UB Vice President for Research and Economic Development markup
Joseph P Balthasar UB Profile
Pharmacy
Professor of Pharmaceutical Sciences
Director of the Center for Protein Therapeutics
Executive Director of University Research Initiatives
452 Kapoor Hall
Buffalo NY 14214
716-645-4807
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Shun Xin Wang-Lin, Ruth Olson, Janet M Beanan, Ulrike MacDonald, Joseph P Balthasar, Thomas A Russo
PubMedId: 28947651
Journal Article: Infection and immunity, vol. 85, 2017
Authors: Yujie Yang, Tommy R. Li, Joseph P. Balthasar
Journal-article: The AAPS Journal, vol. 19, pp. 1791-1803, 2017
Authors: Ming Zhang, Bo An, Yang Qu, Shichen Shen, Wei Fu, Yuan-Ju Chen, Xue Wang, Rebeccah Young, John M Canty, Joseph P Balthasar, Keeley Murphy, Debadeep Bhattacharyya, Jonathan Josephs, Luca Ferrari, Shaolian Zhou, Surendra Bansal, Faye Vazvaei, Jun Qu
Abstract: For LC-MS-based targeted quantification of biotherapeutics and biomarkers in clinical and pharmaceutical environments, high sensitivity, high throughput, and excellent robustness are all essential but remain challenging. For example, though nano-LC-MS has been employed to enhance analytical sensitivity, it falls short because of its low loading capacity, poor throughput, and low operational robustness. Furthermore, high chemical noise in protein bioanalysis typically limits the sensitivity. Here we describe a novel trapping-micro-LC-MS (T-LC-MS) strategy for targeted protein bioanalysis, which achieves high sensitivity with exceptional robustness and high throughput. A rapid, high-capacity trapping of biological samples is followed by LC-MS analysis; dynamic sample trapping and cleanup are performed using pH, column chemistry, and fluid mechanics separate from the LC-MS analysis, enabling orthogonality, which contributes to the reduction of chemical noise and thus results in improved sensitivity. Typically, the selective-trapping and -delivery approach strategically removes >85% of the matrix peptides and detrimental components, markedly enhancing sensitivity, throughput, and operational robustness, and narrow-window-isolation selected-reaction monitoring further improves the signal-to-noise ratio. In addition, unique LC-hardware setups and flow approaches eliminate gradient shock and achieve effective peak compression, enabling highly sensitive analyses of plasma or tissue samples without band broadening. In this study, the quantification of 10 biotherapeutics and biomarkers in plasma and tissues was employed for method development. As observed, a significant sensitivity gain (up to 25-fold) compared with that of conventional LC-MS was achieved, although the average run time was only 8 min/sample. No appreciable peak deterioration or loss of sensitivity was observed after >1500 injections of tissue and plasma samples. The developed method enabled, for the first time, ultrasensitive LC-MS quantification of low levels of a monoclonal antibody and antigen in a tumor and cardiac troponin I in plasma after brief cardiac ischemia. This strategy is valuable when highly sensitive protein quantification in large sample sets is required, as is often the case in typical biomarker validation and pharmaceutical investigations of antibody therapeutics.
PubMedId: 29276835
Journal Article: Analytical chemistry, vol. 90, 2017
Authors: Miao Qu, Bo An, Shichen Shen, Ming Zhang, Xiaomeng Shen, Xiaotao Duan, Joseph P Balthasar, Jun Qu
Keywords: antibody-drug conjugate biotherapeutics liquid chromatography mass spectrometry quantification
Abstract: In the last decade, the advancement of liquid chromatography mass spectrometry (LC/MS) techniques has enabled their broad application in protein characterization, both quantitatively and qualitatively. Owing to certain important merits of LC/MS techniques (e.g., high selectivity, flexibility, and rapid method development), LC/MS assays are often deemed as preferable alternatives to conventional methods (e.g., ligand-binding assays) for the analysis of protein biotherapeutics. At the discovery and development stages, LC/MS is generally employed for two purposes absolute quantification of protein biotherapeutics in biological samples and qualitative characterization of proteins. For absolute quantification of a target protein in bio-matrices, recent work has led to improvements in the efficiency of LC/MS method development, sample treatment, enrichment and digestion, and high-performance low-flow-LC separation. These advances have enhanced analytical sensitivity, specificity, and robustness. As to qualitative analysis, a range of techniques have been developed to characterize intramolecular disulfide bonds, glycosylation, charge variants, primary sequence heterogeneity, and the drug-to-antibody ratio of antibody drug conjugate (ADC), which has enabled a refined ability to assess product quality. In this review, we will focus on the discussion of technical challenges and strategies of LC/MS-based quantification and characterization of biotherapeutics, with the emphasis on the analysis of antibody-based biotherapeutics such as monoclonal antibodies (mAbs) and ADCs. 2016 Wiley Periodicals, Inc. Mass Spec Rev 36:734-754, 2017.
PubMedId: 27097288
Journal Article: Mass spectrometry reviews, vol. 36, 2016
Authors: Patrick M Glassman, Joseph P Balthasar
Keywords: Clinical pharmacokinetics; lymphocyte; monoclonal antibodies; physiologically-based pharmacokinetics; receptor occupancy
Abstract: Many clinically approved and investigational monoclonal antibody (mAb)-based therapeutics are directed against proteins located in the systemic circulation, including cytokines, growth factors, lymphocyte proteins, and shed antigens. Interaction between mAb and target may lead to non-linear pharmacokinetics (PK), characterized by rapid, target-mediated elimination. Several groups have reported that determinants of target-mediated elimination could include mAb-target binding, target expression, and target turnover. Recently, we scaled a physiologically-based pharmacokinetic model for mAb disposition to man and used it to predict the non-linear PK of mAbs directed against tumor epithelial proteins. In this work, we extended the previously described model to account for the influence of lymphocyte proteins on mAb PK in man. To account for the dynamic behavior of lymphocytes in the circulation, lymphocyte cycling between blood and lymphoid organs was described using first-order transfer rate constants. Use of lymphocyte cycling and reported target turnover rates in the model allowed the accurate prediction of the pharmacokinetics and pharmacodynamics (PD) of 4 mAbs (TRX1, MTRX1011a, rituximab, daclizumab) directed against 3 lymphocyte targets (CD4, CD20, CD25). The results described here suggest that the proposed model structure may be useful in the a priori prediction of the PK/PD properties of mAbs directed against antigens in the circulation.
PubMedId: 27892793
Journal Article: mAbs, vol. 9, 2016
Authors: Patrick M Glassman, Joseph P Balthasar
Keywords: Clinical pharmacokinetics Monoclonal antibody Physiologically-based pharmacokinetics (PBPK) Target-mediated drug disposition (TMDD)
Abstract: Accurate prediction of the clinical pharmacokinetics of new therapeutic entities facilitates decision making during drug discovery, and increases the probability of success for early clinical trials. Standard strategies employed for predicting the pharmacokinetics of small-molecule drugs (e.g., allometric scaling) are often not useful for predicting the disposition monoclonal antibodies (mAbs), as mAbs frequently demonstrate species-specific non-linear pharmacokinetics that is related to mAb-target binding (i.e., target-mediated drug disposition, TMDD). The saturable kinetics of TMDD are known to be influenced by a variety of factors, including the sites of target expression (which determines the accessibility of target to mAb), the extent of target expression, the rate of target turnover, and the fate of mAb-target complexes. In most cases, quantitative information on the determinants of TMDD is not available during early phases of drug discovery, and this has complicated attempts to employ mechanistic mathematical models to predict the clinical pharmacokinetics of mAbs. In this report, we introduce a simple strategy, employing physiologically-based modeling, to predict mAb disposition in humans. The approach employs estimates of inter-antibody variability in rate processes of extravasation in tissues and fluid-phase endocytosis, estimates for target concentrations in tissues derived through use of categorical immunohistochemical scores, and in vitro measures of the turnover of target and target-mAb complexes. Monte Carlo simulations were performed for four mAbs (cetuximab, figitumumab, dalotuzumab, trastuzumab) directed against three targets (epidermal growth factor receptor, insulin-like growth factor receptor 1, human epidermal growth factor receptor 2). The proposed modeling strategy was able to predict well the pharmacokinetics of cetuximab, dalotuzumab, and trastuzumab at a range of doses, but trended towards underprediction of figitumumab concentrations, particularly at high doses. The general agreement between model predictions and experimental observations suggests that PBPK modeling may be useful for the a priori prediction of the clinical pharmacokinetics of mAb therapeutics.
PubMedId: 27377311
Journal Article: Journal of pharmacokinetics and pharmacodynamics, vol. 43, 2016
Authors: Veena A Thomas, Joseph P Balthasar
Keywords: anti-angiogenesis drugdrug interaction monoclonal antibodies pharmacokinetics
Abstract: In this investigation, we test the hypothesis that treatment with sorafenib, an anti-angiogenic agent, decreases tumor vascularization and, consequently, hinders the delivery of monoclonal antibodies (mAb) to xenograft tumors. Severe combined immunodeficiency mice bearing carcinoembryonic antigen (CEA) expressing tumor xenografts were divided into control and sorafenib-treated groups. Sorafenib was administered to the latter group at 50mg/kg IP every 48h, starting 4days post-tumor implantation. When tumors attained a size of 200-300mm(3), mice were evaluated for (a) tumor microvessel density (using immunohistochemical analysis), (b) tumor macromolecular extravasation (using Evans Blue Dye (EBD)), (c) pharmacokinetics of an anti-CEA mAb, T84.66, following an intravenous dose of 10mg/kg, and (d) intra-tumoral spatial distribution of T84.66 (using autoradiography). Sorafenib treatment resulted in a substantial reduction in tumor growth rate, a visible reduction in tumor microvessel density, and in a 46.4% decrease in EBD extravasation in tumor tissue (p<0.0455). For control and treated mice, no significant difference was found for the area under the mAb plasma concentration-time curve (AUC(0-7d): 1.6710(3)1.2810(2) vs. 1.7610(3)1.7510(2) nMday, p=0.51). However, tumor AUC(0-7d) was reduced by 40.8% in sorafenib-treated mice relative to that observed in control mice (5.6110(2)4.2710(1) vs. 9.4810(2)5.6110(1) nMday, p<0.001). Sorafenib therapy was also found to markedly alter mAb tumor spatial distribution. The results collectively suggest that sorafenib treatment causes a significant reduction in mAb delivery to, and distribution within, solid tumors.
PubMedId: 27029796
Journal Article: The AAPS journal, vol. 18, 2016
Authors: Patrick M Glassman, Joseph P Balthasar
Keywords: Antibody engineering Monoclonal antibodies Physiologically-based pharmacokinetics (PBPK)
Abstract: The development of 'catch and release', or pH-sensitive, monoclonal antibodies (mAb) has become of interest to groups seeking to reduce the influence of target-mediated elimination on pharmacokinetics and pharmacodynamics. In this work, a catenary physiologically-based pharmacokinetic (PBPK) model is described to predict the pharmacokinetic benefit conferred by engineering mAbs for 'catch and release' binding. Our previously published PBPK model was adapted for consideration of the production and elimination of proprotein convertase subtilisin/kexin type 9 (PCSK9) in mice, and the model was then applied to predict the pharmacokinetics of anti-PCSK9 mAb with pH-stable (J10) and pH-sensitive binding (J17). The model was able to generate reasonable predictions of both J10 and J17 plasma pharmacokinetics. For J10, mean (standard deviation) predicted vs. observed areas under the plasma concentration curve (AUCinf) were: 217 (77.1) vs. 103nMday (1mg/kg), 1.1410(3) (858) vs. 769nMday (3mg/kg), and 6.6010(3) (5.5810(3)) vs. 2.8610(3)nMday (10mg/kg), and for J17 the values were: 838 (161) vs. 818nMday (1mg/kg), 2.3010(3) (441) vs. 2.5710(3)nMday (3mg/kg), and 8.4210(3) (1.5010(3)) vs. 9.1710(3)nMday (10mg/kg). Further simulations with the model predicted that target turnover and the magnitude of change in the complex dissociation rate constant between pH 7.4 and pH 6.0 are key determinants of the improvements in pharmacokinetics associated with 'catch and release' mAbs. The model described here may be useful for prediction of the pharmacokinetics of 'catch and release' mAbs directed against other targets.
PubMedId: 27041125
Journal Article: International journal of pharmaceutics, vol. 505, 2016
Authors: Frank A Engler, Joseph P Balthasar
Keywords: ELISA assay gelonin pharmacokinetics protein toxin ribosome-inactivating protein
Abstract: This article details the development and validation of an enzyme-linked immunosorbent assay (ELISA) for the quantification of gelonin in mouse plasma. The ELISA was validated for intra- and inter-day variability and for accuracy over a standard curve range of 7.5-100 ng/mL. The assay was then applied to assess gelonin pharmacokinetics in mice. Results from the ELISA were compared to data obtained from a parallel study conducted with (125)Iodine-labeled gelonin, with quantification via gamma counting. The ELISA demonstrated good precision, as the percent coefficient of variation of quality control samples in intra-day and inter-day validation ranged from 5.4-9.3% and 2.9-7.3%, respectively. Sample recoveries ranged from 98.3-105% of nominal values. The ELISA method yielded lower plasma concentrations of gelonin than found from the less-specific gamma counting method. Consequently, pharmacokinetic analyses yielded significantly higher estimates for volume of distribution (106 31 vs. 55.8 13 mL/kg) and plasma clearance (34.7 6.6 vs. 10.9 2.1 mL/min/kg) for data determined by ELISA vs. by gamma counting.
PubMedId: 27135787
Journal Article: Journal of immunoassay & immunochemistry, vol. 37, 2016
Authors: Patrick M Glassman, Yang Chen, Joseph P Balthasar
Keywords: Monoclonal antibody Pharmacokinetics Physiologically-based
Abstract: Preclinical assessment of monoclonal antibody (mAb) disposition during drug development often includes investigations in non-human primate models. In many cases, mAb exhibit non-linear disposition that relates to mAb-target binding [i.e., target-mediated disposition (TMD)]. The goal of this work was to develop a physiologically-based pharmacokinetic (PBPK) model to predict non-linear mAb disposition in plasma and in tissues in monkeys. Physiological parameters for monkeys were collected from several sources, and plasma data for several mAbs associated with linear pharmacokinetics were digitized from prior literature reports. The digitized data displayed great variability; therefore, parameters describing inter-antibody variability in the rates of pinocytosis and convection were estimated. For prediction of the disposition of individual antibodies, we incorporated tissue concentrations of target proteins, where concentrations were estimated based on categorical immunohistochemistry scores, and with assumed localization of target within the interstitial space of each organ. Kinetics of target-mAb binding and target turnover, in the presence or absence of mAb, were implemented. The model was then employed to predict concentration versus time data, via Monte Carlo simulation, for two mAb that have been shown to exhibit TMD (2F8 and tocilizumab). Model predictions, performed a priori with no parameter fitting, were found to provide good prediction of dose-dependencies in plasma clearance, the areas under plasma concentration versu time curves, and the time-course of plasma concentration data. This PBPK model may find utility in predicting plasma and tissue concentration versus time data and, potentially, the time-course of receptor occupancy (i.e., mAb-target binding) to support the design and interpretation of preclinical pharmacokinetic-pharmacodynamic investigations in non-human primates.
PubMedId: 26364301
Journal Article: Journal of pharmacokinetics and pharmacodynamics, vol. 42, 2015
Authors: Patrick M Glassman, Lubna Abuqayyas, Joseph P Balthasar
Keywords: biologics biotechnology (BTN) pharmaceutics (PCT) pharmacodynamics (PDY) pharmacokinetics and metabolism
Abstract: Monoclonal antibody (mAb) therapeutics are in use for several disease conditions, and have generally shown excellent clinical benefit, in large part due to their high specificity and affinity for target proteins. As this therapeutic class continues to grow in size, improved understanding of the mechanisms controlling mAb biodistribution and protein binding may be expected to allow better prediction of safety and efficacy. Due to the large size and polarity of antibodies, rates of mAb distribution and elimination are typically much slower than those reported for small molecule drugs. Additionally, high affinity interaction with target proteins will often influence mAb pharmacokinetics, leading to complex, nonlinear tissue distribution and elimination. In this report, we summarize key determinants of mAb disposition, methods for assessing antibody exposure and protein binding, and model-based approaches that may be utilized to predict mAb pharmacokinetics.
PubMedId: 25707961
Journal Article: Journal of clinical pharmacology, vol. 55 Suppl 3, 2014
Authors: Meong Cheol Shin, Jian Zhang, Kyoung Ah Min, Kyuri Lee, Cheol Moon, Joseph P Balthasar, Victor C Yang
Keywords: Anti-CEA monoclonal antibody Cancer Gelonin Heparin Protein transduction domain
Abstract: The bottlenecks of current chemotherapy in the treatment of colorectal cancer lie in the ineffectiveness of the existing anti-cancer small molecule drugs as well as the dose-limiting toxicity caused by the nonselective action on normal tissues by such drugs. To address these problems, we introduce a novel therapeutic strategy based on tumor targeting using a non-internalizing anti-carcinoembryonic antigen (CEA) monoclonal antibody (mAb) and intracellular delivery of the extremely potent yet cell-impermeable protein toxin gelonin via the aid of a cell-penetrating peptide (also termed as protein transduction domain; PTD). A chimeric TAT-gelonin fusion protein was genetically engineered, and it displayed remarkably enhanced anti-cancer activity against human colorectal cancer cells, with IC50 values being several orders of magnitude lower than the unmodified gelonin. On the other hand, a chemically synthesized conjugate of heparin and a murine anti-CEA mAb, T84.66 (termed T84.66-Hep) was found able to bind highly specifically to CEA over-expressing LS174T colorectal cancer cells. When mixing together, TAT-gelonin and T84.66-Hep could associate tightly and automatically through an electrostatic interaction between the cationic TAT and anionic heparin. In preliminary in vivo studies using LS174T s.c. xenograft tumor bearing mouse, selective and significantly augmented (58-fold) delivery of TAT-gelonin to the tumor target was observed, when compared with administration of TAT-gelonin alone. More importantly, efficacy studies also revealed that only the TAT-gelonin/T84.66-Hep complex yielded a significant inhibition of tumor growth (46%) without causing gelonin-induced systemic toxicity. Overall, this study suggested a generic strategy to effectively yet safely deliver potent PTD-modified protein toxins to the tumor.
PubMedId: 25204286
Journal Article: Journal of controlled release : official journal of the Controlled Release Society, vol. 194, 2014
Authors: Kip P Conner, Brooke M Rock, Gayle K Kwon, Joseph P Balthasar, Lubna Abuqayyas, Larry C Wienkers, Dan A Rock
Abstract: The pharmacokinetic (PK) behavior of monoclonal antibodies (mAbs) is influenced by target-mediated drug disposition, off-target effects, antidrug antibody-mediated clearance, and interaction with fragment-crystallizable domain (Fc) receptors such as neonatal Fc receptor. All of these interactions hold the potential to impact mAb biodistribution. Near infrared (NIR) fluorescent probes offer an approach complementary to radionuclides to characterize drug disposition. Notably, the use of FDA-approved IRDye800 (IR800; LI-COR, Lincoln, NE) as a protein-labeling agent in preclinical work holds the potential for quantitative tissue analysis. Here, we tested the utility of the IR800 dye as a quantitative mAb tracer during pharmacokinetic analysis in both plasma and tissues using a model mouse monoclonal IgG1 (8C2) labeled with 1.5 molecules of IR800. The plasma PK parameters derived from a mixture of IR800-8C2 and 8C2 dosed intravenously to C57BL/6 mice at 8 mg/kg exhibited a large discrepancy in exposure depending on the method of quantitation [CLplasma = 8.4 ml/d per kilogram (NIR fluorescence detection) versus 2.5 ml/d per kilogram (enzyme-linked immunosorbent assay)]. The disagreement between measurements suggests that the PK of 8C2 is altered by addition of the IR800 dye. Additionally, direct fluorescence analysis of homogenized tissues revealed several large differences in IR800-8C2 tissue uptake when compared with a previously published study using [(125)I]8C2, most notably an over 4-fold increase in liver concentration. Finally, the utility of IR800 in combination with whole body imaging was examined by comparison of IR800-8C2 levels observed in animal sagittal cross-sections to those measured in homogenized tissues. Our results represent the first PK analysis in both mouse plasma and tissues of an IR800-mAb conjugate and suggest that mAb disposition is significantly altered by IR800 conjugation to 8C2.
PubMedId: 25209366
Journal Article: Drug metabolism and disposition: the biological fate of chemicals, vol. 42, 2014
Authors: Eslam Nouri-Nigjeh, Ming Zhang, Tao Ji, Haoying Yu, Bo An, Xiaotao Duan, Joseph Balthasar, Robert W Johnson, Jun Qu
Abstract: LC-MS provides a promising alternative to ligand-binding assays for quantification of therapeutic proteins and biomarkers. As LC-MS methodology is based on the analysis of proteolytic peptides, calibration approaches utilizing various calibrators and internal standards (I.S.) have been developed. A comprehensive assessment of the accuracy and reliability of these approaches is essential but has yet been reported. Here we performed a well-controlled and systematic comparative study using quantification of monoclonal-antibody in plasma as the model system. Method development utilized a high-throughput orthogonal-array-optimization, and two sensitive and stable signature-peptides (SP) from different domains were selected based on extensive evaluations in plasma matrix. With the purities of all protein/peptide standards corrected by quantitative amino acid analysis (AAA), five calibration approaches using stable-isotope-labeled (SIL) I.S. were thoroughly compared, including those at peptide, extended-peptide, and protein levels and two "hybrid" approaches (i.e., protein calibrator with SIL-peptide or SIL-extended-peptide I.S.). These approaches were further evaluated in parallel for a 15 time point, preclinical pharmacokinetic study. All methods showed good precision (CV% < 20%). When examined with protein-spiked plasma QC, peptide-level calibration exhibited severe negative biases (-23 to -62%), highly discordant results between the two SP (deviations of 38-56%), and misleading pharmacokinetics assessments. Extended-peptide calibration showed significant improvements but still with unacceptable accuracy. Conversely, protein-level and the two hybrid calibrations achieved good quantitative accuracy (error < 10%), concordant results by two SP (deviations < 15%), and correct pharmacokinetic parameters. Hybrid approaches were found to provide a cost-effective means for accurate quantification without the costly SIL-protein. Other key findings include (i) using two SP provides a versatile gauge for method reliability; (ii) evaluation of peptide stability in the matrix before SP selection is critical; and (iii) using AAA to verify purities of protein/peptide calibrators ensures accurate quantitation. These results address fundamental calibration issues that have not been adequately investigated in published studies and will provide valuable guidelines for the "fit for purpose" development of accurate LC-MS assays for therapeutic proteins and biomarkers in biological matrices.
PubMedId: 24611550
Journal Article: Analytical chemistry, vol. 86, 2014
Authors: Patrick M Glassman, Joseph P Balthasar
Keywords: Antibodies monoclonal oncology pharmacodynamics pharmacokinetics
Abstract: Since the approval of rituximab in 1997, monoclonal antibodies (mAbs) have become an increasingly important component of therapeutic regimens in oncology. The success of mAbs as a therapeutic class is a result of great strides that have been made in molecular biology and in biotechnology over the past several decades. Currently, there are 14 approved mAb products for oncology indications, and there are ten additional mAbs in late stages of clinical trials. Compared to traditional chemotherapeutic agents, mAbs have several advantages, including a long circulating half-life and high target specificity. Antibodies can serve as cytotoxic agents when administered alone, exerting a pharmacologic effect through several mechanisms involving the antigen binding (Fab) and/or Fc domains of the molecule, and mAbs may also be utilized as drug carriers, targeting a toxic payload to cancer cells. The extremely high affinity of mAbs for their targets, which is desirable with respect to pharmacodynamics (i.e., contributing to the high therapeutic selectivity of mAb), often leads to complex, non-linear, target-mediated pharmacokinetics. In this report, we summarize the pharmacokinetic and pharmacodynamics of mAbs that have been approved and of mAbs that are near approval for oncology indications, with particular focus on the molecular and cellular mechanisms responsible for their disposition and efficacy.
PubMedId: 24738036
Journal Article: Cancer biology & medicine, vol. 11, 2014
Authors: Lubna Abuqayyas, Xiaoyan Zhang, Joseph P. Balthasar
Journal-article: International Journal of Pharmaceutics, vol. 444, pp. 185-192, 2013
Authors: Yanguang Cao, Joseph P Balthasar, William J Jusko
Abstract: Minimal physiologically-based pharmacokinetic (mPBPK) models provide a sensible modeling approach when fitting only plasma (or blood) data yielding physiologically-relevant PK parameters that may provide more practical value than parameters of mammillary models. We propose a second-generation mPBPK model specifically for monoclonal antibodies (mAb) by including (lumping) several essential components of mAb PK used in full PBPK models. These components include convection as the primary mechanism of antibody movement from plasma into tissues and return to plasma with interstitial fluid as the major extravascular distribution space. The model divides tissue spaces into two groups according to their vascular endothelial structure, leaky and tight, which consequently allows discernment of two types and general sites of distribution. This mPBPK model was applied to two mAbs in mice and ten mAbs with linear kinetics in humans. The model captured their plasma PK profiles well with predictions of concentrations in interstitial fluid for two types of tissues. Predictions of tissue concentrations for mAb 7E3 and 8C2 were consistent with actual measurements in mice, indicating the feasibility of this model in assessing extravascular distribution in the two categories of tissues. The vascular reflection coefficients () of tight tissues (V(tight)) ranged 0.883-0.987 and coefficients () for leaky tissues (V(leaky)) ranged 0.311 to 0.837. The plasma clearance (CL(p)) varied among the mAbs in humans from 0.0054 to 0.03 L/h. In addition, applying this model generates parameters for mAb transcapillary escape rates and assesses major sites of elimination. Four of ten mAbs exhibited better fitting statistics premised on elimination from interstitial fluid than from plasma. This approach allows comparisons of mAb PK when only plasma data are available, provides more realistic parameters and predictions than mammillary models, and may provide an intermediate step towards utilizing full PBPK models for mAbs.
PubMedId: 23996115
Journal Article: Journal of pharmacokinetics and pharmacodynamics, vol. 40, 2013
Authors: Dhaval K Shah, Joseph P Balthasar
Abstract: We are investigating an inverse targeting strategy to reduce the dose limiting systemic toxicities resultant from intraperitoneal administration of topotecan, a model chemotherapeutic drug. This approach utilizes systemic co-administration of anti-topotecan antibodies to alter the plasma and tissue disposition kinetics of topotecan. To better predict the effects of 8C2, a high affinity anti-topotecan monoclonal antibody, on the pharmacokinetics of topotecan, two mathematical models have been developed and evaluated. Model 1 is a hybrid physiologically based pharmacokinetic (PBPK) model that was created by merging a PBPK model for topotecan with a simple two compartment model of 8C2 pharmacokinetics. Model 2 is a comprehensive PBPK model developed by merging a PBPK model for IgG with a PBPK model for topotecan. To help validate the simulation results from both the models, a tissue distribution experiment was conducted, in which topotecan and 8C2 were co-administered in mice. Experimental and simulated data were compared by calculating the median percent prediction error (%PE) for all tissues. For both models, the median %PE values for all the tissues were less than 100%, indicating that the predicted values were, on average, less than twofold the observed plasma and tissue topotecan concentrations values. In general model 2 was found to be more predictive of the data set than model 1, as the overall median %PE value for model 2 (%PE=63) was less than model 1 (%PE=73).
PubMedId: 24368689
Journal Article: Journal of pharmacokinetics and pharmacodynamics, vol. 41, 2013
Authors: Dhaval K Shah, Joseph P Balthasar
Keywords: 8C2 Enzyme linked immunosorbent assay Immunotoxicotherapy Intraperitoneal chemotherapy Inverse targeting Monoclonal antibody Pharmacokinetic/toxicodynamic model Subcutaneous bioavailability Surface plasmon resonance Topotecan
Abstract: To facilitate the development of an inverse targeting strategy, where anti-topotecan antibodies are administered to prevent systemic toxicity following intraperitoneal topotecan, a pharmacokinetic/toxicodynamic (PK/TD) model was developed and evaluated. The pharmacokinetics of 8C2, a monoclonal anti-topotecan antibody, were assessed following IV and SC administration, and the data were characterized using a two compartmental model with nonlinear absorption and elimination. A hybrid PK model was constructed by combining a PBPK model for topotecan with the two-compartment model for 8C2, and the model was employed to predict the disposition of topotecan, 8C2, and the topotecan-8C2 complex. The model was linked to a toxicodynamic model for topotecan-induced weight-loss, and simulations were conducted to predict the effects of 8C2 on the toxicity of topotecan in mice. Increasing the molar dose ratio of 8C2 to topotecan resulted in a dose-dependent decrease in the unbound (i.e., not bound to 8C2) topotecan exposure in plasma (AUCf) and a decrease in the extent of topotecan-induced weight-loss. Consistent with model predictions, toxicodynamic experiments showed substantial reduction in the percent nadir weight loss observed with 30 mg/kg IP topotecan after co-administration of 8C2 (20 8% vs. 10 8%). The investigation supports the use of anti-topotecan mAb to reduce the systemic toxicity of IP topotecan chemotherapy.
PubMedId: 24508555
Journal Article: International journal of pharmaceutics, vol. 465, 2013
Authors: Frank A Engler, Bo Zheng, Joseph P Balthasar
Abstract: This study employed a mouse model to evaluate the effects of diabetic nephropathy on the pharmacokinetics of 8C2, a murine monoclonal antibody (mAb). Streptozotocin (STZ) was administered to mice to induce diabetic nephropathy (125mg/kg/day2). Mice were grouped (n=8-10) based on time after STZ-treatment (control, 1, 2, 3, 4, or 6weeks), and injected intravenously with 10mg/kg 8C2. Blood samples were collected up to 7days, and 8C2 plasma concentrations were determined via immunoassay. Inulin clearance and urinary albumin excretion rate (UAE) were determined to assess renal function. UAE, inulin clearance, and 8C2 clearance increased significantly following STZ. Comparing control and 6week STZ-treatment groups, UAE and inulin clearance increased from 25.73.3 to 99.313.7g/day, and from 42131 to 58478l/min. 8C2 clearance increased from 12112.5 to 22861l/hr/kg (p<0.01). 8C2 clearance was highly correlated with UAE (r(2): 0.731). Inclusion of UAE as a covariate in population modeling explained significant residual variability in 8C2 clearance. The clearance of 8C2 increased significantly in STZ-treated mice. Population pharmacokinetic modeling suggests that UAE has potential for use in predicting mAb clearance in subjects with diabetic nephropathy, possibly assisting in the individualization of mAb dosing.
PubMedId: 24203494
Journal Article: Pharmaceutical research, vol. 31, 2013
Authors: Xiaotao Duan, Lubna Abuqayyas, Lipeng Dai, Joseph P. Balthasar, Jun Qu
Journal-article: Analytical Chemistry, vol. 84, pp. 4373-4382, 2012
Authors: Rong Deng, Joseph P. Balthasar
Book-chapter: Development of Antibody-Based Therapeutics, pp. 391-404, 2012
Authors: Lubna Abuqayyas, Joseph P. Balthasar
Journal-article: Molecular Pharmaceutics, vol. 10, pp. 1505-1513, 2012
Authors: Xiaotao Duan, Lipeng Dai, Shang-Chiung Chen, Joseph P. Balthasar, Jun Qu
Journal-article: Journal of Chromatography A, vol. 1251, pp. 63-73, 2012
Authors: Lubna Abuqayyas, Joseph P. Balthasar
Journal-article: International Journal of Pharmaceutics, vol. 439, pp. 8-16, 2012
Authors: Lubna Abuqayyas, Joseph P. Balthasar
Journal-article: The AAPS Journal, vol. 14, pp. 445-455, 2012
Authors: Sandhya Girish, Steven W Martin, Mark C Peterson, Lei K. Zhang, Hong Zhao, Joseph Balthasar, Raymond Evers, Honghui Zhou, Min Zhu, Lewis Klunk, Chao Han, Eva Gil Berglund, Shiew-Mei Huang, Amita Joshi
Journal-article: The AAPS Journal, vol. 13, pp. 405-416, 2011
Authors: Dhaval K. Shah, Jean Veith, Ralph J. Bernacki, Joseph P. Balthasar
Journal-article: Cancer Chemotherapy and Pharmacology, vol. 68, pp. 951-958, 2011
Authors: Jin Chen, Joseph P. Balthasar
Reference-entry: Pharmaceutical Sciences Encyclopedia, 2010
Authors: Shweta R. Urva, Victor C. Yang, Joseph P. Balthasar
Journal-article: Journal of Pharmaceutical Sciences, vol. 99, pp. 1582-1600, 2010
Authors: Beom Soo Shin, J??rgen B. Bulitta, Joseph P. Balthasar, Minki Kim, Yohan Choi, Sun Dong Yoo
Journal-article: Cancer Chemotherapy and Pharmacology, vol. 68, pp. 465-475, 2010
Authors: Dhaval K. Shah, Joseph P. Balthasar
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 38, pp. 121-142, 2010
Authors: Beom Soo Shin, Chi Ho Yoon, Joseph P. Balthasar, Bu Young Choi, Seok Hyun Hong, Hyoung Jun Kim, Jong Bong Lee, Sang Wook Hwang, Sun Dong Yoo
Journal-article: Biomedical Chromatography, vol. 23, pp. 764-769, 2009
Authors: D. K. Shah, B. S. Shin, J. Veith, K. Toth, R. J. Bernacki, J. P. Balthasar
Journal-article: Journal of Pharmacology and Experimental Therapeutics, vol. 329, pp. 580-591, 2009
Authors: Shweta R. Urva, Victor C. Yang, Joseph P. Balthasar
Journal-article: Journal of Immunoassay and Immunochemistry, vol. 31, pp. 1-9, 2009
Authors: Shweta R. Urva, Beom Soo Shin, Victor C. Yang, Joseph P. Balthasar
Journal-article: Journal of Chromatography B, vol. 877, pp. 837-841, 2009
Authors: Shweta R Urva, Victor C Yang, Joseph P Balthasar
Abstract: Carcinoembryonic antigen (CEA) is a tumor associated antigen that is over-expressed in colorectal cancer and several other cancers of the gastrointestinal system. An enzyme linked immunosorbent assay was developed to determine CEA concentrations in mouse plasma. The assay was validated over the standard curve range of 1-20 ng/mL. The intra-assay recoveries ranged from 93-104% with associated percent coefficients of variation (CV%) ranging between 2.5-12.8%. The inter-assay recoveries were in the range of 98.4-105% and their CV% values were between 4.77-10.1%. The assay was used to detect the presence of circulating CEA in the LS174T adenocarcinoma xenograft model and to study the pharmacokinetics of recombinant CEA in athymic mice.
PubMedId: 19739015
Journal Article: Journal of immunoassay & immunochemistry, vol. 30, 2009
Authors: Amit Garg, Joseph P Balthasar
Abstract: It has been suggested that the neonatal Fc receptor (FcRn) is a primary determinant of the distribution of IgG to the brain. In the present report, (125)I-labeled 7E3, a monoclonal IgG1 antibody, was injected intravenously to groups of FcRn-deficient mice and C57BL/6J control mice. Sub-groups of three mice were sacrificed at several time points. Blood and brain tissue were harvested and radioactivity was assessed. Antibody concentrations in brain were corrected for residual blood using (51)Cr-labeled red blood cells. Data were analyzed via WinNonlin, and areas under plasma and tissue concentration vs. time curves (AUCs) were assessed via the Bailer method. The apparent plasma elimination half-life and clearance of 7E3 were 13.61 +/- 0.61 days and 6.5 +/- 0.10 ml/day/kg in control mice and 0.70 +/- 0.05 days and 63.5 +/- 2.7 ml/day/kg in the knockout mice. Plasma and brain AUCs (0-10 days) were found to be 3,338.7 +/- 50.4 and 7.46 +/- 0.5 nM day in control animals and 781.2 +/- 16.6 and 1.65 +/- 0.1 nM day in FcRn-deficient animals. There was no significant difference between brain-to-plasma AUC ratios in control and FcRn-deficient mice (0.0022 +/- 0.00015 vs. 0.0021 +/- 0.00011, p = 0.3347). Two-way analysis of variance showed no significant differences, at any time point, between brain-to-plasma concentration ratios determined from control and knockout animals. The results suggest that FcRn does not contribute significantly to the "blood-brain barrier" for IgG in mice, and the data suggest that FcRn is not responsible for the low exposure of IgG in the brain relative to plasma.
PubMedId: 19636712
Journal Article: The AAPS journal, vol. 11, 2008
Authors: W Wang, E Q Wang, J P Balthasar
Abstract: More than 20 monoclonal antibodies have been approved as therapeutic drugs by the US Food and Drug Administration, and it is quite likely that the number of approved antibodies will double in the next 7-10 years. Antibody drugs show several desirable characteristics, including good solubility and stability, long persistence in the body, high selectivity and specificity, and low risk for bioconversion to toxic metabolites. However, many antibody drugs demonstrate attributes that complicate drug development, including very poor oral bioavailability, incomplete absorption following intramuscular or subcutaneous administration, nonlinear distribution, and nonlinear elimination. In addition, antibody administration often leads to an endogenous antibody response, which may alter the pharmacokinetics and efficacy of the therapeutic antibody. Antibodies have been developed for a wide range of disease conditions, with effects produced through a complex array of mechanisms. This article attempts to provide a brief overview of the main determinants of antibody pharmacokinetics and pharmacodynamics. Clinical Pharmacology & Therapeutics (2008); 84, 5, 548-558 doi:10.1038/clpt.2008.170.
PubMedId: 18784655
Journal Article: Clinical pharmacology and therapeutics, vol. 84, 2008
Authors: Beom Soo Shin, Jin Won Choi, Joseph P. Balthasar, Deok Ki Hong, Jung Jun Kim, Sun Dong Yoo
Journal-article: Rapid Communications in Mass Spectrometry, vol. 21, pp. 329-335, 2007
Authors: R. Deng, J. P. Balthasar
Journal-article: Blood, vol. 109, pp. 2470-2476, 2007
Authors: Rong Deng, Joseph P. Balthasar
Journal-article: Journal of Pharmaceutical Sciences, vol. 96, pp. 1625-1637, 2007
Authors: Amit Garg, Joseph P. Balthasar
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 34, pp. 687-709, 2007
Authors: Jin Chen, Qiang Lu, Joseph P. Balthasar
Journal-article: Journal of Pharmacokinetics and Pharmacodynamics, vol. 34, pp. 829-847, 2007
Authors: Kate E Getman, Joseph P Balthasar
Abstract: FcRn protects immune gamma globulin (IgG) from intracellular catabolism, and thereby contributes to the long plasma half-life associated with this class of antibody. The present study tested the hypothesis that 4C9, an anti-FcRn antibody, would increase the in vivo systemic clearance of a model antibody, anti-methotrexate IgG (AMI), in rats. Hybridomas secreting 4C9 and AMI were grown in serum free medium, and monoclonal 4C9 and AMI were purified via protein-G chromatography. Rats were instrumented with jugular vein cannulas 2-3 days prior to investigation, and 4C9 was administered intravenously at doses of 3, 15, and 60 mg/kg. AMI was then administered 4, 24, and 48 h after administration of 4C9. Blood samples were collected and assayed to determine AMI concentrations. The anti-FcRn antibody, 4C9, increased AMI systemic clearance in a dose-dependent manner (from 0.99+/-0.14 mg/h/kg in control animals to 1.27+/-0.05, 1.73+/-0.50, and 1.97+/-0.49 mL/h/kg in animals treated with 3, 15, and 60 mg/kg 4C9; p<0.05). These data were well-captured with an indirect-effect pharmacokinetic-pharmacodynamic model. The effect of 4C9 was found to be transient; no significant effects on AMI systemic clearance were observed when pre-treatment time was increased to 24 or 48 h. As such, the data demonstrate that 4C9, a monoclonal anti-FcRn antibody, induces a transient, dose-dependent increase in the elimination of IgG. The results suggest that FcRn inhibitors may have utility in the treatment of antibody-mediated autoimmune and alloimmune conditions.
PubMedId: 15682382
Journal Article: Journal of pharmaceutical sciences, vol. 94, 2005
Authors: Rong Deng, Joseph P Balthasar
Abstract: Immune thrombocytopenia (ITP) is an autoimmune disease that is mediated by anti-platelet antibodies. It is believed that anti-platelet antibody-opsonized platelets are eliminated through Fcgamma receptor-mediated and complement-mediated phagocytosis by macrophages of the reticuloendothelial system (RES). Polyclonal pooled immunoglobulin with high titer for the D-antigen of erythrocytes (i.e., anti-D) has been successfully used to ameliorate ITP. Based on the pathogenesis of ITP and based on the successful application of anti-D for the treatment of ITP, we hypothesized that antibody-coated liposomes may be used to inhibit Fcgamma receptor-mediated and complement-mediated phagocytosis, thereby increasing platelet counts in ITP. To test this hypothesis, we have developed a liposome preparation that is coated with a model monoclonal IgG1 antibody. Antibody-coated liposomes were found to inhibit complement deposition and macrophage phagocytosis in vitro. Furthermore, antibody-coated liposomes were also found to attenuate thrombocytopenia in a rat model of ITP, in a dose-dependent manner. The results suggest that antibody-coated liposomes may be used as 'decoy particles' to competitively inhibit the destruction of antibody-coated platelets; thus, antibody-coated liposomes may have value in the treatment of ITP.
PubMedId: 16171959
Journal Article: International journal of pharmaceutics, vol. 304, 2005
Authors: Emel Emregul, Allan David, Joseph P Balthasar, Victor C Yang
Abstract: Immune thrombocytopenic purpura (ITP) is an autoimmune disease that affects thousands of Americans each year. The resulting thrombocytopenia, which develops from destruction of platelets (PLT) by anti-PLT autoantibodies (APAb), is often associated with hemorrhagic complications. Existing therapies are not effective and are associated with significant morbidity. Recently, a new treatment modality using plasmapheresis with a Protein-A column has shown some clinical promise. Yet, although this method would remove the pathogenic APAb, it would also deplete protective antibodies, thereby weakening the body's self-defense system. Because about 80% of patients with ITP develop APAb against the GPIIb/IIIa antigens on PLT, a novel approach of attaching a GPIIb/IIIa-linked bioreactor with an extracorporeal circuit is suggested herein to achieve highly effective/specific APAb removal and overcome shortcomings of plasmapheresis in treating ITP. A hollow fiber-based bioreactor device was fabricated, and GPIIb/IIIa antigens were immobilized onto the inner lumens of the hollow fibers by using the epichlorohydrin activation method. An optimized bioreactor containing a loading of 1.63 mg GPIIb/IIIa/g fibers and adsorption capacity of 1.9 mg 7E3/g fibers was developed. Preliminary proof-of-concept investigation using a 7E3-induced thrombocytopenic rat model (which mimicked clinical ITP) was carried out. A complete (100%) return of PLT counts to their initial levels was observed in rats within 6 h after the GPIIb/IIIa bioreactor treatment. In addition, a rapid restoration of WBC counts in the treated rats was also found. These preliminary findings shed light of promise of using the GPIIb/IIIa bioreactor approach in achieving highly improved ITP therapy.
PubMedId: 16108027
Journal Article: Journal of biomedical materials research. Part A, vol. 75, 2005
Authors: Tanya Wang, Zhiqiang Yang, Emel Emregul, Allan David, Joseph P Balthasar, Junfeng Liang, Victor C Yang
Abstract: Heparin employed in extracorporeal blood circulation (ECBC) procedures (e.g. open heart operations) often leads to a high incidence of bleeding complications. Protamine employed in heparin neutralization, on the other hand, can cause severe adverse reactions. We previously developed an approach that could prevent both heparin- and protamine-induced toxic side effects concomitantly. This approach consisted of placing a hollow fiber-based bioreactor device containing immobilized protamine (termed a "protamine bioreactor") at the distal end of the ECBC procedure. This protamine bioreactor would remove heparin after heparin served its anticoagulant purpose in the ECBC device, thereby eliminating heparin-induced bleeding risks. In addition, this protamine bioreactor would prevent protamine from entering the patients, thereby aborting any protamine-induced toxic effects. Both in vitro and in vivo studies have successfully demonstrated the feasibility of this approach. Despite promises, early findings also revealed two shortcomings that must be overcome for the protamine bioreactor to be applied clinically. The first drawback was that the cyanate ester linkages, involved in conjugating protamine to the bioreactor device, were unstable and prone to hydrolysis, resulting in the leakage of a significant amount of protamine into circulation during application of the protamine bioreactor. The second deficiency was that the capacity of the protamine bioreactor in heparin removal was rather low, owing to the limited surface area of the hollow fibers for protamine immobilization and subsequently heparin adsorption. In this paper, we present novel strategies to overcome these two limitations. A new conjugation method based on the use of 4-(oxyacetyl)phenoxyacetic acid (OAPA) as the activating reagent was employed to yield stable linkages, via the abundant arginine residues of protamine, onto the hollow fibers. Results showed that while the amount of protamine immobilized on each gram of fibers was relatively comparable between the OAPA and the previous CNBr activation methods (7.45 mg/g versus 7.69 mg/g fibers), there was virtually no detectable leaching of immobilized protamine from the bioreactor by the OAPA method, comparing to 35% leaching of protamine by the previous CNBr method following 72 h of storage of the bioreactor in PBS buffer at 37 degrees C. To improve the capacity and functionality of the protamine bioreactor, two novel approaches were adopted. Long chain and high molecular weight poly-lysine was linked to the hollow fibers, prior to protamine coupling, to create multiple layers of immobilized protamine for subsequent heparin adsorption. In addition, a poly(ethylene glycol) (PEG) chain was inserted between protamine and the hollow fibers to yield a three-dimensional, free dynamic motion for immobilized protamine. Preliminary observations indicated that a four- to five-fold enhancement in heparin adsorption was attained by utilizing each of these new approaches. Aside from their current use, these new strategies can also be employed generically to improve the functionality of any affinity-type bioreactor. Indeed, efforts have been made recently in utilizing these approaches to develop a clinically usable GPIIb/IIIa bioreactor for the treatment of immune thrombocytopenic purpura (ITP)-an autoimmune disease.
PubMedId: 16246511
Journal Article: International journal of pharmaceutics, vol. 306, 2005
Authors: Jin Chen, Joseph P Balthasar
Abstract: A rapid and sensitive high-performance liquid chromatographic (HPLC) assay has been developed to allow determination of total (i.e. bound and unbound) and free (i.e. unbound) topotecan (TPT) in mouse plasma in the presence and absence of anti-TPT antibodies. The chromatographic analysis was carried out using reversed-phase isocratic elution with a Nova-Pak C18 column (3.9 mm x 150 mm, 4 microm) protected by a Nova-Pak C18 guard column (3.9 mm x 20 mm, 4 microm), where 10 mM KH(2)PO(4)-methanol-triethylamine (72:26:2 (v/v/v), pH 3.5) was used as the mobile phase. Topotecan was quantified with fluorescence detection using an excitation wavelength of 361 nm and an emission wavelength of 527 nm. The retention time for the internal standard, acridine, and TPT were 7.4 and 9.0 min, respectively. The lower limit of quantitation (LOQ) for TPT was determined as 0.02 ng in mouse plasma and mouse plasma ultrafiltrate, corresponding to a concentration of 1 ng/ml in 20 microl mouse plasma. The assay was shown to be linear over a concentration range of 1-500 ng/ml. The recoveries of free and total TPT from spiked mouse plasma were within 10% of theoretical values (assessed at 1, 20 and 500 ng/ml). The validated HPLC assay was applied to evaluate TPT pharmacokinetics following administration of TPT to Swiss Webster mice and to hyperimmunized and control BALB/c mice. The assay has been shown to be capable for measuring total and free TPT in mouse plasma with high sensitivity and will allow the testing of the effect of anti-TPT antibodies on the disposition of TPT.
PubMedId: 15664349
Journal Article: Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, vol. 816, 2004
Authors: Evelyn D Lobo, Ryan J Hansen, Joseph P Balthasar
Abstract: The U.S. Food and Drug administration (FDA) has approved several polyclonal antibody preparations and at least 18 monoclonal antibody preparations (antibodies, antibody fragments, antibody fusion proteins, etc.). These drugs, which may be considered as a diverse group of therapeutic proteins, are associated with several interesting pharmacokinetic characteristics. Saturable binding with target antigen may influence antibody disposition, potentially leading to nonlinear distribution and elimination. Independent of antigen interaction, concentration-dependent elimination may be expected for IgG antibodies, due to the influence of the Brambell receptor, FcRn, which protects IgG from catabolism. Antibody administration may induce the development of an endogenous antibody response, which may alter the pharmacokinetics of the therapeutic antibody. Additionally, the pharmacodynamics of antibodies are also complex; these drugs may be used for a wide array of therapeutic applications, and effects may be achieved by a variety of mechanisms. This article provides an overview of many of the complexities associated with antibody pharmacokinetics and pharmacodynamics.
PubMedId: 15389672
Journal Article: Journal of pharmaceutical sciences, vol. 93, 2004
Authors: Zia R. Tayab, Joseph P. Balthasar
Journal-article: Journal of Immunoassay and Immunochemistry, vol. 25, pp. 335-344, 2004
Authors: Feng Jin, Joseph P Balthasar
Abstract: The use of high-dose intravenous gamma immunoglobulin (IVIG) for the treatment of immune thrombocytopenic purpura (ITP) was first reported more than two decades ago. After the therapeutic benefit of IVIG was established in ITP, it was then successfully used to treat many other autoimmune diseases. Although a complete definition of the mechanism of IVIG action is still lacking, extensive research suggests that IVIG may achieve its therapeutic effects through multiple mechanisms. IVIG exerts immunomodulatory effects that may include antiidiotypic neutralization of antiplatelet antibodies, stimulation of Fcgamma receptor IIB expression, and inhibition of Fcgamma receptor-mediated platelet destruction. Recent work suggests that a large fraction of the benefit provided by IVIG may be the result of competitive inhibition of neonatal Fc receptor (FcRn) and IVIG-induced acceleration of antiplatelet antibody elimination. This review provides an overview and critical discussion of mechanisms that may be responsible of IVIG effects in ITP.
PubMedId: 15866704
Journal Article: Human immunology, vol. 66, 2004
Authors: Ellen Q. Wang, Joseph P. Balthasar, Ho-Leung Fung
Journal-article: Pharmaceutical Research, vol. 21, pp. 114-120, 2004
Authors: Zia R Tayab, Joseph P Balthasar
Abstract: This laboratory is investigating the use of anti-methotrexate IgG (AMI) and anti-methotrexate Fab fragments (AMF) within an inverse targeting strategy that is designed to enhance the pharmacokinetic selectivity of intraperitoneal (i.p.) chemotherapy. The goal of this study was to develop enzyme-linked immunosorbent assays (ELISAs) to determine concentrations of AMI and AMF in mouse and rat plasma. An antigen-specific ELISA was developed for AMI and AMF in mouse and rat plasma. The assay was validated with respect to precision and accuracy by evaluating the recovery of AMI and AMF from mouse and rat plasma samples. Preliminary pharmacokinetic studies of AMI and AMF were performed in Sprague-Dawley rats and Swiss Webster mice. The animals were instrumented with a jugular vein cannula and administered AMI or AMF, 15 mg kg(-1) via the cannula. Plasma samples were taken at various time points and analyzed using the ELISA, and the observed concentration vs. time profiles were subjected to non-compartmental pharmacokinetic analyses. Standard curves for the ELISAs were found to be linear over concentration ranges of 0-250 and 0-350 ng mL(-1) for AMI and AMF, respectively. Intra-assay and inter-assay recovery of AMI and AMF from plasma samples were found to be within 15% of theoretical values. Preliminary pharmacokinetic investigations of AMI allowed estimation of AMI clearance to be 0.017 mL kg(-1) min(-1) in the rat and 0.043 mL kg(-1) min(-1) in the mouse. AMF clearance was estimated to be 0.038 and 1.93 mL kg(-1) min(-1) in the mouse and rat, respectively. In conclusion, ELISAs have been developed and validated for quantitation of AMI and AMF in rat and mouse plasma. The assays will allow further investigations of AMI and AMF pharmacokinetics.
PubMedId: 15552588
Journal Article: Journal of immunoassay & immunochemistry, vol. 25, 2004
Authors: Ryan J Hansen, Joseph P Balthasar
Abstract: Intravenous immunoglobulin (IVIG) has been an important therapy of immune thrombocytopenic purpura (ITP) for more than 20 years; however, questions still remain regarding the mechanisms of IVIG action in ITP. Recent reviews have focused on the hypotheses that IVIG effects are mediated via inhibition of Fc-receptor mediated platelet phagocytosis, suppression of anti-platelet antibody production, and anti-idiotypic inhibition of anti-platelet antibodies; however, new research suggests that a significant portion of IVIG benefit may be due to IVIG-mediated acceleration of the elimination of anti-platelet antibodies. It is anticipated that promising new therapies of ITP, and other autoimmune conditions, will be developed based on a better understanding of IVIG mechanism of action.
PubMedId: 15074465
Journal Article: Clinical laboratory, vol. 50, 2004
Authors: Ryan J Hansen, Joseph P Balthasar
Abstract: We have recently shown that intravenous immunoglobulin (IVIG) therapy leads to an increased rate of anti-platelet antibody clearance in an animal model of immune thrombocytopenia. The present study was performed to confirm the importance of the FcRn receptor in mediating this effect of IVIG. The pharmacokinetics of an anti-platelet antibody, 7E3, were studied in mice lacking expression of FcRn and in control mice, both in the presence and absence of IVIG. IVIG increased the clearance of 7E3 in mice with functioning FcRn receptors, with an average clearance value of 14.4 +/- 1.4 ml/d/kg in IVIG treated mice vs. 5.2 +/- 0.3 ml/d/kg in controls (P <0.001). In mice lacking expression of FcRn, IVIG treatment did not increase 7E3 clearance (61.0 +/- 3.6 ml/d/kg vs. 71.5 +/- 4.0 ml/d/kg in controls). Thus, these data support the hypothesis that IVIG increases antibody elimination via saturation of FcRn.
PubMedId: 12529736
Journal Article: Thrombosis and haemostasis, vol. 88, 2003
Authors: Evelyn D Lobo, Joseph P Balthasar
Abstract: The time course of chemotherapeutic effect is often delayed relative to the time course of chemotherapeutic exposure. In many cases, this delay is difficult to characterize mathematically through the use of standard pharmacodynamic models. In the present work, we investigated the relationship between methotrexate (MTX) exposure and the time course of MTX effects on tumor cell growth in culture. Two cancer cell lines, Ehrlich ascites cells and sarcoma 180 cells, were exposed for 24 hours to MTX concentrations that varied more than 700-fold (0.19-140 micro g/mL). Viable cells were counted on days 1, 3, 5, 7, 9, 11, 13, 15, 17, 20, 22, and 24 for Ehrlich ascites cells and on days 1, 2, 3, 5, 7, 9, 11, 13, 14, 15, 17, 19, and 21 for sarcoma 180 cells, through the use of a tetrazolium assay. Although MTX was removed 24 hours after application, cell numbers reached nadir values more than 100 hours after MTX exposure. Data from each cell line were fitted to 3 pharmacodynamic models of chemotherapeutic cell killing: a cell cycle phase-specific model, a phase-nonspecific model, and a transit compartment model (based on the general model recently reported by Mager and Jusko, Clin Pharmacol Ther. 70:210-216, 2001). The transit compartment model captured the data much more accurately than the standard pharmacodynamic models, with correlation coefficients ranging from 0.86 to 0.999. This report shows the successful application of a transit compartment model for characterization of the complex time course of chemotherapeutic effects; such models may be very useful in the development of optimization strategies for cancer chemotherapy.
PubMedId: 12646013
Journal Article: AAPS pharmSci, vol. 4, 2003
Authors: Evelyn D Lobo, Joseph P Balthasar
Abstract: The prediction of chemotherapeutic efficacy is complicated by "protocol dependencies" in dose-effect and dose-toxicity relationships. It has been proposed that pharmacokinetic-pharmacodynamic mathematical models may allow characterization of chemotherapeutic protocol dependencies, and may facilitate the prediction of chemotherapeutic efficacy; however, few demonstrations exist in the literature. The present study examines the pharmacokinetics and toxicodynamics of methotrexate (MTX), a commonly used anticancer agent, after intraperitoneal (i.p.) administration to mice. MTX was administered via bolus or infusion (24, 72, and 168 h), at doses of 2.5-1000 mg/kg. MTX plasma and peritoneal pharmacokinetics were characterized through standard noncompartmental and compartmental techniques. Body weight loss was used as a measure of MTX-induced toxicity. We found that MTX pharmacokinetics were independent of dose (over a range of 3-600 mg/kg) and independent of dosing mode (i.e., i.p. bolus vs. i.p. infusion). However, MTX-induced toxicity was shown to be highly dependent on the dosing protocol used. For example, the maximally tolerated dose (i.e., the dose related to a mean body weight loss of 10%) was 200-fold greater after bolus administration relative to that observed for 72-h infusion (760 mg/kg vs. 3.8 mg/kg). This profound protocol dependence in the relationship between MTX-induced toxicity and MTX exposure was characterized through the use of a time-dissociated pharmacokinetic-pharmacodynamic model (median prediction error: 3.9%).
PubMedId: 12884252
Journal Article: Journal of pharmaceutical sciences, vol. 92, 2003
Authors: Ryan Hansen, Joseph Balthasar
Journal-article: Thrombosis and Haemostasis, vol. 88, pp. 898-899, 2002
Authors: Ryan J Hansen, Joseph P Balthasar
Abstract: Experiments were conducted to investigate the effects of intravenous immunoglobulin (IVIG) in a rat model of immune thrombocytopenia (ITP). Rats were pretreated with 0 to 2 g/kg IVIG and then challenged with an antiplatelet antibody (7E3, 8 mg/kg). IVIG effects on 7E3-induced thrombocytopenia and on 7E3 pharmacokinetics were determined. IVIG pretreatment led to significant changes in the degree and time-course of 7E3-induced thrombocytopenia (P =.031). Nadir percent platelet counts were 121% to 279% greater in animals treated with IVIG (0.4-2 g/kg) than in animals receiving 7E3 alone. IVIG treatment also led to dose-dependent increases in 7E3 clearance (P <.001), with more than 2-fold increases in 7E3 clearance seen following the highest dose of IVIG. In vitro experiments showed that IVIG effects on platelet count are not likely due to anti-idiotypic inhibition of 7E3-platelet binding and that IVIG did not directly bind to 7E3. Consequently, IVIG-7E3 binding cannot explain the increase of 7E3 clearance following IVIG treatment. We propose that the observed increase in 7E3 clearance with IVIG therapy is due to saturation of the FcRn salvage receptor for IgG. The importance of the effect of IVIG on 7E3 clearance to the prevention of thrombocytopenia in these animals is unclear at present; nonetheless, these data provide experimental support for a new mechanism of IVIG action in ITP (ie, IVIG-mediated increases in antiplatelet antibody elimination). This model of ITP will be useful for further investigations of IVIG mechanism of action and for development of new therapies for ITP.
PubMedId: 12200371
Journal Article: Blood, vol. 100, 2002
Authors: R J Hansen, J P Balthasar
Abstract: The pharmacokinetics, pharmacodynamics, and platelet binding of 7E3, an anti-glycoprotein IIb/IIIa (GPIIb/IIIa) monoclonal antibody, were studied in the rat in an attempt to develop a quantitative animal model of immune thrombocytopenia (ITP). 7E3, a murine IgG1 antibody developed against human GPIIb/IIIa, demonstrated cross-reactivity with rat platelets by flow cytometry and via enzyme-linked immunosorbent assay. The apparent affinity (K(A)) of 7E3-rat platelet binding was 1.2 +/- 0.2 x 10(7) M(-1), with 3.3 +/- 0.3 x 10(4) binding sites per platelet. Following intravenous 7E3 administration (0.8, 4, and 8 mg/kg), plasma concentrations declined in a bi-exponential manner, with a terminal half-life of 61 +/- 5 h and a steady-state volume of distribution of 62 +/- 15 ml/kg. Clearance was dose-dependent, with values ranging from 0.64 +/- 0.08 ml/h/kg (8 mg/kg) to 1.01 +/- 0.08 ml/h/kg (0.8 mg/kg). 7E3 induced a reproducible, severe thrombocytopenia in rats and extended bleeding in a manner consistent with human ITP. Nadir platelet counts were 79 +/- 33, 25 +/- 6, and 17 +/- 2 x 10(6)/ml, for 7E3 doses of 0.8, 4, and 8 mg/kg, respectively. Bleeding times after a 10-mm tail incision ranged from 5 +/- 3 min in control animals to 15 +/- 0 min (the maximum allowed time in this study) in animals receiving 8 mg/kg. Blood volumes lost during bleeding experiments ranged from 30 +/- 24 microl (control) to 349 +/- 358 microl (8 mg/kg). A reproducible, quantitative rat model of ITP has been created; this model is expected to facilitate the evaluation of new treatments for this disease.
PubMedId: 11408538
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 298, 2001
Authors: R J Hansen, J P Balthasar
Abstract: An enzyme-linked immunosorbent assay (ELISA) has been developed to determine concentrations of murine IgG in rat plasma. Specifically, the assay was developed to measure a murine anti-glycoprotein IIIa antibody (AP-3) in rat plasma to facilitate future investigations of AP-3 pharmacokinetics and pharmacodynamics in the rat. The working range of the assay is 15-100 ng ml(-1), corresponding to a limit of quantification of 1.5 microg ml(-1) in rat plasma. The assay was validated with respect to accuracy, precision, and cross-reactivity with both pooled rat and mouse IgG. Intra-assay recoveries of AP-3 in rat plasma ranged from 93 to 103%, with CV% values ranging from 5.2 to 8.5%. Inter-assay recoveries of the plasma AP-3 samples ranged from 107 to 119% with CV% values ranging from 17.7 to 25.1%. The assay has no appreciable cross reactivity with pooled rat IgG and full cross reactivity with pooled mouse IgG, making this an ideal assay to determine plasma pharmacokinetics of mouse antibodies in the rat. The assay was used to determine the pharmacokinetics of AP-3 in a Sprague-Dawley rat.
PubMedId: 10703969
Journal Article: Journal of pharmaceutical and biomedical analysis, vol. 21, 2000
Authors: E D Lobo, J P Balthasar
Abstract: Recently, Balthasar and Fung have proposed that anti-methotrexate antibody fragments may be employed to enhance the selectivity of intraperitoneal methotrexate (MTX) therapy. This current work presents a sensitive high-performance liquid chromatographic method for measuring plasma concentrations of total (i.e., bound and unbound) MTX and free (unbound) MTX in rat and mouse plasma, in the presence or absence of therapeutic anti-MTX antibody fragments. The assay involves pre-column derivatization of MTX by sodium hydrosulfite to 2,4-diamino-6-methylpteridine. The limit of quantitation for MTX by this assay was 1.25 ng in rat plasma, mouse plasma and mouse plasma ultrafiltrate, which corresponds to a concentration of 25 ng/ml for a 50 microl sample. The limit of quantitation was found to be 2.5 ng in rat plasma ultrafiltrate (i.e., 50 ng/ml in 50 microl rat plasma ultrafiltrate). The method was shown to be quite accurate, as the mean assayed concentration of quality control samples was within 10% of theoretical values. We have applied the method to the investigation of MTX pharmacokinetics in mice and rats, following the administration of MTX alone or following simultaneous administration of MTX and anti-MTX Fab fragments. The method has been shown to be suitable for the assay of total and free methotrexate in the plasma of these species and will enable the testing of pharmacokinetic hypotheses regarding the influence of anti-MTX Fab fragments on the disposition of MTX.
PubMedId: 10676999
Journal Article: Journal of chromatography. B, Biomedical sciences and applications, vol. 736, 2000
Authors: Ryan J Hansen, Joseph P Balthasar
Journal-article: Journal of Pharmaceutical and Biomedical Analysis, vol. 21, pp. 1011-1016, 1999
Authors: Evelyn D Lobo, Joseph P Balthasar
Journal-article: Journal of Chromatography B: Biomedical Sciences and Applications, vol. 736, pp. 191-199, 1999
Authors: J A Bauer, J P Balthasar, H L Fung
Abstract: Prolonged continuous administration of nitroglycerin (NTG) leads to hemodynamic tolerance. We used a previously developed pharmacokinetic-pharmacodynamic (PK/PD) model of NTG tolerance in experimental heart failure to test whether dosage regimens, designed from this model, may allow avoidance of tolerance development upon continuous NTG infusion. Simulation experiments (using ADAPT II) were performed to evolve a time-variant infusion regimen that would theoretically provide sustained hemodynamic effect (30% reduction in left ventricular end-diastolic pressure, LVEDP) throughout 10 hours of drug dosing. A computer controlled infusion pump was utilized to deliver this time-variant input. Infusion experiments were then conducted in CHF rats to challenge the predictability of the applied PK/PD model. Simulations showed that exponentially increasing input functions provided more sustained LVEDP effects when compared to linear or hyperbolic input functions delivering the same total NTG dose. A computer-selected infusion regimen of 6.56e0.00156 x minutes micrograms/min was anticipated to provide the desired hemodynamic profile in our animal model. Experiments conducted in rats with congestive heart failure (n = 4) confirmed the prediction of sustained hemodynamic effect without tolerance (28 +/- 4% reduction in LVEDP at 10 hrs). These findings support the utility of our PK/PD model of NTG tolerance in predicting NTG action, and serve as an example of therapeutic optimization through PK/PD considerations.
PubMedId: 9327439
Journal Article: Pharmaceutical research, vol. 14, 1997
Authors: Joseph P. Balthasar, Ho-Leung Fung
Journal-article: Life Sciences, vol. 58, pp. 535-543, 1996
Authors: J P Balthasar, H L Fung
Abstract: We have hypothesized that antidrug antibodies (ADAb) may be employed to impart site-specific alterations in the disposition of drug molecules, potentially allowing for targeted drug therapy. We are specifically interested in minimizing systemic exposure to free drug and systemic toxicities resultant from regional chemotherapy through the intravenous administration of ADAb. In this report, we present the production and purification of anti-methotrexate Fab fragments, and we present investigations of the effects of anti-methotrexate Fab and anti-methotrexate immunoglobulin G on the disposition of methotrexate in the rat. Pharmacokinetic studies revealed that intravenous anti-methotrexate immunoglobulin G (anti-MTX IgG) and anti-methotrexate Fab (anti-MTX Fab) administration produced dramatic alterations in the plasma pharmacokinetics of methotrexate (MTX), following intraperitoneal MTX administration (area under the total MTX concentration vs time curve for anti-MTX IgG relative to control, 420 +/- 90 (p < 0.05); for anti-MTX Fab relative to control, 46 +/- 6.1 (p < 0.05); area under the free MTX concentration vs time curve for anti-MTX IgG relative to control, 0.64 +/- 0.16; for anti-MTX Fab relative to control, 0.45 +/- 0.20 (p < 0.05)). Additional studies conducted in anesthetized rats revealed no significant alterations in the area under the total peritoneal MTX concentration vs time curves, free MTX peritoneal concentration vs time curves, or peritoneal exit rate of MTX in anti-MTX Fab treated animals relative to controls. Therefore, our pharmacokinetic studies demonstrate that ADAb may produce site-specific alterations in drug pharmacokinetics, potentially enhancing the site specificity of drug distribution and drug action following regional chemotherapy.
PubMedId: 8897267
Journal Article: Journal of pharmaceutical sciences, vol. 85, 1996
Authors: J P Balthasar, H L Fung
Abstract: Intraperitoneal drug administration has been utilized to increase chemotherapeutic exposure to tumors confined to the peritoneal cavity, such as those found in ovarian, gastric, colo-rectal and mesotheliomal cancers. Extensive pre-clinical and clinical experimentation has been conducted to assess the pharmacokinetic and therapeutic benefits of this mode of therapy. Pharmacokinetic studies have shown that the barrier function of the peritoneal membrane may be utilized to produce large, favorable concentration gradients between peritoneal perfusate and blood. However, most clinical studies so far have demonstrated minimal increases in drug efficacy or decreases in drug toxicities from intraperitoneal drug therapy alone. This paper reviews the application of adjunctive therapies that have been rationally conceived to optimize intraperitoneal drug therapy through the alteration of antineoplastic pharmacokinetics and pharmacodynamics.
PubMedId: 8632706
Journal Article: Life sciences, vol. 58, 1996
Authors: Joseph P. Balthasar, Ho???Leung Fung
Journal-article: Journal of Pharmaceutical Sciences, vol. 84, pp. 2-6, 1995
Authors: J P Balthasar, H L Fung
Abstract: We describe the production, purification, characterization, and disposition of rabbit polyclonal methotrexate antibodies. These antibodies are prepared for subsequent testing of a drug delivery approach to reduce systemic toxicity upon regional administration of methotrexate. The polyclonal antibodies were raised in New Zealand white female rabbits immunized with a methotrexate-bovine serum albumin conjugate. The anti-methotrexate antibodies were sequestered from rabbit serum through the use of a protein-G affinity column which allowed for purification of up to 100 mg of rabbit IgG in 30-40 min. The extent of purification was demonstrated through SDS-PAGE and calculation of specific binding activity relative to total protein concentration. The purified antibodies have been shown to have high affinity (Keq range: 1.8 x 10(8) to 8.75 x 10(9) M-1) and high selectivity for methotrexate. Preliminary pharmacokinetic studies of the purified antibodies in the rat following a 6 mg/kg intravenous infusion (n = 4) indicate a steady state volume of distribution of 38.0 +/- 11.2 mL kg-1, a systemic clearance of 0.92 +/- 0.67 mL kg-1 h-1 and an elimination half life of 28.9 +/- 7.9 h (mean +/- SD).
PubMedId: 7714737
Journal Article: Journal of pharmaceutical sciences, vol. 84, 1995
Authors: J Balthasar, H L Fung
Abstract: The direct administration of chemotherapeutic agents into the peritoneal cavity has been investigated as a method to treat cancers residing within the peritoneum. The benefits of i.p. drug administration are limited, however, by the systemic toxicity of antineoplastic drugs which diffuse out of the peritoneum and into the general circulation. We propose that antidrug antibody fragments may be useful in binding chemotherapeutics in the general circulation, thereby reducing the systemic tissue exposure and toxicity resulting from such i.p. therapy. Inasmuch as antibody fragments directed against antineoplastic agents are not available, we tested our hypothesis by using i.v. administered ovine antidigoxin Fab fragments and determined their ability to limit digoxin tissue exposure and toxicity in mice after an i.p. digoxin injection. The rate of digoxin disappearance from the peritoneal cavity and the fraction of digoxin unbound in the peritoneal cavity were also assessed to determine the effect of the antibody fragments on peritoneal exposure. Our results showed that the antidigoxin antibody fragments can greatly decrease digoxin tissue exposure and toxicity without affecting peritoneal exposure, unbound fraction of digoxin in the peritoneum or peritoneal digoxin disappearance rate. Although the utility of drug-binding antibodies and antibody fragments for the treatment of drug intoxication is well known, these results demonstrated the potential ability of antidrug antibody fragments to improve the site-specificity of drug therapy.
PubMedId: 8113985
Journal Article: The Journal of pharmacology and experimental therapeutics, vol. 268, 1994
Authors: Jin Chen, Joseph P. Balthasar
Book-chapter: Handbook of Pharmaceutical Biotechnology, pp. 835-850,
Presented by CUBS © 2024