Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2681: 383-398, 2023.
Article in English | MEDLINE | ID: mdl-37405660

ABSTRACT

To select the most promising screening hits from antibody and VHH display campaigns for subsequent in-depth profiling and optimization, it is highly desirable to assess and select sequences on properties beyond only their binding signals from the sorting process. In addition, developability risk criteria, sequence diversity, and the anticipated complexity for sequence optimization are relevant attributes for hit selection and optimization. Here, we describe an approach for the in silico developability assessment of antibody and VHH sequences. This method not only allows for ranking and filtering multiple sequences with regard to their predicted developability properties and diversity, but also visualizes relevant sequence and structural features of potentially problematic regions and thereby provides rationales and starting points for multi-parameter sequence optimization.


Subject(s)
Antibodies
2.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35858707

ABSTRACT

BACKGROUND: Bintrafusp alfa (BA) is a bifunctional fusion protein designed for colocalized, simultaneous inhibition of two immunosuppressive pathways, transforming growth factor-ß (TGF-ß) and programmed death-ligand 1 (PD-L1), within the tumor microenvironment (TME). We hypothesized that targeting PD-L1 to the tumor by BA colocalizes the TGF-ß trap (TGF-ßRII) to the TME, enabling it to sequester TGF-ß in the tumor more effectively than systemic TGF-ß blockade, thereby enhancing antitumor activity. METHODS: Multiple technologies were used to characterize the TGF-ß trap binding avidity. BA versus combinations of anti-PD-L1 and TGF-ß trap or the pan-TGF-ß antibody fresolimumab were compared in proliferation and two-way mixed lymphocyte reaction assays. Immunophenotyping of tumor-infiltrating lymphocytes (TILs) and RNA sequencing (RNAseq) analysis assessing stromal and immune landscape following BA or the combination therapy were performed in MC38 tumors. TGF-ß and PD-L1 co-expression and their associated gene signatures in MC38 tumors and human lung carcinoma tissue were studied with single-cell RNAseq (scRNAseq) and immunostaining. BA-induced internalization, degradation, and depletion of TGF-ß were investigated in vitro. RESULTS: BA and fresolimumab had comparable intrinsic binding to TGF-ß1, but there was an ~80× avidity-based increase in binding affinity with BA. BA inhibited cell proliferation in TGF-ß-dependent and PD-L1-expressing cells more potently than TGF-ß trap or fresolimumab. Compared with the combination of anti-PD-L1 and TGF-ß trap or fresolimumab, BA enhanced T cell activation in vitro and increased TILs in MC38 tumors, which correlated with efficacy. BA induced distinct gene expression in the TME compared with the combination therapy, including upregulation of immune-related gene signatures and reduced activities in TGF-ß-regulated pathways, such as epithelial-mesenchymal transition, extracellular matrix deposition, and fibrosis. Regulatory T cells, macrophages, immune cells of myeloid lineage, and fibroblasts were key PD-L1/TGF-ß1 co-expressing cells in the TME. scRNAseq analysis suggested BA modulation of the macrophage phenotype, which was confirmed by histological assessment. PD-L1/TGF-ß1 co-expression was also seen in human tumors. Finally, BA induced TGF-ß1 internalization and degradation in the lysosomes. CONCLUSION: BA more effectively blocks TGF-ß by targeting TGF-ß trap to the tumor via PD-L1 binding. Such colocalized targeting elicits distinct and superior antitumor responses relative to single agent combination therapy.


Subject(s)
Lung Neoplasms , Transforming Growth Factor beta , B7-H1 Antigen , Humans , Immunologic Factors , Programmed Cell Death 1 Receptor , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tumor Microenvironment
3.
MAbs ; 13(1): 1932230, 2021.
Article in English | MEDLINE | ID: mdl-34116620

ABSTRACT

Understanding the pharmacokinetic (PK) properties of a drug, such as clearance, is a crucial step for evaluating efficacy. The PK of therapeutic antibodies can be complex and is influenced by interactions with the target, Fc-receptors, anti-drug antibodies, and antibody intrinsic factors. A growing body of literature has linked biophysical properties of antibodies, particularly nonspecific-binding propensity, hydrophobicity and charged regions to rapid clearance in preclinical species and selected human PK studies. A clear understanding of the connection between biophysical properties and their impact on PK would allow for early selection and optimization of antibodies and reduce costly attrition during clinical trials due to sub-optimal human clearance. Due to the difficulty in obtaining large and unbiased human PK data, previous studies have focused mostly on preclinical PK. For this study, we obtained and curated the most comprehensive clinical PK dataset to date and calculated accurate estimates of linear clearance for 64 monoclonal antibodies ranging from investigational candidates in Phase 2 trials to marketed products. This allows for the first time a deep analysis of the influence of biophysical and sequence-based in silico properties directly on human clearance. We use statistical analysis and a Random Forest classifier to identify properties that have the greatest influence in our dataset. Our findings indicate that in vitro poly-specificity assay and in silico estimated isoelectric point can discriminate fast and slow clearing antibodies, extending previous observations on preclinical clearance. This provides a simple yet powerful approach to select antibodies with desirable PK during early-stage screening.


Subject(s)
Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Blood Chemical Analysis/methods , Humans , Machine Learning
6.
Oncoimmunology ; 9(1): 1744921, 2020.
Article in English | MEDLINE | ID: mdl-32313722

ABSTRACT

T cell immunoglobulin and mucin domain-3 (TIM-3) is an immune checkpoint that regulates normal immune responses but can be exploited by tumor cells to evade immune surveillance. TIM-3 is primarily expressed on immune cells, particularly on dysfunctional and exhausted T cells, and engagement of TIM-3 with its ligands promotes TIM-3-mediated T cell inhibition. Antagonistic ligand-blocking anti-TIM-3 antibodies have the potential to abrogate T cell inhibition, activate antigen-specific T cells, and enhance anti-tumor immunity. Here we describe M6903, a fully human anti-TIM-3 antibody without effector function and with high affinity and selectivity to TIM-3. We demonstrate that M6903 blocks the binding of TIM-3 to three of its ligands, phosphatidylserine (PtdSer), carcinoembryonic antigen cell adhesion-related molecule 1 (CEACAM1), and galectin 9 (Gal-9). These results are supported by an atomic resolution crystal structure and functional assays, which demonstrate that M6903 monotherapy enhanced T cell activation. This activation was further enhanced by the combination of M6903 with bintrafusp alfa, a bifunctional fusion protein that simultaneously blocks the transforming growth factor-ß (TGF-ß) and programmed death ligand 1 (PD-L1) pathways. M6903 and bintrafusp alfa combination therapy also enhanced anti-tumor efficacy in huTIM-3 knock-in mice, relative to either monotherapy. These in vitro and in vivo data, along with favorable pharmacokinetics in marmoset monkeys, suggest that M6903 as a monotherapy warrants further pre-clinical assessment and that M6903 and bintrafusp alfa may be a promising combination therapy in the clinic.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Animals , Antibodies, Monoclonal , Lymphocyte Activation , Mice , T-Lymphocytes
7.
J Biol Chem ; 291(48): 25106-25119, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27694443

ABSTRACT

Bispecific antibodies (bsAbs) and antibody-drug conjugates (ADCs) have already demonstrated benefits for the treatment of cancer in several clinical studies, showing improved drug selectivity and efficacy. In particular, simultaneous targeting of prominent cancer antigens, such as EGF receptor (EGFR) and c-MET, by bsAbs has raised increasing interest for potentially circumventing receptor cross-talk and c-MET-mediated acquired resistance during anti-EGFR monotherapy. In this study, we combined the selectivity of EGFR × c-MET bsAbs with the potency of cytotoxic agents via bispecific antibody-toxin conjugation. Affinity-attenuated bispecific EGFR × c-MET antibody-drug conjugates demonstrated high in vitro selectivity toward tumor cells overexpressing both antigens and potent anti-tumor efficacy. Due to basal EGFR expression in the skin, ADCs targeting EGFR in general warrant early safety assessments. Reduction in EGFR affinity led to decreased toxicity in keratinocytes. Thus, the combination of bsAb affinity engineering with the concept of toxin conjugation may be a viable route to improve the safety profile of ADCs targeting ubiquitously expressed antigens.


Subject(s)
Antibodies, Bispecific/immunology , ErbB Receptors/immunology , Immunotoxins/immunology , Proto-Oncogene Proteins c-met/immunology , A549 Cells , Antibodies, Bispecific/therapeutic use , Hep G2 Cells , Humans , Immunotoxins/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology
8.
J Mol Recognit ; 28(4): 269-76, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25664688

ABSTRACT

The binding of programmed death ligand 1 protein (PD-L1) to its receptor programmed death protein 1 (PD-1) mediates immunoevasion in cancer and chronic viral infections, presenting an important target for therapeutic intervention. Several monoclonal antibodies targeting the PD-L1/PD-1 signaling axis are undergoing clinical trials; however, the epitopes of these antibodies have not been described. We have combined orthogonal approaches to localize and characterize the epitope of a monoclonal antibody directed against PD-L1 at good resolution and with high confidence. Limited proteolysis and mass spectrometry were applied to reveal that the epitope resides in the first immunoglobulin domain of PD-L1. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) was used to identify a conformational epitope comprised of discontinuous strands that fold to form a beta sheet in the native structure. This beta sheet presents an epitope surface that significantly overlaps with the PD-1 binding interface, consistent with a desired PD-1 competitive mechanism of action for the antibody. Surface plasmon resonance screening of mutant PD-L1 variants confirmed that the region identified by HDX-MS is critical for the antibody interaction and further defined specific residues contributing to the binding energy. Taken together, the results are consistent with the observed inhibitory activity of the antibody on PD-L1-mediated immune evasion. This is the first report of an epitope for any antibody targeting PD-L1 and demonstrates the power of combining orthogonal epitope mapping techniques.


Subject(s)
Antibodies, Monoclonal/chemistry , B7-H1 Antigen/immunology , Epitopes/analysis , Deuterium Exchange Measurement/methods , Epitope Mapping/methods , Humans , Mass Spectrometry/methods , Proteolysis
9.
RNA ; 14(7): 1284-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18495940

ABSTRACT

Archaeal class I CCA-adding enzymes use a ribonucleoprotein template to build and repair the universally conserved 3'-terminal CCA sequence of the acceptor stem of all tRNAs. A wealth of structural and biochemical data indicate that the Archaeoglobus fulgidus CCA-adding enzyme binds primarily to the tRNA acceptor stem through a long, highly conserved alpha-helix that lies nearly parallel to the acceptor stem and makes many contacts with its sugar-phosphate backbone. Although the geometry of this alpha-helix is nearly ideal in all available cocrystal structures, the helix contains a highly conserved, potentially helix-breaking proline or glycine near the N terminus. We performed a mutational analysis to dissect the role of this residue in CCA-addition activity. We found that the phylogenetically permissible P295G mutant and the phylogenetically absent P295T had little effect on CCA addition, whereas P295A and P295S progressively interfered with CCA addition (C74>C75>A76 addition). We also examined the effects of these mutations on tRNA binding and the kinetics of CCA addition, and performed a computational analysis using Rosetta Design to better understand the role of P295 in nucleotide transfer. Our data indicate that CCA-adding activity does not correlate with the stability of the pre-addition cocrystal structures visualized by X-ray crystallography. Rather, the data are consistent with a transient conformational change involving P295 of the tRNA-binding alpha-helix during or between one or more steps in CCA addition.


Subject(s)
Archaeoglobus fulgidus/metabolism , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/metabolism , RNA, Transfer/metabolism , Amino Acid Substitution , Archaeoglobus fulgidus/enzymology , Computer Simulation , Crystallography, X-Ray , Models, Molecular , Mutagenesis , Nucleic Acid Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , RNA Nucleotidyltransferases/genetics , Software
10.
J Biol Chem ; 282(20): 15159-69, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17355978

ABSTRACT

The conserved dystroglycan-dystrophin (Dg.Dys) complex connects the extracellular matrix to the cytoskeleton. In humans as well as Drosophila, perturbation of this complex results in muscular dystrophies and brain malformations and in some cases cellular polarity defects. However, the regulation of the Dg.Dys complex is poorly understood in any cell type. We now find that in loss-of-function and overexpression studies more than half (34 residues) of the Dg proline-rich conserved C-terminal regions can be truncated without significantly compromising its function in regulating cellular polarity in Drosophila. Notably, the truncation eliminates the WW domain binding motif at the very C terminus of the protein thought to mediate interactions with dystrophin, suggesting that a second, internal WW binding motif can also mediate this interaction. We confirm this hypothesis by using a sensitive fluorescence polarization assay to show that both WW domain binding sites of Dg bind to Dys in humans (K(d) = 7.6 and 81 microM, respectively) and Drosophila (K(d) = 16 and 46 microM, respectively). In contrast to the large deletion mentioned above, a single proline to an alanine point mutation within a predicted Src homology 3 domain (SH3) binding site abolishes Dg function in cellular polarity. This suggests that an SH3-containing protein, which has yet to be identified, functionally interacts with Dg.


Subject(s)
Cell Polarity/physiology , Drosophila Proteins/metabolism , Dystroglycans/metabolism , Dystrophin/metabolism , src Homology Domains , Amino Acid Motifs , Amino Acid Sequence/genetics , Amino Acid Substitution , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Dystroglycans/genetics , Dystrophin/genetics , Humans , Point Mutation , Protein Binding/genetics , Sequence Deletion , Sequence Homology, Amino Acid , src Homology Domains/genetics
11.
EMBO J ; 26(2): 481-93, 2007 Jan 24.
Article in English | MEDLINE | ID: mdl-17215867

ABSTRACT

Perturbation in the Dystroglycan (Dg)-Dystrophin (Dys) complex results in muscular dystrophies and brain abnormalities in human. Here we report that Drosophila is an excellent genetically tractable model to study muscular dystrophies and neuronal abnormalities caused by defects in this complex. Using a fluorescence polarization assay, we show a high conservation in Dg-Dys interaction between human and Drosophila. Genetic and RNAi-induced perturbations of Dg and Dys in Drosophila cause cell polarity and muscular dystrophy phenotypes: decreased mobility, age-dependent muscle degeneration and defective photoreceptor path-finding. Dg and Dys are required in targeting glial cells and neurons for correct neuronal migration. Importantly, we now report that Dg interacts with insulin receptor and Nck/Dock SH2/SH3-adaptor molecule in photoreceptor path-finding. This is the first demonstration of a genetic interaction between Dg and InR.


Subject(s)
Disease Models, Animal , Drosophila , Dystroglycans/metabolism , Dystrophin/metabolism , Muscular Dystrophy, Animal/genetics , Nervous System Malformations/genetics , Adaptor Proteins, Signal Transducing , Animals , Animals, Genetically Modified , Cell Movement/genetics , Cell Polarity , Dystroglycans/genetics , Dystroglycans/physiology , Dystrophin/genetics , Dystrophin/physiology , Humans , Models, Biological , Models, Molecular , Muscular Dystrophy, Animal/etiology , Mutation , Oncogene Proteins/metabolism
12.
J Mol Biol ; 357(3): 917-27, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16473368

ABSTRACT

An important objective of computational protein design is the generation of high affinity peptide inhibitors of protein-peptide interactions, both as a precursor to the development of therapeutics aimed at disrupting disease causing complexes, and as a tool to aid investigators in understanding the role of specific complexes in the cell. We have developed a computational approach to increase the affinity of a protein-peptide complex by designing N or C-terminal extensions which interact with the protein outside the canonical peptide binding pocket. In a first in silico test, we show that by simultaneously optimizing the sequence and structure of three to nine residue peptide extensions starting from short (1-6 residue) peptide stubs in the binding pocket of a peptide binding protein, the approach can recover both the conformations and the sequences of known binding peptides. Comparison with phage display and other experimental data suggests that the peptide extension approach recapitulates naturally occurring peptide binding specificity better than fixed backbone design, and that it should be useful for predicting peptide binding specificities from crystal structures. We then experimentally test the approach by designing extensions for p53 and dystroglycan-based peptides predicted to bind with increased affinity to the Mdm2 oncoprotein and to dystrophin, respectively. The measured increases in affinity are modest, revealing some limitations of the method. Based on these in silico and experimental results, we discuss future applications of the approach to the prediction and design of protein-peptide interactions.


Subject(s)
Peptides/chemical synthesis , Peptides/metabolism , Protein Engineering , Proteins/chemical synthesis , Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Drug Design , Humans , Molecular Sequence Data , Peptides/genetics , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proteins/genetics , Proto-Oncogene Proteins c-mdm2/chemical synthesis , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , beta Catenin/chemical synthesis , beta Catenin/genetics , beta Catenin/metabolism , src Homology Domains/physiology
13.
EMBO J ; 21(17): 4691-8, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12198171

ABSTRACT

To identify nucleotides in or near the active site, we have used a circularly permuted version of the VS ribozyme capable of cleavage and ligation to incorporate a single photoactive nucleotide analog, 4-thio- uridine, immediately downstream of the scissile bond. Exposure to UV light produced two cross-linked RNAs, in which the 4-thio-uridine was cross-linked to A756 in the 730 loop of helix VI. The cross-links formed only under conditions that support catalytic activity, suggesting that they reflect functionally relevant conformations of the RNA. One of the cross-linked RNAs contains a lariat, indicative of intramolecular cross-linking in the ligated RNA; the other is a branched molecule in which the scissile phosphodiester bond is cleaved, but occupies the same site in the ribozyme-substrate complex. These are the two forms of the RNA expected to be the ground state structures on either side of the transition state. This localization of the active site is consistent with previous mutational, biochemical and biophysical data, and provides direct evidence that the cleavage site in helix I interacts with the 730 loop in helix VI.


Subject(s)
Cross-Linking Reagents/pharmacology , Endoribonucleases/chemistry , Fungal Proteins/chemistry , Neurospora crassa/enzymology , RNA, Catalytic/chemistry , RNA, Fungal/chemistry , Thiouridine/pharmacology , Base Sequence , Binding Sites , Catalytic Domain , Endoribonucleases/drug effects , Endoribonucleases/radiation effects , Fungal Proteins/drug effects , Fungal Proteins/radiation effects , Molecular Sequence Data , Nucleic Acid Conformation , Photochemistry , RNA, Catalytic/drug effects , RNA, Catalytic/radiation effects , RNA, Fungal/drug effects , RNA, Fungal/radiation effects , Ultraviolet Rays
14.
J Mol Biol ; 320(3): 443-54, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12096902

ABSTRACT

We show here that the ribozyme domain of the Neurospora VS ribozyme consists of separable upper and lower subdomains. Deletion analysis demonstrates that the entire upper subdomain (helices III/IV/V) is dispensable for site-specific cleavage activity, providing experimental evidence that the active site is contained within the lower subdomain and within the substrate itself. We demonstrate an important role in cleavage activity for a region of helix VI called the 730 loop. Surprisingly, several loop sequences, sizes, and structures at this position can support site-specific cleavage, suggesting that a variety of non-Watson-Crick structures, rather than a specific loop structure, in this region of the ribozyme can contribute to formation of the active site.


Subject(s)
Endoribonucleases/chemistry , Endoribonucleases/genetics , Neurospora/enzymology , Neurospora/genetics , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , RNA, Fungal/chemistry , RNA, Fungal/genetics , Base Sequence , Catalytic Domain/genetics , Endoribonucleases/metabolism , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleic Acid Conformation , RNA, Catalytic/metabolism , RNA, Fungal/metabolism , Sequence Deletion , Substrate Specificity
15.
Nucleic Acids Res ; 30(5): 1132-8, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11861903

ABSTRACT

We have used nucleotide analog interference mapping and site-specific substitution to determine the effect of 2'-deoxynucleotide substitution of each nucleotide in the VS ribozyme on the self-cleavage reaction. A large number of 2'-hydroxyls (2'-OHs) that contribute to cleavage activity of the VS ribozyme were found distributed throughout the core of the ribozyme. The locations of these 2'-OHs in the context of a recently developed helical orientation model of the VS ribozyme suggest roles in multi-stem junction structure, helix packing, internal loop structure and catalysis. The functional importance of three separate 2'-OHs supports the proposal that three uridine turns contribute to local and long-range tertiary structure formation. A cluster of important 2'-OHs near the loop that is the candidate region for the active site and one very important 2'-OH in the loop that contains the cleavage site confirm the functional importance of these two loops. A cluster of important 2'-OHs lining the minor groove of stem-loop I and helix II suggests that these regions of the backbone may play an important role in positioning helices in the active structure of the ribozyme.


Subject(s)
Endoribonucleases/chemistry , Endoribonucleases/metabolism , Neurospora/enzymology , RNA, Catalytic/chemistry , RNA, Catalytic/metabolism , Base Sequence , Deoxyribonucleotides/chemistry , Hydroxides/chemistry , Molecular Sequence Data , Nucleic Acid Conformation , Thionucleotides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...