Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Eur J Clin Pharmacol ; 79(1): 137-147, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36416938

ABSTRACT

PURPOSE: Antibodies that bind soluble targets such as cytokines belong to an important class of immunotherapies. Target levels can significantly accumulate after antibody administration due to formation of antibody-target complex, accompanied with suppression in free target which is often difficult to measure. Being a surrogate for pharmacodynamic activity, free target suppression is often predicted using in silico tools. The objective of this work is to illustrate the utility of modelling and to compare static versus dynamic models in the prediction of free target suppression. METHODS: Using binding principles, we have derived a static equation to predict free target suppression at steady state (FTSS). This equation operates with five input parameters and accounts for target accumulation over time. Its predictivity was compared to a dynamic model and to other existing metrics in literature via simulations and assumptions were illustrated. RESULTS: We demonstrated the utility of in silico tools in prediction of free target suppression using static and dynamic models and clarified the assumptions in key input parameters and their limitations. Predicted values using the FTSS equation correlate very well with those from the dynamic model at level > 20% target suppression, relevant for antagonistic antibodies. CONCLUSION: In silico tools are needed to predict target suppression by antibody drugs. Static or dynamic models can be used dependant on the scope, available data and undertaken assumptions. These tools can be used to guide discovery and development of antibodies and has the potential to reduce clinical failure.


Subject(s)
Antibodies, Monoclonal , Models, Biological , Humans , Antibodies, Monoclonal/pharmacology , Cytokines , Computer Simulation
2.
Eur J Drug Metab Pharmacokinet ; 46(2): 163-172, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33386550

ABSTRACT

The development of peptide therapeutics has increased enormously in recent decades. Many of the peptide drugs and antibody fragments that lack Fc backbone have a short half-life in circulation. In general, the half-life supports the design of the dosing regimen and frequency of administration, which are key aspects in the discovery of peptide drugs intended for long duration of action. Less frequent administration such as weekly or monthly can improve compliance and adherence to therapy. Serum albumin binding is a key approach to extend the half-life of peptide drugs. Despite the evidence of half-life prolongation of a variety of peptide drugs via albumin, quantitative prediction for humans is still a key question. Challenges in the measurement of albumin binding and in understanding the clearance mechanisms can limit quantitative prediction. We integrated pharmacokinetic concepts and albumin binding across species in a quantitative model to be used as a tool for prediction of half-life. Preliminary validation on a limited dataset indicated a good correlation between predicted and observed values. Further development of more quantitative models is warranted.


Subject(s)
Models, Biological , Serum Albumin/metabolism , Animals , Drug Administration Schedule , Drug Development , Half-Life , Humans , Peptides/pharmacokinetics
3.
Sci Rep ; 10(1): 16758, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028880

ABSTRACT

Unimolecular dual agonists for the glucagon-like peptide 1 receptor (GLP1R) and glucagon receptor (GCGR) are emerging as a potential new class of important therapeutics in type 2 diabetes (T2D). Reliable and quantitative assessments of in vivo occupancy on each receptor would improve the understanding of the efficacy of this class of drugs. In this study we investigated the target occupancy of the dual agonist SAR425899 at the GLP1R in pancreas and GCGR in liver by Positron Emission Tomography/Computed Tomography (PET/CT). Patients with T2D were examined by [68Ga]Ga-DO3A-Tuna-2 and [68Ga]Ga-DO3A-Exendin4 by PET, to assess the GCGR in liver and GLP1R in pancreas, respectively. Follow up PET examinations were performed after 17 (GCGR) and 20 (GLP-1R) days of treatment with SAR425899, to assess the occupancy at each receptor. Six out of 13 included patients prematurely discontinued the study due to adverse events. SAR425899 at a dose of 0.2 mg daily demonstrated an average GCGR occupancy of 11.2 ± 14.4% (SD) in N = 5 patients and a GLP1R occupancy of 49.9 ± 13.3%. Fasting Plasma Glucose levels (- 3.30 ± 1.14 mmol/L) and body weight (- 3.87 ± 0.87%) were lowered under treatment with SAR425899. In conclusion, SAR425899 demonstrated strong interactions at the GLP1R, but no clear occupancy at the GCGR. The study demonstrates that quantitative target engagement of dual agonists can be assessed by PET.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacology , Liver/drug effects , Pancreas/drug effects , Receptors, Glucagon/agonists , Aged , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Type 2/diagnostic imaging , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Hypoglycemic Agents/therapeutic use , Liver/diagnostic imaging , Liver/metabolism , Male , Middle Aged , Pancreas/diagnostic imaging , Pancreas/metabolism , Positron Emission Tomography Computed Tomography
4.
Diabetes Obes Metab ; 21(1): 120-128, 2019 01.
Article in English | MEDLINE | ID: mdl-30091218

ABSTRACT

AIMS: To evaluate the safety, pharmacokinetics and pharmacodynamics of SAR425899, a novel polypeptide, active as an agonist at both the glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCR), in healthy volunteers and in overweight/obese patients with type 2 diabetes (T2D). METHODS: Subcutaneous administrations of SAR425899 were tested in two randomized, placebo-controlled, double-blind clinical trials. In the first trial, healthy overweight volunteers (body mass index [BMI] 25-30 kg/m2 ; n = 32) received single-ascending doses (0.01-0.1 mg) of SAR425899 or placebo. In the second, a multiple-ascending-dose trial (NCT02411825), healthy normal- to overweight volunteers (BMI 20-30 kg/m2 ; n = 40) and overweight/obese patients with T2D (BMI 28-42 kg/m2 ; n = 36) received daily doses of SAR425899 or placebo over 21 or 28 days, respectively. RESULTS: The most frequently reported adverse events were gastrointestinal; gastrointestinal side effects were less pronounced in patients with T2D compared with healthy volunteers. SAR425899 significantly reduced levels of fasting plasma glucose (P < 0.05 vs. placebo) and glycated haemoglobin (P < 0.001 versus placebo) in patients with T2D. Additionally, SAR425899 led to reductions in body weight, with a maximal reduction of 5.32 kg in healthy volunteers and 5.46 kg in patients with T2D (P < 0.001 vs. placebo) at end of treatment. CONCLUSIONS: SAR425899 was well tolerated and led to favourable glycaemic effects in patients with T2D and weight reduction in both healthy volunteers and patients. Whether dual GLP-1R/GCR agonism represents a treatment method that is superior to pure GLP-1R agonists for obesity and diabetes treatment remains to be confirmed.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents , Receptors, Glucagon/agonists , Adolescent , Adult , Blood Glucose/analysis , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Type 2/complications , Female , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Lipids/blood , Male , Middle Aged , Obesity/complications , Placebos , Young Adult
5.
Curr Clin Pharmacol ; 13(1): 55-64, 2018.
Article in English | MEDLINE | ID: mdl-29773068

ABSTRACT

BACKGROUND: Blinatumomab is a bispecific T-cell engager (BiTE®) antibody construct targeting CD3ε on T cells and CD19 on B cells. We describe the relationship between pharmacokinetics (PK) of blinatumomab and pharmacodynamic (PD) changes in peripheral lymphocytes, serum cytokines, and tumor size in patients with non-Hodgkin lymphoma (NHL). METHODS: In a phase 1 study, 76 patients with relapsed/refractory NHL received blinatumomab by continuous intravenous infusion at various doses (0.5 to 90 µg/m2/day). PD changes were analyzed with respect to dose, blinatumomab concentration at steady state (Css), and cumulative area under the concentration-versus-time curve (AUCcum). RESULTS: B-cell depletion occurred within 48 hours at doses ≥5 µg/m2/day, followed first-order kinetics, and was blinatumomab exposure-dependent. Change in tumor size depended on systemic blinatumomab exposure and treatment duration and could be fitted to an Emax model, which predicted a 50% reduction in tumor size at AUCcum of ≥1,340 h×µg/L and Css of ≥1,830 pg/mL, corresponding to a blinatumomab dose of 47 µg/m2/day for 28 days. The magnitude of transient cytokine elevation, observed within 1-2 days of infusion start, was dose-dependent, with less pronounced elevation at low starting doses. CONCLUSION: B-lymphocyte depletion following blinatumomab infusion was exposure-dependent. Transient cytokine elevation increased with dose; it was less pronounced at low starting doses. Tumor response was a function of exposure, suggesting utility for the PK/PD relationship in dose selection for future studies, including NHL and other malignant settings.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents/administration & dosage , B-Lymphocytes/metabolism , Lymphoma, Non-Hodgkin/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Area Under Curve , Cytokines/blood , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Treatment Outcome , Young Adult
6.
Exp Hematol Oncol ; 6: 14, 2017.
Article in English | MEDLINE | ID: mdl-28533941

ABSTRACT

BACKGROUND: Blinatumomab has shown a remission rate of 69% in an exploratory single-arm, phase II dose-escalation study in adult patients with relapsed/refractory B-precursor acute lymphoblastic leukemia (ALL). We evaluated changes in laboratory parameters and immunopharmacodynamic markers in patients who received blinatumomab in the exploratory phase II study. METHODS: Data from 36 adults with relapsed/refractory ALL receiving blinatumomab as 4-week continuous IV infusions in various dose cohorts were analyzed for changes in liver enzymes, first-dose parameters, peripheral blood cell subpopulations, and cytokine/granzyme B release. Associations with clinical response were evaluated. RESULTS: Liver enzymes and inflammatory parameters transiently increased primarily during the first treatment week without clinical symptoms and reversed to baseline levels thereafter. B and T cells showed expected depletion and redistribution kinetics, respectively. Similarly, thrombocytes and T cells displayed an initial decline in cell counts, whereas neutrophils peaked during the first days after infusion start. T-cell redistribution coincided with upregulation of LFA-1 and CD69. Patients who responded to blinatumomab had more pronounced T-cell expansion, which was associated with proliferation of CD4+ and CD8+ T cells and memory subsets. Release of cytokines and granzyme B primarily occurred during the first week of cycle 1, except for IL-10, which was released in subsequent cycles. Blinatumomab step-dosing was associated with lower cytokine release and lower body temperature. CONCLUSIONS: In this study of relapsed/refractory ALL, blinatumomab-induced changes in laboratory parameters were transient and reversible. The evaluated PD markers demonstrated blinatumomab activity, and the analysis of cytokines supported the rationale for stepwise dosing. (ClinicalTrials.gov Identifier NCT01209286.).

7.
Blood ; 119(26): 6226-33, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22592608

ABSTRACT

T cell-engaging CD19/CD3-bispecific BiTE Ab blinatumomab has shown an 80% complete molecular response rate and prolonged leukemia-free survival in patients with minimal residual B-lineage acute lymphoblastic leukemia (MRD(+) B-ALL). Here, we report that lymphocytes in all patients of a phase 2 study responded to continuous infusion of blinatumomab in a strikingly similar fashion. After start of infusion, B-cell counts dropped to < 1 B cell/µL within an average of 2 days and remained essentially undetectable for the entire treatment period. By contrast, T-cell counts in all patients declined to a nadir within < 1 day and recovered to baseline within a few days. T cells then expanded and on average more than doubled over baseline within 2-3 weeks under continued infusion of blinatumomab. A significant percentage of reappearing CD8(+) and CD4(+) T cells newly expressed activation marker CD69. Shortly after start of infusion, a transient release of cytokines dominated by IL-10, IL-6, and IFN-γ was observed, which no longer occurred on start of a second treatment cycle. The response of lymphocytes in leukemic patients to continuous infusion of blinatumomab helps to better understand the mode of action of this and other globally T cell-engaging Abs. The trial is registered with www.clinicaltrials.gov identifier NCT00560794.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antigens, CD19/immunology , CD3 Complex/immunology , Immune System/drug effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/pharmacokinetics , Antilymphocyte Serum/administration & dosage , Antilymphocyte Serum/adverse effects , Antilymphocyte Serum/metabolism , Cytotoxicity, Immunologic/drug effects , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Immunotherapy/adverse effects , Immunotherapy/methods , Infusion Pumps , Lymphocyte Activation/drug effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome
8.
J Clin Invest ; 118(9): 3151-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688284

ABSTRACT

PKC isoforms tau, alpha, and beta play fundamental roles in the activation of T cells and other immune cell functions. Here we show that the PKC inhibitor AEB071 both abolishes the production of several cytokines by activated human T cells, keratinocytes, and macrophages in vitro and inhibits an acute allergic contact dermatitis response in rats. To translate these findings into humans, single and multiple ascending oral doses of AEB071 were administered to healthy volunteers and patients with psoriasis, respectively. AEB071 was well tolerated with no clinically relevant laboratory abnormalities. Ex vivo stimulation of lymphocytes from subjects exposed to single doses of AEB071 resulted in a dose-dependent inhibition of both lymphocyte proliferation and IL2 mRNA expression. Clinical severity of psoriasis was reduced up to 69% compared with baseline after 2 weeks of treatment, as measured by the Psoriasis Area Severity Index (PASI) score. The improvement in psoriasis patients was accompanied by histological improvement of skin lesions and may be partially explained by a substantial reduction of p40+ dermal cells, which are known to mediate psoriasis. These data suggest that AEB071 could be an effective novel treatment regimen for psoriasis and other autoimmune diseases, and that AEB071 warrants long-term studies to establish safety and efficacy.


Subject(s)
Lymphocytes/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Psoriasis/drug therapy , Animals , Dermatitis/drug therapy , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Hypersensitivity/drug therapy , Interleukin-2/biosynthesis , Lymphocyte Activation/drug effects , Placebos , Protein Isoforms , Protein Kinase Inhibitors/therapeutic use , Rats , Skin/drug effects
10.
J Clin Pharmacol ; 44(12): 1368-78, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15545307

ABSTRACT

R411 is a dual alpha4beta1-alpha4beta7 integrin antagonist under development for the treatment of chronic asthma. The objective of this study was to investigate the pharmacokinetics and safety of R411 and its active metabolite, RO0270608, in humans. A 3-part phase I trial was conducted in 132 healthy volunteers: (1) 12 subjects received 200 mg R411 as a single oral dose or 100 mg RO0270608 as an intravenous infusion in a 1-sequence crossover design; (2) 7 groups of 10 subjects received 1 of 7 single oral doses of R411 (10-1200 mg) in a parallel, placebo-controlled, ascending adaptive dose design; and (3) 5 groups of 10 subjects each received repeated oral qd doses of R411 (50-900 mg) for up to 3 weeks in a parallel, placebo-controlled, ascending adaptive dose design. The absolute bioavailability of RO0270608 (mean +/- standard deviation) after oral administration of R411 was 27% +/- 4%, and the terminal half-life was 7.33 +/- 2.29 hours. After IV infusion of RO0270608, total clearance (mean +/- standard deviation) was 19.4 +/- 7.1 L/h, and the volume of distribution was 93.1 +/- 36.1 L. After single ascending oral doses of R411, area under the concentration-time curve from 0 to infinity of active metabolite RO0270608 increased proportionally from 150 to 1200 mg (P > .05). Following repeated administration, the oral clearance was independent of time. No drug accumulation was observed, and no safety concerns were revealed up to a dose of 900 mg after up to 3 weeks of treatment.


Subject(s)
Anthraquinones/pharmacokinetics , Integrin alpha4beta1/antagonists & inhibitors , Integrins/antagonists & inhibitors , Administration, Oral , Adult , Anthraquinones/metabolism , Anthraquinones/therapeutic use , Asthma/drug therapy , Biological Availability , Chronic Disease , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Half-Life , Humans , Infusions, Intravenous , Integrin alpha4beta1/administration & dosage , Integrin alpha4beta1/therapeutic use , Integrins/administration & dosage , Integrins/therapeutic use , Male
12.
Drug Metab Dispos ; 30(7): 853-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065445

ABSTRACT

Ketamine is a widely used drug for its anesthetic and analgesic properties; it is also considered as a drug of abuse, as many cases of ketamine illegal consumption were reported. Ketamine is N-demethylated by liver microsomal cytochrome P450 into norketamine. The identification of the enzymes responsible for ketamine metabolism is of great importance in clinical practice. In the present study, we investigated the metabolism of ketamine in human liver microsomes at clinically relevant concentrations. Liver to plasma concentration ratio of ketamine was taken into consideration. Pooled human liver microsomes and human lymphoblast-expressed P450 isoforms were used. N-demethylation of ketamine was correlated with nifedipine oxidase activity (CYP3A4-specific marker reaction), and it was also correlated with S-mephenytoin N-demethylase activity (CYP2B6-specific marker reaction). Orphenadrine, a specific inhibitor to CYP2B6, and ketoconazole, a specific inhibitor to CYP3A4, inhibited the N-demethylation of ketamine in human liver microsomes. In human lymphoblast-expressed P450, the activities of CYP2B6 were higher than those of CYP3A4 and CYP2C9 at three concentrations of ketamine, 0.005, 0.05, and 0.5 mM. When these results were extrapolated using the average relative content of these P450 isoforms in human liver, CYP3A4 was the major enzyme involved in ketamine N-demethylation. The present study demonstrates that CYP3A4 is the principal enzyme responsible for ketamine N-demethylation in human liver microsomes and that CYP2B6 and CYP2C9 have a minor contribution to ketamine N-demethylation at therapeutic concentrations of the drug.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Ketamine/metabolism , Microsomes, Liver/enzymology , Oxidoreductases, N-Demethylating/metabolism , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/chemistry , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/chemistry , Dose-Response Relationship, Drug , Half-Life , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Ketamine/chemistry , Kinetics , Lymphocyte Activation , Lymphocytes/enzymology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/chemistry
13.
Eur J Clin Pharmacol ; 58(1): 37-40, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11956671

ABSTRACT

Ketamine is an anaesthetic agent extensively used in intensive care patients, and it has proved its efficacy in the management of burned patients. In these patients, alterations in serum protein binding occur that may have significant clinical implications. Scarce data were observed in the literature about the binding of ketamine to human plasma proteins, and no data about the binding of its active metabolites, norketamine (NK) and dehydronorketamine (DHNK) were found. In this study, protein binding of ketamine, NK and DHNK in human serum were determined using the ultrafiltration technique. The percentage of drug bound to serum proteins at 30 degrees C was found to be 69%, 60% and 50% for DHNK, ketamine and NK, respectively, while these percentages were 75%, 64% and 54% for DHNK, ketamine and NK respectively at 20 degrees C. The binding of ketamine and its metabolites was independent of drug concentration.


Subject(s)
Anesthetics, Dissociative/metabolism , Blood Proteins/metabolism , Ketamine/metabolism , Anesthetics, Dissociative/blood , Humans , Ketamine/analogs & derivatives , Ketamine/blood , Protein Binding , Ultrafiltration
SELECTION OF CITATIONS
SEARCH DETAIL
...