Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Biopharm Drug Dispos ; 44(4): 344-347, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37345420

ABSTRACT

Today real word data (RWD) are playing a greater role in informing health care decisions. A physiologically based pharmacokinetic model (PBPK) and observed exposure-risk relationship predicted an increased bleeding risk induced by rivaroxaban (RXB) in patients with mild to moderate chronic kidney disease (CKD) taking concomitant medications that are combined Pgp-CYP3A inhibitors. In this commentary, we explore the potential use of RWD to assess the clinical consequence of this complex drug-drug interaction predicted from PBPK. This is a retrospective, case control, pilot study using a RWD dataset of 896,728 patients with mild to moderate chronic kidney disease and rivaroxaban use that was refined based upon combined Pgp-CYP3A inhibitor exposure and report of drug-induced bleeding (DIB). The odds ratio of patients with mild to moderate chronic kidney disease taking rivaroxaban with or without concurrent Pgp-CYP3A inhibitor use having a DIB was calculated. The odds ratio for DIB was 2.04 (CI95 1.82, 2.3; p < 0.001) suggesting an approximate doubling of bleeding risk which is consistent with the rivaroxaban exposure changes predicted by the published PBPK model and observed exposure-risk relationship. This exploratory analysis demonstrated the potential utility of RWD to assess model-based predictions as part of a drugs life cycle management.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Renal Insufficiency, Chronic , Humans , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Rivaroxaban/pharmacokinetics , Retrospective Studies , Pilot Projects , Drug Interactions , Models, Biological , Cytochrome P-450 CYP3A , Computer Simulation
3.
J Clin Pharmacol ; 60 Suppl 1: S160-S178, 2020 10.
Article in English | MEDLINE | ID: mdl-33205429

ABSTRACT

Since 2016, results from physiologically based pharmacokinetic (PBPK) analyses have been routinely found in the clinical pharmacology section of regulatory applications submitted to the US Food and Drug Administration (FDA). In 2018, the Food and Drug Administration's Office of Clinical Pharmacology published a commentary summarizing the application of PBPK modeling in the submissions it received between 2008 and 2017 and its impact on prescribing information. In this commentary, we provide an update on the application of PBPK modeling in submissions received between 2018 and 2019 and highlight a few notable examples.


Subject(s)
Computer Simulation , Drug Approval/statistics & numerical data , Models, Biological , Pharmacokinetics , Pharmacology, Clinical/statistics & numerical data , United States Food and Drug Administration/statistics & numerical data , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Decision Making , Drug Interactions , United States
5.
J Pharm Sci ; 108(1): 21-25, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30385284

ABSTRACT

This commentary provides an update on the status of physiologically based pharmacokinetic modeling and simulation at the U.S. Food and Drug Administration's Office of Clinical Pharmacology. Limitations and knowledge gaps in integration of physiologically based pharmacokinetic approach to inform regulatory decision making, as well as the importance of scientific engagement with drug developers who intend to use this approach, are highlighted.


Subject(s)
Pharmaceutical Preparations/standards , Pharmacology, Clinical/legislation & jurisprudence , Computer Simulation/legislation & jurisprudence , Humans , Models, Biological , Pharmacokinetics , United States , United States Food and Drug Administration/legislation & jurisprudence
6.
Drug Discov Today Technol ; 21-22: 67-73, 2016.
Article in English | MEDLINE | ID: mdl-27978990

ABSTRACT

This paper focuses on the role of clinical and translational pharmacology in the drug development and the regulatory process. Contemporary regulatory issues faced by FDA's Office of Clinical Pharmacology (OCP) in fulfilling its mission to advance the science of drug response and translate patient diversity into optimal drug therapy are discussed. Specifically current focus of the following key aspects of the drug development and regulatory science processes are discussed: the OCP vision and mission, two key OCP initiatives (i.e. guidance modernization, labeling and health communications), and translational and clinical pharmacology related regulatory science issues in (i.e. uncertainty, breakthrough therapies, individualization).


Subject(s)
Drug Discovery/legislation & jurisprudence , Legislation, Drug , Pharmacology, Clinical/legislation & jurisprudence , Translational Research, Biomedical/legislation & jurisprudence , Humans , United States , United States Food and Drug Administration
7.
J Clin Pharmacol ; 56 Suppl 7: S122-31, 2016 07.
Article in English | MEDLINE | ID: mdl-27385170

ABSTRACT

Transporters play an important role in drug absorption, disposition, and drug action. The evaluation of drug transporters requires a comprehensive understanding of transporter biology and pharmacology. Physiologically based pharmacokinetic (PBPK) models may offer an integrative platform to quantitatively evaluate the role of drug transporters and its interplay with other drug disposition processes such as passive drug diffusion and elimination by metabolizing enzymes. To date, PBPK modeling and simulations integrating drug transporters lag behind that for drug-metabolizing enzymes. In addition, predictive performance of PBPK has not been well established for predicting the role of drug transporters in the pharmacokinetics of a drug. To enhance overall predictive performance of transporter-based PBPK models, it is necessary to have a detailed understanding of transporter biology for proper representation in the models and to have a quantitative understanding of the contribution of transporters in the absorption and metabolism of a drug. This article summarizes PBPK-based submissions evaluating the role of drug transporters to the Office of Clinical Pharmacology of the US Food and Drug Administration.


Subject(s)
Membrane Transport Proteins/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , United States Food and Drug Administration/legislation & jurisprudence , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Forecasting , Humans , Pharmaceutical Preparations/administration & dosage , United States
8.
AAPS J ; 18(3): 573-7, 2016 05.
Article in English | MEDLINE | ID: mdl-26912182

ABSTRACT

Precision medicine promises to improve both the efficacy and safety of therapeutic products by better informing why some patients respond well to a drug, and some experience adverse reactions, while others do not. Pharmacogenomics is a key component of precision medicine and can be utilized to select optimal doses for patients, more precisely identify individuals who will respond to a treatment and avoid serious drug-related toxicities. Since pharmacogenomic biomarker information can help inform drug dosing, efficacy, and safety, pharmacogenomic data are critically reviewed by FDA staff to ensure effective use of pharmacogenomic strategies in drug development and appropriate incorporation into product labels. Pharmacogenomic information may be provided in drug or biological product labeling to inform health care providers about the impact of genotype on response to a drug through description of relevant genomic markers, functional effects of genomic variants, dosing recommendations based on genotype, and other applicable genomic information. The format and content of labeling for biologic drugs will generally follow that of small molecule drugs; however, there are notable differences in pharmacogenomic information that might be considered useful for biologic drugs in comparison to small molecule drugs. Furthermore, the rapid entry of biologic drugs for treatment of rare genetic diseases and molecularly defined subsets of common diseases will likely lead to increased use of pharmacogenomic information in biologic drug labels in the near future. In this review, we outline the general principles of therapeutic product labeling and discuss the utilization of pharmacogenomic information in biologic drug labels.


Subject(s)
Biological Products/standards , Drug Labeling/standards , Pharmacogenetics/standards , Precision Medicine/standards , United States Food and Drug Administration/standards , Biological Products/metabolism , Biomarkers/metabolism , Drug Labeling/methods , Humans , Pharmacogenetics/methods , Precision Medicine/methods , United States
10.
Clin Pharmacokinet ; 54(1): 117-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25260695

ABSTRACT

BACKGROUND AND OBJECTIVE: The US Food and Drug Administration (FDA) has seen a recent increase in the application of physiologically based pharmacokinetic (PBPK) modeling towards assessing the potential of drug-drug interactions (DDI) in clinically relevant scenarios. To continue our assessment of such approaches, we evaluated the predictive performance of PBPK modeling in predicting cytochrome P450 (CYP)-mediated DDI. METHODS: This evaluation was based on 15 substrate PBPK models submitted by nine sponsors between 2009 and 2013. For these 15 models, a total of 26 DDI studies (cases) with various CYP inhibitors were available. Sponsors developed the PBPK models, reportedly without considering clinical DDI data. Inhibitor models were either developed by sponsors or provided by PBPK software developers and applied with minimal or no modification. The metric for assessing predictive performance of the sponsors' PBPK approach was the R predicted/observed value (R predicted/observed = [predicted mean exposure ratio]/[observed mean exposure ratio], with the exposure ratio defined as [C max (maximum plasma concentration) or AUC (area under the plasma concentration-time curve) in the presence of CYP inhibition]/[C max or AUC in the absence of CYP inhibition]). RESULTS: In 81 % (21/26) and 77 % (20/26) of cases, respectively, the R predicted/observed values for AUC and C max ratios were within a pre-defined threshold of 1.25-fold of the observed data. For all cases, the R predicted/observed values for AUC and C max were within a 2-fold range. CONCLUSION: These results suggest that, based on the submissions to the FDA to date, there is a high degree of concordance between PBPK-predicted and observed effects of CYP inhibition, especially CYP3A-based, on the exposure of drug substrates.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Models, Biological , Pharmacokinetics , Computer Simulation , Drug Interactions , Humans , United States , United States Food and Drug Administration
11.
Clin Cancer Res ; 20(15): 3902-7, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24824310

ABSTRACT

On November 1, 2013, the U.S. Food and Drug Administration (FDA) approved obinutuzumab (GAZYVA; Genentech, Inc.), a CD20-directed cytolytic antibody, for use in combination with chlorambucil for the treatment of patients with previously untreated chronic lymphocytic leukemia (CLL). In stage 1 of the trial supporting approval, patients with previously untreated CD20-positive CLL were randomly allocated (2:2:1) to obinutuzumab + chlorambucil (GClb, n = 238), rituximab + chlorambucil (RClb, n = 233), or chlorambucil alone (Clb, n = 118). The primary endpoint was progression-free survival (PFS), and secondary endpoints included overall response rate (ORR). Only the comparison of GClb to Clb was relevant to this approval and is described herein. A clinically meaningful and statistically significant improvement in PFS with medians of 23.0 and 11.1 months was observed in the GClb and Clb arms, respectively (HR, 0.16; 95% CI, 0.11-0.24; P < 0.0001, log-rank test). The ORRs were 75.9% and 32.1% in the GClb and Clb arms, respectively, and the complete response rates were 27.8% and 0.9% in the GClb and Clb arms, respectively. The most common adverse reactions (≥10%) reported in the GClb arm were infusion reactions, neutropenia, thrombocytopenia, anemia, pyrexia, cough, and musculoskeletal disorders. Obinutuzumab was the first Breakthrough Therapy-designated drug to receive FDA approval.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Approval , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Chlorambucil/administration & dosage , Female , Follow-Up Studies , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Prognosis , Survival Rate , United States , United States Food and Drug Administration
12.
Oncologist ; 19(1): 94-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24309980

ABSTRACT

On October 26, 2012, the U.S. Food and Drug Administration (FDA) granted accelerated approval to omacetaxine mepesuccinate (Synribo; Teva Pharmaceuticals USA, Inc., North Wales, PA, http://www.tevausa.com) for the treatment of adult patients with chronic phase (CP) or accelerated phase (AP) chronic myeloid leukemia (CML) with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs). The approval was based on the FDA review of data from 111 patients with CML in CP or in AP who had received two or more prior TKIs, including imatinib. Major cytogenetic response was achieved in 18% of patients with CP, with a median response duration of 12.5 months. Major hematologic response was achieved in 14% of patients with AP, with a median response duration of 4.7 months. The FDA safety evaluation was based on submitted data from 163 patients with CP or AP CML who had received at least one dose of omacetaxine mepesuccinate. The safety evaluation was limited by the single-arm design of the clinical trials as conducted in a small number of previously treated patients. The most common (≥20%) adverse reactions of any grade in enrolled patients included thrombocytopenia, anemia, neutropenia, diarrhea, nausea, fatigue, asthenia, injection site reaction, pyrexia, and infection. The FDA concluded that omacetaxine mepesuccinate has shown activity and a favorable benefit-to-risk profile for the studied population of adult patients with CML (CP or AP) with resistance and/or intolerance to two or more TKIs. Further evidence of response durability to verify clinical benefit is pending.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Harringtonines/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Aged , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacology , Clinical Trials, Phase II as Topic , Drug Approval , Female , Harringtonines/adverse effects , Harringtonines/pharmacology , Homoharringtonine , Humans , Male , Middle Aged , United States , United States Food and Drug Administration
13.
Clin Cancer Res ; 18(12): 3212-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22544377

ABSTRACT

On November 16, 2011, the U.S. Food and Drug Administration (FDA) granted full approval to ruxolitinib, (Jakafi; Incyte Corp.), an inhibitor of the Janus kinases 1 and 2, for the treatment of patients with intermediate- or high-risk myelofibrosis, including primary myelofibrosis, postpolycythemia vera myelofibrosis, and postessential thrombocythemia myelofibrosis. This approval was based on the results of 2 large randomized phase III trials that enrolled patients with intermediate-2 or high-risk myelofibrosis and compared ruxolitinib with placebo (study 1) or best available therapy (study 2). The primary efficacy endpoint was the proportion of patients who experienced a reduction in spleen volume of ≥ 35% at 24 weeks (study 1) or 48 weeks (study 2). The key secondary endpoint in study 1 was the proportion of patients who experienced a ≥ 50% improvement from baseline in myelofibrosis total symptom score at 24 weeks. The results of these studies showed that a greater proportion of patients treated with ruxolitinib experienced a ≥ 35% reduction in spleen volume as compared with those treated with placebo (42% vs. 1%, P < 0.0001) or best available therapy (29% vs. 0%, P < 0.0001). A greater proportion of patients in study 1 experienced a ≥ 50% reduction in the myelofibrosis total symptom score during treatment with ruxolitinib than with placebo (46% vs. 5%, P < 0.0001). Ruxolitinib treatment was associated with an increased incidence of grades III and IV anemia, thrombocytopenia, and neutropenia. This is the first drug approved for myelofibrosis.


Subject(s)
Drug Approval , Primary Myelofibrosis/drug therapy , Pyrazoles/therapeutic use , Anemia/chemically induced , Clinical Trials, Phase III as Topic , Female , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Male , Neutropenia/chemically induced , Nitriles , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Pyrimidines , Randomized Controlled Trials as Topic , Thrombocytopenia/chemically induced , United States , United States Food and Drug Administration
14.
J Clin Pharmacol ; 52(1 Suppl): 91S-108S, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22232759

ABSTRACT

Chronic kidney disease, or renal impairment (RI) can increase plasma levels for drugs that are primarily renally cleared and for some drugs whose renal elimination is not a major pathway. We constructed physiologically based pharmacokinetic (PBPK) models for 3 nonrenally eliminated drugs (sildenafil, repaglinide, and telithromycin). These models integrate drug-dependent parameters derived from in vitro, in silico, and in vivo data, and system-dependent parameters that are independent of the test drugs. Plasma pharmacokinetic profiles of test drugs were simulated in subjects with severe RI and normal renal function, respectively. The simulated versus observed areas under the concentration versus time curve changes (AUCR, severe RI/normal) were comparable for sildenafil (2.2 vs 2.0) and telithromycin (1.6 vs 1.9). For repaglinide, the initial, simulated AUCR was lower than that observed (1.2 vs 3.0). The underestimation was corrected once the estimated changes in transporter activity were incorporated into the model. The simulated AUCR values were confirmed using a static, clearance concept model. The PBPK models were further used to evaluate the changes in pharmacokinetic profiles of sildenafil metabolite by RI and of telithromycin by RI and co-administration with ketoconazole. The simulations demonstrate the utility and challenges of the PBPK approach in evaluating the pharmacokinetics of nonrenally cleared drugs in subjects with RI.


Subject(s)
Carbamates/pharmacokinetics , Ketolides/pharmacokinetics , Kidney Diseases/metabolism , Models, Biological , Piperazines/pharmacokinetics , Piperidines/pharmacokinetics , Sulfones/pharmacokinetics , Area Under Curve , Carbamates/blood , Chronic Disease , Computer Simulation , Drug Interactions , Humans , Ketolides/blood , Piperazines/blood , Piperidines/blood , Purines/blood , Purines/pharmacokinetics , Sildenafil Citrate , Sulfones/blood
15.
Biopharm Drug Dispos ; 33(2): 99-110, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22270945

ABSTRACT

BACKGROUND: Rivaroxaban is an oral Factor Xa inhibitor. The primary objective of this communication was to quantitatively predict changes in rivaroxaban exposure when individuals with varying degrees of renal impairment are co-administered with another drug that is both a P-gp and a moderate CYP3A4 inhibitor. METHODS: A physiologically based pharmacokinetic (PBPK) model was developed to simulate rivaroxaban pharmacokinetics in young (20-45 years) or older (55-65 years) subjects with normal renal function, mild, moderate and severe renal impairment, with or without concomitant use of the combined P-gp and moderate CYP3A4 inhibitor, erythromycin. RESULTS: The simulations indicate that combined factors (i.e., renal impairment and the use of erythromycin) have a greater impact on rivaroxaban exposure than expected when the impact of these factors are considered individually. Compared with normal young subjects taking rivaroxaban, concurrent mild, moderate or severe renal impairment plus erythromycin resulted in 1.9-, 2.4- or 2.6-fold increase in exposure, respectively in young subjects; and 2.5-, 2.9- or 3.0-fold increase in exposure in older subjects. CONCLUSIONS: These simulations suggest that a drug-drug-disease interaction is possible, which may significantly increase rivaroxaban exposure and increase bleeding risk. These simulations render more mechanistic insights as to the possible outcomes and allow one to reach a decision to add cautionary language to the approved product labeling for rivaroxaban.


Subject(s)
Models, Biological , Morpholines/pharmacokinetics , Physiological Phenomena/physiology , Thiophenes/pharmacokinetics , Adult , Aged , Drug Interactions/physiology , Evaluation Studies as Topic , Forecasting , Humans , Middle Aged , Rivaroxaban , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...