Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Pharmacokinet ; 62(1): 89-99, 2023 01.
Article in English | MEDLINE | ID: mdl-36633811

ABSTRACT

BACKGROUND AND OBJECTIVE: Givosiran, approved for the treatment of acute hepatic porphyria (AHP), is the first subcutaneously administered RNAi therapeutic. This analysis was undertaken to describe the plasma pharmacokinetics (PK) of givosiran and its active metabolite, AS(N-1)3' givosiran, and to identify factors that contribute to intersubject PK variability. METHODS: A population PK model was developed using data from givosiran clinical trials that enrolled patients with AHP or who were asymptomatic chronic high excreters (CHEs) of toxic heme intermediates. Givosiran and AS(N-1)3' givosiran PK were modeled simultaneously using non-linear mixed-effects modeling. RESULTS: Plasma PK of givosiran was best described by a two-compartment model. Givosiran absorption after subcutaneous administration and conversion of givosiran to AS(N-1)3' givosiran were incorporated as first-order processes. Hepatic clearance was the major route of elimination from the central compartment, with renal clearance accounting for < 20% of the total clearance. Body weight, East Asian ethnicity, and renal impairment were significant covariates in the model; however, none of the covariates evaluated resulted in clinically meaningful differences in plasma exposures of givosiran and AS(N-1)3' givosiran. The model adequately described observed concentrations and variability across a wide range of dose levels. Model-derived simulations showed similar exposures for givosiran and its active metabolite in adults and adolescents. CONCLUSIONS: The PK of givosiran and its active metabolite were not significantly affected by demographic or clinical parameters that would require adjustment from the approved body weight-based dose of givosiran 2.5 mg/kg once monthly.


Subject(s)
Porphyrias, Hepatic , RNAi Therapeutics , Adult , Adolescent , Humans , Body Weight
2.
Clin Pharmacol Drug Dev ; 11(11): 1264-1272, 2022 11.
Article in English | MEDLINE | ID: mdl-35908210

ABSTRACT

Triheptanoin is an odd-carbon, medium-chain triglyceride consisting of three fatty acids with seven carbons each on a glycerol backbone, indicated for the treatment of adult and pediatric patients with long-chain fatty acid oxidation disorders (LC-FAOD). A total of 562 plasma concentrations of heptanoate, the most abundant and pharmacologically active metabolite of triheptanoin, from 13 healthy adult subjects and 30 adult and pediatric subjects with LC-FAOD were included in the population pharmacokinetic (PK) analyses. Multiple peaks of heptanoate observed in several subjects were characterized by dual first-order absorption with a lag time in the second absorption compartment. The disposition of heptanoate in human plasma was adequately described by one-compartmental distribution with a linear elimination. The apparent clearance (CL/F) and apparent volume of distribution were allometrically scaled with body weight to describe PK data across a wide range of age groups in subjects with LC-FAOD. The typical CL/F in adult subjects with LC-FAOD was ≈19% lower than that in healthy subjects. Model-estimated elimination half-life for LC-FAOD patients was ∼1.7 hours, supporting a recommended dosing frequency of ≥4 times per day. Covariate analyses indicate that age, race, and sex did not lead to clinically meaningful changes in the exposure of heptanoate.


Subject(s)
Heptanoates , Lipid Metabolism, Inborn Errors , Adult , Humans , Child , Lipid Metabolism, Inborn Errors/metabolism , Healthy Volunteers , Triglycerides , Fatty Acids/metabolism
3.
Clin Transl Sci ; 14(6): 2497-2509, 2021 11.
Article in English | MEDLINE | ID: mdl-34402197

ABSTRACT

Teduglutide is a recombinant analog of human glucagon-like peptide-2 that regulates the functional and structural integrity of the cells lining the gastrointestinal tract. Teduglutide is approved for the treatment of patients with short bowel syndrome (SBS) who are dependent on parenteral support (PS). Population pharmacokinetic (PK) and exposure-response analyses were performed to support teduglutide dosing in patients with SBS. The analysis included 219 patients with SBS (aged <1 year, 5 patients; 1-11 years, 86 patients; 12-17 years, 8 patients; 18-79 years, 120 patients), and 259 non-SBS subjects (including healthy volunteers and subjects with renal or liver impairment). A one-compartment model with first-order absorption and linear elimination adequately characterized the PKs of teduglutide. In patients with SBS, the apparent clearance (CL/F), volume of distribution (V/F), and elimination half-life of teduglutide were 16.0 L/h, 33.9 L, and 1.47 h, respectively. CL/F depended on body weight and renal function, and V/F depended on body weight and age. Maximum concentration (Cmax ) of teduglutide was similar in adult and pediatric patients, and in Japanese and non-Japanese patients. A time- and exposure-response model dependent on the Cmax of teduglutide adequately characterized the reduction in PS over more than 2 years of treatment. Daily dosing of 0.05 mg/kg teduglutide resulted in a maximum reduction in PS of 5.76 L/week. Higher Cmax values were associated with a more important reduction in PS over time. Adult and pediatric patients with SBS presented similar PKs and response to teduglutide.


Subject(s)
Gastrointestinal Agents/pharmacokinetics , Peptides/pharmacokinetics , Short Bowel Syndrome/drug therapy , Adolescent , Adult , Aged , Child , Child, Preschool , Humans , Infant , Japan/ethnology , Middle Aged , Parenteral Nutrition , Young Adult
4.
J Clin Pharmacol ; 61(4): 555-564, 2021 04.
Article in English | MEDLINE | ID: mdl-33091166

ABSTRACT

Elevated bradykinin levels are responsible for the development of clinical symptoms in patients with hereditary angioedema (HAE). Icatibant is a bradykinin type 2 receptor antagonist indicated for the acute treatment of HAE attacks. A population modeling and simulation approach was used to examine sources of variability impacting icatibant pharmacokinetics (PK) and provide guidance on icatibant dosing in pediatric patients with HAE. An exposure-response analysis was performed for the time to onset of symptom relief (TOSR). Data from 141 adults (133 healthy, 8 with HAE) who received subcutaneous icatibant 30 mg and 31 pediatric patients with HAE who received 0.4 mg/kg (capped at 30 mg) were included in the analysis. Icatibant PK was described by a 2-compartment model with linear elimination. Complete absorption of icatibant was expected within 1 hour of dosing. The apparent clearance and central volume of distribution were 15.4 L/h and 20.4 L, respectively. Icatibant PK was mainly dependent on body weight. The mean TOSR was very short (1.38 hours). A flat exposure-response was observed, confirming that the relationship plateaued at the level of exposure observed in pediatric patients. Simulations confirmed that weight band-based dosing regimens (10 mg [12-25 kg], 15 mg [26-40 kg], 20 mg [41-50 kg], 25 mg [51-65 kg], and 30 mg [>65 kg]) resulted in exposure similar to the 0.4-mg/kg dose. This analysis showed that icatibant undergoes rapid absorption, reaches levels required for therapeutic response, and promptly relieves HAE symptoms. A weight band-based dosing regimen is appropriate in pediatric patients with HAE.


Subject(s)
Angioedemas, Hereditary/drug therapy , Bradykinin B2 Receptor Antagonists/pharmacokinetics , Bradykinin B2 Receptor Antagonists/therapeutic use , Bradykinin/analogs & derivatives , Adolescent , Adult , Body Weight , Bradykinin/pharmacokinetics , Bradykinin/therapeutic use , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Models, Biological , Young Adult
5.
Nucleic Acid Ther ; 30(3): 143-152, 2020 06.
Article in English | MEDLINE | ID: mdl-32175804

ABSTRACT

Hereditary transthyretin-mediated amyloidosis is an inherited, rapidly progressive, life-threatening disease caused by mutated transthyretin (TTR) protein. Patisiran is a small interfering RNA (siRNA) formulated in a lipid nanoparticle that inhibits hepatic TTR protein synthesis by RNA interference. We have developed an indirect-response pharmacokinetic-pharmacodynamic model relating plasma siRNA (ALN-18328) levels to serum TTR reduction across five clinical studies. A sigmoidal function described this relationship, with estimated Hill coefficient of 0.548, and half maximal inhibitory concentration (IC50), IC80, and IC90 values of 9.45, 118.5, and 520.5 ng/mL, respectively. Following patisiran 0.3 mg/kg every 3 weeks (q3w), steady-state plasma ALN-18328 exposures were between IC80 and IC90, yielding average serum TTR reductions of 80%-90% from baseline. Covariate analysis indicated similar TTR reduction across evaluated intrinsic and extrinsic factors, obviating the need for dose adjustment. Modeling results support the recommended patisiran dosing schedule of 0.3 mg/kg q3w, with a maximum dose of 30 mg for patients weighing ≥100 kg.


Subject(s)
Amyloid Neuropathies, Familial/blood , Models, Statistical , Neuroprotective Agents/pharmacokinetics , Prealbumin/antagonists & inhibitors , RNA, Small Interfering/pharmacokinetics , Adult , Aged , Aged, 80 and over , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Amyloid Neuropathies, Familial/therapy , Case-Control Studies , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Dosage Calculations , Female , Gene Expression , Humans , Male , Middle Aged , Mutation , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neuroprotective Agents/blood , Prealbumin/genetics , Prealbumin/metabolism , RNA Interference , RNA, Small Interfering/blood
6.
Mol Pain ; 6: 90, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21143971

ABSTRACT

BACKGROUND: The CCR2/CCL2 system has been identified as a regulator in the pathogenesis of neuropathy-induced pain. However, CCR2 target validation in analgesia and the mechanism underlying antinociception produced by CCR2 antagonists remains poorly understood. In this study, in vitro and in vivo pharmacological approaches using a novel CCR2 antagonist, AZ889, strengthened the hypothesis of a CCR2 contribution to neuropathic pain and provided confidence over the possibilities to treat neuropathic pain with CCR2 antagonists. RESULTS: We provided evidence that dorsal root ganglia (DRG) cells harvested from CCI animals responded to stimulation by CCL2 with a concentration-dependent calcium rise involving PLC-dependent internal stores. This response was associated with an increase in evoked neuronal action potentials suggesting these cells were sensitive to CCR2 signalling. Importantly, treatment with AZ889 abolished CCL2-evoked excitation confirming that this activity is CCR2-mediated. Neuronal and non-neuronal cells in the spinal cord were also excited by CCL2 applications indicating an important role of spinal CCR2 in neuropathic pain. We next showed that in vivo spinal intrathecal injection of AZ889 produced dose-dependent analgesia in CCI rats. Additionally, application of AZ889 to the exposed spinal cord inhibited evoked neuronal activity and confirmed that CCR2-mediated analgesia involved predominantly the spinal cord. Furthermore, AZ889 abolished NMDA-dependent wind-up of spinal withdrawal reflex pathway in neuropathic animals giving insight into the spinal mechanism underlying the analgesic properties of AZ889. CONCLUSIONS: Overall, this study strengthens the important role of CCR2 in neuropathic pain and highlights feasibility that interfering on this mechanism at the spinal level with a selective antagonist can provide new analgesia opportunities.


Subject(s)
Hyperalgesia/drug therapy , Neuralgia/drug therapy , Piperazines/therapeutic use , Receptors, CCR2/antagonists & inhibitors , Spinal Cord/pathology , Animals , Calcium Signaling , Drug Delivery Systems , Ganglia, Spinal/pathology , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, CCR2/physiology , Signal Transduction
7.
Basic Clin Pharmacol Toxicol ; 102(1): 59-65, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17983423

ABSTRACT

Chondroitin sulfate is referred as a symptomatic slow-acting drug for osteoarthritis because it improves articular function, and reduces joint swelling and effusion. In addition, chondroitin sulfate prevents joint space narrowing of the knee. We hypothesized that the anti-inflammatory effect of chondroitin sulfate is associated to a decrease in the activation of mitogen-activated protein kinases (MAPK) and of the transcription factors nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1). Cultured rabbit chondrocytes were stimulated with interleukin-1beta (IL-1beta) in presence of chondroitin sulfate. Nuclear translocation of NF-kappaB and AP-1, and nitrite concentrations (as an index for nitric oxide) was assessed 48 hr later. The effect of chondroitin sulfate on IL-1beta activation of extracellular signal-regulated kinase 1/2 (Erk1/2) and p38MAPK was documented by immunoblot. The effect of chondroitin sulfate on sodium nitroprusside-induced apoptosis was evaluated with the terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labelling assay. Chondroitin sulfate reduced IL-1beta-induced NF-kappaB nuclear translocation, but not AP-1 translocation, it decreased IL-1beta-induced phosphorylation of Erk1/2 and abrogated p38MAPK phosphorylation, but did not prevent IL-1beta-induced increase in nitrite. Finally, chondroitin sulfate decreased nitroprusside-induced apoptosis of the chondrocytes. These results suggest that some of the biological activities of chondroitin sulfate may be associated to the reduction in Erk1/2 and p38MAPK phosphorylation and nuclear transactivation of NF-kappaB.


Subject(s)
Cell Nucleus/drug effects , Chondrocytes/drug effects , Chondroitin Sulfates/pharmacology , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Animals , Apoptosis/drug effects , Bromodeoxyuridine/metabolism , Cartilage, Articular/cytology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Drug Antagonism , Drug Therapy, Combination , Flavonoids/pharmacology , Male , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Nitrites/metabolism , Nitroprusside/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Rabbits , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Psychiatry Neurosci ; 31(4): 229-45, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16862241

ABSTRACT

The peptide neurotensin has been studied for more than 30 years. Although it is widely distributed in the central and peripheral nervous systems, neurotensin has been more intensely studied with regard to its interactions with the central dopamine system. A number of claims have been made regarding its possible implication in many diseases of the central nervous system, including schizophrenia. In this review, we describe briefly the basic biology of this neuropeptide, and then we consider the strengths and the weaknesses of the data that suggest a role for neurotensin in schizophrenia, drug abuse, Parkinson's disease, pain, central control of blood pressure, eating disorders, cancer, neurodegenerative disorders and inflammation.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System/physiopathology , Neurotensin/physiology , Animals , Biogenic Monoamines/metabolism , Biogenic Monoamines/physiology , Humans , Neurotensin/biosynthesis , Pain/physiopathology , Parkinson Disease/physiopathology , Schizophrenia/physiopathology , Substance-Related Disorders/physiopathology , Synaptic Transmission
9.
Neuropharmacology ; 50(5): 595-605, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16412480

ABSTRACT

Alternative splicing of the dopamine D2 receptor gene produces two distinct isoforms referred to as D2long (D2L) and D2short (D2S). In mesencephalic dopamine neurons, inhibition of the firing rate through activation of somatodendritic D2 receptors and blockade of neurotransmitter release through stimulation of terminal D2 receptors represent major roles of D2 autoreceptors. Recently, data obtained from D2L-deficient mice suggested that D2S acts as the preferential D2 autoreceptor. In the present study, we investigate whether this D2 isoform-specific autoreceptor function is linked to differences in the subcellular localization and/or signaling properties of the D2S and D2L using mesencephalic neurons transfected with enhanced green fluorescent protein (EGFP)-tagged receptors. Our results show that EGFP-tagged D2S and D2L are localized to the axonal and somatodendritic compartments of mesencephalic neurons. In addition, we demonstrate that EGFP-tagged D2S and D2L regulate cellular excitability, neurotransmitter release and basal levels of intracellular calcium with similar effectiveness. Overall, our morphological and electrophysiological studies suggest that the major D2 autoreceptor function attributed to D2S is likely explained by the predominant expression of this isoform in dopamine neurons rather than by distinct subcellular localization and signaling properties of D2S and D2L.


Subject(s)
Autoreceptors/physiology , Gene Expression/physiology , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Autoreceptors/drug effects , Calcium/metabolism , Dose-Response Relationship, Drug , Electric Stimulation/methods , Gene Expression/drug effects , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mesencephalon/cytology , Patch-Clamp Techniques/methods , Protein Isoforms/genetics , Protein Isoforms/metabolism , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transfection/methods
10.
Biochem Pharmacol ; 71(3): 366-76, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16332358

ABSTRACT

In vivo, the 21-aminosteroid U74389G prevents the decrease in cytochrome P450 (P450) activity produced by a turpentine-induced inflammatory reaction (TIIR). To investigate the underlying mechanism of action, four groups of rabbits were used, controls receiving or not U74389G, and rabbits with the inflammatory reaction receiving or not U74389G. Hepatocytes were isolated 48h later and incubated for 4 and 24h with the serum of the rabbits. In vivo, the TIIR diminished CYP1A1/2 and 3A6 expression, and enhanced hepatic malondialdehyde (MDA) and nitric oxide (NO*) concentrations (p<0.05). U74389G prevented the increase in MDA, as well as the decrease in CYP1A1/2 amounts and activity, but increased CYP3A6 expression by 40% (p<0.05). In vitro, compared with serum from control rabbits (S(CONT)), incubation of serum from rabbits with TIIR (S(TIIR)) for 4 and 24h with hepatocytes from rabbits with TIIR (H(TIIR)) reduced CYP1A2 and CYP3A6 activity (p<0.05) and increased the formation of NO* and MDA. In rabbits with TIIR pretreated with U74389G, the S(TIIR+U) failed to reduce CYP1A2 activity or to increase MDA, although increased NO* and further reduced CYP3A6 activity. On the other hand, in hepatocytes harvested from rabbits with TIIR pretreated with U74389G, S(TIIR) did not decrease CYP1A2 activity and did not enhance MDA, but still increased NO*. In vitro, the reduction of CYP1A2 and CYP3A6 activity by S(TIIR) is not associated to NF-kappaB activation. In conclusion, U74389G prevents CYP1A1/2 down-regulation and decrease in activity by a double mechanism: hindering the release of serum mediators and by averting intracellular events, effect possibly associated with its antioxidant activity. On the other hand, U74389G up-regulates CYP3A6 but inhibits its catalytic activity.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Inflammation/metabolism , Pregnatrienes/pharmacology , Protective Agents/pharmacology , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Down-Regulation , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Inflammation/chemically induced , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Rabbits , Serum/metabolism , Turpentine
SELECTION OF CITATIONS
SEARCH DETAIL