35 results on '"Amita Datta-Mannan"'
Search Results
2. Reducing target binding affinity improves the therapeutic index of anti-MET antibody-drug conjugate in tumor bearing animals
- Author
-
Amita Datta-Mannan, Hiuwan Choi, Zhaoyan Jin, Ling Liu, Jirong Lu, David J. Stokell, Anthony T. Murphy, Kenneth W. Dunn, Michelle M. Martinez, and Yiqing Feng
- Subjects
Medicine ,Science - Published
- 2024
3. Physiologically Based Modeling to Predict Monoclonal Antibody Pharmacokinetics in Humans from in vitro Physiochemical Properties
- Author
-
Shihao Hu, Amita Datta-Mannan, and David Z. D’Argenio
- Subjects
FcRn interaction ,nonspecific binding ,antibody convective transport ,two-pore theory ,in vitro-in vivo prediction ,PBPK model ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
A model-based framework is presented to predict monoclonal antibody (mAb) pharmacokinetics (PK) in humans based on in vitro measures of antibody physiochemical properties. A physiologically based pharmacokinetic (PBPK) model is used to explore the predictive potential of 14 in vitro assays designed to measure various antibody physiochemical properties, including nonspecific cell-surface interactions, FcRn binding, thermal stability, hydrophobicity, and self-association. Based on the mean plasma PK time course data of 22 mAbs from humans reported in the literature, we found a significant positive correlation (R = 0.64, p = .0013) between the model parameter representing antibody-specific vascular to endothelial clearance and heparin relative retention time, an in vitro measure of nonspecific binding. We also found that antibody-specific differences in paracellular transport due to convection and diffusion could be partially explained by antibody heparin relative retention time (R = 0.52, p = .012). Other physiochemical properties, including antibody thermal stability, hydrophobicity, cross-interaction and self-association, in and of themselves were not predictive of model-based transport parameters. In contrast to other studies that have reported empirically derived expressions relating in vitro measures of antibody physiochemical properties directly to antibody clearance, the proposed PBPK model-based approach for predicting mAb PK incorporates fundamental mechanisms governing antibody transport and processing, informed by in vitro measures of antibody physiochemical properties, and can be expanded to include more descriptive representations of each of the antibody processing subsystems, as well as other antibody-specific information.
- Published
- 2022
- Full Text
- View/download PDF
4. Influence of physiochemical properties on the subcutaneous absorption and bioavailability of monoclonal antibodies
- Author
-
Amita Datta-Mannan, Selina Estwick, Chen Zhou, Hiuwan Choi, Nicole E. Douglass, Derrick R. Witcher, Jirong Lu, Catherine Beidler, and Rohn Millican
- Subjects
Subcutaneous absorption ,subcutaneous bioavailability ,monoclonal antibody ,physiochemical characterization ,hydrophobicity ,thermal stability ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Many therapeutic monoclonal antibodies (mAbs) were initially developed for intravenous (IV) administration. As a means to improve mAb drug-ability and the patient experience, subcutaneous (SC) administration is an increasingly important delivery route for mAbs. Unlike IV administration, bioavailability limitations for antibodies have been reported following SC injection and can dictate whether a mAb is administered via this parenteral route. The SC bioavailability of antibodies has been difficult to predict, and it can be variable and partial, with values ranging from ~50% to 100%. The mechanisms leading to the incomplete bioavailability of some mAbs relative to others are not well understood. There are some limited data that suggest the physiochemical properties inherent to a mAb can contribute to its SC absorption, bioavailability, and in vivo fate. In this study, we evaluated the integrated influence of multiple mAb physiochemical factors on the SC absorption and bioavailability of six humanized mAbs in both rats and cynomolgus monkeys. We demonstrate the physiochemical properties of mAbs are critical to their rate and extent of SC absorption. The combination of high positive charge and hydrophobic interaction significantly reduced the rate of the evaluated mAb’s SC absorption and bioavailability. Reduction or balancing of both these attributes via re-engineering the mAbs restored desirable properties of the molecules assessed. This included reduced association with SC tissue, improvements in mAb absorption from the SC space and overall SC bioavailability. Our findings point to the importance of evaluating the relative balance between various physiochemical factors, including charge, hydrophobicity, and stability, to improve the SC drug-ability of mAbs for selecting or engineering mAbs with enhanced in vivo absorption and bioavailability following SC administration.
- Published
- 2020
- Full Text
- View/download PDF
5. Pharmacokinetic Developability and Disposition Profiles of Bispecific Antibodies: A Case Study with Two Molecules
- Author
-
Amita Datta-Mannan, Robin Brown, Stephanie Key, Paul Cain, and Yiqing Feng
- Subjects
bispecific antibody ,monoclonal antibody ,scFv ,single-chain variable fragment ,pharmacokinetic ,FcRn neonatal Fc receptor ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Bispecific antibodies (BsAb) that engage multiple pathways are a promising therapeutic strategy to improve and prolong the efficacy of biologics in complex diseases. In the early stages of discovery, BsAbs often exhibit a broad range of pharmacokinetic (PK) behavior. Optimization of the neonatal Fc receptor (FcRn) interactions and removal of undesirable physiochemical properties have been used to improve the ‘pharmacokinetic developability’ for various monoclonal antibody (mAb) therapeutics, yet there is a sparsity of such information for BsAbs. The present work evaluated the influence of FcRn interactions and inherent physiochemical properties on the PK of two related single chain variable fragment (scFv)-based BsAbs. Despite their close relation, the two BsAbs exhibit disparate PK in cynomolgus monkeys with BsAb-1 having an aberrant clearance of ~2 mL/h/kg and BsAb-2 displaying a an ~10-fold slower clearance (~0.2 mL/h/kg). Evaluation of the physiochemical characteristics of the molecules, including charge, non-specific binding, thermal stability, and hydrophobic properties, as well as FcRn interactions showed some differences. In-depth drug disposition results revealed that a substantial disparity in the complete release from FcRn at a neutral pH is a primary factor contributing to the rapid clearance of the BsAb-1 while other biophysical characteristics were largely comparable between molecules.
- Published
- 2021
- Full Text
- View/download PDF
6. Antibody Conjugates-Recent Advances and Future Innovations
- Author
-
Donmienne Leung, Jacqueline M. Wurst, Tao Liu, Ruben M. Martinez, Amita Datta-Mannan, and Yiqing Feng
- Subjects
antibodies ,site-specific conjugation ,bioconjugates ,adc ,antibody-drug conjugates ,payloads ,linkers ,nucleic acids ,adme ,developability ,formulation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Monoclonal antibodies have evolved from research tools to powerful therapeutics in the past 30 years. Clinical success rates of antibodies have exceeded expectations, resulting in heavy investment in biologics discovery and development in addition to traditional small molecules across the industry. However, protein therapeutics cannot drug targets intracellularly and are limited to soluble and cell-surface antigens. Tremendous strides have been made in antibody discovery, protein engineering, formulation, and delivery devices. These advances continue to push the boundaries of biologics to enable antibody conjugates to take advantage of the target specificity and long half-life from an antibody, while delivering highly potent small molecule drugs. While the “magic bullet” concept produced the first wave of antibody conjugates, these entities were met with limited clinical success. This review summarizes the advances and challenges in the field to date with emphasis on antibody conjugation, linker-payload chemistry, novel payload classes, absorption, distribution, metabolism, and excretion (ADME), and product developability. We discuss lessons learned in the development of oncology antibody conjugates and look towards future innovations enabling other therapeutic indications.
- Published
- 2020
- Full Text
- View/download PDF
7. Intravital Microscopy Reveals Unforeseen Biodistribution Within the Liver and Kidney Mechanistically Connected to the Clearance of a Bifunctional Antibody
- Author
-
Amita Datta-Mannan, Bruce A. Molitoris, Yiqing Feng, Michelle M. Martinez, Ruben M. Sandoval, Robin M. Brown, Daniel Merkel, Johnny E. Croy, and Kenneth W. Dunn
- Subjects
Pharmacology ,Pharmaceutical Science - Published
- 2022
- Full Text
- View/download PDF
8. Identification of a Multi-Component Formulation for Intestinal Delivery of a GLP-1/Glucagon Co-agonist Peptide
- Author
-
Huyen, Tran, Phenil J, Patel, Aktham, Aburub, Andrea, Sperry, Selina, Estwick, Mohamed E H, ElSayed, and Amita Datta, -Mannan
- Subjects
Pharmacology ,Swine ,Organic Chemistry ,Pharmaceutical Science ,Capsules ,Glucagon ,Citric Acid ,Rats ,Intestinal Absorption ,Glucagon-Like Peptide 1 ,Animals ,Swine, Miniature ,Molecular Medicine ,Protease Inhibitors ,Pharmacology (medical) ,Peptides ,Peptide Hydrolases ,Biotechnology - Abstract
Oral delivery of therapeutic peptides has been challenging due to multiple physiological factors and physicochemical properties of peptides. We report a systematic approach to identify formulation compositions combining a permeation enhancer and a peptidase inhibitor that minimize proteolytic degradation and increase absorption of a peptide across the small intestine.An acylated glucagon-like peptide-1/glucagon co-agonist peptide (4.5 kDa) was selected as a model peptide. Proteolytic stability of the peptide was investigated in rat and pig SIF. Effective PEs and multiple component formulations were identified in rats. Relative bioavailability of the peptide was determined in minipigs via intraduodenal administration (ID) of enteric capsules.The peptide degraded rapidly in the rat and pig SIF. Citric acid, SBTI, and SBTCI inhibited the enzymatic degradation. The peptide self-associated into trimers in solution, however, addition of PEs monomerized the peptide. C10 was the most effective PE among tested PEs (DPC, LC, rhamnolipid, C12-maltosides, and SNAC) to improve intestinal absorption of the peptide in the rat IJ-closed loop model. A combination of C10 and SBTI or SBTCI increased the peptide exposure 5-tenfold compared to the exposure with the PE alone in the rat IJ-cannulated model, and achieved 1.06 ± 0.76% bioavailability in minipigs relative to subcutaneous via ID administration using enteric capsules.We identified SBTI and C10 as an effective peptidase inhibitor and PE for intestinal absorption of the peptide. The combination of SBTI and C10 addressed the peptide physiochemical properties and provides a formulation strategy to achieve intestinal delivery of this peptide.
- Published
- 2022
- Full Text
- View/download PDF
9. Monoclonal Antibody Pharmacokinetics in Cynomolgus Monkeys Following Subcutaneous Administration: Physiologically Based Model Predictions from Physiochemical Properties
- Author
-
Shihao Hu, Amita Datta-Mannan, and David Z. D’Argenio
- Subjects
Macaca fascicularis ,Injections, Subcutaneous ,Animals ,Antibodies, Monoclonal ,Biological Availability ,Pharmaceutical Science ,Immunotherapy - Abstract
An integrated physiologically based modeling framework is presented for predicting pharmacokinetics and bioavailability of subcutaneously administered monoclonal antibodies in cynomolgus monkeys, based on in silico structure-derived metrics characterizing antibody size, overall charge, local charge, and hydrophobicity. The model accounts for antibody-specific differences in pinocytosis, transcapillary transport, local lymphatic uptake, and pre-systemic degradation at the subcutaneous injection site and reliably predicts the pharmacokinetics of five different wild-type mAbs and their Fc variants following intravenous and subcutaneous administration. Significant associations were found between subcutaneous injection site degradation rate and the antibody's local positive charge of its complementarity-determining region (R = 0.56, p = 0.0012), antibody pinocytosis rate and its overall positive charge (R = 0.59, p = 0.00063), and antibody paracellular transport and its overall charge together with hydrophobicity (R = 0.63, p = 0.00096). Based on these results, population simulations were performed to predict the relationship between bioavailability and antibody local positive charge. In addition, model simulations were conducted to calculate the relative contribution of absorption pathways (lymphatic and blood), pre-systemic degradation pathways (interstitial and lysosomal), and the influence of injection site lymph flow on antibody bioavailability and pharmacokinetics. The proposed physiologically based modeling framework integrates fundamental mechanisms governing antibody subcutaneous absorption and disposition, with structured-based physiochemical properties, to predict antibody bioavailability and pharmacokinetics in vivo.
- Published
- 2022
- Full Text
- View/download PDF
10. Physiologically Based Modeling to Predict Monoclonal Antibody Pharmacokinetics in Humans from in vitro Physiochemical Properties
- Author
-
David D'Argenio, Amita Datta-Mannan, and Shihao Hu
- Subjects
Kinetics ,Heparin ,Immunology ,Antibodies, Monoclonal ,Humans ,Immunology and Allergy ,Tissue Distribution ,Models, Biological - Abstract
A model-based framework is presented to predict monoclonal antibody (mAb) pharmacokinetics (PK) in humans based on
- Published
- 2022
- Full Text
- View/download PDF
11. Modulation of the Biophysical Properties of Bifunctional Antibodies as a Strategy for Mitigating Poor Pharmacokinetics
- Author
-
J.R. Fitchett, Johnny E. Croy, Robin M Brown, Amita Datta-Mannan, Deepa Balasubramaniam, Aik Roy Heng, and Jennifer Pereira
- Subjects
Male ,genetic structures ,medicine.drug_class ,Protein domain ,CHO Cells ,Computational biology ,Monoclonal antibody ,Biochemistry ,Biophysical Phenomena ,Protein Structure, Secondary ,Mice ,Cricetulus ,Protein structure ,In vivo ,Cricetinae ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Immunologic Factors ,Tissue Distribution ,Receptor ,Mice, Knockout ,biology ,Chemistry ,Chinese hamster ovary cell ,HEK 293 cells ,Protein Structure, Tertiary ,Macaca fascicularis ,HEK293 Cells ,biology.protein ,Antibody - Abstract
Interest in the development of bi- or multispecific antibody (BsAbs)-based biotherapeutics is growing rapidly due to their inherent ability to interact with many targets simultaneously, thereby potentially protracting their functionality relative to monoclonal antibodies (mAbs). Biophysical property assays have been used to improve the probability of clinical success for various mAb therapeutics; however, there is a paucity of such data for BsAbs. This work evaluates a fusion of an IgG with an isolated protein domain (deemed ECD) and serves to understand how molecular architecture influences biophysical and biochemical properties and, in turn, how these relate to drug disposition. The biophysical characteristics of the molecules (charge, nonspecific binding, FcRn and Fcγ receptor interactions, thermal stability, structure-dynamics, and hydrophobic properties) indicated preferred orientations of ECD and IgG, which supported better pharmacokinetic outcomes. In certain instances, in which ECD-IgG configurations led to suboptimal biophysical behavior in the form of increased hydrophobicity and global ECD instability, drug clearance was found to be increased by ≥2-fold, driven by endothelial cell-based association/clearance mechanisms in the liver, kidneys, and spleen. Improvements in the pharmacokinetic properties were afforded by positional modulation of ECD that was able to bring the disposition characteristics in line with those of the parental mAb. The findings provide some pragmatic, broadly applicable strategies and guidance for the design considerations and evaluation of ECD-BsAb constructs. Additional studies, delineating the precise interactions involved in the clearance of the ECD-BsAb constructs, remain an opportunistic area for improving their in vivo kinetic properties.
- Published
- 2019
- Full Text
- View/download PDF
12. Influence of physiochemical properties on the subcutaneous absorption and bioavailability of monoclonal antibodies
- Author
-
Rohn Lee Millican, Chen Zhou, Catherine Brautigam Beidler, Nicole E Douglass, Jirong Lu, Derrick R. Witcher, Amita Datta-Mannan, Selina Estwick, and Hiuwan Choi
- Subjects
medicine.drug_class ,Injections, Subcutaneous ,Immunology ,Biological Availability ,Bioengineering ,Absorption (skin) ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,thermal stability ,03 medical and health sciences ,0302 clinical medicine ,Drug Development ,Subcutaneous Absorption ,charge ,Pharmacokinetics ,In vivo ,Report ,medicine ,Animals ,Humans ,Immunology and Allergy ,subcutaneous bioavailability ,hydrophobicity ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry, Physical ,Protein Stability ,Chemistry ,drug-ability ,Rats ,Bioavailability ,Macaca fascicularis ,nonspecific binding ,monoclonal antibody ,disposition ,030220 oncology & carcinogenesis ,biology.protein ,Parenteral route ,Antibody ,physiochemical characterization ,Hydrophobic and Hydrophilic Interactions ,pharmacokinetics ,chemical properties ,Protein Binding - Abstract
Many therapeutic monoclonal antibodies (mAbs) were initially developed for intravenous (IV) administration. As a means to improve mAb drug-ability and the patient experience, subcutaneous (SC) administration is an increasingly important delivery route for mAbs. Unlike IV administration, bioavailability limitations for antibodies have been reported following SC injection and can dictate whether a mAb is administered via this parenteral route. The SC bioavailability of antibodies has been difficult to predict, and it can be variable and partial, with values ranging from ~50% to 100%. The mechanisms leading to the incomplete bioavailability of some mAbs relative to others are not well understood. There are some limited data that suggest the physiochemical properties inherent to a mAb can contribute to its SC absorption, bioavailability, and in vivo fate. In this study, we evaluated the integrated influence of multiple mAb physiochemical factors on the SC absorption and bioavailability of six humanized mAbs in both rats and cynomolgus monkeys. We demonstrate the physiochemical properties of mAbs are critical to their rate and extent of SC absorption. The combination of high positive charge and hydrophobic interaction significantly reduced the rate of the evaluated mAb’s SC absorption and bioavailability. Reduction or balancing of both these attributes via re-engineering the mAbs restored desirable properties of the molecules assessed. This included reduced association with SC tissue, improvements in mAb absorption from the SC space and overall SC bioavailability. Our findings point to the importance of evaluating the relative balance between various physiochemical factors, including charge, hydrophobicity, and stability, to improve the SC drug-ability of mAbs for selecting or engineering mAbs with enhanced in vivo absorption and bioavailability following SC administration.
- Published
- 2020
- Full Text
- View/download PDF
13. Mechanisms Influencing the Pharmacokinetics and Disposition of Monoclonal Antibodies and Peptides
- Author
-
Amita Datta-Mannan
- Subjects
medicine.drug_class ,Metabolic Clearance Rate ,In silico ,Injections, Subcutaneous ,Druggability ,Pharmaceutical Science ,Self Administration ,Bioinformatics ,Monoclonal antibody ,Health outcomes ,030226 pharmacology & pharmacy ,Models, Biological ,Medication Adherence ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,medicine ,Animals ,Humans ,Computer Simulation ,Tissue Distribution ,Pharmacology ,Biological Products ,Drug disposition ,business.industry ,Antibodies, Monoclonal ,Disposition ,Therapeutic modalities ,030220 oncology & carcinogenesis ,Drug Design ,Injections, Intravenous ,business ,Peptides - Abstract
Monoclonal antibodies (mAbs) and peptides are an important class of therapeutic modalities that have brought improved health outcomes in areas with limited therapeutic optionality. Presently, there more than 90 mAb and peptide therapeutics on the United States market, with over 600 more in various clinical stages of development in a broad array of therapeutic areas, including diabetes, autoimmune disorders, oncology, neuroscience, and cardiovascular and infectious diseases. Notwithstanding this potential, there is high clinical rate of attrition, with approximately 10% reaching patients. A major contributor to the failure of the molecules is often times an incomplete or poor understanding of the pharmacokinetics (PK) and disposition profiles leading to limited or diminished efficacy. Increased and thorough characterization efforts directed at disseminating mechanisms influencing the PK and disposition of mAbs and peptides can aid in improving the design for their intended pharmacological activity, and thereby their clinical success. The PK and disposition factors for mAbs and peptides are broadly influenced by target-mediated drug disposition and nontarget-related clearance mechanisms related to the interplay between the relationship of the structure and physiochemical properties of mAbs and peptides with physiologic processes. This review focuses on nontarget-related factors influencing the disposition and PK of mAbs and peptides. Contemporary considerations around the increasing in silico approaches to identify nontarget-related molecule limitations and enhancing the druggability of mAbs and peptides, including parenteral and nonparenteral delivery strategies that are geared toward improving patient experience and compliance, are also discussed.
- Published
- 2019
14. Influence of FcRn binding properties on the gastrointestinal absorption and exposure profile of Fc molecules
- Author
-
Scott A. Lawrence, Bernice Ellis, Amita Datta-Mannan, Robin M Brown, Selina Estwick, Ross Blankenship, and Arunkumar Thangaraju
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Receptors, Fc ,Absorption (skin) ,01 natural sciences ,Biochemistry ,Intestinal absorption ,Gastrointestinal absorption ,Neonatal Fc receptor ,Drug Discovery ,Humans ,Molecular Biology ,Cells, Cultured ,Binding Sites ,010405 organic chemistry ,Chemistry ,Histocompatibility Antigens Class I ,Organic Chemistry ,Binding properties ,Hydrogen-Ion Concentration ,Immunoglobulin Fc Fragments ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,HEK293 Cells ,Intestinal Absorption ,Transcytosis ,Gastrointestinal Absorption ,Caco-2 ,Biophysics ,Molecular Medicine ,Transport system - Abstract
The neonatal Fc receptor (FcRn) represents a transport system with the potential to facilitate absorption of biologics across the gastrointestinal barrier. How biologics interact with FcRn to enable their gastrointestinal absorption, and how these interactions might be optimized in a biological therapeutic are not well understood. Thus, we studied the absorption of Fc molecules from the intestine using three IgG4-derived Fc variants with different, pH-dependent FcRn binding and release profiles. Using several different intestinal models, we consistently observed that FcRn binding affinity correlated with transcytosis. Our findings support targeting FcRn to enable intestinal absorption of biologics and highlight additional strategic considerations for future work.
- Published
- 2021
- Full Text
- View/download PDF
15. Aberrant bispecific antibody pharmacokinetics linked to liver sinusoidal endothelium clearance mechanism in cynomolgus monkeys
- Author
-
Johnny E. Croy, Stacy Torgerson, Matthew D. Breyer, Victor J. Wroblewski, Amita Datta-Mannan, and Linda Schirtzinger
- Subjects
0301 basic medicine ,Biodistribution ,Bispecific antibody ,Metabolic Clearance Rate ,medicine.drug_class ,Immunology ,Receptors, Fc ,Pharmacology ,Monoclonal antibody ,03 medical and health sciences ,0302 clinical medicine ,Neonatal Fc receptor ,Pharmacokinetics ,In vivo ,Report ,Antibodies, Bispecific ,medicine ,Extracellular ,Animals ,Humans ,Immunology and Allergy ,Mechanism (biology) ,Histocompatibility Antigens Class I ,Capillaries ,Macaca fascicularis ,030104 developmental biology ,Liver ,030220 oncology & carcinogenesis ,Half-Life - Abstract
Bispecific antibodies (BsAbs) can affect multiple disease pathways, thus these types of constructs potentially provide promising approaches to improve efficacy in complex disease indications. The specific and non-specific clearance mechanisms/biology that affect monoclonal antibody (mAb) pharmacokinetics are likely involved in the disposition of BsAbs. Despite these similarities, there are a paucity of studies on the in vivo biology that influences the biodistribution and pharmacokinetics of BsAbs. The present case study evaluated the in vivo disposition of 2 IgG-fusion BsAb formats deemed IgG-ECD (extracellular domain) and IgG-scFv (single-chain Fv) in cynomolgus monkeys. These BsAb molecules displayed inferior in vivo pharmacokinetic properties, including a rapid clearance (> 0.5 mL/hr/kg) and short half-life relative to their mAb counterparts. The current work evaluated factors in vivo that result in the aberrant clearance of these BsAb constructs. Results showed the rapid clearance of the BsAbs that was not attributable to target binding, reduced neonatal Fc receptor (FcRn) interactions or poor molecular/biochemical properties. Evaluation of the cellular distribution of the constructs suggested that the major clearance mechanism was linked to binding/association with liver sinusoidal endothelial cells (LSECs) versus liver macrophages. The role of LSECs in facilitating the clearance of the IgG-ECD and IgG-scFv BsAb constructs described in these studies was consistent with the minimal influence of clodronate-mediated macrophage depletion on the pharmacokinetics of the constructs in cynomolgus monkeys The findings in this report are an important demonstration that the elucidation of clearance mechanisms for some IgG-ECD and IgG-scFv BsAb molecules can be unique and complicated, and may require increased attention due to the proliferation of these more complex mAb-like structures.
- Published
- 2016
- Full Text
- View/download PDF
16. The Properties of Cysteine-Conjugated Antibody-Drug Conjugates Are Impacted by the IgG Subclass
- Author
-
Yiqing Feng, Anthony Murphy, David J. Stokell, Hiuwan Choi, Jason Tang, Amita Datta-Mannan, and Wrobleski Aaron D
- Subjects
0301 basic medicine ,Male ,Antibody-drug conjugate ,Immunoconjugates ,Immunoglobulin Variable Region ,Pharmaceutical Science ,CHO Cells ,Subclass ,03 medical and health sciences ,Cricetulus ,Pharmacokinetics ,Drug Stability ,Animals ,Cysteine ,biology ,Drug discovery ,Chemistry ,Antibodies, Monoclonal ,Rats ,body regions ,Macaca fascicularis ,030104 developmental biology ,Biochemistry ,Solubility ,Immunoglobulin G ,biology.protein ,Antibody ,Linker ,Conjugate - Abstract
Among the numerous antibody-drug conjugate (ADC) clinical candidates, one of the most prevalent types utilizes the interchain cysteines in antibodies to conjugate auristatin via a maleimide-containing linker. In this class of ADCs, there are a paucity of systematic studies characterizing how IgG subclass influences the biophysical properties and in vivo pharmacokinetics of the ADC molecules. In the current investigation, we studied cysteine-conjugated ADCs using a model system consisting of human IgG1, IgG2, and IgG4 antibodies with the same variable region. Our findings identified some unforeseen differences among the three ADCs. Drug conjugation profiling by LC-MS revealed that 50% of inter heavy-light chain disulfide bonds are disrupted to conjugate drugs in IgG1 antibody while only 10% in IgG2 antibody and 20% in IgG4 antibody. The solution behavior of the ADCs was interrogated in concentrating experiments and diffusion interaction parameter measurements. We found that drug conjugation affected the solution property of the three antibodies differently, with the IgG2-based ADC having the most increased propensity to aggregate. Rat PK studies using a sensitive LC-MS-based bioanalytical method showed that the IgG1-based ADC has poor peripheral linker-payload stability while the IgG2- and IgG4-based ADCs are stable. The conjugate stability of the IgG2-based ADC was further confirmed in a cynomolgus monkey PK study. Overall, the IgG2-based ADC exhibited the best PK/conjugate stability but also the most deterioration in stability among the three ADCs. Our findings provide important information and present multifactorial considerations for the selection of IgG subclass during ADC drug discovery when employing stochastic cysteine conjugation.
- Published
- 2018
17. Insights into the Impact of Heterogeneous Glycosylation on the Pharmacokinetic Behavior of Follistatin-Fc–Based Biotherapeutics
- Author
-
Benjamin C. Yaden, Jennifer Pereira, Andrew Korytko, Amita Datta-Mannan, Lihua Huang, and Johnny E. Croy
- Subjects
Male ,Follistatin ,Glycosylation ,Pharmaceutical Science ,CHO Cells ,Mice, SCID ,Pharmacology ,Biology ,Mice ,chemistry.chemical_compound ,Cricetulus ,Pharmacokinetics ,Cricetinae ,Animals ,Humans ,Receptor ,Mice, Knockout ,Chinese hamster ovary cell ,HEK 293 cells ,Sialic acid ,Biological Therapy ,Macaca fascicularis ,HEK293 Cells ,chemistry ,biology.protein ,Asialoglycoprotein receptor ,human activities - Abstract
Follistatin 315 heparan sulfate-binding deficient mutant human IgG4 Fc fusion (FST-ΔHBS-Fc) is a follistatin (FST) based Fc fusion protein currently being developed as a novel therapy for several potential indications, including muscle wasting. Previous assessments of the pharmacokinetics and therapeutic activity of FST-ΔHBS-Fc have shown a close association of the exposure-response relationship. The current work builds upon these initial studies by investigating the glycosylation characteristics of FST-ΔHBS-Fc after recombinant expression and its impact on the pharmacokinetics in mice and Cynomolgus monkeys. The data presented indicate that FST-ΔHBS-Fc is heterogeneously glycosylated at the three putative sites in FST when recombinantly expressed in stably transfected Chinese hamster ovary cells. Such carbohydrate heterogeneity, especially with regards to sialic acid incorporation, directly results in sugar-dependent clearance in both mice and Cynomolgus monkeys. Examination of the pharmacokinetics of FST-ΔHBS-Fc molecules containing variable sialic acid content in asialoglycoprotein receptor 1 (ASPGR-1) knockout mice supports the receptor's role as part of the clearance mechanism of the molecules. Based on the evaluation of several variably sialylated lots of material in pharmacokinetic assessments, we define specifications for average sialic acid incorporation into FST-ΔHBS-Fc that result in limited sugar-mediated clearance. Taken together, these studies highlight the importance of establishing an early understanding of the glycosylation/pharmacokinetic relationships of FST-ΔHBS-Fc, which will provide a basis for future application toward optimal systemic drug delivery and dosing strategies.
- Published
- 2015
- Full Text
- View/download PDF
18. Balancing charge in the complementarity-determining regions of humanized mAbs without affecting pI reduces non-specific binding and improves the pharmacokinetics
- Author
-
Victor J. Wroblewski, Donmienne Doen Mun Leung, Jirong Lu, Arunkumar Thangaraju, Ying Tang, Derrick R. Witcher, and Amita Datta-Mannan
- Subjects
Models, Molecular ,medicine.drug_class ,Stereochemistry ,Immunology ,Complementarity determining region ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Mice ,Antibody Specificity ,In vivo ,Pi ,medicine ,Animals ,Humans ,Immunology and Allergy ,Distribution (pharmacology) ,Isoelectric Point ,Catabolism ,Chemistry ,Complementarity Determining Regions ,In vitro ,HEK293 Cells ,Isoelectric point ,Immunoglobulin G ,Biophysics ,Reports - Abstract
Lowering the isoelectric point (pI) through engineering the variable region or framework of an IgG can improve its exposure and half-life via a reduction in clearance mediated through non-specific interactions. As such, net charge is a potentially important property to consider in developing therapeutic IgG molecules having favorable pharmaceutical characteristics. Frequently, it may not be possible to shift the pI of monoclonal antibodies (mAbs) dramatically without the introduction of other liabilities such as increased off-target interactions or reduced on-target binding properties. In this report, we explored the influence of more subtle modifications of molecular charge on the in vivo properties of an IgG1 and IgG4 monoclonal antibody. Molecular surface modeling was used to direct residue substitutions in the complementarity-determining regions (CDRs) to disrupt positive charge patch regions, resulting in a reduction in net positive charge without affecting the overall pI of the mAbs. The effect of balancing the net positive charge on non-specific binding was more significant for the IgG4 versus the IgG1 molecule that we examined. This differential effect was connected to the degree of influence on cellular degradation in vitro and in vivo clearance, distribution and metabolism in mice. In the more extreme case of the IgG4, balancing the charge yielded an ∼7-fold improvement in peripheral exposure, as well as significantly reduced tissue catabolism and subsequent excretion of proteolyzed products in urine. Balancing charge on the IgG1 molecule had a more subtle influence on non-specific binding and yielded only a modest alteration in clearance, distribution and elimination. These results suggest that balancing CDR charge without affecting the pI can lead to improved mAb pharmacokinetics, the magnitude of which is likely dependent on the relative influence of charge imbalance and other factors affecting the molecule's disposition.
- Published
- 2015
- Full Text
- View/download PDF
19. Inhibition of Activin A Ameliorates Skeletal Muscle Injury and Rescues Contractile Properties by Inducing Efficient Remodeling in Female Mice
- Author
-
Guoli Dai, Venkatesh Krishnan, Johnny E. Croy, Pamela K. Shetler, Amita Datta-Mannan, Alexander E. Culver, Andrea Milner, Jonathan M. Wilson, Benjamin C. Yaden, and Yan X. Wang
- Subjects
medicine.medical_specialty ,animal structures ,Enzyme-Linked Immunosorbent Assay ,Inflammation ,Biology ,Pathology and Forensic Medicine ,Fibrosis ,Internal medicine ,medicine ,Animals ,Regeneration ,Muscle, Skeletal ,Myogenesis ,Skeletal muscle ,medicine.disease ,Immunohistochemistry ,Muscle atrophy ,Activins ,Mice, Inbred C57BL ,Electroporation ,Endocrinology ,medicine.anatomical_structure ,embryonic structures ,Female ,Tumor necrosis factor alpha ,medicine.symptom ,PAX7 ,Transcriptome ,hormones, hormone substitutes, and hormone antagonists ,Muscle Contraction ,Transforming growth factor - Abstract
Activin A, a member of the transforming growth factor-β superfamily, provides pleiotropic regulation of fibrosis and inflammation. We aimed at determining whether selective inhibition of activin A would provide a regenerative benefit. The introduction of activin A into normal muscle increased the expression of inflammatory and muscle atrophy genes Tnf, Tnfrsf12a, Trim63, and Fbxo32 by 3.5-, 10-, 2-, and 4-fold, respectively. The data indicate a sensitive response of muscle to activin A. Two hours after cardiotoxin-induced muscle damage, local activin A protein expression increased by threefold to ninefold. Neutralization of activin A with a specific monoclonal antibody in this muscle injury model decreased the muscle protein levels of lymphotoxin α and Il17a by 32% and 42%, respectively. Muscle histopathological features showed that activin A antibody-treated mice displayed an increase in muscle degradation, with the concomitant 9.2-fold elevation in F4/80-positive cells 3 days after injury. At the same time, the number of Pax7/Myod1-positive cells also increased, indicative of potentiated muscle precursor activation. Ultimately, activin A inhibition resulted in rapid recovery of muscle contractile properties indicated by a restoration of maximum and specific force. In summary, selective inhibition of activin A with a monoclonal antibody in muscle injury leads to the early onset of tissue degradation and subsequent enhanced myogenesis, thereby accelerating muscle repair and functional recovery.
- Published
- 2014
- Full Text
- View/download PDF
20. Abstract 353: A novel molecule with profound tumor killing activity
- Author
-
Amita Datta-Mannan, David J. Stokell, Ling Liu, Yiqing Feng, Xianming Chen, Sheng-Bin Peng, Philip W. Iversen, Philip Arthur Hipskind, Wrobleski Aaron D, Gregory P. Donoho, Yin Yin, and Wei Zeng
- Subjects
Cancer Research ,Antibody-drug conjugate ,biology ,Chemistry ,Cell ,Cancer ,medicine.disease_cause ,medicine.disease ,medicine.anatomical_structure ,Oncology ,In vivo ,Cancer research ,medicine ,biology.protein ,Erlotinib ,KRAS ,Antibody ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
LY3343544: A novel MET antibody drug conjugate that shows profound pre-clinical in vivo anti-tumor activities, irrespective of MET pathway dependence MET is over-expressed in many types of human tumors. Due to the heterogeneity of human tumors, MET antibodies or small molecule inhibitors have benefited only small subsets of patients with tumors driven by signaling through the c-Met pathway. The patient selection strategies to identify those tumors with MET activation dependence are helpful in predicting sensitivity to many of these inhibitors. It was reported previously that Lilly’s MET antibody, emibetuzumab, showed clinical activity in selective NSCLC patients with high MET IHC staining (90% to 100% 3+ positive) when it was combined with erlotinib in Phase II clinical Trials. In searching for a better treatment for patients carrying the MET overexpression tumors regardless other co-existing mutations, we developed LY3343544, a novel antibody drug conjugate (ADC) molecule that consists of emibetuzumab conjugated with the potent microtubule inhibitor MMAE using a unique lysine conjugation approach. Upon binding to MET, LY3343544 is internalized via receptor-mediated endocytosis. LY3343544 maintains the similar binding and internalization activities to the cell surface MET as compared to emibetuzumab in the competitive cell binding assay and the internalization assay. We reported here that LY3343544 showed profound anti-tumor activity in a preclinical mouse models, and overcome intrinsic resistance mechanisms including KRAS, BRAF, PI3K and TP53 mutations. LY3343544 kills tumor cells expressing a wide range of MET levels on the cell surface and is capable of killing a variety of MET-overexpressing tumor cells including pancreatic, cholanglocarcinoma, colorectal, NSCLC, gastric, head and neck tumor cells in vitro. In contrast, LY3343544 does not kill human normal endothelial cells and normal epithelial cells, no activity on human peripheral blood mononuclear cells with or without activation as well as in cell-based assays. Moreover, LY3343544 is more stable in rodent PK studies than typical inter chain Cys VC-MMAE conjugates and showed tumor regressions in colorectal, NSCLC, gastric and pancreatic mouse xenograft models. Furthermore, LY3343544 shows profound tumor regression in >50% of PDAC PDX models (n=40): 20% complete response (CR); 22.5% partial response (PR); and 17.5% stable disease (SD); overall disease control rate (DCR) is 60%. In addition, LY3343544 shows tumor growth inhibition in cholangiocarcinoma PDX model that is resistant to emibetuzumab. In summary, LY3343544 is highly potent in killing a variety of tumor cells in cell-based killing assays. It demonstrated good stability in vivo and profound anti-tumor efficacy in multiple mouse xenograft models and patient-derived xenograft models thus is a promising agent to treat many types of cancers. Citation Format: Ling Liu, Aaron D. Wrobleski, Yin Yin, Wei Zeng, Xianming Chen, David J. Stokell, Sheng-bin Peng, Amita Datta-Mannan, Gregory P. Donoho, Philip W. Iversen, Philip Hipskind, Yiqing Feng. A novel molecule with profound tumor killing activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 353.
- Published
- 2019
- Full Text
- View/download PDF
21. Engineered FcRn Binding Fusion Peptides Significantly Enhance the Half-Life of a Fab Domain in Cynomolgus Monkeys
- Author
-
Nicole E Douglass, Zhaoyan Y. Jin, Anthony T. Murphy, Jeffrey S. Boyles, Bernice Ellis, Amita Datta-Mannan, Fariba Norouziyan-Cooper, Amber Peariso, Lihua Huang, and Derrick R. Witcher
- Subjects
Male ,0106 biological sciences ,medicine.drug_class ,Peptide ,Receptors, Fc ,Monoclonal antibody ,01 natural sciences ,Applied Microbiology and Biotechnology ,Cell Line ,Immunoglobulin Fab Fragments ,Neonatal Fc receptor ,Pharmacokinetics ,Cell Line, Tumor ,010608 biotechnology ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,Histocompatibility Antigens Class I ,010401 analytical chemistry ,Antibodies, Monoclonal ,Half-life ,General Medicine ,Fusion protein ,Cyclic peptide ,0104 chemical sciences ,Kinetics ,Macaca fascicularis ,HEK293 Cells ,chemistry ,Pharmacodynamics ,Biophysics ,Molecular Medicine ,Caco-2 Cells ,Peptides ,Half-Life ,Protein Binding - Abstract
There is a rapidly growing reinvigoration of the investigation of small proteins, cyclic peptides, and mAb derived domains as biotherapies. The drugability of these structures are challenged by fast peripheral clearance properties that can reduce their potential to be realized as medicines. Engineering strategies have been of limited value because mechanistically the half-life benefit is manifested by increasing the molecular weight and/or the hydrodyanimc radius which slows the molecule's renal elimination, but can result in the inherent loss of activity and target accessibility. The present work evaluated an alternative approach using smaller peptide sequences which bind to the neonatal Fc receptor (FcRn). Results revealed, small linear and cyclic FcRn binding peptides (FcRnBPs) fused to a combination of the N- and C-termini of a Fab can significantly improve the pharmacokinetics of the protein in cynomolgus monkeys relative to the parental Fab. The linear and cyclic conformations, as well as, the number of FcRnBPs fused to the Fab both influence the clearance and the extent of pharmacokinetic benefit. FcRnBP fusion protein kinetics were also affected by a combination of post-translation modifications and non-specific binding properties. The results in this report lay some foundation in fostering the advent of newer technologies toward successfully improving the pharmacokinetics of proteins, peptides, and mAb-derived domains. Additional work in the integration of a variety of factors including the intended site of action, tissue disposition, metabolism, toxicity and pharmacokinetic, and pharmacodynamics relationship of the intended therapeutic modality are key areas for advancement of these approaches.
- Published
- 2018
- Full Text
- View/download PDF
22. Monoclonal Antibody Clearance
- Author
-
Jeffry D. Watkins, Derrick R. Witcher, Ying Tang, Victor J. Wroblewski, and Amita Datta-Mannan
- Subjects
biology ,medicine.drug_class ,Chemistry ,Cell Biology ,Plasma protein binding ,Pharmacology ,Monoclonal antibody ,Biochemistry ,Receptor–ligand kinetics ,Neonatal Fc receptor ,Pharmacokinetics ,In vivo ,medicine ,biology.protein ,Antibody ,Receptor ,Molecular Biology - Abstract
The neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. There are mixed reports on whether modification of the interaction with FcRn can be used as an engineering strategy to improve the pharmacokinetic and pharmacodynamic properties of monoclonal antibodies. We tested whether the T250Q/M428L mutations, which improved the pharmacokinetics of humanized IgGs in the rhesus monkey, would translate to a pharmacokinetic benefit in both cynomolgus monkeys and mice when constructed on a different humanized IgG framework (anti-tumor necrosis factor-α (TNFα)). The T250Q/M428L anti-TNFα variant displayed an ∼40-fold increase in binding affinity to cynomolgus monkey FcRn (C-FcRn) at pH 6.0, with maintenance of the pH binding dependence. We also constructed another anti-TNFα variant (P257I/Q311I) whose binding kinetics with the C-FcRn was similar to that of the T250Q/M428L variant. The binding affinity of the T250Q/M428L variant for murine FcRn was increased ∼500-fold, with maintenance of pH dependence. In contrast to the interaction with C-FcRn, this interaction was driven mainly by a decrease in the rate of dissociation. Despite the improved in vitro binding properties of the anti-TNFα T250Q/M428L and P257I/Q311I variants to C-FcRn, the pharmacokinetic profiles of these molecules were not differentiated from the wild-type antibody in cynomolgus monkeys after intravenous administration. When administered intravenously to mice, the T250Q/M428L anti-TNFα variant displayed improved pharmacokinetics, characterized by an ∼2-fold slower clearance than the wild-type antibody. The discrepancy between these data and previously reported benefits in rhesus monkeys and the inability of these mutations to translate to improved kinetics across species may be related to a number of factors. We propose extending consideration to differences in the absolute IgG-FcRn affinity, the kinetics of the IgG/FcRn interaction, and differences in the relative involvement of this pathway in the context of other factors influencing the disposition or elimination of monoclonal antibodies.
- Published
- 2007
- Full Text
- View/download PDF
23. Current Approaches for Absorption, Distribution, Metabolism, and Excretion Characterization of Antibody-Drug Conjugates: An Industry White Paper
- Author
-
Johnny J. Yang, Weirong Wang, Nagendra Chemuturi, Ramaswamy A. Iyer, Eugenia Kraynov, Markus Walles, Priya Sriraman, David J Moore, Amita Datta-Mannan, Edit Tarcsa, Guangqing Xiao, Amrita V. Kamath, Enrique Escandón, Matthew Barfield, Michaela Bairlein, Antoine Deslandes, and Dan A. Rock
- Subjects
0301 basic medicine ,Drug ,Immunoconjugates ,Drug Industry ,media_common.quotation_subject ,Pharmaceutical Science ,Computational biology ,Pharmacology ,Drug molecule ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,White paper ,Antibodies monoclonal ,Distribution (pharmacology) ,Animals ,Humans ,Hardware_ARITHMETICANDLOGICSTRUCTURES ,Drug industry ,ADME ,media_common ,Chemistry ,Antibodies, Monoclonal ,body regions ,030104 developmental biology ,Pharmaceutical Preparations - Abstract
An antibody-drug conjugate (ADC) is a unique therapeutic modality composed of a highly potent drug molecule conjugated to a monoclonal antibody. As the number of ADCs in various stages of nonclinical and clinical development has been increasing, pharmaceutical companies have been exploring diverse approaches to understanding the disposition of ADCs. To identify the key absorption, distribution, metabolism, and excretion (ADME) issues worth examining when developing an ADC and to find optimal scientifically based approaches to evaluate ADC ADME, the International Consortium for Innovation and Quality in Pharmaceutical Development launched an ADC ADME working group in early 2014. This white paper contains observations from the working group and provides an initial framework on issues and approaches to consider when evaluating the ADME of ADCs.
- Published
- 2015
24. The interplay of non-specific binding, target-mediated clearance and FcRn interactions on the pharmacokinetics of humanized antibodies
- Author
-
Donmienne Doen Mun Leung, Jirong Lu, Derrick R. Witcher, Victor J. Wroblewski, Ying Tang, and Amita Datta-Mannan
- Subjects
medicine.drug_class ,Metabolic Clearance Rate ,Immunology ,Antibody Affinity ,Plasma protein binding ,Complementarity determining region ,Receptors, Fc ,Pharmacology ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Protein Engineering ,Immunoglobulin G ,Mice ,Neonatal Fc receptor ,Pharmacokinetics ,Report ,medicine ,Immunology and Allergy ,Animals ,Humans ,biology ,Chemistry ,Histocompatibility Antigens Class I ,Protein engineering ,Complementarity Determining Regions ,In vitro ,Cell biology ,Macaca fascicularis ,biology.protein ,Protein Binding - Abstract
The application of protein engineering technologies toward successfully improving antibody pharmacokinetics has been challenging due to the multiplicity of biochemical factors that influence monoclonal antibody (mAb) disposition in vivo. Physiological factors including interactions with the neonatal Fc receptor (FcRn) and specific antigen binding properties of mAbs, along with biophysical properties of the mAbs themselves play a critical role. It has become evident that applying an integrated approach to understand the relative contribution of these factors is critical to rationally guide and apply engineering strategies to optimize mAb pharmacokinetics. The study presented here evaluated the influence of unintended non-specific interactions on the disposition of mAbs whose clearance rates are governed predominantly by either non-specific (FcRn) or target-mediated processes. The pharmacokinetics of 8 mAbs representing a diverse range of these properties was evaluated in cynomolgus monkeys. Results revealed complementarity-determining region (CDR) charge patch engineering to decrease charge-related non-specific binding can have a significant impact on improving the clearance. In contrast, the influence of enhanced in vitro FcRn binding was mixed, and related to both the strength of charge interaction and the general mechanism predominant in governing the clearance of the particular mAb. Overall, improved pharmacokinetics through enhanced FcRn interactions were apparent for a CDR charge-patch normalized mAb which was affected by non-specific clearance. The findings in this report are an important demonstration that mAb pharmacokinetics requires optimization on a case-by-case basis to improve the design of molecules with increased therapeutic application.
- Published
- 2015
25. Chemokine-Binding Specificity of Soluble Chemokine-Receptor Analogues: Identification of Interacting Elements by Chimera Complementation
- Author
-
Amita Datta-Mannan and Martin J. Stone
- Subjects
Chemokine CCL11 ,Models, Molecular ,CCR1 ,CCR2 ,Receptors, CCR2 ,Receptors, CCR3 ,Recombinant Fusion Proteins ,Molecular Mimicry ,Molecular Sequence Data ,CCR3 ,C-C chemokine receptor type 7 ,Biology ,CCL7 ,Biochemistry ,Chemokine receptor ,Solubility ,Chemokine binding ,Chemokines, CC ,Mutagenesis, Site-Directed ,Receptors, Chemokine ,Amino Acid Sequence ,CC chemokine receptors ,Chemokine CCL2 ,Protein Binding - Abstract
The specificity of chemokine-receptor interactions plays a central role in the regulation of leukocyte migration in inflammatory responses. Herein, we describe a soluble mimic of CC chemokine receptor 2 (CCR2), dubbed CROSS-N(2)E3(2), which incorporates the N-terminal region (N) and third extracellular loop (E3) elements of CCR2 displayed on the surface of a soluble protein scaffold. CROSS-N(2)E3(2) binds to the CCR2 ligand monocyte chemoattractant protein-1 (MCP-1) with a dissociation equilibrium constant of 1.1 +/- 0.1 microM but does not bind to the cognate chemokines of the receptor CCR3 (eotaxin-1, -2, and -3). Similarly, a soluble analogue of CCR3 (CROSS(5)-N(3)E3(3)) binds to eotaxin-1, -2, and -3 but not to MCP-1. Thus, these receptor analogues have the same specificity as the natural receptors. Using soluble proteins containing N and E3 elements from different receptors (CROSS-N(2)E3(3) and CROSS-N(3)E3(2)), we demonstrate that both receptor elements are required for optimal binding to the cognate chemokines. In addition, we report the binding affinities of all four CROSS proteins to a panel of two wild-type and six chimeric chemokines. These complementation studies indicate the regions of the chemokines that interact with each element of the receptors, allowing us to deduce the orientations of the receptor extracellular elements relative to the bound chemokines.
- Published
- 2004
- Full Text
- View/download PDF
26. Application of FcRn binding assays to guide mAb development
- Author
-
Amita Datta-Mannan and Victor J. Wroblewski
- Subjects
Pharmacology ,biology ,medicine.drug_class ,Chemistry ,Histocompatibility Antigens Class I ,Pharmaceutical Science ,Antibodies, Monoclonal ,Context (language use) ,Computational biology ,Plasma protein binding ,Receptors, Fc ,Monoclonal antibody ,In vitro ,Neonatal Fc receptor ,In vivo ,medicine ,biology.protein ,Animals ,Humans ,Pharmacokinetics ,Antibody ,Receptor ,Protein Binding - Abstract
Monoclonal antibodies (mAbs) represent an important class of therapeutic modalities. To optimize their pharmaceutical properties, studies have focused on improving mAb pharmacokinetic/pharmacodynamic profiles by modulating their interactions with the neonatal Fc receptor (FcRn). The influence of both the chemical and physical properties of IgGs has been examined in the context of FcRn interactions. In this regard, a variety of FcRn binding assays and tools have been developed and used to characterize the interaction with IgGs. However, a predictive relationship between the FcRn binding interaction of IgGs in vitro and their pharmacokinetics in vivo broadly across mAbs remains elusive. Many studies have increasingly suggested that the interplay between the characteristics of the mAb and the nature of its target can influence disposition and elimination. Thus, it is becoming increasingly evident that along with FcRn interactions, consideration of the non-FcRn-based biologic processes active in mAb disposition should be integrated into mAb development and optimization. Herein, we describe how the pharmacokinetics of mAbs can be modulated through FcRn interactions and provide perspectives on interpreting the receptor binding parameters in relation to other mechanisms involved in antibody disposition to aid in guiding mAb development.
- Published
- 2014
27. Follistatin: a novel therapeutic for the improvement of muscle regeneration
- Author
-
Guoli Dai, Yan Wang, Johnny E. Croy, Pamela K. Shetler, Andrea Milner, Amita Datta-Mannan, Benjamin C. Yaden, Jonathan M. Wilson, Henry Uhlman Bryant, Jessica Andrews, and Venkatesh Krishnan
- Subjects
medicine.medical_specialty ,Follistatin ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Myostatin ,Pharmacology ,Protein Engineering ,Mice ,Atrophy ,Internal medicine ,medicine ,Macrophage ,Animals ,Regeneration ,Muscle, Skeletal ,biology ,Growth factor ,Regeneration (biology) ,medicine.disease ,Immunoglobulin Fc Fragments ,Endocrinology ,Immunoglobulin G ,GDF11 ,biology.protein ,Systemic administration ,Molecular Medicine - Abstract
Follistatin (FST) is a member of the tissue growth factor β family and is a secreted glycoprotein that antagonizes many members of the family, including activin A, growth differentiation factor 11, and myostatin. The objective of this study was to explore the use of an engineered follistatin therapeutic created by fusing FST315 lacking heparin binding activity to the N terminus of a murine IgG1 Fc (FST315-ΔHBS-Fc) as a systemic therapeutic agent in models of muscle injury. Systemic administration of this molecule was found to increase body weight and lean muscle mass after weekly administration in normal mice. Subsequently, we tested this agent in several models of muscle injury, which were chosen based on their severity of damage and their ability to reflect clinical settings. FST315-ΔHBS-Fc treatment proved to be a potent inducer of muscle remodeling and regeneration. FST315-ΔHBS-Fc induced improvements in muscle repair after injury/atrophy by modulating the early inflammatory phase allowing for increased macrophage density, and Pax7-positive cells leading to an accelerated restoration of myofibers and muscle function. Collectively, these data demonstrate the benefits of a therapeutically viable form of FST that can be leveraged as an alternate means of ameliorating muscle regeneration.
- Published
- 2014
28. Generation and activity of a humanized monoclonal antibody that selectively neutralizes the epidermal growth factor receptor ligands transforming growth factor-α and epiregulin
- Author
-
Catherine Brautigam Beidler, Shannon M Harlan, Derrick R. Witcher, Jeffrey S. Boyles, Bernice Ellis, Robert L. Johnson, Josef G. Heuer, Shaoyou Chu, Elaine M Conner, Ramona J Petrovan, Amita Datta-Mannan, Anja Stauber, Derek D. Yang, and Matthew D. Breyer
- Subjects
Male ,TGF alpha ,medicine.drug_class ,Molecular Sequence Data ,Humanized antibody ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Epiregulin ,Cell Line ,Epidermal growth factor ,medicine ,Animals ,Humans ,Epidermal growth factor receptor ,Amino Acid Sequence ,Myofibroblasts ,EGFR inhibitors ,Cell Proliferation ,Pharmacology ,Mice, Knockout ,biology ,Epidermal Growth Factor ,Transforming Growth Factor alpha ,Molecular biology ,Antibodies, Neutralizing ,ErbB Receptors ,Mice, Inbred C57BL ,Macaca fascicularis ,Animals, Newborn ,Immunoglobulin G ,biology.protein ,Molecular Medicine ,Transforming growth factor ,Protein Binding - Abstract
At least seven distinct epidermal growth factor (EGF) ligands bind to and activate the EGF receptor (EGFR). This activation plays an important role in the embryo and in the maintenance of adult tissues. Importantly, pharmacologic EGFR inhibition also plays a critical role in the pathophysiology of diverse disease states, especially cancer. The roles of specific EGFR ligands are poorly defined in these disease states. Accumulating evidence suggests a role for transforming growth factor α (TGFα) in skin, lung, and kidney disease. To explore the role of Tgfa, we generated a monoclonal antibody (mAb41) that binds to and neutralizes human Tgfa with high affinity (KD = 36.5 pM). The antibody also binds human epiregulin (Ereg) (KD = 346.6 pM) and inhibits ligand induced myofibroblast cell proliferation (IC50 values of 0.52 and 1.12 nM for human Tgfa and Ereg, respectively). In vivo, a single administration of the antibody to pregnant mice (30 mg/kg s.c. at day 14 after plug) or weekly administration to neonate mice (20 mg/kg s.c. for 4 weeks) phenocopy Tgfa knockout mice with curly whiskers, stunted growth, and expansion of the hypertrophic zone of growth plate cartilage. Humanization of this monoclonal antibody to a human IgG4 antibody (LY3016859) enables clinical development. Importantly, administration of the humanized antibody to cynomolgus monkeys is absent of the skin toxicity observed with current EGFR inhibitors used clinically and no other pathologies were noted, indicating that neutralization of Tgfa could provide a relatively safe profile as it advances in clinical development.
- Published
- 2014
29. An engineered human follistatin variant: insights into the pharmacokinetic and pharmocodynamic relationships of a novel molecule with broad therapeutic potential
- Author
-
Johnny E. Croy, Benjamin C. Yaden, Amita Datta-Mannan, Venkatesh Krishnan, and Bryan Edward Jones
- Subjects
Male ,endocrine system ,Follistatin ,Biology ,Pharmacology ,Ligands ,Protein Engineering ,Cell Line ,Mice ,Pharmacokinetics ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Heparin ,Sepharose ,Skeletal muscle ,Protein engineering ,Muscle atrophy ,Recombinant Proteins ,Mice, Inbred C57BL ,Muscular Atrophy ,medicine.anatomical_structure ,HEK293 Cells ,chemistry ,Pharmacodynamics ,Immunoglobulin G ,embryonic structures ,biology.protein ,Molecular Medicine ,Heparan sulfate binding ,medicine.symptom ,Glycoprotein ,hormones, hormone substitutes, and hormone antagonists ,Half-Life ,Protein Binding - Abstract
Human follistatin is a regulatory glycoprotein with widespread biological functions including anti-inflammatory activities, wound healing properties and muscle stimulating effects. The role of follistatin in a wide range of biological activities shows promise for potential clinical application, which has prompted considerable interest in the investigation of the protein as a potential disease modifying agent. In spite of this potential, the development of follistatin as a broad use biotherapeutic has been severely hindered by a poor understanding and characterization of its pharmacokinetic and pharmacodynamic (PK/PD) relationships. Therefore, to better define these relationships, we performed in-depth analyses of the PK/PD relationships of native follistatin-315. Our data indicates that the intrinsic PK/PD properties of native follistatin-315 are poorly suited for acting as a parentally administered biotherapeutic with broad systemic effects. Herein, we leveraged protein engineering to modify the PK characteristics of the native molecule by fusing follistatin-315 to a murine IgG1 Fc and removing the intrinsic heparan sulfate binding activity of follistatin. The engineered variant molecule had ~100- and ~1600-fold improvements in terminal half-life and exposure, respectively. In contrast to the native follistatin-315, the variant showed a robust, dose-dependent pharmacological effect when administered subcutaneously on a weekly basis in mouse models of muscle atrophy and degeneration. These studies highlight the underappreciated and critical relationship between optimizing multiple physical and chemical properties of follistatin on its overall PK/PD profile. Moreover, our findings provide the first documented strategy towards the development of a follistatin therapeutic with potential utility in patients affected with skeletal muscle diseases.
- Published
- 2012
30. FcRn affinity-pharmacokinetic relationship of five human IgG4 antibodies engineered for improved in vitro FcRn binding properties in cynomolgus monkeys
- Author
-
Amita Datta-Mannan, Victor J. Wroblewski, Derrick R. Witcher, David Driver, Craig Duane Dickinson, Chi-Kin Chow, and Jirong Lu
- Subjects
medicine.drug_class ,Pharmaceutical Science ,Plasma protein binding ,Receptors, Fc ,Pharmacology ,In Vitro Techniques ,Monoclonal antibody ,Cell Line ,Neonatal Fc receptor ,In vivo ,medicine ,Animals ,Humans ,Receptor ,biology ,Chemistry ,Histocompatibility Antigens Class I ,Protein engineering ,In vitro ,Cell biology ,Macaca fascicularis ,Immunoglobulin G ,biology.protein ,Antibody ,Protein Binding - Abstract
The pH-dependent binding of IgGs to the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. Enhancing interactions between Fc and FcRn via protein engineering has been successfully used as an approach for improving the pharmacokinetics of monoclonal antibodies (mAbs). Although the quantitative translatability of the in vitro FcRn affinity enhancement to an in vivo pharmacokinetic benefit has been supported by several studies, there are also published reports indicating a disconnect in this relation. The body of literature suggests there are likely additional biochemical and biophysical properties of the mAbs along with their FcRn affinity that influence the in vivo pharmacokinetics. Herein, we more broadly evaluate the in vitro Fc-FcRn interactions and biochemical properties of five humanized IgG4 antibodies each with two Fc variant sequences (T250Q/M428L and V308P) and their corresponding pharmacokinetics in cynomolgus monkeys. Our findings indicate that the FcRn affinity-pharmacokinetic relationship does not show a direct correlation either across different IgGs or between the two variant sequences within a platform. Other parameters that have been suggested to contribute to mAb pharmacokinetic properties, such as the pH-dependent dissociation of the FcRn-IgG complexes, mAb biophysical properties, and nonspecific/charge binding characteristics of the mAbs, also did not independently explain the differing pharmacokinetic behaviors. Our results suggest that there is likely not a single in vitro parameter that readily predicts in vivo pharmacokinetics, but that the relative contribution and interplay of several factors along with the FcRn binding affinity are important determinants of mAb pharmacokinetic properties.
- Published
- 2012
31. Influence of improved FcRn binding on the subcutaneous bioavailability of monoclonal antibodies in cynomolgus monkeys
- Author
-
Jirong Lu, Derrick R. Witcher, Amita Datta-Mannan, and Victor J. Wroblewski
- Subjects
medicine.drug_class ,Short Communication ,Immunology ,Mutation, Missense ,Biological Availability ,Receptors, Fc ,Pharmacology ,Monoclonal antibody ,Infusions, Subcutaneous ,Cell Line ,Subcutaneous injection ,Antibodies, Monoclonal, Murine-Derived ,Mice ,Neonatal Fc receptor ,Pharmacokinetics ,In vivo ,Immunology and Allergy ,Medicine ,Animals ,Humans ,biology ,business.industry ,Histocompatibility Antigens Class I ,Recombinant Proteins ,Bioavailability ,Macaca fascicularis ,Amino Acid Substitution ,Monoclonal ,biology.protein ,Antibody ,business ,Half-Life - Abstract
Engineering monoclonal antibodies (mAbs) with improved binding to the neonatal Fc receptor (FcRn) is a strategy that can extend their in vivo half-life and slow their systemic clearance. Published reports have predominantly characterized the pharmacokinetics of mAbs after intravenous administration. Recently, studies in mice suggest FcRn may also play a role in affecting the subcutaneous bioavailability of mAbs. Herein, we examined whether five mAbs engineered with the T250Q/M428L Fc mutations that improved their FcRn interactions, and subsequently their in vivo pharmacokinetics after intravenous administration, had improved subcutaneous bioavailability compared with their wild-type counterparts in cynomolgus monkeys. Similar to the intravenous administration findings, the pharmacokinetic profiles of our variant mAbs after subcutaneous injection showed improved half-life or clearance. In contrast, a clear effect was not observed on the subcutaneous bioavailability. We expect that while FcRn may play a role in determining mAb subcutaneous bioavailability, multiple biopharmaceutical and physiological factors are likely to influence the success of engineering strategies aimed at targeting this pathway for improving bioavailability.
- Published
- 2012
32. Neonatal Fc receptor mediates internalization of Fc in transfected human endothelial cells
- Author
-
Kenneth W. Dunn, Nancy Goebl, Clifford M. Babbey, Derrick R. Witcher, Victor J. Wroblewski, and Amita Datta-Mannan
- Subjects
media_common.quotation_subject ,Recombinant Fusion Proteins ,Endocytic cycle ,education ,Endosomes ,Receptors, Fc ,Biology ,Cell Line ,Mice ,Neonatal Fc receptor ,Low affinity ,Animals ,Humans ,Internalization ,Molecular Biology ,Nucleotide salvage ,media_common ,Histocompatibility Antigens Class I ,Transferrin ,Endothelial Cells ,Dextrans ,Cell Biology ,Transfection ,Articles ,Hydrogen-Ion Concentration ,Molecular biology ,Endocytosis ,Cell biology ,Immunoglobulin Fc Fragments ,Immunoglobulin G ,Lysosomes ,Homeostasis - Abstract
The neonatal Fc receptor, FcRn mediates an endocytic salvage pathway that prevents degradation of IgG, thus contributing to the homeostasis of circulating IgG. Based on the low affinity of IgG for FcRn at neutral pH, internalization of IgG by endothelial cells is generally believed to occur via fluid-phase endocytosis. To investigate the role of FcRn in IgG internalization, we used quantitative confocal microscopy to characterize internalization of fluorescent Fc molecules by HULEC-5A lung microvascular endothelia transfected with GFP fusion proteins of human or mouse FcRn. In these studies, cells transfected with FcRn accumulated significantly more intracellular Fc than untransfected cells. Internalization of FcRn-binding forms of Fc was proportional to FcRn expression level, was enriched relative to dextran internalization in proportion to FcRn expression level, and was blocked by incubation with excess unlabeled Fc. Because we were unable to detect either surface expression of FcRn or surface binding of Fc, these results suggest that FcRn-dependent internalization of Fc may occur through sequestration of Fc by FcRn in early endosomes. These studies indicate that FcRn-dependent internalization of IgG may be important not only in cells taking up IgG from an extracellular acidic space, but also in endothelial cells participating in homeostatic regulation of circulating IgG levels.
- Published
- 2008
33. Monoclonal antibody clearance. Impact of modulating the interaction of IgG with the neonatal Fc receptor
- Author
-
Amita, Datta-Mannan, Derrick R, Witcher, Ying, Tang, Jeffry, Watkins, and Victor J, Wroblewski
- Subjects
Tumor Necrosis Factor-alpha ,Antibodies, Monoclonal ,Receptors, Fc ,Hydrogen-Ion Concentration ,Surface Plasmon Resonance ,Macaca mulatta ,Cell Line ,Kinetics ,Macaca fascicularis ,Mice ,Immunoglobulin G ,Mutation ,Animals ,Humans ,Protein Binding - Abstract
The neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. There are mixed reports on whether modification of the interaction with FcRn can be used as an engineering strategy to improve the pharmacokinetic and pharmacodynamic properties of monoclonal antibodies. We tested whether the T250Q/M428L mutations, which improved the pharmacokinetics of humanized IgGs in the rhesus monkey, would translate to a pharmacokinetic benefit in both cynomolgus monkeys and mice when constructed on a different humanized IgG framework (anti-tumor necrosis factor-alpha (TNFalpha)). The T250Q/M428L anti-TNFalpha variant displayed an approximately 40-fold increase in binding affinity to cynomolgus monkey FcRn (C-FcRn) at pH 6.0, with maintenance of the pH binding dependence. We also constructed another anti-TNFalpha variant (P257I/Q311I) whose binding kinetics with the C-FcRn was similar to that of the T250Q/M428L variant. The binding affinity of the T250Q/M428L variant for murine FcRn was increased approximately 500-fold, with maintenance of pH dependence. In contrast to the interaction with C-FcRn, this interaction was driven mainly by a decrease in the rate of dissociation. Despite the improved in vitro binding properties of the anti-TNFalpha T250Q/M428L and P257I/Q311I variants to C-FcRn, the pharmacokinetic profiles of these molecules were not differentiated from the wild-type antibody in cynomolgus monkeys after intravenous administration. When administered intravenously to mice, the T250Q/M428L anti-TNFalpha variant displayed improved pharmacokinetics, characterized by an approximately 2-fold slower clearance than the wild-type antibody. The discrepancy between these data and previously reported benefits in rhesus monkeys and the inability of these mutations to translate to improved kinetics across species may be related to a number of factors. We propose extending consideration to differences in the absolute IgG-FcRn affinity, the kinetics of the IgG/FcRn interaction, and differences in the relative involvement of this pathway in the context of other factors influencing the disposition or elimination of monoclonal antibodies.
- Published
- 2006
34. Humanized IgG1 variants with differential binding properties to the neonatal Fc receptor: relationship to pharmacokinetics in mice and primates
- Author
-
Weidong Jiang, Victor J. Wroblewski, Derrick R. Witcher, Ying Tang, Jeffry D. Watkins, and Amita Datta-Mannan
- Subjects
Male ,medicine.drug_class ,Pharmaceutical Science ,Plasma protein binding ,CHO Cells ,Receptors, Fc ,Monoclonal antibody ,Transfection ,Antibodies ,Mice ,Neonatal Fc receptor ,Cricetulus ,In vivo ,Cricetinae ,medicine ,Animals ,Humans ,Receptor ,Pharmacology ,biology ,Tumor Necrosis Factor-alpha ,Histocompatibility Antigens Class I ,biology.organism_classification ,Cell biology ,Mice, Inbred C57BL ,Macaca fascicularis ,Immunoglobulin G ,Immunology ,Mutation ,biology.protein ,Antibody ,Protein Binding - Abstract
It is well established that the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. As such, modification of the interaction of IgG with FcRn has been the focus of protein-engineering strategies designed to generate therapeutic antibodies with improved pharmacokinetic properties. In the current work, we characterized differences in interaction of IgG between mouse and primate receptors using three humanized anti-tumor necrosis factor alpha antibodies with variant IgG(1) Fc regions. The wild-type and variant IgG showed a differential combination of improved affinity, modified dissociation kinetics, and altered pH-dependent complex dissociation when evaluated on the primate and murine receptors. The observed in vitro binding differences within and between species allowed us to more completely relate these parameters to their influence on the in vivo pharmacokinetics in mice and cynomolgus monkeys. The variant antibodies have different pharmacokinetic behavior in cynomolgus monkeys and mice, which appears to be related to the unique binding characteristics observed with the murine receptor. However, we did not observe a direct relationship between increased binding affinity to the receptor and improved pharmacokinetic properties for these molecules in either species. This work provides further insights into how the FcRn/IgG interaction may be modulated to develop monoclonal antibodies with improved therapeutic properties.
- Published
- 2006
35. Specificity determinants for chemokine recognition identified using eotaxin-MCP-1 chimeras
- Author
-
Todd Parody, Amita Datta-Mannan, Martin J. Stone, and Michael R. Mayer
- Subjects
CCR1 ,Chemokine CCL11 ,Models, Molecular ,CCR2 ,Chemokine receptor CCR5 ,Chemokine receptor ,Protein Conformation ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Biophysics ,Eotaxin ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,Biochemistry ,Polymerase Chain Reaction ,Sensitivity and Specificity ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,Genetics ,Amino Acid Sequence ,Cloning, Molecular ,Molecular Biology ,Chemokine CCL2 ,030304 developmental biology ,0303 health sciences ,Binding Sites ,biology ,Chimera ,hemic and immune systems ,Cell Biology ,Cell biology ,Monocyte chemoattractant protein-1 ,Chemokine ,030220 oncology & carcinogenesis ,Chemokines, CC ,biology.protein ,Specificity ,XCL2 ,Receptors, Chemokine ,Chemokines ,CCL21 - Abstract
To identify the elements of two chemokines [monocyte chemoattractant protein-1 (MCP-1) and eotaxin] that control their differential recognition by their respective receptors (CCR2 and CCR3), we have studied the receptor interactions of MCP-1-eotaxin chimeras. Each receptor was found to exhibit a distinct binding preference for proteins containing the amino-terminal region of the cognate chemokine for that receptor. However, other elements dictating chemokine preference were different for the two receptors. In some cases, the influence of replacing a particular region was dependent on the identities of neighboring regions, indicating a complex network of cooperative and/or compensating interactions.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.