UB Vice President for Research and Economic Development markup
Donald E Mager UB Profile
Pharmacy
Professor and Vice Chair
431 Kapoor Hall
Buffalo NY 14214
716-645-2903
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Brenda Cirincione, Jeffrey Edwards, Donald E Mager
Keywords: exenatide pharmacokinetics subcutaneous extended release transit compartments type 2 diabetes mellitus
Abstract: Exenatide is a glucagon-like peptide-1 receptor agonist with both immediate- and extended-release (ER) formulations that are approved for the treatment of type 2 diabetes mellitus. Long-term exposure from the ER formulation is achieved through slow peptide release from a degradable microsphere formulation. The goal of this analysis was to develop a pharmacokinetic model for the ER formulation following single and once-weekly multiple-dose administration. Pharmacokinetic data were collected from two clinical trials-one that evaluated single-dose administration of 2.5, 5, 7, and 10mg of ER exenatide and a second that included weekly administration of 0.8 and 2mg for 15weeks. A population pharmacokinetic model, describing 1586 exenatide concentrations from 64 patients, was developed in the nonlinear mixed-effects modeling software program NONMEM. Pharmacokinetics of the ER formulation was described by a two-compartment model with linear and nonlinear elimination. The complex absorption profile was quantified using three simultaneous processes: a first-order process characterizing a rapid initial release and two series of transit compartments to describe the second (3weeks postinjection) and third phases of drug release (7weeks postinjection). Estimation of the combined single- and multiple-dose data adequately described the rise to steady-state (8-10weeks) and decline to undetectable concentrations that occurred about 10weeks after treatment cessation. Thus, a population-based pharmacokinetic model was developed that provides a platform for future pharmacodynamic analyses with the ER formulation of exenatide.
PubMedId: 27896683
Journal Article: The AAPS journal, vol. 19, 2016
Authors: Charvi Nanavati, Donald E Mager
Keywords: Bortezomib pharmacodynamic modeling synergistic combination vorinostat
Abstract: To examine the combination of bortezomib and vorinostat in multiple myeloma cells (U266) and xenografts, and to assess the nature of their potential interactions with semi-mechanistic pharmacodynamic models and biomarkers. U266 proliferation was examined for a range of bortezomib and vorinostat exposure times and concentrations (alone and in combination). A non-competitive interaction model was used with interaction parameters that reflect the nature of drug interactions after simultaneous and sequential exposures. p21 and cleaved PARP were measured using immunoblotting to assess critical biomarker dynamics. For xenografts, data were extracted from literature and modeled with a PK/PD model with an interaction parameter. Estimated model parameters for simultaneous in vitro and xenograft treatments suggested additive drug effects. The sequence of bortezomib preincubation for 24 hours, followed by vorinostat for 24 hours, resulted in an estimated interaction term significantly less than 1, suggesting synergistic effects. p21 and cleaved PARP were also up-regulated the greatest in this sequence. Semi-mechanistic pharmacodynamic modeling suggests synergistic pharmacodynamic interactions for the sequential administration of bortezomib followed by vorinostat. Increased p21 and cleaved PARP expression can potentially explain mechanisms of their enhanced effects, which require further PK/PD systems analysis to suggest an optimal dosing regimen.
PubMedId: 28101809
Journal Article: Pharmaceutical research, vol. 34, 2016
Authors: Hong Li, Donald E Mager, Brenda M Sandmaier, Barry E Storer, Michael J Boeckh, Meagan J Bemer, Brian R Phillips, Linda J Risler, Jeannine S McCune
Keywords: Albumin Covariates Cyclosporine Hematopoietic cell transplantation Limited sampling schedule Mycophenolic acid Population pharmacokinetics Therapeutic drug monitoring
Abstract: A novel approach to personalizing postgrafting immunosuppression in hematopoietic cell transplantation (HCT) recipients is evaluating inosine monophosphate dehydrogenase (IMPDH) activity as a drug-specific biomarker of mycophenolic acid (MPA)-induced immunosuppression. This prospective study evaluated total MPA, unbound MPA, and total MPA glucuronide plasma concentrations and IMPDH activity in peripheral blood mononuclear cells (PMNCs) at 5 time points after the morning dose of oral mycophenolate mofetil(MMF) on day+21 in 56 nonmyeloablative HCT recipients. Substantial interpatient variability in pharmacokinetics and pharmacodynamics was observed and accurately characterized by the population pharmacokinetic-dynamic model. IMPDH activity decreased with increasing MPA plasma concentration, with maximum inhibition coinciding with maximum MPA concentration in most patients. The overall relationship between MPA concentration and IMPDH activity was described by a direct inhibitory maximum effect model with an IC50 of 3.23mg/L total MPA and 57.3ng/mL unbound MPA. The day+21 IMPDH area under the effect curve (AUEC) was associated with cytomegalovirus reactivation, nonrelapse mortality, and overall mortality. In conclusion, a pharmacokinetic-dynamic model was developed that relates plasma MPA concentrations with PMNC IMPDH activity after an MMF dose in HCT recipients. Future studies should validate this model and confirm that day+21 IMPDH AUEC is a predictive biomarker.
PubMedId: 24727337
Journal Article: Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, vol. 20, 2014
Authors: R Bruno, F Mentr??, S Tannenbaum, Y Wang, B Corrigan, D E Mager
Journal-article: Clinical Pharmacology & Therapeutics, vol. 95, pp. 581-582, 2014
Authors: Zhi-Jian Liu, Karin M Hoffmeister, Zhongbo Hu, Donald E Mager, Sihem Ait-Oudhia, Marlyse A Debrincat, Irina Pleines, Emma C Josefsson, Benjamin T Kile, Joseph Italiano, Haley Ramsey, Renata Grozovsky, Peter Veng-Pedersen, Chaitanya Chavda, Martha Sola-Visner
Abstract: The fetal/neonatal hematopoietic system must generate enough blood cells to meet the demands of rapid growth. This unique challenge might underlie the high incidence of thrombocytopenia among preterm neonates. In this study, neonatal platelet production and turnover were investigated in newborn mice. Based on a combination of blood volume expansion and increasing platelet counts, the platelet mass increased sevenfold during the first 2 weeks of murine life, a time during which thrombopoiesis shifted from liver to bone marrow. Studies applying in vivo biotinylation and mathematical modeling showed that newborn and adult mice had similar platelet production rates, but neonatal platelets survived 1 day longer in circulation. This prolonged lifespan fully accounted for the rise in platelet counts observed during the second week of murine postnatal life. A study of pro-apoptotic and anti-apoptotic Bcl-2 family proteins showed that neonatal platelets had higher levels of the anti-apoptotic protein Bcl-2 and were more resistant to apoptosis induced by the Bcl-2/Bcl-xL inhibitor ABT-737 than adult platelets. However, genetic ablation or pharmacologic inhibition of Bcl-2 alone did not shorten neonatal platelet survival or reduce platelet counts in newborn mice, indicating the existence of redundant or alternative mechanisms mediating the prolonged lifespan of neonatal platelets.
PubMedId: 24599546
Journal Article: Blood, vol. 123, 2014
Authors: Leonid Kagan, Jie Zhao, Donald E Mager
Abstract: To investigate the effect of dose level and anatomical site of injection on the pharmacokinetics of rituximab in mice, and to evaluate the utility of a pharmacokinetic model for describing interspecies differences in subcutaneous absorption between mice and rats. Rituximab serum concentrations were measured following intravenous and subcutaneous administration at the back and abdomen of mice. Several approaches were compared for scaling model parameters from estimated values in rats. The bioavailability of rituximab following subcutaneous injection was inversely related to the dose level and was dependent on the site of injection in mice. The overall rate of absorption was faster in mice as compared to rats. Subcutaneous absorption profiles were well described using the proposed structural model, in which the total receptor concentration, the affinity of rituximab to the receptor, and the degradation rate constant were assumed to be species independent. Subcutaneous absorption processes show similar trends in rats and mice, although the magnitude differs between species. A mathematical model that combines the absorption of free and bound antibody with presystemic degradation successfully captured rituximab pharmacokinetics in both species, and approaches for sharing and scaling parameters between species were identified.
PubMedId: 24852895
Journal Article: Pharmaceutical research, vol. 31, 2014
Authors: Krithika A Shetty, Matthew P Kosloski, Donald E Mager, Sathy V Balu-Iyer
Keywords: B-domain deleted factor VIII inhibitor development lipids next-generation FVIII pharmacodynamics pharmacokinetics phosphatidylinositol nanoparticles protein delivery simulations
Abstract: Factor VIII (FVIII) replacement therapy in hemophilia A (HA) is complicated by a short half-life and high incidence of inhibitory antibody response against the protein. Phosphatidylinositol (PI) containing lipidic nanoparticles have previously been shown to reduce the immunogenicity and prolong the half-life of full length FVIII. It has not been established whether this prolongation in half-life improves hemostatic efficacy and whether this approach could be extended to the B-domain deleted form of FVIII (BDD FVIII). In the current study, we evaluated the pharmacokinetics (PK), hemostatic efficacy, and immunogenicity of BDD FVIII associated with PI nanoparticles (PI-BDD FVIII) in HA mice. Comparative human PK was predicted using an "informed scaling" approach. PI-BDD FVIII showed an approximate 1.5-fold increase in terminal half-life compared with free BDD FVIII following i.v. bolus doses of 40 IU/kg. PI-BDD FVIII-treated animals retained hemostatic efficacy longer than the free FVIII-treated group in a tail vein transection model of hemostasis. PI association reduced the development of inhibitory and binding antibodies against BDD FVIII after a series of i.v. injections. The combined improvements in circulating half-life and hemostatic efficacy could significantly prolong the time above clinically established therapeutic thresholds of prophylactic FVIII replacement therapy in humans.
PubMedId: 24700333
Journal Article: Journal of pharmaceutical sciences, vol. 104, 2014
Authors: M R Birtwistle, D E Mager, J M Gallo
Abstract: Declining success rates coupled with increased costs is leading to an inevitable breaking point in the drug development pipeline. Can we avoid it by incorporating the vast mechanistic understanding of drug action? A recent review highlights this dilemma and proposes "quantitative logic gate" modeling as a solution.(1) The goal of this commentary is to contrast this approach with mechanistic biochemical network models, which, although alluded to by Kiruoac and Onsum, requires a closer analysis.
PubMedId: 24448020
Journal Article: CPT: pharmacometrics & systems pharmacology, vol. 2, 2014
Authors: Ting Chen, Leonid Kagan, Donald E Mager
Keywords: Type 2 diabetes exenatide mathematical modeling pharmacodynamics pharmacokinetics
Abstract: The purpose of this study is to investigate the effect of exenatide on glycemic control following two administration routes in a streptozotocin/nicotinamide (STZ/NA)-induced diabetic rat model, and to develop a pharmacodynamic model to better understand the disease progression and the action of exenatide in this experimental system. Two groups of STZ/NA-induced diabetic rats were treated for 2 weeks with 20 (g/kg/day) of exenatide, either by continuous subcutaneous (SC) infusion or two SC injections daily. Disease progression was associated with slower glucose utilization. Fasting blood glucose was significantly reduced by 30 mg/dL in both treatment groups at the end of 2 weeks. A subsequent intravenous glucose tolerance test (IVGTT) confirmed an improved glucose tolerance in both treatment groups; however, overall glycemic control was similar between groups, likely due to the relatively low and short-term drug exposure. A population indirect response model was successfully developed to simultaneously describe the STZ/NA-induced disease progression, responses to an IVGTT, and exenatide effects on these systemic challenges. The unified model includes a single set of parameters, and the cumulative area under the drug-receptor concentration curve was used as a unique driving force to account for systemic effects long after drug elimination.
PubMedId: 23897494
Journal Article: Journal of pharmaceutical sciences, vol. 102, 2013
Authors: Matthew P. Kosloski, Dipak S. Pisal, Donald E. Mager, Sathy V. Balu-Iyer
Journal-article: Journal of Pharmaceutical Sciences, vol. 102, pp. 2380-2394, 2013
Authors: X.-L. Jiang, H.-W. Shen, D. E. Mager, A.-M. Yu
Journal-article: Drug Metabolism and Disposition, vol. 41, pp. 975-986, 2013
Authors: H. Li, D.E. Mager, B.M. Sandmaier, D.G. Maloney, M.J. Bemer, J.S. McCune
Journal-article: The Journal of Clinical Pharmacology, vol. 53, pp. 393-402, 2013
Authors: Cara L McDermott, Brenda M Sandmaier, Barry Storer, Hong Li, Donald E Mager, Michael J Boeckh, Meagan J Bemer, Jennifer Knutson, Jeannine S McCune
Keywords: Bayesian Graft versus host disease Hematopoietic cell transplant Mycophenolic acid Pharmacokinetics
Abstract: We evaluated the pharmacodynamic relationships between mycophenolic acid (MPA), the active metabolite of mycophenolate mofetil (MMF), and outcomes in 308 patients after nonmyeloablative hematopoietic cell transplantation. Patients were conditioned with total body irradiationfludarabine, received grafts from HLA-matched related (n=132) or unrelated (n=176) donors, and received postgrafting immunosuppression with MMF and a calcineurin inhibitor. Total and unbound MPA pharmacokinetics were determined to day 25; maximum a posteriori Bayesian estimators were used to estimate total MPA concentration at steady state (Css). Rejection occurred in 9 patients, 8 of whom had a total MPA Css less than 3g/mL. In patients receiving a related donor graft, MPA Css was not associated with clinical outcomes. In patients receiving an unrelated donor graft, low total MPA Css was associated with increased grades III to IV acute graft-versus-host disease and increased nonrelapse mortality but not with day 28T cell chimerism, disease relapse, cytomegalovirus reactivation, or overall survival. We conclude that higher initial oral MMF doses and subsequent targeting of total MPA Css to greater than 2.96g/mL could lower grades III to IV acute graft-versus-host disease and nonrelapse mortality in patients receiving an unrelated donor graft.
PubMedId: 23660171
Journal Article: Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation, vol. 19, 2013
Authors: Scott A Van Wart, Susan E Shoaf, Suresh Mallikaarjun, Donald E Mager
Keywords: cirrhosis heart failure hyponatremia pharmacokinetics tolvaptan
Abstract: Tolvaptan is a selective V2 -receptor antagonist used to treat hypervolemic and euvolemic hyponatremia. A population pharmacokinetic (PK) analysis was performed for tolvaptan in NONMEM based upon data obtained from three trials conducted in 93 healthy subjects and six trials conducted in 628 congestive heart failure (CHF) patients or 24 hepatic cirrhosis patients receiving oral tolvaptan (5 to 240 mg). A two-compartment model with first-order absorption and elimination best described tolvaptan PK. Relative oral bioavailability was modeled relative to 100% for a 30 mg dose and ranged from 79.4% to 122%. Body weight and the impact of CHF or hepatic cirrhosis relative to healthy subjects were statistically significant (p < 0.001) predictors of both the apparent oral clearance (CL/F) and apparent central volume of distribution (Vc /F). The CL/F was reduced to 58.2% for New York Heart Association (NYHA) Class 1 or 2 CHF, 45.5% for NYHA Class 3 or 4 CHF, and 58.0% for hepatic cirrhosis relative to healthy subjects. Vc /F was reduced to 59.9% for NYHA Class 1 or 2 CHF and 51.3% for NYHA Class 3 or 4 CHF, and was 64.8% larger for severe hepatic cirrhosis (Child-Pugh score 10) relative to healthy subjects. A slight additional decrease in CL/F of 18.3% was also detected for patients with moderate hyponatremia (serum sodium of 115-130 mEq/l) after adjusting for CHF or cirrhosis (p < 0.001). This population PK model enabled assessment of tolvaptan PK with varying degrees of CHF and hepatic cirrhosis with fluid overload and may be used to explore PK-PD relationships with respect to fluid and electrolyte balance.
PubMedId: 23794414
Journal Article: Biopharmaceutics & drug disposition, vol. 34, 2013
Authors: Ying Hong, Jasper Dingemanse, Patricia Sidharta, Donald E Mager
Abstract: In this study, glucose and insulin concentration-time profiles in subjects with type 2 diabetes mellitus (T2DM) under meal tolerance test (MTT) and hyperglycemic clamp (HGC) conditions were co-modeled simultaneously. Blood glucose and insulin concentrations were obtained from 20 subjects enrolled in a double-blind, placebo-controlled, randomized, two-way crossover study. Patients were treated with palosuran or placebo twice daily for 4 weeks and then switched to the alternative treatment after a 4-week washout period. The MTT and HGC tests were performed 1 h after drug administration on days 28 and 29 of each treatment period. Population data analysis was performed using NONMEM. The HGC model incorporates insulin-dependent glucose clearance and glucose-induced insulin secretion. This model was extended for the MTT, in which glucose absorption was described using a transit compartment with a mean transit time of 62.5 min. The incretin effect (insulin secretion triggered by oral glucose intake) was also included, but palosuran did not influence insulin secretion or sensitivity. Glucose clearance was 0.164 L/min with intersubject and interoccasion variability of 9.57% and 31.8%. Insulin-dependent glucose clearance for the HGC was about 3-fold greater than for the MTT (0.0111 vs. 0.00425 L/min/[mU/L]). The maximal incretin effect was estimated to enhance insulin secretion 2-fold. The lack of palosuran effect coupled with a population-based analysis provided quantitative insights into the variability of glucose and insulin regulation in patients with T2DM following multiple glucose tolerance tests. Application of these models may also prove useful in antihyperglycemic drug development and assessing glucose-insulin homeostasis.
PubMedId: 23904152
Journal Article: The AAPS journal, vol. 15, 2013
Authors: J M Harrold, M Ramanathan, D E Mager
Abstract: Identification of novel targets is a critical first step in the drug discovery and development process. Most diseases such as cancer, metabolic disorders, and neurological disorders are complex, and their pathogenesis involves multiple genetic and environmental factors. Finding a viable drug target-drug combination with high potential for yielding clinical success within the efficacy-toxicity spectrum is extremely challenging. Many examples are now available in which network-based approaches show potential for the identification of novel targets and for the repositioning of established targets. The objective of this article is to highlight network approaches for identifying novel targets with greater chances of gaining approved drugs with maximal efficacy and minimal side effects. Further enhancement of these approaches may emerge from effectively integrating computational systems biology with pharmacodynamic systems analysis. Coupling genomics, proteomics, and metabolomics databases with systems pharmacology modeling may aid in the development of disease-specific networks that can be further used to build confidence in target identification.
PubMedId: 24025802
Journal Article: Clinical pharmacology and therapeutics, vol. 94, 2013
Authors: Scott A Van Wart, Susan E Shoaf, Suresh Mallikaarjun, Donald E Mager
Keywords: heart failure hydrochlorothiazide population pharmacokinetics
Abstract: Hydrochlorothiazide (HCTZ) is a thiazide diuretic used for the treatment of hypertension and edema associated with fluid overload conditions such as congestive heart failure (CHF). A population-based meta-analysis approach in NONMEM was used to develop a PK model to characterize the time-course of HCTZ concentrations in plasma and excretion into the urine for healthy subjects and CHF patients. Data from healthy subjects receiving 100mg of oral HCTZ were supplemented with additional plasma concentration and urinary excretion versus time data published in the literature following administration of oral HCTZ doses ranging from 10 to 500mg to healthy subjects or patients with renal failure, CHF or hypertension. A two-compartment model with first-order oral absorption, using a Weibull function, and first-order elimination best described HCTZ PK. Creatinine clearance (CLCR ) was a statistically significant predictor of renal clearance (CLR ). Non-renal clearance was estimated to be 2.44l/h, CLR was 18.3l/h and T1/2, was 1.6h and T1/2, was 14.8h for a typical individual with normal renal function (CLCR =120ml/min). However, CLR was reduced to 10.5, 5.47 and 2.70l/h in mild (CLCR =80ml/min), moderate (CLCR =50ml/min) and severe (CLCR =30ml/min) renal impairment, respectively. Model diagnostics helped to demonstrate that the population PK model reasonably predicts the rate of urinary HCTZ excretion over time using dosing history and estimated CLCR , allowing for the convenient assessment of PK-PD relationships for HCTZ when given alone or in combination with other agents used to treat fluid overload conditions.
PubMedId: 24123104
Journal Article: Biopharmaceutics & drug disposition, vol. 34, 2013
Authors: Leonid Kagan, Pavel Gershkovich, Kishor M Wasan, Donald E Mager
Abstract: To investigate the biodistribution of amphotericin B (AmB) in mice and rats following administration of liposomal AmB (AmBisome) using a physiologically-based pharmacokinetic (PBPK) modeling framework and to utilize this approach for predicting AmBisome pharmacokinetics in human tissues. AmB plasma and tissue concentration-time data, following single and multiple intravenous administration of nonliposomal and liposomal AmB to mice and rats, were extracted from literature. The whole-body PBPK model was constructed and incorporated nonliposomal and liposomal subcompartments. Various structural models for individual organs were evaluated. Allometric relationships were incorporated into the model to scale parameters based on species body weight. A non-Michaelis-Menten mechanism was included into the structure of the liver and spleen liposomal compartments to describe saturable uptake of particles by the reticuloendothelial system. The model successfully described plasma and tissue pharmacokinetics of AmB after administration of AmBisome to rats and mice. The dual PBPK model demonstrated good predictive performance by reasonably simulating AmB exposure in human tissues. This modeling framework can be potentially utilized for optimizing AmBisome therapy in humans and for investigating pathophysiological factors controlling AmB pharmacokinetics and pharmacodynamics.
PubMedId: 23793994
Journal Article: Pharmaceutical research, vol. 31, 2013
Authors: Scott A Van Wart, Susan E Shoaf, Suresh Mallikaarjun, Donald E Mager
Keywords: cirrhosis furosemide heart failure population pharmacokinetics
Abstract: Furosemide is a loop diuretic frequently used to treat fluid overload conditions such as hepatic cirrhosis and congestive heart failure (CHF). A population-based meta-analysis approach in NONMEM was used to develop a PK model characterizing the time-course of furosemide in plasma and excretion into the urine for healthy subjects and fluid overload patients. Furosemide PK data from healthy subjects receiving 80mg of oral furosemide were supplemented with additional individual and aggregate plasma concentration and urinary excretion versus time data from the literature after intravenous (i.v.) or oral furosemide administration (10-500mg) to healthy subjects or fluid overload patients. A three-compartment model with zero-order input following i.v. administration (or first-order absorption using a Weibull function after oral administration) and first-order elimination best described furosemide PK. A covariate analysis identified creatinine clearance (CL(CR)) as a statistically significant predictor of renal clearance (CL(R)), with a population mean CL(R) of 4.67, 3.11, 1.95 and 1.17l/h for a subject with normal renal function (CL(CR)=120ml/min) or mild (CL(CR)=80ml/min), moderate (CL(CR)=50ml/min) or severe (CLCR =30ml/min) renal impairment. Oral bioavailability was 59.1% and non-renal clearance was 2.02l/h. A PC-VPC and other model diagnostics demonstrated that the population PK model can reasonably predict the rate of urinary furosemide excretion over time using dosing history and commonly available demographic data, allowing for convenient assessment of PK-PD relationships for furosemide when given alone or in combination with other agents used to treat fluid overload conditions.
PubMedId: 24151207
Journal Article: Biopharmaceutics & drug disposition, vol. 35, 2013
Authors: Matthew P Kosloski, Dipak S Pisal, Donald E Mager, Sathy V Balu-Iyer
Keywords: factor VIII hemophilia A lipid particles nonlinear pharmacokinetics phosphatidylinositol
Abstract: Factor VIII (FVIII) is an important cofactor in the blood coagulation cascade and its deficiency or dysfunction causes hemophilia A (HA), a bleeding disorder. Replacement with recombinant FVIII is limited by a short half-life and the development of inhibitory antibodies. A phosphatidylinositol (PI) containing lipid nanoparticle was developed that, when associated with FVIII, reduces immunogenicity and prolongs circulation of the therapeutic protein in HA mice. A multiple dose level pharmacokinetic (PK) study of human free FVIII and its FVIII-PI complex over a clinically relevant range of doses (20, 40 and 200IU/kg) was conducted in HA mice to investigate linearity of the PK and to determine if the reduced catabolism of FVIII following association with PI particles, previously only observed in the terminal phase following 400IU/kg, could be extendable over a range of doses. The findings suggest that the disposition of FVIII is best characterized by a two-compartment model with saturable Michaelis-Menten elimination. Spontaneous complexation of FVIII with PI particles significantly increases plasma survival of the protein at 20 and 40IU/kg doses. Human simulations at 40IU/kg project an increase in the terminal half-life and the time to reach a minimum therapeutic threshold of 0.01IU/ml of 5.4h and 40h, respectively, compared with free FVIII. Formulation with PI containing lipid particles may represent a viable delivery strategy for improving FVIII therapy.
PubMedId: 24259187
Journal Article: Biopharmaceutics & drug disposition, vol. 35, 2013
Authors: Hong Li, Donald E. Mager, Meagan J. Bemer, David H. Salinger, Paolo Vicini, Brenda M. Sandmaier, Richard Nash, Jeannine S. McCune
Journal-article: The Journal of Clinical Pharmacology, vol. 52, pp. 1654-1664, 2012
Authors: Anshu Marathe, Suhasini Iyer, Zhihua Julia Qiu, Jennifer Visich, Donald E. Mager
Journal-article: Pharmaceutical Research, vol. 29, pp. 3180-3187, 2012
Authors: V L Chudasama, F Schaedeli Stark, J M Harrold, J Tibbitts, S R Girish, M Gupta, N Frey, D E Mager
Journal-article: Clinical Pharmacology & Therapeutics, vol. 92, pp. 520-527, 2012
Authors: Donald E. Mager, Vidhi Mody, Chao Xu, Alan Forrest, Wojciech G. Lesniak, Shraddha S. Nigavekar, Muhammed T. Kariapper, Leah Minc, Mohamed K. Khan, Lajos P. Balogh
Journal-article: Pharmaceutical Research, vol. 29, pp. 2534-2542, 2012
Authors: Xiaochen Zhao, Swarajya Lakshmi Vattem Venkata, Ruin Moaddel, Dave A. Luckenbaugh, Nancy E. Brutsche, Lobna Ibrahim, Carlos A. Zarate Jr, Donald E. Mager, Irving W. Wainer
Journal-article: British Journal of Clinical Pharmacology, vol. 74, pp. 304-314, 2012
Authors: Donald E. Mager
Book-chapter: Development of Antibody-Based Therapeutics, pp. 285-302, 2012
Authors: John M Harrold, Robert M Straubinger, Donald E Mager
Abstract: Combination chemotherapy represents the standard-of-care for non-Hodgkin lymphoma. However, the development of new therapeutic regimens is empirical and this approach cannot be used prospectively to identify novel or optimal drug combinations. Quantitative system pharmacodynamic models could promote the discovery and development of combination regimens based upon first principles. In this study, we developed a mathematical model that integrates temporal patterns of drug exposure, receptor occupancy, and signal transduction to predict the effects of the CD20 agonist rituximab in combination with rhApo2L/TNF-related apoptosis inducing ligand or fenretinide, a cytotoxic retinoid, upon growth kinetics in non-Hodgkin lymphoma xenografts. The model recapitulated major regulatory mechanisms, including target-mediated disposition of rituximab, modulation of proapoptotic intracellular signaling induced by CD20 occupancy, and the relative efficacy of death receptor isoforms. The multiscale model coupled tumor responses to individual anticancer agents with their mechanisms of action in vivo, and the changes in Bcl-xL and Fas induced by CD20 occupancy were linked to explain the synergy of these drugs. Tumor growth profiles predicted by the model agreed with cell and xenograft data, capturing the apparent pharmacologic synergy of these agents with fidelity. Together, our findings provide a mechanism-based platform for exploring new regimens with CD20 agonists.
PubMedId: 22350416
Journal Article: Cancer research, vol. 72, 2012
Authors: Ting Chen, Donald E. Mager, Leonid Kagan
Journal-article: Pharmaceutical Research, vol. 30, pp. 751-760, 2012
Authors: S. Ait-Oudhia, R. M. Straubinger, D. E. Mager
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 344, pp. 103-112, 2012
Authors: Sihem Ait-Oudhia, Robert M. Straubinger, Donald E. Mager
Journal-article: Pharmaceutical Research, vol. 29, pp. 2833-2844, 2012
Authors: Helen Kastrissios, Joseph R. Walker, Timothy J. Carrothers, Smita Kshirsagar, Tatiana Khariton, Bahru Habtemariam, Donald E. Mager, Shashank Rohatagi
Journal-article: The Journal of Clinical Pharmacology, vol. 52, pp. 404-415, 2012
Authors: Melanie A Felmlee, Marilyn E Morris, Donald E Mager
Abstract: Pharmacodynamic modeling is based on a quantitative integration of pharmacokinetics, pharmacological systems, and (patho-) physiological processes for understanding the intensity and time-course of drug effects on the body. Application of such models to the analysis of meaningful experimental data allows for the quantification and prediction of drug-system interactions for both therapeutic and adverse drug responses. In this chapter, commonly used mechanistic pharmacodynamic models are presented with respect to their important features, operable equations, and signature profiles. In addition, literature examples showcasing the utility of these models to adverse drug events are highlighted. Common model types that are covered include simple direct effects, biophase distribution, indirect effects, signal transduction, and irreversible effects.
PubMedId: 23007443
Journal Article: Methods in molecular biology (Clifton, N.J.), vol. 929, 2012
Authors: Ravi Iyengar, Shan Zhao, Seung-Wook Chung, Donald E Mager, James M Gallo
Abstract: The emerging discipline of systems pharmacology aims to combine analysis and computational modeling of cellular regulatory networks with quantitative pharmacology approaches to drive the drug discovery processes, predict rare adverse events, and catalyze the practice of personalized precision medicine. Here, we introduce the concept of enhanced pharmacodynamic (ePD) models, which synergistically combine the desirable features of systems biology and current PD models within the framework of ordinary or partial differential equations. ePD models that analyze regulatory networks involved in drug action can account for a drug's multiple targets and for the effects of genomic, epigenomic, and posttranslational changes on the drug efficacy. This new knowledge can drive drug discovery and shape precision medicine.
PubMedId: 22440734
Journal Article: Science translational medicine, vol. 4, 2012
Authors: Leonid Kagan, Donald E Mager
Abstract: Absorption of monoclonal antibodies (mAbs) after s.c. injection results from the interplay among several kinetic processes. The aims of this study were to investigate the absorption mechanisms of rituximab in rats by using slow s.c. infusion and coadministration with nonspecific IgG or hyaluronidase, and to evaluate the predictive performance of the pharmacokinetic model previously developed to describe the nonlinear absorption behavior of mAbs. Rituximab serum concentrations were measured after s.c. coadministration with nonspecific IgG and hyaluronidase to rats. Several dose levels and different injection sites were evaluated. For the back site, 6.5- and 2.6-fold decreases in the area under the concentration-time curve were obtained after coadministration with IgG for 1 and 10 mg/kg doses compared with administration of rituximab alone. For the abdomen, only a minor reduction in concentrations was observed. Hyaluronidase increased the rate of s.c. absorption and the bioavailability (1.9- and 1.6-fold for the back and the abdomen injection of 10 mg/kg). Our previously established pharmacokinetic model provided excellent predictions of the effect of nonspecific IgG on rituximab absorption. In conclusion, the magnitude of the effect of absorption modifiers is dependent on the site of injection and the dose level of rituximab. Pharmacokinetic profiles further support the hypothesis that neonatal Fc receptor-mediated transport is a major determinant of s.c. absorption of mAbs.
PubMedId: 23129212
Journal Article: Drug metabolism and disposition: the biological fate of chemicals, vol. 41, 2012
Authors: Leonid Kagan, Michael R. Turner, Sathy V. Balu-Iyer, Donald E. Mager
Journal-article: Pharmaceutical Research, vol. 29, pp. 490-499, 2011
Authors: Dhananjay D. Marathe, Anshu Marathe, Donald E. Mager
Journal-article: Biopharmaceutics & Drug Disposition, vol. 32, pp. 471-481, 2011
Authors: Leonid Kagan, Pavel Gershkovich, Kishor M. Wasan, Donald E. Mager
Journal-article: The AAPS Journal, vol. 13, pp. 255-264, 2011
Authors: Anson K. Abraham, Tristan S. Maurer, Amit S. Kalgutkar, Xiang Gao, Mei Li, David R. Healy, Donna N. Petersen, David A. Griffith, Donald E. Mager
Journal-article: The AAPS Journal, vol. 13, pp. 265-273, 2011
Authors: David Z. D'Argenio, Donald E. Mager
Journal-article: The Journal of Clinical Pharmacology, vol. 50, pp. 56S-57S, 2010
Authors: L. Kagan, T. Dreifinger, D. E. Mager, A. Hoffman
Journal-article: Drug Metabolism and Disposition, vol. 38, pp. 1560-1566, 2010
Authors: Pascal Houz??, Donald E. Mager, Patricia Ris??de, Fr??d??ric J. Baud
Journal-article: Toxicological Sciences, vol. 116, pp. 660-672, 2010
Authors: S. Dhindsa, M. G. Miller, C. L. McWhirter, D. E. Mager, H. Ghanim, A. Chaudhuri, P. Dandona
Abstract: To determine the prevalence of subnormal testosterone concentrations in patients with obesity and with type 2 diabetes in a primary care clinic population. Free testosterone concentrations of 1,849 men (1,451 nondiabetic and 398 diabetic) in the Hypogonadism In Males (HIM) study were analyzed. The HIM study was a U.S.-based cross-sectional study designed to define the prevalence of hypogonadism in men aged >45 years. Free testosterone was measured by equilibrium dialysis. The prevalence of subnormal free testosterone concentrations in lean, overweight, and obese nondiabetic men was 26% (n = 275), 29% (n = 687), and 40% (n = 489), respectively (P < 0.001 for trend), and 44% (n = 36), 44% (n = 135), and 50% (n = 227), respectively, in diabetic men (P = 0.46 for trend within group and P < 0.05 compared with nondiabetic men). The mean free testosterone concentration of diabetic men was significantly lower than that of nondiabetic men. Free testosterone concentrations were negatively and significantly (P < 0.001) related to age (r = -0.37), BMI (r = -0.18), and sex hormone-binding globulin (r = -0.11) in multiple regression analysis. The average decline of free testosterone concentrations was 7.8 pg/ml per decade in nondiabetic men and 8.4 pg/ml per decade in diabetic men. Forty percent of obese nondiabetic men and 50% of obese diabetic men aged >or=45 years have subnormal free testosterone concentrations. In view of its high prevalence, obesity is probably the condition most frequently associated with subnormal free testosterone concentrations in males. The concomitant presence of diabetes is associated with an additional increase in the prevalence of subnormal free testosterone concentrations.
PubMedId: 20200299
Journal-article: Diabetes Care, vol. 33, pp. 1186-1192, 2010
Authors: Donald E. Mager, Murali Ramanathan
Reference-entry: Pharmaceutical Sciences Encyclopedia, 2010
Authors: Anshu Marathe, Scott Van Wart, Donald E. Mager
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 38, pp. 223-235, 2010
Authors: A. K. Abraham, L. Kagan, S. Kumar, D. E. Mager
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 334, pp. 327-332, 2010
Authors: Leonid Kagan, Anson K. Abraham, John M. Harrold, Donald E. Mager
Journal-article: Pharmaceutical Research, vol. 27, pp. 920-932, 2010
Authors: Matthew P. Kosloski, Aaron Peng, Prashant R. Varma, Anas M. Fathallah, Razvan D. Miclea, Donald E. Mager, Sathy V. Balu-iyer
Journal-article: Drug Delivery, vol. 18, pp. 246-254, 2010
Authors: Michael E. Goldberg, Marc C. Torjman, Robert J. Schwartzman, Donald E. Mager, Irving W. Wainer
Journal-article: Chirality, vol. 23, pp. 138-143, 2010
Authors: Chao Xu, Donald E Mager
Abstract: Quantitative structure-pharmacokinetic relationships (QSPKR) modeling is a valuable tool in drug discovery and development. It can provide insights into the molecular determinants of processes governing the time course of drug exposure and response. Empirical and mechanism-based QSPKR models are discussed, including specific examples for oral absorption, nonspecific protein binding, volume of distribution, total metabolic stability and specific interactions with drug metabolizing enzymes. Emphasis is placed on state-of-the-art techniques, including new approaches for the direct simulation of concentration-time profiles from molecular descriptors (temporal QSPKR). Reviewing the application of current QSPKR modeling techniques will place these methods in context and highlight their respective advantages and limitations, as well as opportunities for further refinement. The expansion of readily available molecular descriptors and advanced algorithms has improved empirical models and enabled the development of robust models for non-congeneric series. Empirical models focus on point estimates of global PK processes and physiologically-based models may be more desirable than data-driven methods. Further integration of relevant biological and pharmacological mechanisms will improve the ability to predict the full time course of drug concentration and effect profiles for diverse compounds and experimental conditions.
PubMedId: 21105864
Journal Article: Expert opinion on drug metabolism & toxicology, vol. 7, 2010
Authors: Xiaoyu Yan, Donald E. Mager, Wojciech Krzyzanski
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 37, pp. 25-47, 2009
Authors: Anson K Abraham, Donald E Mager, Xiang Gao, Mei Li, David R Healy, Tristan S Maurer
Abstract: The purpose of this study was to develop a mechanism-based pharmacokinetic/pharmacodynamic model that describes the regulation of the parathyroid hormone (PTH)-Ca(2+) system in rats and humans. Temporal concentration data for endogenous PTH and Ca(2+) were extracted from literature for rats (normal adult males) and humans. In addition, exogenous PTH was administered subcutaneously to male Sprague-Dawley rats with jugular vein catheters, and plasma concentrations were measured over time. A mathematical model was developed and fitted simultaneously to endogenous PTH, Ca(2+), and exogenous PTH concentrations in rats. Ca(2+) concentrations were described using a turnover model, with its depletion being induced by a chelating agent, and PTH concentrations were characterized using a precursor-dependent indirect response model. The same structural model was used for fitting data obtained in humans. PTH stimulation was driven by occupancy of the Ca(2+) sensing receptor, and lowering of physiological Ca(2+) concentrations increased PTH secretion, with PTH profiles being adequately described by the model. PTH stimulatory capacity was baseline-dependent in rats [S(max_rats) = 34.8 x PTH(0)] and humans [S(max_humans) = 392/PTH(0)]. Modeling results suggest that normal rats are twice as sensitive to Ca(2+)-induced PTH stimulation compared with humans. In conclusion, the developed model adequately characterizes the PTH-Ca(2+) regulation across species and may be useful in the development of therapeutic drugs targeting this system.
PubMedId: 19386792
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 330, 2009
Authors: Donald E Mager, Sukyung Woo, William J Jusko
Abstract: An important feature of mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) models is the identification of drug- and system-specific factors that determine the intensity and time-course of pharmacological effects. This provides an opportunity to integrate information obtained from in vitro bioassays and preclinical pharmacological studies in animals to anticipate the clinical and adverse responses to drugs in humans. The fact that contemporary PK/PD modeling continues to evolve and seeks to emulate systems level properties should provide enhanced capabilities to scale-up pharmacodynamic data. Critical steps in drug discovery and development, such as lead compound and first in human dose selection, may become more efficient with the implementation and further refinement of translational PK/PD modeling. In this review, we highlight fundamental principles in pharmacodynamics and the basic expectations for in vitro bioassays and traditional allometric scaling in PK/PD modeling. Discussion of PK/PD modeling efforts for recombinant human erythropoietin is also included as a case study showing the potential for advanced systems analysis to facilitate extrapolations and improve understanding of inter-species differences in drug responses.
PubMedId: 19252333
Journal Article: Drug metabolism and pharmacokinetics, vol. 24, 2009
Authors: Chao Xu, Thomas M. Barchet, Donald E. Mager
Journal-article: Cancer Chemotherapy and Pharmacology, vol. 65, pp. 325-333, 2009
Authors: Jun Yang, Donald E Mager, Robert M Straubinger
Abstract: Semi-mechanistic pharmacodynamic (PD) models that capture tumor responses to anticancer agents with fidelity can provide valuable insights that could aid in the optimization of dosing regimens and the development of drug delivery strategies. This study evaluated the utility and potential interchangeability of two transduction-type PD models: a cell distribution model (CDM) and a signal distribution model (SDM). The evaluation was performed by simulating dense and sparse tumor response data with one model and analyzing it using the other. Performance was scored by visual inspection and precision of parameter estimation. Capture of tumor response data was also evaluated for a liposomal formulation of paclitaxel in the paclitaxel-resistant murine Colon-26 model. A suitable PK model was developed by simultaneous fitting of literature data for paclitaxel formulations in mice. Analysis of the simulated tumor response data revealed that the SDM was more flexible in describing delayed drug effects upon tumor volume progression. Dense and sparse data simulated using the CDM were fit very well by the SDM, but under some conditions, data simulated using the SDM were fitted poorly by the CDM. Although both models described the dose-dependent therapeutic responses of Colon-26 tumors, the fit by the SDM contained less bias. The CDM and SDM are both useful transduction models that recapitulate, with fidelity, delayed drug effects upon tumor growth. However, they are mechanistically distinct and not interchangeable. Both fit some types of tumor growth data well, but the SDM appeared more robust, particularly where experimental data are sparse.
PubMedId: 19902363
Journal Article: The AAPS journal, vol. 12, 2009
Authors: Rong Zhou, Richard V Mazurchuk, Judith H Tamburlin, John M Harrold, Donald E Mager, Robert M Straubinger
Abstract: Nano- and microparticulate carriers can exert a beneficial impact on the pharmacodynamics of anticancer agents. To investigate the relationships between carrier and antitumor pharmacodynamics, paclitaxel incorporated in liposomes (L-pac) was compared with the clinical standard formulated in Cremophor-EL/ethanol (Cre-pac) in a rat model of advanced primary brain cancer. Three maximum-tolerated-dose regimens given by intravenous administration were investigated: 50 mg/kg on day 8 (d8) after implantation of 9L gliosarcoma tumors; 40 mg/kg on d8 and d15; 20 mg/kg on d8, d11, and d15. Body weight change and neutropenia were assessed as pharmacodynamic markers of toxicity. The pharmacodynamic markers of antitumor efficacy were increase in lifespan (ILS) and tumor volume progression, measured noninvasively by magnetic resonance imaging. At equivalent doses, neutropenia was similar for both formulations, but weight loss was more severe for Cre-pac. No regimen of Cre-pac extended survival, whereas L-pac at 40 mg/kg x2 doses was well tolerated and mediated 26% ILS (p < 0.0002) compared with controls. L-pac at a lower cumulative dose (20 mg/kg x3) was even more effective (40% ILS; p < 0.0001). In striking contrast, the identical regimen of Cre-pac was lethal. Development of a novel semimechanistic pharmacodynamic model permitted quantitative hypothesis testing with the tumor volume progression data, and suggested the existence of a transient treatment effect that was consistent with sensitization or "priming" of tumors by more frequent L-pac dosing schedules. Therefore, improved antitumor responses of carrier-based paclitaxel formulations can arise both from dose escalation, because of reduced toxicity, and from novel carrier-mediated alterations of antitumor pharmacodynamic effects.
PubMedId: 19861574
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 332, 2009
Authors: Anshu Marathe, Wojciech Krzyzanski, Donald E. Mager
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 36, pp. 199-219, 2009
Authors: Donald E. Mager, Sukyung Woo, William J. Jusko
Journal-article: Drug Metabolism and Pharmacokinetics, vol. 24, pp. 16-24, 2009
Authors: D Siluk, D E Mager, H S Kim, Y Wang, A M Furimsky, A Ta, L V Iyer, C E Green, I W Wainer
Abstract: (R,R)-fenoterol (Fen), a beta(2)-adrenoceptor agonist, is under clinical investigation in the treatment of congestive heart disease. The pharmacokinetics and metabolism of the 4-methoxyphenyl derivative of (R,R)-Fen, (R,R)-MFen, have been determined following intravenous and oral administration to the rat and compared with corresponding results obtained with (R,R)-Fen. Results from the study suggest that (R,R)-MFen can offer pharmacokinetic and metabolic advantages in comparison to an earlier (R,R)-Fen. The oral administration revealed that the net exposure of (R,R)-MFen was about three-fold higher than that of (R,R)-Fen (7.2 versus 2.3 min x nmol ml(-1)), while intravenous administration proved that the clearance was significantly reduced, 48 versus 146 ml min(-1) kg(-1), the T(1/2) was significantly longer, 152.9 versus 108.9 min, and the area under the curve (AUC) was significantly increased, 300 versus 119 min x nmol ml(-1). (R,R)-MFen was primarily cleared by glucuronidation associated with significant presystemic glucuronidation of the compound. After intravenous and oral administration of (R,R)-MFen, (R,R)-Fen and (R,R)-Fen-G were detected in the urine samples indicating that (R,R)-MFen was O-demethylated and subsequently conjugated to (R,R)-Fen-G. The total (R,R)-Fen and (R,R)-Fen-G as a percentage of the dose after intravenous administration was 3.6%, while after oral administration was 0.3%, indicating that only a small fraction of the drug escaped presystemic glucuronidation and was available for O-demethylation. The glucuronidation pattern was confirmed by the results from in vitro studies where incubation of (R,R)-MFen with rat hepatocytes produced (R,R)-MFen-G, (R,R)-Fen and (R,R)-Fen-G, while incubation with rat intestinal microsomes only resulted in the formation of (R,R)-MFen-G.
PubMedId: 20039779
Journal Article: Xenobiotica; the fate of foreign compounds in biological systems, vol. 40, 2009
Authors: A. Marathe, M. C. Peterson, D. E. Mager
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 326, pp. 555-562, 2008
Authors: Anson K Abraham, Murali Ramanathan, Bianca Weinstock-Guttman, Donald E Mager
Abstract: Restoration of dysregulated bone homeostasis is a therapeutic goal in many diseases including osteoporosis, rheumatoid arthritis and metastatic cancer. The molecular pathways regulating bone remodeling are major therapeutic targets, and studies continue to reveal endogenous factors that may be pathologically up- or down-regulated and lead to an uncoupling of bone formation and resorption. The purpose of this commentary is to highlight new mechanisms of bone homeostatic regulation mediated through the induction of endogenous interferon-beta (IFN-beta). The receptor activator of nuclear factor-kappaB (RANK) ligand (RANKL) is an important factor in the bone resorption cascade, and the RANK-RANKL interaction has been shown to induce IFN-beta and osteoclastogenesis via induction of the c-fos gene. Subsequent binding of IFN-beta to its biological receptor initiates a signal transduction cascade through the classic JAK/STAT pathway, causing an inhibition of c-fos protein production and osteoclast proliferation and differentiation (negative feedback). Another mechanism pertinent to the anti-resorptive effect of IFN-beta is the induction of nitric oxide which has been shown to inhibit osteoclast formation. The role of IFN-beta in bone metabolism could warrant its systematic evaluation as a potential adjunct to therapeutic regimens of osteolytic diseases. Here we also provide discussion of the potential challenges to optimizing IFN-beta pharmacotherapy for such purposes.
PubMedId: 19428330
Journal-article: Biochemical pharmacology, vol. 77, 2008
Authors: DE Mager, WJ Jusko
Journal-article: Clinical Pharmacology & Therapeutics, vol. 83, pp. 909-912, 2008
Authors: Y Hong, J Dingemanse, DE Mager
Journal-article: Clinical Pharmacology & Therapeutics, vol. 84, pp. 136-143, 2008
Authors: Ying Hong, Shashank Rohatagi, Bahru Habtemariam, Joseph R. Walker, Sherwyn L. Schwartz, Donald E. Mager
Journal-article: The Journal of Clinical Pharmacology, vol. 48, pp. 696-707, 2008
Authors: Ying Hong, Fran M Gengo, Michelle M Rainka, Vernice E Bates, Donald E Mager
Journal-article: Clinical Pharmacokinetics, vol. 47, pp. 129-137, 2008
Authors: Ying Hong, Donald E. Mager, Robert A. Blum, William J. Jusko
Journal-article: Pharmaceutical Research, vol. 24, pp. 1088-1097, 2007
Authors: Francis M Gengo, Lisa Rubin, Matthew Robson, Michelle Rainka, Michael F Gengo, Donald E Mager, Vernice Bates
Abstract: This study was designed to measure the magnitude and duration of inhibition of platelet aggregation following doses of aspirin or ibuprofen alone or taken in combination in a group of healthy volunteers. Ten normal volunteer subjects underwent 3 randomized treatment sessions: aspirin 325 mg alone, ibuprofen 400 mg alone, and ibuprofen 400 mg, followed by dosing with aspirin 325 mg 2 hours thereafter. In addition, a confirmatory study was performed in patients. Over 27 months, a cohort of patients treated with aspirin for secondary stroke prophylaxis while concomitantly taking a nonsteroidal anti-inflammatory drug (NSAID) was identified. A significant reduction was found in both the magnitude and duration of aspirin's inhibitory effect on platelet aggregation when ibuprofen was given prior to aspirin administration in normal volunteer subjects. During a 27-month period, a cohort of 28 patients took regular daily doses of ibuprofen or naproxen. Of these 28 patients, 18 returned for follow-up testing in the absence of this pharmacodynamic interaction. None of these 18 patients demonstrated inhibition of platelet aggregation while on both NSAID and aspirin; however, all showed inhibition of aggregation following discontinuation of the NSAID. Notably, 13 of these 18 patients (72%) had experienced a recurrent ischemic episode while taking aspirin and NSAIDs concomitantly. These data suggest that ibuprofen prevents the irreversible inhibition of platelet aggregation produced by aspirin needed for secondary stroke prophylaxis, and this interaction can have clinical consequences for patients taking aspirin.
PubMedId: 18094224
Journal Article: Journal of clinical pharmacology, vol. 48, 2007
Authors: Anson K Abraham, Wojciech Krzyzanski, Donald E Mager
Abstract: Sensitivity analysis is commonly used to characterize the effects of parameter perturbations on model output. One use for the approach is the optimization of an experimental design enabling estimation of model parameters with improved accuracy. The primary objective of this study is to conduct a sensitivity analysis of selected target-mediated pharmacokinetic models, ascertain the effect of parameter variations on model predictions, and identify influential model parameters. One linear model (Model 1, control) and 2 target-mediated models (Models 2 and 3) were evaluated over a range of dose levels. Simulations were conducted with model parameters being perturbed at the higher and lower ends from literature mean values. Profiles of free plasma drug concentrations and their partial derivatives with respect to each parameter vs time were analyzed. Perturbations resulted in altered outputs, the extent of which reflected parameter influence. The model outputs were highly sensitive to perturbations of linear disposition parameters in all 3 models. The equilibrium dissociation constant (K(D)) was less influential in Model 2 but was influential in the terminal phase in Model 3, highlighting the role of K(D) in this region. An equation for Model 3 in support of the result for K(D) was derived. Changes in the initial receptor concentration [R(tot) (0)] paralleled the observed effects of initial plasma volume (V(c)) perturbations, with increased influence at higher values. Model 3 was also sensitive to the rates of receptor degradation and internalization. These results suggest that informed sampling may be essential to accurately estimate influential parameters of target-mediated models.
PubMedId: 17614360
Journal Article: The AAPS journal, vol. 9, 2007
Authors: Danuta Siluk, Donald E. Mager, Naomi Gronich, Darrell Abernethy, Irving W. Wainer
Journal-article: Journal of Chromatography B, vol. 859, pp. 213-221, 2007
Authors: Donald E. Mager, Evgeny Kobrinsky, Amirali Masoudieh, Anna Maltsev, Darrell R. Abernethy, Nikolai M. Soldatov
Journal-article: Biophysical Journal, vol. 93, pp. 2900-2910, 2007
Authors: D. E. Mager, D. R. Abernethy
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 321, pp. 423-430, 2007
Authors: Donald E. Mager
Journal-article: Advanced Drug Delivery Reviews, vol. 58, pp. 1326-1356, 2006
Authors: Donald E. Mager
Journal-article: Biochemical Pharmacology, vol. 72, pp. 1-10, 2006
Authors: Donald E. Mager, Ruiqian Wan, Martin Brown, Aiwu Cheng, Przemyslaw Wareski, Darrell R. Abernethy, Mark P. Mattson
Journal-article: The FASEB Journal, vol. 20, pp. 631-637, 2006
Authors: Donald E. Mager, Jason D. Shirey, Dermot Cox, Desmond J. Fitzgerald, Darrell R. Abernethy
Journal-article: Journal of Pharmaceutical Sciences, vol. 94, pp. 2475-2486, 2005
Authors: Donald E. Mager, Berend Neuteboom, William J. Jusko
Journal-article: Pharmaceutical Research, vol. 22, pp. 58-61, 2005
Authors: Donald E Mager, Wojciech Krzyzanski
Abstract: The aim of this study is to derive and evaluate an equilibrium model of a previously developed general pharmacokinetic model for drugs exhibiting target-mediated drug disposition (TMDD). A quasi-equilibrium solution to the system of ordinary differential equations that describe the kinetics of TMDD was obtained. Computer simulations of the equilibrium model were carried out to generate plasma concentration-time profiles resulting from a large range of intravenous bolus doses. Additionally, the final model was fitted to previously published pharmacokinetic profiles of leukemia inhibitory factor (LIF), a cytokine that seems to exhibit TMDD, following intravenous administration of 12.5, 25, 100, 250, 500, or 750 microg/kg in sheep. Simulations show that pharmacokinetic profiles display steeper distribution phases for lower doses and similar terminal disposition phases, but with slight underestimation at early time points as theoretically expected. The final model well-described LIF pharmacokinetics, and the final parameters, which were estimated with relatively good precision, were in good agreement with literature values. An equilibrium model of TMDD is developed that recapitulates the essential features of the full general model and eliminates the need for estimating drug-binding microconstants that are often difficult or impossible to identify from typical in vivo pharmacokinetic data.
PubMedId: 16180117
Journal Article: Pharmaceutical research, vol. 22, 2005
Authors: Evgeny Kobrinsky, Donald E Mager, Sarah A Bentil, Shin-Ichi Murata, Darrell R Abernethy, Nikolai M Soldatov
Abstract: In this study, we sought to characterize functional signaling domains by applying the multiresolution properties of the continuous wavelet transform to fluorescence resonance energy transfer (FRET) microscopic images of plasma membranes. A genetically encoded FRET reporter of protein kinase C (PKC)-dependent phosphorylation was expressed in COS1 cells. Differences between wavelet coefficient matrices revealed several heterogeneous domains (typically ranging from 1 to 5 microm), reflecting the dynamic balance between PKC and phosphatase activity during stimulation with phorbol-12,13-dibutyrate or acetylcholine. The balance in these domains was not necessarily reflected in the overall plasma membrane changes, and observed heterogeneity was absent when cells were exposed to a phosphatase or PKC inhibitor. Prolonged exposure to phorbol-12,13-dibutyrate and acetylcholine yielded more homogeneous FRET distribution in plasma membranes. The proposed wavelet-based image analysis provides, for the first time, a basis and a means of detecting and quantifying dynamic changes in functional signaling domains, and may find broader application in studying fine aspects of cellular signaling by various imaging reporters.
PubMedId: 15722423
Journal Article: Biophysical journal, vol. 88, 2005
Authors: Alicia M Segrave, Donald E Mager, Susan A Charman, Glenn A Edwards, Christopher J H Porter
Abstract: The pharmacokinetics of recombinant human leukemia inhibitory factor (rhLIF) were investigated following i.v. and s.c. administration of a wide range of dose levels. Parallel studies were conducted where single i.v. bolus doses of 12.5, 25, 100, 250, 500, or 750 microg/kg rhLIF (n = 2) or s.c. doses of 10, 20, or 50 microg/kg rhLIF (n = 4) were administered to sheep. Blood samples were collected for up to 24 h postdosing, and the plasma concentrations of rhLIF were analyzed by enzyme-linked immunosorbent assay. Noncompartmental analysis demonstrated an increase in the terminal elimination half-life (from 0.27 to 2.29 h) and a decrease in systemic clearance (from 5.18 to 1.09 ml/min/kg) with increasing i.v. doses of rhLIF, suggesting nonlinear pharmacokinetic behavior. A greater than proportional increase in the area under the plasma concentration-time curve with dose also indicated significantly nonlinear pharmacokinetics after s.c. administration. A mechanistic compartmental model was developed to characterize the pharmacokinetics of rhLIF. The key feature of the model accounting for the nonlinear pharmacokinetic behavior of rhLIF was high-affinity, saturable receptor binding and subsequent cellular internalization and degradation. The apparent total density of LIF cell surface receptors and receptor turnover dynamics were included in the model, along with nonspecific binding and linear elimination from the systemic circulation. The absorption of rhLIF from the s.c. injection site into the systemic circulation was characterized by a first-order absorption process via a delay compartment. The proposed model satisfactorily captured the complex pharmacokinetic profiles of rhLIF following both i.v. and s.c. administration.
PubMedId: 14872093
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 309, 2004
Authors: D. E. Mager
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 311, pp. 830-835, 2004
Authors: Donald E Mager, Berend Neuteboom, Constantinos Efthymiopoulos, Alain Munafo, William J Jusko
Abstract: A pharmacokinetic/pharmacodynamic (PK/PD) model was developed to simultaneously characterize interferon after i.v. and s.c. dosing at various dose levels. A sequential study in monkeys (n = 18) was conducted, where single doses of 1, 3, and 10 MIU/kg of recombinant-human interferon-beta (IFN-beta) 1a were given i.v. and then s.c. Plasma concentrations of IFN-beta were determined and biphasic neopterin concentrations were used as the pharmacodynamic (PD) endpoint. Multiple dosing also was evaluated by giving 1 MIU/kg s.c. doses once daily for 7 days (n = 3). The integrated model uses target-mediated drug disposition to describe drug elimination by receptor binding and internalization, and well characterizes the observed nonlinear pharmacokinetic (PK) profiles. The s.c. doses exhibited an absorption phase (Tmax = 3 h) and incomplete bioavailability (F = 0.3-0.7). An indirect response model for stimulation of neopterin triphosphate production by activated receptor complex followed by conversion to neopterin was used to jointly model the formation and loss of neopterin with a capacity factor Smax = 23.8. Greater relative neopterin response after s.c. dosing was accounted for by prolonged receptor activation relative to the SC50 value. Repeated daily s.c. dosing produced modestly elevated IFN-beta1a concentrations and neopterin concentrations that were lower than simulated from single-dose modeling. Although several mechanisms could be involved, these phenomena were simply remodeled as down-regulation of Smax and receptors. The PK/PD model for IFN-beta1a depicts receptor binding as a key feature controlling nonlinear elimination, nonstationary kinetics, and neopterin induction in a manner consistent with known processes controlling its disposition and pharmacological effects.
PubMedId: 12660309
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 306, 2003
Authors: Donald E Mager, Elzbieta Wyska, William J Jusko
Abstract: Pharmacodynamics is the study of the time course of pharmacological effects of drugs. The field of pharmacodynamic modeling has made many advances, due in part to the relatively recent development of basic and extended mechanism-based models. The purpose of this article is to describe the classic as well as contemporary approaches, with an emphasis on pertinent equations and salient model features. In addition, current methods of integrating various system complexities into these models are discussed. Future pharmacodynamic models will most likely reflect an assembly of the basic components outlined in this review.
PubMedId: 12695336
Journal Article: Drug metabolism and disposition: the biological fate of chemicals, vol. 31, 2003
Authors: Donald E. Mager, Nancy A. Pyszczynski, William J. Jusko
Journal-article: Journal of Pharmaceutical Sciences, vol. 92, pp. 881-889, 2003
Authors: D. E. Mager
Journal-article: Drug Metabolism and Disposition, vol. 31, pp. 510-518, 2003
Authors: Donald E. Mager, Nasir Moledina, William J. Jusko
Journal-article: Journal of Pharmaceutical Sciences, vol. 92, pp. 1521-1525, 2003
Authors: Elzbieta Wyska, Donald E. Mager, Wojciech Krzyzanski
Journal-article: Journal of Pharmaceutical Sciences, vol. 92, pp. 1438-1454, 2003
Authors: Donald E. Mager, Sheren X. Lin, Robert A. Blum, Christian D. Lates, William J. Jusko
Journal-article: The Journal of Clinical Pharmacology, vol. 43, pp. 1216-1227, 2003
Authors: D. E. Mager
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 307, pp. 969-976, 2003
Authors: Donald E Mager, William J Jusko
Abstract: An integrated receptor-based pharmacokinetic/pharmacodynamic (PK/PD) model of interferon-beta la (IFN-beta la) previously developed for monkeys was used to capture the time-course of drug and induced neopterin concentrations after intravenous (IV) and subcutaneous (SC) dosing in humans. Data were extracted from the literature by digitalization. Single-dose (3 IV doses and I SC dose) PK/PD profiles were simultaneously fitted using the basic model and the ADAPT II computer program. Additional submodels incorporating neutralizing antibody formation and negative feedback inhibition were applied to account for drug accumulation and lower than expected neopterin concentrations encountered after multiple-dosing (1 SC dose every 48 hs). The basic model jointly-captured the nonlinear PK behavior of the drug and induced neopterin concentrations after all single doses. Slow and incomplete absorption (F = 0.33) of the SC dose resulted in prolonged drug concentrations reflective of flip-flop kinetics. Despite lower drug concentrations, SC dosing produced a similar neopterin profile as compared with the IV doses; however, with a longer time to peak effect and slightly higher neopterin concentrations at later time points. The PD component of the model represents a modified precursor-dependent indirect response model driven by the amount of internalized drug-receptor complex. The latter stimulated a 6-fold increase in the production of the neopterin precursor (Smax = 5.89). Drug accumulation and lower than expected neopterin concentrations after multiple dosing were also captured after the inclusion of the submodels. The present integrated PK/PD model for IFN-beta 1a is mechanistic in nature with receptor-mediated disposition and dynamics and was successfully applied to human clinical data.
PubMedId: 12425473
Journal Article: Pharmaceutical research, vol. 19, 2002
Authors: Donald E. Mager, William J. Jusko
Journal-article: Journal of Pharmaceutical Sciences, vol. 91, pp. 2441-2451, 2002
Authors: D Mager
Journal-article: Clinical Pharmacology & Therapeutics, vol. 70, pp. 210-216, 2001
Authors: Donald E. Mager, Murali Ramanthan
Book-chapter: Handbook of Pharmaceutical Biotechnology, pp. 253-278,
Presented by CUBS © 2024