifampridine exposure were observed between acetylation phenotypes across renal function levels. Mean amifampridine exposure values of AUC0-∞ and Cmax were up to 8.8-fold higher in the SA group compared with the RA group across renal function levels. By comparison, mean AUC0-∞ was less affected by renal function within an acetylator group, only 2- to 3-fold higher in individuals with severe renal impairment (RI) compared with those with normal renal function. Exposure to amifampridine in the SA group with normal renal function was higher (AUC0-∞, approximately 1.8-fold; Cmax, approximately 4.1-fold) than the RA group with severe RI. Exposure to the inactive 3-N-acetyl metabolite was higher than amifampridine in both acetylator groups, independent of renal function level. The metabolite is cleared by renal excretion, and exposure was clearly dependent on renal function with 4.0- to 6.8-fold increases in AUC0-∞ from normal to severe RI. No new tolerability findings were observed. IMPLICATIONS: A single dose of 10 mg of amifampridine phosphate was well tolerated, independent of renal function and acetylator status. The results indicate that the PK profile of amifampridine is affected by metabolic acetylator phenotype to a greater extent than by renal function level, supporting Firdapse™ administration in individuals with RI in line with current labeling recommendations. Amifampridine should be dosed to effect per the individual patient need, altering administration frequency and dose in normal through severe RI. The therapeutic dose of amifampridine phosphate should be tailored to the individual patient needs by gradual dose titration up to the present maximum recommended dose (60-80 mg/day) or until dose-limiting AEs intervene to avoid overdosing and underdosing. EudraCT identifier: 2013-005349-35.


Subject(s)
4-Aminopyridine/analogs & derivatives , Kidney/metabolism , Potassium Channel Blockers/pharmacokinetics , Renal Insufficiency/metabolism , 4-Aminopyridine/adverse effects , 4-Aminopyridine/pharmacokinetics , Acetylation , Adult , Aged , Amifampridine , Female , Humans , Male , Middle Aged , Potassium Channel Blockers/adverse effects
8.
Farm. hosp ; 41(2): 283-291, mar.-abr. 2017. tab
Article in English | IBECS | ID: ibc-160954

ABSTRACT

Objective: Assessment of the efficacy and safety of fampridine for walking improvement in adult patients with multiple sclerosis. Method: A descriptive retrospective study of all patients who initiated treatment with fampridine between March, 2014 and February, 2015. Efficacy was assessed through the 25- foot walk test and the 12-item walking scale for multiple sclerosis. It was reviewed whether patients had suffered any of the most frequent adverse effects described in the pivotal clinical trial. Results: Six patients were included, with a 66.7% response rate. At 3-6 months, the mean change in walking speed (compared to baseline) was 39.32% and there was a mean improvement of 15 points in the walking scale. Only one patient presented adverse effects. Conclusions: The results obtained are encouraging, particularly when fampridine is the only drug currently approved to control such a disabling symptom as instability while walking (AU)


Objetivo: Evaluación de la efectividad y seguridad de la fampridina en la mejoría de la marcha de pacientes adultos con esclerosis múltiple. Método: Estudio descriptivo retrospectivo de todos los pacientes que iniciaron tratamiento con fampridina entre marzo de 2014 y febrero de 2015. Se evaluó la efectividad utilizando el test de marcha de los 25 pies y la escala de movilidad de 12 ítems de la esclerosis múltiple. Se revisó si los pacientes habían sufrido alguno de los efectos adversos más frecuentes descritos en el ensayo pivotal. Resultados: Se incluyeron 6 pacientes, con una tasa de respuesta del 66,7%. A los 3-6 meses, el cambio medio en la velocidad de la marcha (comparado con el basal) fue del 39,32% y hubo una mejora media de 15 puntos en la escala de movilidad. Solo uno de los pacientes presentó efectos adversos. Conclusiones: Los resultados obtenidos son esperanzadores, sobre todo cuando la fampridina es el único fármaco autorizado actualmente para controlar un síntoma tan discapacitante como es la inestabilidad de la marcha (AU)


Subject(s)
Humans , Gait Disorders, Neurologic/drug therapy , Multiple Sclerosis/drug therapy , 4-Aminopyridine/pharmacokinetics , Retrospective Studies , Patient Safety , Treatment Outcome
9.
Neurorehabil Neural Repair ; 31(4): 387-396, 2017 04.
Article in English | MEDLINE | ID: mdl-28107804

ABSTRACT

BACKGROUND: 4-Aminopyridine (4-AP) is a Food and Drug Administration-approved drug to improve motor function in people with multiple sclerosis. Preliminary results suggest the drug may act on intact neural circuits and not just on demyelinated ones. OBJECTIVE: To determine if 4-AP at clinically relevant levels alters the excitability of intact motor circuits. METHODS: In anesthetized rats, electrodes were placed over motor cortex and the dorsal cervical spinal cord for electrical stimulation, and electromyogram electrodes were inserted into biceps muscle to measure responses. The motor responses to brain and spinal cord stimulation were measured before and for 5 hours after 4-AP administration both in uninjured rats and rats with a cut lesion of the pyramidal tract. Blood was collected at the same time as electrophysiology to determine drug plasma concentration with a goal of 20 to 100 ng/mL. RESULTS: We first determined that a bolus infusion of 0.32 mg/kg 4-AP was optimal: it produced on average 61.5 ± 1.8 ng/mL over the 5 hours after infusion. This dose of 4-AP increased responses to spinal cord stimulation by 1.3-fold in uninjured rats and 3-fold in rats with pyramidal tract lesion. Responses to cortical stimulation also increased by 2-fold in uninjured rats and up to 4-fold in the injured. CONCLUSION: Clinically relevant levels of 4-AP strongly augment physiological responses in intact circuits, an effect that was more robust after partial injury, demonstrating its broad potential in treating central nervous system injuries.


Subject(s)
4-Aminopyridine/pharmacology , Central Nervous System Agents/pharmacology , Cervical Cord/drug effects , Motor Cortex/drug effects , Pyramidal Tracts/drug effects , Spinal Cord Injuries/drug therapy , 4-Aminopyridine/blood , 4-Aminopyridine/pharmacokinetics , Animals , Central Nervous System Agents/blood , Central Nervous System Agents/pharmacokinetics , Cervical Cord/injuries , Cervical Cord/physiology , Cervical Cord/physiopathology , Drug Evaluation, Preclinical , Electric Stimulation , Electromyography , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Female , Forelimb/drug effects , Forelimb/physiology , Forelimb/physiopathology , Microelectrodes , Motor Cortex/physiology , Motor Cortex/physiopathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Pyramidal Tracts/injuries , Pyramidal Tracts/physiology , Pyramidal Tracts/physiopathology , Random Allocation , Rats, Sprague-Dawley , Spinal Cord Injuries/physiopathology
10.
Biomed Chromatogr ; 30(9): 1371-7, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26849839

ABSTRACT

This study describes a comparison between LC-UV and LC-MS method for the simultaneous analyses of a few disease-modifying agents of multiple sclerosis. Quantitative determination of fampridine (FAM), teriflunomide (TFM) and dimethyl fumarate (DMF) was performed in human plasma with the recovery values in the range of 85-115%. A reversed-phase high-performance liquid chromatography (HPLC) with UV as well as MS detection is used. The method utilizes an XBridge C18 silica column and a gradient elution with mobile phase consisting of ammonium formate and acetonitrile at a flow rate of 0.5 mL min(-1) . The method adequately resolves FAM, TFM and DMF within a run time of 15 min. Owing to low molecular weights, the estimation of DMF and FAM is more versatile in UV than MS detection. With LC-UV, the detection limits of FAM, TFM and DMF were 0.1, 0.05, 0.05 µg and the quantification limit for all the analytes was 1 µg. With LC-MS, the detection and quantification limits for all of the analytes were 1 and 5 ng, respectively. The two techniques were completely validated and shown to be reproducible and sensitive. They were applied to a pharmacokinetic study in rats by a single oral dose. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
4-Aminopyridine/blood , Chromatography, Liquid/methods , Crotonates/blood , Dimethyl Fumarate/blood , Mass Spectrometry/methods , Spectrophotometry, Ultraviolet/methods , Toluidines/blood , 4-Aminopyridine/pharmacokinetics , Animals , Crotonates/pharmacokinetics , Dimethyl Fumarate/pharmacokinetics , Humans , Hydroxybutyrates , Nitriles , Rats , Reference Standards , Reproducibility of Results , Toluidines/pharmacokinetics
11.
Chem Commun (Camb) ; 52(22): 4187-90, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26906286

ABSTRACT

3,4-Diaminopyridine has shown promise in reversing botulinum intoxication, but poor pharmacokinetics and a narrow therapeutic window limit its clinical utility. Thus, we developed a pH-dependent oral delivery platform using club moss spore exines. These exine microcapsules slowed 3,4-diaminopyridine absorption, limited its seizure activity, and enabled delivery of doses which prolonged mouse survival after botulism neurotoxin A intoxication.


Subject(s)
4-Aminopyridine/analogs & derivatives , Botulinum Toxins, Type A/poisoning , Capsules , Lycopodium/chemistry , 4-Aminopyridine/administration & dosage , 4-Aminopyridine/pharmacokinetics , Administration, Oral , Amifampridine , Microscopy, Electron, Scanning
12.
Oncotarget ; 7(3): 2329-42, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26295308

ABSTRACT

CCT245737 is the first orally active, clinical development candidate CHK1 inhibitor to be described. The IC50 was 1.4 nM against CHK1 enzyme and it exhibited>1,000-fold selectivity against CHK2 and CDK1. CCT245737 potently inhibited cellular CHK1 activity (IC50 30-220 nM) and enhanced gemcitabine and SN38 cytotoxicity in multiple human tumor cell lines and human tumor xenograft models. Mouse oral bioavailability was complete (100%) with extensive tumor exposure. Genotoxic-induced CHK1 activity (pS296 CHK1) and cell cycle arrest (pY15 CDK1) were inhibited both in vitro and in human tumor xenografts by CCT245737, causing increased DNA damage and apoptosis. Uniquely, we show CCT245737 enhanced gemcitabine antitumor activity to a greater degree than for higher doses of either agent alone, without increasing toxicity, indicating a true therapeutic advantage for this combination. Furthermore, development of a novel ELISA assay for pS296 CHK1 autophosphorylation, allowed the quantitative measurement of target inhibition in a RAS mutant human tumor xenograft of NSCLC at efficacious doses of CCT245737. Finally, CCT245737 also showed significant single-agent activity against a MYC-driven mouse model of B-cell lymphoma. In conclusion, CCT245737 is a new CHK1 inhibitor clinical development candidate scheduled for a first in man Phase I clinical trial, that will use the novel pS296 CHK1 ELISA to monitor target inhibition.


Subject(s)
4-Aminopyridine/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , Checkpoint Kinase 1/drug effects , Lung Neoplasms/drug therapy , Lymphoma, B-Cell/drug therapy , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazines/pharmacology , Xenograft Model Antitumor Assays , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , HT29 Cells , Humans , Irinotecan , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Pyrazines/pharmacokinetics , Gemcitabine
13.
Int J Clin Pharmacol Ther ; 53(8): 674-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26152130

ABSTRACT

OBJECTIVE: 3,4-diaminopyridine (3,4-DAP) is commonly used for treating neuromuscular diseases, such as the Lambert-Eaton myasthenic syndrome, but the pharmacokinetics of 3,4-DAP base have not been investigated. We therefore studied 3,4-DAP base pharmacokinetics in healthy Japanese volunteers. MATERIALS AND METHODS: In this crossover study, we administered a single oral dose of 10 or 20 mg 3,4-DAP base to healthy Japanese volunteers (n = 5) after food intake, or 10 mg 3,4-DAP to fasting individuals. We measured serum 3,4-DAP concentrations, performed electrocardiography (ECG), and administered questionnaires. RESULTS: After administration of 10 or 20 mg 3,4-DAP following food intake, the maximum serum concentrations (Cmax) were 8.09 ± 4.47 ng/mL and 35.8 ± 15.7 ng/mL, respectively (mean ± standard deviation; SD), and the areas under the serum concentration-time curve (extrapolated to infinity) were 639 ± 213 ng x min/mL and 2,097 ± 936 ng x min/mL (mean ± SD), respectively. Administration to fasted individuals indicated that food intake did not significantly alter 3,4-DAP pharmacokinetics. ECG showed no clinically significant changes, but PR intervals were prolonged in all cases. Two out of 5 subjects showed perioral paresthesia symptoms after administration of 20 mg 3,4-DAP. CONCLUSION: This study indicated that 3,4-DAP base pharmacokinetics were non-linear. Although no clinically significant changes in ECG were observed, it is advisable to perform ECG periodically during 3,4-DAP administration in order to monitor cardiac function. Moreover, the development of perioral paresthesia may be dependent on the dose of 3,4-DAP used.


Subject(s)
4-Aminopyridine/analogs & derivatives , Asian People , Neuromuscular Agents/pharmacokinetics , 4-Aminopyridine/administration & dosage , 4-Aminopyridine/adverse effects , 4-Aminopyridine/blood , 4-Aminopyridine/pharmacokinetics , Administration, Oral , Adult , Amifampridine , Area Under Curve , Cross-Over Studies , Electrocardiography , Fasting , Half-Life , Healthy Volunteers , Heart Rate/drug effects , Humans , Japan , Metabolic Clearance Rate , Models, Biological , Neural Conduction/drug effects , Neuromuscular Agents/administration & dosage , Neuromuscular Agents/adverse effects , Neuromuscular Agents/blood , Nonlinear Dynamics , Postprandial Period , Risk Assessment , Surveys and Questionnaires , Treatment Outcome , Young Adult
14.
Clin Ther ; 37(7): 1555-63, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26101174

ABSTRACT

PURPOSE: Amifampridine (3,4-diaminopyridine) has been approved in the European Union for the treatment of Lambert-Eaton myasthenic syndrome. Amifampridine has a narrow therapeutic index, and supratherapeutic exposure has been associated with dose-dependent adverse events, including an increased risk for seizure. This study assessed the effect of food on the relative bioavailability of amifampridine in healthy subjects and informed on conditions that can alter exposure. METHODS: This randomized, open-labeled, 2-treatment, 2-period crossover study enrolled 47 healthy male and female subjects. Subjects were randomly assigned to receive 2 single oral doses of amifampridine phosphate salt (20 mg base equivalents per dose) under fed or fasted conditions separated by a washout period. Blood and urine samples for pharmacokinetic analyses were taken before and after dosing. Plasma concentrations of amifampridine and an inactive 3-N-acetyl metabolite were determined. The relative bioavailability values of amifampridine and metabolite were assessed based on the plasma PK parameters AUC0-∞, AUC0-t, and Cmax in the fed and fasted states using noncompartmental pharmacokinetic analysis. Parent drug and metabolite excretion were calculated from urinary concentrations. A food effect on bioavailability would be established if the 90% CI of the ratio of population geometric mean value of AUC0-∞, AUC0-t, or Cmax between fed and fasted administration was not within the bioequivalence range of 80% to 125%. Tolerability was assessed based on adverse-event reporting, clinical laboratory assessments, physical examination including vital sign measurements, 12-lead ECG, and concurrent medication use. FINDINGS: Food slowed and somewhat decreased the absorption of amifampridine. There was a decrease in exposure (Cmax, 44%; AUC, 20%) after oral administration of amifampridine phosphate salt in the presence of food, and mean Tmax was 2-fold longer in the fed state. The extent of exposure and plasma elimination half-life of the major metabolite was greater than those of amifampridine in the fed and fasted conditions. Mean AUCs in the fed and fasted states were slightly greater in women than men, with no difference in mean Cmax. Orally administered amifampridine was renally eliminated (>93%) as the parent compound and metabolite within 24 hours. Single oral doses of 20 mg of amifampridine phosphate salt were considered well tolerated in both the fed and fasted conditions. High intersubject variability (%CVs, >30%) in amifampridine pharmacokinetic parameter values was observed. IMPLICATIONS: At the intended dose under fasting conditions, amifampridine exposure may be increased. European Union Drug Regulating Authorities Clinical Trials identifier: 2011-000596-13.


Subject(s)
4-Aminopyridine/analogs & derivatives , Eating/physiology , Food-Drug Interactions/physiology , 4-Aminopyridine/adverse effects , 4-Aminopyridine/pharmacokinetics , Administration, Oral , Adult , Amifampridine , Area Under Curve , Biological Availability , Cross-Over Studies , Fasting/metabolism , Female , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Phosphates/pharmacokinetics , Therapeutic Equivalency , Young Adult
15.
ACS Chem Neurosci ; 5(8): 632-6, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25000171

ABSTRACT

Botulinum neurotoxicity is characterized by peripheral neuromuscular blockade/flaccid paralysis that can lead to respiratory failure and ultimately death. Current therapeutic options provide relief in a pre-exposure scenario, but there are no clinically approved postexposure medical countermeasures. Here, we introduce a platform that utilizes a combination of a toxin sequestering agent and a pharmacological antagonist to ablate botulinum neurotoxicity in a well-defined mouse lethality assay. The platform was constructed to allow for ready exchange of sequestering agent and/or pharmacological antagonist for therapeutic optimization. As such, we attempted to improve upon the pharmacological antagonist, a potassium channel blocker, 3,4-diaminopyridine, through a prodrug approach; thus, a complete kinetic decomposition pathway is described. These experiments provide the first proof-of-principle that a synergistic combination strategy can be used to reduce toxin burden in the peripheral using a sequestering antibody, while restoring muscle action via a pharmacological small molecule antagonist.


Subject(s)
4-Aminopyridine/analogs & derivatives , Botulinum Toxins/antagonists & inhibitors , Botulinum Toxins/toxicity , Neurotoxicity Syndromes/drug therapy , Potassium Channel Blockers/pharmacology , Sequestering Agents/pharmacology , 4-Aminopyridine/chemistry , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Amifampridine , Animals , Botulinum Antitoxin/pharmacology , Drug Therapy, Combination , Female , Kinetics , Mice , Neurotoxicity Syndromes/blood , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacokinetics , Survival Analysis
16.
J Clin Pharmacol ; 54(1): 53-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24150835

ABSTRACT

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.


Subject(s)
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
17.
J Med Chem ; 56(11): 4521-36, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23668484

ABSTRACT

Herein we report our lead optimization effort to identify potent, selective, and orally bioavailable TYK2 inhibitors, starting with lead molecule 3. We used structure-based design to discover 2,6-dichloro-4-cyanophenyl and (1R,2R)-2-fluorocyclopropylamide modifications, each of which exhibited improved TYK2 potency and JAK1 and JAK2 selectivity relative to 3. Further optimization eventually led to compound 37 that showed good TYK2 enzyme and interleukin-12 (IL-12) cell potency, as well as acceptable cellular JAK1 and JAK2 selectivity and excellent oral exposure in mice. When tested in a mouse IL-12 PK/PD model, compound 37 showed statistically significant knockdown of cytokine interferon-γ (IFNγ), suggesting that selective inhibition of TYK2 kinase activity might be sufficient to block the IL-12 pathway in vivo.


Subject(s)
4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemical synthesis , Aminopyridines/chemical synthesis , Benzamides/chemical synthesis , TYK2 Kinase/antagonists & inhibitors , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Benzamides/pharmacokinetics , Benzamides/pharmacology , Biological Availability , Crystallography, X-Ray , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-12/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Mice , Microsomes, Liver/metabolism , Models, Molecular , Protein Binding , Rats , STAT4 Transcription Factor/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
18.
Curr Med Res Opin ; 29(12): 1637-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23157466

ABSTRACT

OBJECTIVE: Using data pooled from several studies of dalfampridine extended release (ER), a population pharmacokinetic model was developed for the purposes of characterizing the population pharmacokinetics and pharmacodynamics of dalfampridine-ER with respect to variability in pharmacokinetics, covariates affecting the pharmacokinetics, and whether the current therapeutic dosage represents an optimal dosage. Studies were conducted in healthy volunteers and multiple sclerosis (MS) patients over the course of development and registration of dalfampridine extended release tablets (dalfampridine-ER [Ampyra *]; prolonged-, modified- or sustained-release fampridine [Fampyra †] in some countries). METHODS: The model used to best describe the population pharmacokinetics of dalfampridine-ER was an open, one-compartment model with first-order absorption, first-order elimination and an absorption lag time. RESULTS: The population median estimated oral clearance was 36 L/h for a 50-year-old woman with a creatinine clearance of 105 mL/min and 42 L/h for a comparable man. The typical volume of distribution was 304 L for women and 403 L for men. The estimated absorption rate constant was 1.22 hours(-1) in the fasted state and 2.22 hours(-1) when given with food. The covariates identified as having a significant effect (p < 0.01) on model fit were food and gender on absorption rate, and gender, age and creatinine clearance on oral clearance. Only creatinine clearance and age are of clinical relevance. Concomitant medications did not affect any of the parameters in the model. Exposure-response relationships for both efficacy and safety were consistent with what has been observed in clinical trials. Limitations of this study include some reliance on unpublished data, and the limited effectiveness of the model for determining the likelihood of the efficacy and safety of dalfampridine-ER in clinical practice. CONCLUSIONS: The approved therapeutic dosage regimen of dalfampridine-ER 10 mg twice daily was identified as the optimum dosing regimen based on model-predicted exposure response relationships for efficacy and adverse events. A limitation of this study is the limited effectiveness of the models used to predict long-term efficacy and safety of dalfampridine-ER in clinical use.


Subject(s)
4-Aminopyridine/pharmacokinetics , Models, Biological , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Sex Characteristics , 4-Aminopyridine/administration & dosage , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Time Factors
19.
Curr Med Res Opin ; 29(12): 1627-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23157467

ABSTRACT

BACKGROUND: In January 2010, dalfampridine extended release tablets (dalfampridine-ER [Ampyra *]; prolonged-, modified- or sustained-release fampridine [Fampyra †] in some countries), 10 mg to be administered twice daily approximately 12 hours apart, were approved by the US Food and Drug Administration. This was the first drug indicated to improve walking in patients with MS. SCOPE: Publications describing the pharmacokinetics of dalfampridine-ER or the immediate release formulation were identified from a search of PubMed through June 2012 using the search terms 'dalfampridine OR fampridine OR 4-aminopyridine' AND 'pharmacokinetics' and were supplemented with unpublished studies made available by Acorda Therapeutics Inc. FINDINGS: Pharmacokinetic studies show dose proportionality, with dalfampridine-ER having a more favorable profile than immediate-release dalfampridine. With twice-daily dosing of dalfampridine-ER, time to peak plasma concentration (3.2-3.9 hours) and apparent terminal plasma half-life (5.6-6.4 hours) are approximately twice those of immediate-release formulations, with comparable overall exposure and peak plasma concentrations (21.6 ng/mL) that were maintained at levels approximately 50% lower than immediate release. Steady state is achieved within 39 hours; pharmacokinetics are predictable based on single dosing. Trough plasma concentrations of 13-15 ng/mL are required to maintain efficacy. Renal excretion is predominantly as unchanged compound, and renal clearance in healthy individuals exceeds the glomerular filtration rate. Since dalfampridine-ER exposure increases with renal impairment, it is contraindicated in patients with moderate or severe impairment in the US, and in patients with any renal impairment in the European Union. CONCLUSIONS: Dalfampridine-ER has low protein binding, is not a substrate for p-glycoprotein and does not affect CYP450 enzymes, suggesting a low potential for drug-drug interactions. Because of the narrow therapeutic range and risk of adverse events, including seizure, with increasing plasma concentrations, the recommended dose and regimen of dalfampridine-ER should not be exceeded and not be used with other dalfampridine formulations. A limitation of this review is that it includes some data that have not yet been published.


Subject(s)
4-Aminopyridine/pharmacokinetics , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/pharmacokinetics , 4-Aminopyridine/therapeutic use , Female , Humans , Male , Potassium Channel Blockers/therapeutic use , PubMed
20.
Expert Rev Clin Pharmacol ; 5(6): 649-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23146002

ABSTRACT

Multiple sclerosis is a progressive demyelinating neurological disease resulting in long-term disability, commonly manifesting in walking impairment and reduced quality of life. The use of the potassium channel blocker fampridine, chemically 4-aminopyridine, as an immediate-release formulation to improve action potential conduction in demyelinated axons was hampered by adverse events, including seizures. The prolonged-release formulation of fampridine (known as modified- or sustained-release fampridine in some countries and dalfampridine extended release in the USA) has a longer apparent half-life and a lower peak plasma concentration versus immediate-release fampridine formulations, sustaining its duration of action and reducing the incidence of adverse events. Prolonged-release fampridine is the first drug specifically licensed to improve walking in patients with multiple sclerosis, and has been shown to consistently improve walking ability in a third of patients. Prolonged-release fampridine Timed-Walk Responders showed both clinically significant improvements in walking speed and in patient-reported walking ability.


Subject(s)
4-Aminopyridine/therapeutic use , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/therapeutic use , 4-Aminopyridine/chemistry , 4-Aminopyridine/pharmacokinetics , Clinical Trials as Topic , Delayed-Action Preparations , Humans , Multiple Sclerosis/metabolism , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacokinetics , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...