Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 12(1): 1763138, 2020.
Article in English | MEDLINE | ID: mdl-32432964

ABSTRACT

During the development of a therapeutic monoclonal antibody (mAb-1), the charge variant profile obtained by pH-gradient cation exchange chromatography (CEX) contained two main peaks, each of which exhibited a unique intrinsic fluorescence profile and demonstrated inter-convertibility upon reinjection of isolated peak fractions. Domain analysis of mAb-1 by CEX and liquid chromatography-mass spectrometry indicated that the antigen-binding fragment chromatographed as two separate peaks that had identical mass. Surface plasmon resonance binding analysis to antigen demonstrated comparable kinetics/affinity between these fractionated peaks and unfractionated starting material. Subsequent molecular modeling studies revealed that the relatively long and flexible complementarity-determining region 3 (CDR3) loop on the heavy chain could adopt two discrete pH-dependent conformations: an "open" conformation at neutral pH where the HC-CDR3 is largely solvent exposed, and a "closed" conformation at lower pH where the solvent exposure of a neighboring tryptophan in the light chain is reduced and two aspartic acid residues near the ends of the HC-CDR3 loop have atypical pKa values. The pH-dependent equilibrium between "open" and "closed" conformations of the HC-CDR3, and its proposed role in the anomalous charge variant profile of mAb-1, were supported by further CEX and hydrophobic interaction chromatography studies. This work is an example of how pH-dependent conformational changes and conformation-dependent changes to net charge can unexpectedly contribute to perceived instability and require thorough analytical, biophysical, and functional characterization during biopharmaceutical drug product development.


Subject(s)
Antibodies, Monoclonal/chemistry , Complementarity Determining Regions/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions/immunology , CHO Cells , Chromatography, Liquid/methods , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Cricetinae , Cricetulus , Humans , Hydrogen-Ion Concentration , Mass Spectrometry/methods , Models, Molecular , Peptide Mapping/methods , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Surface Plasmon Resonance/methods
2.
Nature ; 574(7779): 565-570, 2019 10.
Article in English | MEDLINE | ID: mdl-31645726

ABSTRACT

Co-inhibitory immune receptors can contribute to T cell dysfunction in patients with cancer1,2. Blocking antibodies against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1) partially reverse this effect and are becoming standard of care in an increasing number of malignancies3. However, many of the other axes by which tumours become inhospitable to T cells are not fully understood. Here we report that V-domain immunoglobulin suppressor of T cell activation (VISTA) engages and suppresses T cells selectively at acidic pH such as that found in tumour microenvironments. Multiple histidine residues along the rim of the VISTA extracellular domain mediate binding to the adhesion and co-inhibitory receptor P-selectin glycoprotein ligand-1 (PSGL-1). Antibodies engineered to selectively bind and block this interaction in acidic environments were sufficient to reverse VISTA-mediated immune suppression in vivo. These findings identify a mechanism by which VISTA may engender resistance to anti-tumour immune responses, as well as an unexpectedly determinative role for pH in immune co-receptor engagement.


Subject(s)
B7 Antigens/chemistry , B7 Antigens/metabolism , Membrane Glycoproteins/metabolism , T-Lymphocytes/metabolism , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , B7 Antigens/antagonists & inhibitors , B7 Antigens/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Crystallography, X-Ray , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Female , Histidine/metabolism , Humans , Hydrogen-Ion Concentration , Ligands , Male , Membrane Glycoproteins/immunology , Mice , Models, Molecular , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Binding/drug effects , Protein Domains , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
3.
J Pharm Sci ; 104(5): 1632-40, 2015 May.
Article in English | MEDLINE | ID: mdl-25757872

ABSTRACT

In this study, an automated high-throughput relative chemical stability (RCS) assay was developed in which various therapeutic proteins were assessed to determine stability based on the resistance to denaturation post introduction to a chaotrope titration. Detection mechanisms of both intrinsic fluorescence and near UV circular dichroism (near-UV CD) are demonstrated. Assay robustness was investigated by comparing multiple independent assays and achieving r(2) values >0.95 for curve overlays. The complete reversibility of the assay was demonstrated by intrinsic fluorescence, near-UV CD, and biologic potency. To highlight the method utility, we compared the RCS assay with differential scanning calorimetry and dynamic scanning fluorimetry methodologies. Utilizing C1/2 values obtained from the RCS assay, formulation rank-ordering of 12 different mAb formulations was performed. The prediction of long-term stability on protein aggregation is obtained by demonstrating a good correlation with an r(2) of 0.83 between RCS and empirical aggregation propensity data. RCS promises to be an extremely useful tool to aid in candidate formulation development efforts based on the complete reversibility of the method to allow for multiple assessments without protein loss and the strong correlation between the C1/2 data obtained and accelerated stability under stressed conditions.


Subject(s)
Circular Dichroism/methods , High-Throughput Screening Assays/methods , Protein Aggregates , Recombinant Proteins/chemistry , Antibodies, Monoclonal/chemistry , Chemistry, Pharmaceutical , Drug Evaluation, Preclinical/methods , Protein Conformation , Spectrometry, Fluorescence/methods
4.
J Biomol Screen ; 20(2): 212-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25336354

ABSTRACT

As a label-free technology, mass spectrometry (MS) enables assays to be generated that monitor the conversion of substrates with native sequences to products without the requirement for substrate modifications or indirect detection methods. Although traditional liquid chromatography (LC)-MS methods are relatively slow for a high-throughput screening (HTS) paradigm, with cycle times typically ≥ 60 s per sample, the Agilent RapidFire High-Throughput Mass Spectrometry (HTMS) System, with a cycle time of 5-7 s per sample, enables rapid analysis of compound numbers compatible with HTS. By monitoring changes in mass directly, HTMS assays can be used as a triaging tool by eliminating large numbers of false positives resulting from fluorescent compound interference or from compounds interacting with hydrophobic fluorescent dyes appended to substrates. Herein, HTMS assays were developed for multiple protease programs, including cysteine, serine, and aspartyl proteases, and applied as a confirmatory assay. The confirmation rate for each protease assay averaged <30%, independent of the primary assay technology used (i.e., luminescent, fluorescent, and time-resolved fluorescent technologies). Importantly, >99% of compounds designed to inhibit the enzymes were confirmed by the corresponding HTMS assay. Hence, HTMS is an effective tool for removing detection-based false positives from ultrahigh-throughput screening, resulting in hit lists enriched in true actives for downstream dose response titrations and hit-to-lead efforts.


Subject(s)
High-Throughput Screening Assays/methods , Mass Spectrometry/methods , Peptide Hydrolases/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Enzyme Assays/methods , Enzyme Assays/standards , High-Throughput Screening Assays/standards , Humans , Mass Spectrometry/standards , Protease Inhibitors/pharmacology , Reproducibility of Results , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...