Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
CPT Pharmacometrics Syst Pharmacol ; 12(12): 1846-1858, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37002644

ABSTRACT

This study aimed to support dosing regimen selection for isatuximab as a single agent or in combination with dexamethasone for Japanese patients with relapsed/refractory multiple myeloma (RRMM). A joint model characterizing the dynamics of serum M-protein kinetics and its association with progression-free survival (PFS) was developed using data from 201 evaluable Japanese and non-Japanese patients with RRMM enrolled in two monotherapy phase I/II trials, where Japanese patients (n = 31) received isatuximab at 10 or 20 mg/kg once weekly (qw) for 4 weeks then every 2 weeks (q2w) in subsequent cycles (10 or 20 mg/kg qw-q2w). Among non-Japanese patients, 38 received isatuximab 20 mg/kg qw-q2w in combination with dexamethasone. Trial simulations were then performed to evaluate the effect of the isatuximab dosing regimens on both serum M-protein and PFS with and without dexamethasone. The model identified instantaneous changes in serum M-protein as the best on-treatment predictor for PFS. Trial simulations demonstrated that 20 mg/kg qw-q2w induced a greater decrease (30% vs. 22%) of serum M-protein at week 8 and prolonged median PFS by 2.4 weeks compared with 10 mg/kg qw-q2w. Although Japanese patients did not receive isatuximab plus dexamethasone in the phase I/II trial, simulations predicted that isatuximab 20 mg/kg qw-q2w plus dexamethasone would induce a greater decrease (67% vs. 43%) of serum M-protein and a prolonged median PFS by 7.2 weeks compared with isatuximab alone. Trial simulations support the approved isatuximab 20 mg/kg qw-q2w regimen when administered as a single agent and in combination with dexamethasone in Japanese patients.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Japan , Antibodies, Monoclonal, Humanized/therapeutic use , Dexamethasone , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
CPT Pharmacometrics Syst Pharmacol ; 11(6): 766-777, 2022 06.
Article in English | MEDLINE | ID: mdl-35355430

ABSTRACT

Isatuximab is an approved anti-CD38 monoclonal antibody with multiple antitumor modes of action. An exposure-response (E-R) analysis using data from patients with relapsed/refractory multiple myeloma (RRMM) enrolled in a phase Ib clinical study who received isatuximab at doses from 5 to 20 mg/kg weekly for 1 cycle (4 weeks) followed by every 2 weeks thereafter (qw/q2w) in combination with pomalidomide/dexamethasone (n = 44) was first used to determine the optimal dose/schedule for the phase III ICARIA-MM study. It was complemented by an E-R analysis from a second phase Ib study of patients who received isatuximab at doses from 3 to 10 mg/kg q2w or 10 or 20 mg/kg qw/q2w in combination with lenalidomide/dexamethasone (n = 52). Plasma trough concentration at week 4 (CT4W) was the best predictor for response, and the benefit of the initial 4-weekly administration was confirmed. Although the predicted overall response rate (ORR) was higher at 20 mg/kg vs. 10 mg/kg, the 95% confidence intervals were overlapping. Considering the high probability of success to reach the targeted ORR of greater than or equal to 60%, 10 mg/kg qw/q2w was selected. Results of the E-R analysis from the lenalidomide/dexamethasone study and published disease modeling using data from both phase Ib clinical studies reinforced 10 mg/kg qw/q2w as the optimal dose/schedule for the phase III ICARIA-MM study. E-R analysis showed that higher CT4W was associated with higher ORR. Developed models supported the phase III isatuximab dosing regimen selection/confirmation of 10 mg/kg qw/q2w for use in combination with pomalidomide/dexamethasone in patients with RRMM.


Subject(s)
Multiple Myeloma , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dexamethasone/therapeutic use , Humans , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Thalidomide/analogs & derivatives
3.
CPT Pharmacometrics Syst Pharmacol ; 10(8): 928-940, 2021 08.
Article in English | MEDLINE | ID: mdl-34185964

ABSTRACT

This analysis describes the pharmacokinetic/pharmacodynamic (PK/PD) modeling framework that supported selection of the isatuximab (anti-CD38 monoclonal antibody) dosing regimen alongside its early clinical development in patients with relapsed/refractory multiple myeloma (RRMM). The PK/PD mathematical model characterized the variations of patient serum M-protein concentrations, the primary marker of tumor burden in multiple myeloma (MM). Three separate PK/PD models were built sequentially as data became available from phase I clinical trials. The primary PK/PD analysis was initiated using monotherapy phase I study data (n = 122), followed by analysis of data collected from phase Ib combination studies with lenalidomide and dexamethasone (Rd, n = 40) and then with pomalidomide and dexamethasone (Pd, n = 31). Using the PK/PD model, abnormal "myeloma" protein (M-protein) profiles under different isatuximab dosing regimens were simulated. Overall, simulations revealed that regimens which included a loading period of four weekly administrations followed by administration every 2 weeks thereafter (QW4-Q2W), reduced M-protein levels more than a Q2W regimen without a loading period. For isatuximab monotherapy, a 20 mg/kg dose induced greater reduction in serum M-protein levels compared with doses equal or lower than 10 mg/kg. For isatuximab in combination with either Rd or Pd, simulations yielded no substantial benefit in terms of M-protein reduction between isatuximab 10 mg/kg and 20 mg/kg. These PK/PD analyses supported the use of isatuximab 10 mg/kg QW4-Q2W in combination with Pd in the phase III trial.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Models, Biological , Multiple Myeloma/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Myeloma Proteins/metabolism
4.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 649-658, 2020 11.
Article in English | MEDLINE | ID: mdl-33021075

ABSTRACT

Isatuximab, a monoclonal antibody (mAb) of immunoglobulin G (IgG) isotype, specifically targets the cluster of differentiation 38 antigen overexpressed in malignant plasma cells. Isatuximab is used to treat multiple myeloma (MM), characterized by the excessive production of abnormal "myeloma proteins" (M-proteins) that may interact with therapeutic IgG mAb on the neonatal Fc receptor (FcRn)-mediated recycling pathway. The clinical pharmacology profile of isatuximab was investigated by population pharmacokinetics (PKs) modeling in 476 patients with MM who received 1-20 mg/kg isatuximab either as single agent or in combination with pomalidomide-dexamethasone in 4 clinical trials. Isatuximab PKs were characterized by a two-compartment model with parallel time-varying linear clearance (CL) and nonlinear elimination. Due to a mechanism-based drug-disease interaction, patients secreting IgG M-protein exhibited a twofold lower drug exposure compared with patients with non-IgG MM. No dose adjustment was required based on MM immunoglobulin type because efficacy and safety profiles were comparable between IgG and non-IgG MM subpopulations. ß2-microglobulin, body weight, sex, drug material, and race have a limited effect on drug exposure and do not require any dose adjustment. A typical 50% decrease in linear CL from initial treatment to steady-state was predicted, and this decrease correlated with the best overall response rate and was slower for patients with IgG MM. These findings suggest that the time-dependent effect of isatuximab is likely mediated by a combined factor of both disease state evolution and the perturbation of the FcRn-mediated recycling pathway.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Multiple Myeloma/drug therapy , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Body Weight/drug effects , Case-Control Studies , Clinical Trials as Topic , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Drug Interactions , Female , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Male , Multiple Myeloma/immunology , Myeloma Proteins/drug effects , Neoplasm Recurrence, Local/drug therapy , Pharmaceutical Preparations , Receptors, Fc/drug effects , Receptors, Fc/metabolism , Recurrence , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Time Factors , beta 2-Microglobulin/drug effects , beta 2-Microglobulin/metabolism
5.
J Clin Pharmacol ; 57(7): 865-875, 2017 07.
Article in English | MEDLINE | ID: mdl-28138963

ABSTRACT

This work proposes a model-based approach to help select the phase 1 dosing regimen for the antibody-drug conjugate (ADC) SAR408701 leveraging the available data for 2 other ADCs of the same construct: SAR3419 and SAR566658. First, monkey and human pharmacokinetic (PK) data of SAR566658 and SAR3419 were used to establish the appropriate allometric approach to be applied to SAR408701 monkey PK data. Second, a population pharmacokinetics-pharmacodynamics (PK-PD) model was developed to describe tumor volume evolution following SAR408701 injection in mice. Third, allometric approaches identified for SAR566658 and SAR3419 were applied to SAR408701 monkey PK data to predict the human PK profile. Both SAR566658 and SAR3419 human and monkey PK were best described by a 2-compartment linear model. The relative difference was less than 10% between predicted and observed clearance using allometric exponents of 0.75 and 1, respectively. Tumor volume evolution following SAR408701 injection was best described by a full Simeoni model with a plasma concentration threshold of 4.6 µg/mL for eradication in mice. Both allometric exponents were used to predict SAR408701 PK in human from PK in monkey and to identify the potential effective dosing regimens. This translational strategy may be a valuable tool to design future clinical studies for ADCs, to support selection of the most appropriate dosing regimen, and to estimate the minimal dose required to assure antitumor activity, according to the schedule used.


Subject(s)
Immunoconjugates/administration & dosage , Models, Biological , Translational Research, Biomedical , Animals , Computer Simulation , Dose-Response Relationship, Drug , Haplorhini , Humans , Mice
6.
Lancet Infect Dis ; 15(12): 1409-19, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26342427

ABSTRACT

BACKGROUND: Artemisinin-based combination therapies (ACTs) are the recommended first-line treatment for uncomplicated Plasmodium falciparum malaria. Ferroquine is a new combination partner for fast-acting ACTs such as artesunate. We aimed to assess different doses of ferroquine in combination with artesunate against uncomplicated P falciparum malaria in a heterogeneous population in Africa. METHODS: We did a phase 2, multicentre, parallel-group, double-blind, randomised, dose-ranging non-inferiority trial at eight African hospitals (two in Gabon, three in Burkina Faso, one in Benin, and two in Kenya). We recruited patients presenting with acute P falciparum monoinfection (1000-200,000 parasites per µL), and a central body temperature of at least 37·5°C or history of fever in the past 24 h. We assessed patients in two sequential cohorts: cohort 1 contained adults (bodyweight >50 kg) and adolescents (aged ≥14 years, >30 kg), and cohort 2 contained children (aged 2-13 years, 15-30 kg). We randomly assigned patients (1:1:1:1) to receive artesunate 4 mg/kg per day plus ferroquine 2 mg/kg, 4 mg/kg, or 6 mg/kg, given double-blind once per day for 3 days, or ferroquine monotherapy 4 mg/kg per day given single-blind (ie, allocation was only masked from the patient) once per day for 3 days. We did 14 patient visits (screening, 3 treatment days and 48 h post-treatment surveillance, a visit on day 7, then one follow-up visit per week until day 63). The primary endpoint was non-inferiority of treatment in terms of PCR-corrected cure rate against a reference value of 90%, with a 10% non-inferiority margin, assessed in patients treated without major protocol deviations for parasitologically confirmed malaria. We assessed safety in all treated patients. This study is registered with ClinicalTrials.gov, number NCT00988507, and is closed. FINDINGS: Between Oct 16, 2009, and Sept 22, 2010, we randomly assigned 326 eligible patients to treatment groups, with last follow-up visit on Dec 1, 2010. 284 patients (87%) were available for per-protocol analyses. At day 28, PCR-confirmed cure was noted in 68 (97%, 95% CI 90-100) of 70 patients treated with ferroquine 2 mg/kg plus artesunate, 73 (99%, 93-100) of 74 with ferroquine 4 mg/kg plus artesunate, 71 (99%, 93-100) of 72 with ferroquine 6 mg/kg plus artesunate, and 54 (79%, 68-88) of 68 with ferroquine 4 mg/kg monotherapy. The three dose groups of ferroquine plus artesunate met the non-inferiority hypothesis. The most common adverse events were headache in cohort 1 (30 [19%] of 162 patients) and worsening malaria in cohort 2 (23 [14%] of 164 patients); occurrences were similar between treatment groups. INTERPRETATION: Ferroquine combined with artesunate was associated with high cure rates and was safe at all doses tested, and could be a promising new drug combination for the treatment of P falciparum malaria. Ferroquine could also partner other drugs to establish a new generation of antimalarial combinations, especially in regions that have developed resistance to ACTs. FUNDING: Sanofi.


Subject(s)
Aminoquinolines/therapeutic use , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Ferrous Compounds/therapeutic use , Malaria, Falciparum/drug therapy , Adolescent , Adult , Aged , Artesunate , Child , Child, Preschool , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Male , Metallocenes , Middle Aged , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Treatment Outcome
7.
Clin Chim Acta ; 377(1-2): 144-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17097625

ABSTRACT

BACKGROUND: Cytosine arabinoside (1-beta-D-arabinofuranosylcytosine;Ara-C) is the most important antimetabolite used for acute leukemia. We established Ara-C (0.003-1 micromol/l)-resistant NALM-6 leukemia cells, and attempted the characterization of their resistance. METHODS: The Ara-C-resistant cell lines were developed by stepwise increases in the drug. The mRNA expressions were analyzed by reverse transcription-polymerase chain reaction (RT-PCR). The uptake of Ara-C, deoxycytidine kinase (dCK) activity and cytidine deaminase (CDA) activity were measured using radioisotope methods. Cytotoxicity was evaluated using the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay. RESULTS: The mRNA expression of human equilibrative nucleoside transporter-1 (hENT-1), which is an uptake transporter of Ara-C, was initially decreased during the acquisition of resistance to Ara-C. The expression of dCK, an activation enzyme, and of CDA, an inactivation enzyme, was decreased and increased in the late phase, respectively. The cytotoxic effect of Ara-C on parental NALM-6 cells was ameliorated by hENT-1 inhibitors. There were no differences in the cytotoxic effect of other anticancer drugs, but there was similar resistance to nucleoside analogues via hENT-1 between the parental and resistant cells. CONCLUSIONS: Decreased hENT-1 expression and function is causatively responsible for the acquisition of Ara-C resistance and alterations in dCK and CDA contribute to the higher concentration range.


Subject(s)
Cytarabine/pharmacology , Drug Resistance, Neoplasm , Leukemia/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cytarabine/toxicity , Cytidine Deaminase/metabolism , Cytoprotection/drug effects , Deoxycytidine Kinase/metabolism , Dipyridamole/pharmacology , Gene Expression/drug effects , Humans , Leukemia/enzymology , RNA, Messenger/genetics , Thioinosine/analogs & derivatives , Thioinosine/pharmacology
8.
Toxicol Lett ; 166(2): 131-9, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16860949

ABSTRACT

Naringenin (NGEN), a flavonoid, has shown cytotoxicity in various human cancer cell lines and inhibitory effects on tumor growth. In this study, we investigated the apoptosis induced by NGEN via the activation of NF-kappaB and necrosis involving the loss of ATP in human promyeloleukemia HL-60 cells. Exposure to NGEN induced apoptosis dose-dependently up until 0.5mM, but not at 1mM as demonstrated by a quantitative analysis of nuclear morphological change and flow cytometric analysis. An extensive inhibitor for caspases, abolished the NGEN-induced apoptosis. The apoptosis-triggering concentration of NGEN was shown to markedly promote the activation of caspase-3, and slightly promote that of caspase-9, but had no effect on caspase-8. NGEN-induced apoptosis caused by induction of specific NF-kappaB-binding activity and involving the degradation of IkappaBalpha. Incubation with a high concentration of NGEN (1mM) reduced intracellular ATP levels, but no change was observed at lower concentrations. NGEN increased dose-dependently hyperpolarization of mitochondrial membrane potential. This result indicates a common pathway to apoptosis and necrosis by NGEN. One of the mechanisms by NGEN-induced apoptosis may relate to the activation of NF-kappaB that correlates with degradation of IkappaBalpha. Induction of necrosis by NGEN suggests causing by intracellular ATP depletion and mitochondria dysfunctions.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Flavanones/pharmacology , NF-kappa B/metabolism , Necrosis/drug therapy , Blotting, Western , Caspases/metabolism , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , HL-60 Cells , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Leukemia, Promyelocytic, Acute , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism
9.
Biol Pharm Bull ; 29(3): 472-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508148

ABSTRACT

The pineal gland and its main hormone, melatonin (MLT), are involved in a variety of physiological processes. MLT is a member of the indolamine family and has significant antioxidative activity. Acetaminophen (AA) is the most widely used medication in the world, both by prescription and over the counter. In large doses, AA is hepatotoxic causing oxidative stress and lipid peroxidation. Therefore, antioxidants have been used to protect against the toxicity of AA. Here, we examined in vitro and in vivo the protective effects of MLT against AA-induced toxicity in mice. MLT (100 microM) had a significant protective effect on the AA (7 mM)-induced loss of cell viability in mouse primary cultured hepatocytes as determined using the 3H-thymidine incorporation assay and MTT assay. The AA-induced generation of reactive oxygen species (ROS) peaked at 6 h and was followed by an increase in lipid peroxidation at 12 h in hepatocytes. MLT (0.1, 1, 10 or 100 microM) dose-dependently attenuated the increase in both production of ROS and lipid peroxidation by AA. Similarly, in vivo, AA (400, 600 or 800 mg/kg, intraperitoneally)-induced mortality and hepatotoxicity were significantly decreased by MLT (10 mg/kg, subcutaneously). Pretreatment with MLT had a greater protective effect on the hepatotoxicity of AA than post-treatment. However, MLT had no protective effect on the antipyretic effect or antinociception caused by AA. These results suggest that MLT is potentially useful for preventing AA-induced toxicity, but not the antipyretic effect or antinociception caused by AA.


Subject(s)
Acetaminophen/antagonists & inhibitors , Acetaminophen/toxicity , Analgesics, Non-Narcotic/antagonists & inhibitors , Analgesics, Non-Narcotic/toxicity , Antioxidants/pharmacology , Melatonin/pharmacology , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Body Temperature/drug effects , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Hepatocytes/drug effects , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Pain Measurement/drug effects , Reactive Oxygen Species/metabolism , Thymidine/metabolism
10.
Biol Pharm Bull ; 28(12): 2338-41, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16327178

ABSTRACT

We found that octylcaffeate, a semisynthetic caffeic acid derivative, strongly inhibited the growth of human histiolytic lymphoma U937 cells in a dose- and time-dependent manner via apoptosis. Octylcaffeate induced the fragmentation of DNA into multiples of 180 bp (an apoptotic DNA ladder) and condensation of chromatin, and increased the percentage of hypodiploid cells detected with a flow cytometer. DNA fragmentation induced by octylcaffeate was inhibited by pretreatment with Z-DEVD-FMK and Z-Asp-CH(2)D-CB, an inhibitor of caspase, clearly showing that the mode of cell death is apoptotic. These findings suggest that the cytotoxicity of octylcaffeate involves the induction of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caffeic Acids/pharmacology , Leukemia, Lymphoid/drug therapy , Caffeic Acids/antagonists & inhibitors , Caspase 3 , Caspases/metabolism , Caspases/toxicity , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , K562 Cells/drug effects , Time Factors , U937 Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL