5 results on '"Alison Betts"'
Search Results
2. A Translational Quantitative Systems Pharmacology Model for CD3 Bispecific Molecules: Application to Quantify T Cell-Mediated Tumor Cell Killing by P-Cadherin LP DART®
- Author
-
Alison Betts, Lindsay King, Frank Barletta, Piet H. van der Graaf, Dhaval K. Shah, Xiaoying Chen, Tracey Clark, Andrea T. Hooper, Nahor Haddish-Berhane, Cris Kamperschroer, and Adam Root
- Subjects
CD3 Complex ,CD3 bispecific ,medicine.medical_treatment ,CD3 ,T cell ,Pharmaceutical Science ,Antineoplastic Agents ,Mice, SCID ,Lymphocyte Activation ,Models, Biological ,030226 pharmacology & pharmacy ,Immunological synapse ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,PK/PD models ,biology ,Chemistry ,PK/PD ,Correction ,Immunotherapy ,Cadherins ,HCT116 Cells ,Xenograft Model Antitumor Assays ,In vitro ,Macaca fascicularis ,Cytolysis ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,quantitative systems pharmacology ,translational modeling ,immunotherapy ,T-Lymphocytes, Cytotoxic ,Research Article - Abstract
CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species. Electronic supplementary material The online version of this article (10.1208/s12248-019-0332-z) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
3. Preclinical to Clinical Translation of Antibody-Drug Conjugates Using PK/PD Modeling: a Retrospective Analysis of Inotuzumab Ozogamicin
- Author
-
Lindsay King, Boris Shor, Joseph Boni, Alison Betts, Subramanyam Chakrapani, John E. Tolsma, Nahor Haddish-Berhane, Theodore R. Johnson, Paul Jasper, and Yongliang Sun
- Subjects
0301 basic medicine ,Antibody-drug conjugate ,Sialic Acid Binding Ig-like Lectin 2 ,Receptor expression ,Drug Evaluation, Preclinical ,Mice, Nude ,Pharmaceutical Science ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Translational Research, Biomedical ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Cell Line, Tumor ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,medicine ,Animals ,Humans ,Computer Simulation ,Inotuzumab Ozogamicin ,PK/PD models ,Retrospective Studies ,Inotuzumab ozogamicin ,Clinical Trials as Topic ,business.industry ,CD22 ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Immunoglobulin G ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Female ,business ,medicine.drug - Abstract
A mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) model was used for preclinical to clinical translation of inotuzumab ozogamicin, a CD22-targeting antibody-drug conjugate (ADC) for B cell malignancies including non-Hodgkin's lymphoma (NHL) and acute lymphocytic leukemia (ALL). Preclinical data was integrated in a PK/PD model which included (1) a plasma PK model characterizing disposition and clearance of inotuzumab ozogamicin and its released payload N-Ac-γ-calicheamicin DMH, (2) a tumor disposition model describing ADC diffusion into the tumor extracellular environment, (3) a cellular model describing inotuzumab ozogamicin binding to CD22, internalization, intracellular N-Ac-γ-calicheamicin DMH release, binding to DNA, or efflux from the tumor cell, and (4) tumor growth and inhibition in mouse xenograft models. The preclinical model was translated to the clinic by incorporating human PK for inotuzumab ozogamicin and clinically relevant tumor volumes, tumor growth rates, and values for CD22 expression in the relevant patient populations. The resulting stochastic models predicted progression-free survival (PFS) rates for inotuzumab ozogamicin in patients comparable to the observed clinical results. The model suggested that a fractionated dosing regimen is superior to a conventional dosing regimen for ALL but not for NHL. Simulations indicated that tumor growth is a highly sensitive parameter and predictive of successful outcome. Inotuzumab ozogamicin PK and N-Ac-γ-calicheamicin DMH efflux are also sensitive parameters and would be considered more useful predictors of outcome than CD22 receptor expression. In summary, a multiscale, mechanism-based model has been developed for inotuzumab ozogamicin, which can integrate preclinical biomeasures and PK/PD data to predict clinical response.
- Published
- 2016
- Full Text
- View/download PDF
4. Quantitative Prediction of Human Pharmacokinetics for mAbs Exhibiting Target-Mediated Disposition
- Author
-
Pratap Singh, Alison Betts, Gregory Weber, Alaa Ahmad, Anson K. Abraham, John C. Lin, Wojciech Krzyzanski, Anup Zutshi, Aman P. Singh, and Steven W. Martin
- Subjects
Drug disposition ,Chemistry ,medicine.drug_class ,Drug Evaluation, Preclinical ,Antibodies, Monoclonal ,Pharmaceutical Science ,Disposition ,Pharmacology ,Monoclonal antibody ,Models, Biological ,Preclinical data ,In vitro ,Translational Research, Biomedical ,Macaca fascicularis ,Model parameter ,Nonlinear Dynamics ,Species Specificity ,Pharmacokinetics ,In vivo ,medicine ,Animals ,Humans ,Research Article - Abstract
Prediction of human pharmacokinetics (PK) can be challenging for monoclonal antibodies (mAbs) exhibiting target-mediated drug disposition (TMDD). In this study, we performed a quantitative analysis of a diverse set of six mAbs exhibiting TMDD to explore translational rules that can be utilized to predict human PK. A TMDD model with rapid-binding approximation was utilized to fit PK and PD (i.e., free and/or total target levels) data, and average absolute fold error (AAFE) was calculated for each model parameter. Based on the comparative analysis, translational rules were developed and applied to a test antibody not included in the original analysis. AAFE of less than two-fold was observed between monkey and human for baseline target levels (R 0), body-weight (BW) normalized central elimination rate (K el/BW(-0.25)) and central volume (V c/BW(1.0)). AAFE of less than three-fold was estimated for the binding affinity constant (K D). The other four parameters, i.e., complex turnover rate (K int), target turnover rate (K deg), central to peripheral distribution rate constant (K pt) and peripheral to central rate constant (K tp) were poorly correlated between monkey and human. The projected human PK of test antibody based on the translation rules was in good agreement with the observed nonlinear PK. In conclusion, we recommend a TMDD model-based prediction approach that integrates in vitro human biomeasures and in vivo preclinical data using translation rules developed in this study.
- Published
- 2014
- Full Text
- View/download PDF
5. Evolution of Antibody-Drug Conjugate Tumor Disposition Model to Predict Preclinical Tumor Pharmacokinetics of Trastuzumab-Emtansine (T-DM1)
- Author
-
K. Dane Wittrup, Alison Betts, Chethana Kulkarni, Lindsay King, Antari Khot, Katie F. Maass, Aman P. Singh, and Dhaval K. Shah
- Subjects
Antibody-drug conjugate ,Immunoconjugates ,Receptor, ErbB-2 ,Cell ,Pharmaceutical Science ,Antineoplastic Agents ,Breast Neoplasms ,Pharmacology ,Antibodies, Monoclonal, Humanized ,030226 pharmacology & pharmacy ,Models, Biological ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,In vivo ,Neoplasms ,medicine ,Animals ,Humans ,Maytansine ,business.industry ,Disposition ,Trastuzumab ,body regions ,medicine.anatomical_structure ,chemistry ,Trastuzumab emtansine ,030220 oncology & carcinogenesis ,Cancer research ,Cellular model ,business ,Intracellular - Abstract
A mathematical model capable of accurately characterizing intracellular disposition of ADCs is essential for a priori predicting unconjugated drug concentrations inside the tumor. Towards this goal, the objectives of this manuscript were to: (1) evolve previously published cellular disposition model of ADC with more intracellular details to characterize the disposition of T-DM1 in different HER2 expressing cell lines, (2) integrate the improved cellular model with the ADC tumor disposition model to a priori predict DM1 concentrations in a preclinical tumor model, and (3) identify prominent pathways and sensitive parameters associated with intracellular activation of ADCs. The cellular disposition model was augmented by incorporating intracellular ADC degradation and passive diffusion of unconjugated drug across tumor cells. Different biomeasures and chemomeasures for T-DM1, quantified in the companion manuscript, were incorporated into the modified model of ADC to characterize in vitro pharmacokinetics of T-DM1 in three HER2+ cell lines. When the cellular model was integrated with the tumor disposition model, the model was able to a priori predict tumor DM1 concentrations in xenograft mice. Pathway analysis suggested different contribution of antigen-mediated and passive diffusion pathways for intracellular unconjugated drug exposure between in vitro and in vivo systems. Global and local sensitivity analyses revealed that non-specific deconjugation and passive diffusion of the drug across tumor cell membrane are key parameters for drug exposure inside a cell. Finally, a systems pharmacokinetic model for intracellular processing of ADCs has been proposed to highlight our current understanding about the determinants of ADC activation inside a cell.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.