Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Pharmacol ; 63(4): 435-444, 2023 04.
Article in English | MEDLINE | ID: mdl-36453450

ABSTRACT

Systemic lupus erythematosus (SLE) is a systemic, autoimmune disease characterized by chronic inflammation and organ damage. Dapirolizumab pegol inhibits CD40 ligand (CD40L) and is currently undergoing phase 3 trials for the treatment of SLE. To describe the pharmacokinetic characteristics of dapirolizumab pegol and the relationship between exposure and probability of achieving a British Isles Lupus Assessment Group-based Composite Lupus Assessment (BICLA) response, a population pharmacokinetic (popPK) model and an exposure-response model were developed, based on results of the phase 2b trial (RISE; NCT02804763) of dapirolizumab pegol in SLE. Dapirolizumab pegol pharmacokinetics were found to be dose proportional and well described by a 2-compartment model with first-order elimination from the central compartment. In the popPK model, body weight was the only significant covariate. The average concentration of dapirolizumab pegol, derived from the popPK model, was incorporated into the exposure-response model. Overall, the exposure-response model showed that treatment with dapirolizumab pegol increased the probability of transitioning from BICLA "Nonresponder" to "Responder." No significant covariates on BICLA responder status were identified. Notably, the half maximal effective concentration was greater for the transition from "Responder" to "Nonresponder" (150 µg/mL) than the transition from "Nonresponder" to "Responder" (12 µg/mL), indicating that sustained dapirolizumab pegol concentrations may be required to maintain BICLA response. In conclusion, dapirolizumab pegol pharmacokinetics were as expected for a PEGylated molecule and results from the exposure-response model indicate that a favorable dapirolizumab pegol effect was identified for both BICLA "Nonresponder" to "Responder" and "Responder" to "Nonresponder" transition probabilities.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Immunoglobulin Fab Fragments/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Polyethylene Glycols/therapeutic use , Severity of Illness Index , Treatment Outcome
2.
EBioMedicine ; 35: 67-75, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30146344

ABSTRACT

BACKGROUND: Interleukin-13 (IL-13) is a key mediator of T-helper-cell-type-2 (Th-2)-driven asthma, the inhibition of which may improve treatment outcomes. We examined the safety, pharmacokinetics, pharmacodynamics, and immunogenicity of VR942, a dry-powder formulation containing CDP7766, a high-affinity anti-human-IL-13 antigen-binding antibody fragment being developed for the treatment of asthma. METHODS: We conducted a phase 1, randomized, double-blind, placebo-controlled, ascending-dose study at Hammersmith Medicines Research, London, UK, which is now complete. Healthy adults aged 18-50 years (n = 40) were randomized 3:1 to a single inhaled dose of VR942 0.5, 1.0, 5.0, 10, or 20 mg, or placebo. Adults aged 18-50 years who were diagnosed with asthma for ≥6 months before screening, and had forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) values ≥70% of the predicted values at screening (n = 45), were randomized to once-daily inhaled VR942 0.5 or 10 mg, or placebo (2:2:1), or VR942 20 mg or placebo (3:2), for 10 days. All participants were randomized to receive VR942 or placebo based on a randomization list prepared by an independent HMR statistician using SAS® software (SAS Institute, Cary, NC). The primary outcome was safety and tolerability of VR942 (safety population, defined as all who received at least one dose of VR942 or placebo). This study is listed on ClinicalTrials.gov (NCT02473939). FINDINGS: In the VR942 and placebo groups, treatment-emergent adverse events (TEAEs) were reported in 10/30 (33%) and 0/10 (0%) healthy participants, and in 16/29 (55%) and 9/16 (56%) participants with asthma, respectively. Mild intermittent wheezing occurred in 7 participants (VR942 20 mg, n = 4; corresponding placebo, n = 3), resolving spontaneously within 1 h. All TEAEs were mild or moderate; there were no deaths, serious adverse events, or clinically significant changes in vital signs, electrocardiograms, or laboratory parameters. There was no clinically significant immunogenicity, with only one participant with asthma considered positive for treatment-related immunogenicity for CDP7766. INTERPRETATION: This study, considered to be the only example of a dry powder anti-IL-13 fragment antibody being administered via inhalation, demonstrated that single and repeat doses were well tolerated over a period of up to 10 days in duration. Rapid and durable inhibition of fractional exhaled nitric oxide (FeNO) (secondary outcome) provided evidence of pharmacological engagement with the IL-13 target in the airways of participants diagnosed with mild asthma. These data, together with the numerical improvements observed for predose FEV1, justify further clinical evaluation of VR942 in a broader population of patients with asthma, and continue to support the development of an inhaled anti-IL-13 antibody fragment as a potential future treatment that is alternative to monoclonal antibodies delivered via the parenteral route. FUNDING: Study funding and funding for the medical writing and editorial support for preparation of the manuscript were split equally between the two study co-funders (Vectura Ltd and UCB Pharma).


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Interleukin-13/immunology , Administration, Inhalation , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Asthma/metabolism , Asthma/physiopathology , Exhalation , Female , Humans , Least-Squares Analysis , Male , Nitric Oxide/metabolism
3.
Ann Rheum Dis ; 76(11): 1837-1844, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28780512

ABSTRACT

OBJECTIVES: Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease associated with diffuse immune cell dysfunction. CD40-CD40 ligand (CD40L) interaction activates B cells, antigen-presenting cells and platelets. CD40L blockade might provide an innovative treatment for systemic autoimmune disorders. We investigated the safety and clinical activity of dapirolizumab pegol, a polyethylene glycol conjugated anti-CD40L Fab' fragment, in patients with SLE. METHODS: This 32-week randomised, double-blind, multicentre study (NCT01764594) evaluated repeated intravenous administration of dapirolizumab pegol in patients with SLE who were positive for/had history of antidouble stranded DNA/antinuclear antibodies and were on stable doses of immunomodulatory therapies (if applicable). Sixteen patients were randomised to 30 mg/kg dapirolizumab pegol followed by 15 mg/kg every 2 weeks for 10 weeks; eight patients received a matched placebo regimen. Randomisation was stratified by evidence of antiphospholipid antibodies. Patients were followed for 18 weeks after the final dose. RESULTS: No serious treatment-emergent adverse events, thromboembolic events or deaths occurred. Adverse events were mild or moderate, transient and resolved without intervention. One patient withdrew due to infection.Efficacy assessments were conducted only in patients with high disease activity at baseline. Five of 11 (46%) dapirolizumab pegol-treated patients achieved British Isles Lupus Assessment Group-based Composite Lupus Assessment response (vs 1/7; 14% placebo) and 5/12 (42%) evaluable for SLE Responder Index-4 responded by week 12 (vs 1/7; 14% placebo). Mechanism-related gene expression changes were observed in blood RNA samples. CONCLUSIONS: Dapirolizumab pegol could be an effective biological treatment for SLE. Further studies are required to address efficacy and safety. TRIAL REGISTRATION NUMBER: NCT01764594.


Subject(s)
Immunoglobulin Fab Fragments/administration & dosage , Immunologic Factors/administration & dosage , Lupus Erythematosus, Systemic/drug therapy , Polyethylene Glycols/administration & dosage , Transcriptome/drug effects , Administration, Intravenous , Adolescent , Adult , Aged , CD40 Ligand/drug effects , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , RNA/blood , Severity of Illness Index , Treatment Outcome , Young Adult
4.
MAbs ; 8(7): 1319-1335, 2016 10.
Article in English | MEDLINE | ID: mdl-27532598

ABSTRACT

An antibody format, termed Fab-dsFv, has been designed for clinical indications that require monovalent target binding in the absence of direct Fc receptor (FcR) binding while retaining substantial serum presence. The variable fragment (Fv) domain of a humanized albumin-binding antibody was fused to the C-termini of Fab constant domains, such that the VL and VH domains were individually connected to the Cκ and CH1 domains by peptide linkers, respectively. The anti-albumin Fv was selected for properties thought to be desirable to ensure a durable serum half-life mediated via FcRn. The Fv domain was further stabilized by an inter-domain disulfide bond. The bispecific format was shown to be thermodynamically and biophysically stable, and retained good affinity and efficacy to both antigens simultaneously. In in vivo studies, the serum half-life of Fab-dsFv, 2.6 d in mice and 7.9 d in cynomolgus monkeys, was equivalent to Fab'-PEG.


Subject(s)
Antibodies, Bispecific/blood , Immunoglobulin Fab Fragments , Immunoglobulin Variable Region , Serum Albumin/metabolism , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Half-Life , Humans , Immunoglobulin Fab Fragments/blood , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Variable Region/blood , Immunoglobulin Variable Region/chemistry , Mice , Serum Albumin/immunology
5.
Basic Clin Pharmacol Toxicol ; 106(3): 256-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20102365

ABSTRACT

Animal-to-man extrapolation and therapeutic dose prediction are illustrated with two molecules designed to treat epilepsy. Synaptic vesicle protein 2A (SV2A) is the primary molecular target for their anticonvulsive effect, but additional mechanisms may also contribute. Brivaracetam (BRV), currently in phase 3 of clinical development, was used as the benchmark compound. A pharmacokinetic/pharmacodynamic model was built in NONMEM, relating the brain tissue concentrations of BRV in mice and the proportion of animals protected against convulsions in the pharmacological model of audiogenic seizures. Brain concentrations were linked with ex vivo binding to predict brain SV2A occupancy. A physiologically based pharmacokinetic model was developed for predicting BRV concentrations in human plasma and brain tissue. Predicted plasma profiles were in good agreement with observations. Predicted human brain concentrations of BRV and the mouse ex vivo binding pharmacokinetic/pharmacodynamic model were used to extrapolate brain SV2A occupancy at the human therapeutic dose. Secondly, for another compound also exhibiting selective affinity for the same target, similar pharmacokinetic/pharmacodynamic models were built from audiogenic seizure mouse data. Various dosing regimens of the new compound were simulated in order to reach the same brain SV2A occupancy as for the reference compound. These estimations support early development. Assumptions and limitations of the approach are discussed.


Subject(s)
Anticonvulsants , Disease Models, Animal , Epilepsy/drug therapy , Pharmacology , Physiology , Pyrrolidinones , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Epilepsy/blood , Epilepsy/metabolism , Humans , Logistic Models , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Predictive Value of Tests , Protein Binding , Pyrrolidinones/administration & dosage , Pyrrolidinones/pharmacokinetics , Pyrrolidinones/therapeutic use , Tissue Distribution
6.
Invest New Drugs ; 27(2): 140-52, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18712503

ABSTRACT

The aim of this study was to assess the population pharmacokinetics (PopPK) of the novel oral anti-cancer agent E7820. Both a non-linear mixed effects modeling analysis and a non-compartmental analysis (NCA) were performed and results were compared. Data were obtained from a phase I dose escalation study in patients with malignant solid tumors or lymphomas. E7820 was administered daily for 28 days, followed by a washout period of 7 days prior to the start of subsequent cycles. A one compartment model with linear elimination from the central compartment was shown to give adequate fit, while absorption was described using a turnover model. Final population parameter estimates of basic PK parameters obtained with the PopPK method were (RSE): clearance, 6.24 L/h (7.1%), volume of distribution, 66.0 L (8.5%), mean transit time to the absorption compartment, 0.638 h (6.5%). The intake of food prior to dose administration slowed absorption (2.8-fold, RSE 13%) and increased relative bioavailability of E7820 by 36% (RSE 14%), although the effect on C (max) and AUC was not significant. Comparison with the NCA approach showed approximately equal PK parameter estimates and food effect measures, although specific advantages of PopPK included efficiency in use of data and more appropriate assessment of variability.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Indoles/pharmacokinetics , Models, Biological , Sulfonamides/pharmacokinetics , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Female , Food , Humans , Indoles/administration & dosage , Male , Middle Aged , Sulfonamides/administration & dosage
7.
J Pharmacokinet Pharmacodyn ; 31(5): 345-80, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15669772

ABSTRACT

The assessment of drug interactions for a simple turnover system when the basic pharmacodynamic response is governed by indirect mechanisms was explored. This report describes a diverse array of possible in vivo pharmacodynamic effects from a combination of two drugs acting by similar or different indirect mechanisms. Various conditions of pharmacodynamic drug combinations were explored mathematically and by simulation: (a) interactions of two drugs acting simultaneously either on the production (k(in)) or on the dissipation (k(out)) processes controlling the in vivo response by competitive (four cases) or non-competitive interaction (six cases); and (b) combinations of two drugs acting on separate k(in) and k(out) processes simultaneously (four cases). A range of different combinations of drug doses was used. Plasma concentration time profiles were generated according to monoexponential disposition. Pharmacodynamic response profiles were simulated using the above conditions and characterized by descriptors such as Area Between Effect (and Baseline) Curve (ABEC) values. The interaction of agents by competitive mechanisms produced net responses that were additive in nature. Response profiles for non-competitive interactions on the same process were both antagonistic (for two drugs with effects that oppose each other) and synergistic (for two drugs that produce the same response). On the other hand, response signatures for agents acting non-competitively on the production and dissipation factors by opposing mechanisms (e.g. inhibiting k(in) plus stimulating k(out)) showed dramatic changes in net effects and produced apparent drug synergy. Net indirect response profiles for joint use of two or more drugs measured by ABEC values may look "additive", "antagonistic", or "synergistic" depending on doses, their intrinsic potencies, the nature of interaction (competitive or non-competitive) as well as their mechanisms of action. These models may help explain changes in pharmacologic responses to two agents in a more rational and mechanistic fashion than older empirical methods.


Subject(s)
Computer Simulation , Drug Interactions/physiology , Models, Biological , Pharmaceutical Preparations/administration & dosage , Models, Chemical , Pharmaceutical Preparations/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...