Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Exp Immunol ; 215(2): 105-119, 2024 02 07.
Article in English | MEDLINE | ID: mdl-37930865

ABSTRACT

T-cell-engaging bispecifics have great clinical potential for the treatment of cancer and infectious diseases. The binding affinity and kinetics of a bispecific molecule for both target and T-cell CD3 have substantial effects on potency and specificity, but the rules governing these relationships are not fully understood. Using immune mobilizing monoclonal TCRs against cancer (ImmTAC) molecules as a model, we explored the impact of altering affinity for target and CD3 on the potency and specificity of the redirected T-cell response. This class of bispecifics binds specific target peptides presented by human leukocyte antigen on the cell surface via an affinity-enhanced T-cell receptor and can redirect T-cell activation with an anti-CD3 effector moiety. The data reveal that combining a strong affinity TCR with an intermediate affinity anti-CD3 results in optimal T-cell activation, while strong affinity of both targeting and effector domains significantly reduces maximum cytokine release. Moreover, by optimizing the affinity of both parts of the molecule, it is possible to improve the selectivity. These results could be effectively modelled based on kinetic proofreading with limited signalling. This model explained the experimental observation that strong binding at both ends of the molecules leads to reduced activity, through very stable target-bispecific-effector complexes leading to CD3 entering a non-signalling dark state. These findings have important implications for the design of anti-CD3-based bispecifics with optimal biophysical parameters for both activity and specificity.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Receptors, Antigen, T-Cell , T-Lymphocytes , Cytokines , CD3 Complex
2.
JCI Insight ; 6(20)2021 10 22.
Article in English | MEDLINE | ID: mdl-34491911

ABSTRACT

The PD-1/PD-L1 pathway is a key immune checkpoint that regulates T cell activation. There is strong rationale to develop PD-1 agonists as therapeutics against autoimmunity, but progress in this area has been limited. Here, we generated T cell receptor (TCR) targeting, PD-1 agonist bispecifics called ImmTAAI molecules that mimic the ability of PD-L1 to facilitate the colocalization of PD-1 with the TCR complex at the target cell-T cell interface. PD-1 agonist ImmTAAI molecules specifically bound to target cells and were highly effective in activating the PD-1 receptor on interacting T cells to achieve immune suppression. Potent PD-1 antibody ImmTAAI molecules closely mimicked the mechanism of action of endogenously expressed PD-L1 in their localization to the target cell-T cell interface, inhibition of proximal TCR signaling events, and suppression of T cell function. At picomolar concentrations, these bispecifics suppressed cytokine production and inhibited CD8+ T cell-mediated cytotoxicity in vitro. Crucially, in soluble form, the PD-1 ImmTAAI molecules were inactive and, hence, could avoid systemic immunosuppression. This study outlines a promising new route to generate more effective, potent, tissue-targeted PD-1 agonists that can inhibit T cell function locally with the potential to treat autoimmune and chronic inflammatory diseases of high unmet need.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/antagonists & inhibitors , Humans
3.
J Immunol Res ; 2018: 4089459, 2018.
Article in English | MEDLINE | ID: mdl-30417018

ABSTRACT

Lymphocyte costimulation plays a central role in immunology, inflammation, and immunotherapy. The inducible T cell costimulator (ICOS) is expressed on T cells following peptide: MHC engagement with CD28 costimulation. The interaction of ICOS with its sole ligand, the inducible T cell costimulatory ligand (ICOSL; also known as B7-related protein-1), triggers a number of key activities of T cells including differentiation and cytokine production. Suppression of T cell activation can be achieved by blocking this interaction and has been shown to be an effective means of ameliorating disease in models of autoimmunity. In this study, we isolated specific anti-ICOSL new antigen receptor domains from a synthetic phage display library and demonstrated their ability to block the ICOS/ICOSL interaction and inhibit T cell proliferation. Anti-mouse ICOSL domains, considered here as surrogates for the use of anti-human ICOSL domains in patient therapy, were tested for efficacy in a collagen-induced mouse model of rheumatoid arthritis where they significantly decreased the inflammation of joints and delayed and reduced overall disease progression and severity.


Subject(s)
Arthritis, Experimental/therapy , Arthritis, Rheumatoid/immunology , Immunotherapy/methods , Inflammation/therapy , Receptors, Antigen, B-Cell/therapeutic use , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/drug effects , Animals , Arthritis, Experimental/immunology , CHO Cells , Cell Proliferation , Cell Surface Display Techniques , Cricetulus , Disease Models, Animal , Female , Humans , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Inflammation/immunology , Lymphocyte Activation , Mice , Mice, Inbred DBA , Protein Binding , Receptors, Antigen, B-Cell/genetics , Single-Chain Antibodies/genetics , T-Lymphocytes/immunology
4.
Methods Mol Biol ; 907: 177-94, 2012.
Article in English | MEDLINE | ID: mdl-22907351

ABSTRACT

The drive to exploit novel targets and biological pathways has lead to the expansion of classical antibody research into innovative fragment adaptations and novel scaffolds. The hope being that alternative or cryptic epitopes may be targeted, tissue inaccessibility may be overcome, and easier engineering options will facilitate multivalent, multi-targeting approaches. To this end, we have been isolating shark single domains to gain a greater understanding of their potential as therapeutic agents. Their unique shape, small size, inherent stability, and simple molecular architecture make them attractive candidates from a drug discovery perspective. Here we describe protocols to capture the immune repertoire of an immunized shark species and to build and select via phage-display target-specific IgNAR variable domains (VNARs).


Subject(s)
Molecular Biology/methods , Sharks/immunology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/isolation & purification , Animals , Bacteriophages/metabolism , Cell Surface Display Techniques , DNA, Complementary/biosynthesis , Epitopes/immunology , Immunization , Protein Structure, Tertiary , RNA/blood , Single-Domain Antibodies/blood , Single-Domain Antibodies/chemistry
5.
MAbs ; 4(6): 673-85, 2012.
Article in English | MEDLINE | ID: mdl-23676205

ABSTRACT

Advances in recombinant antibody technology and protein engineering have provided the opportunity to reduce antibodies to their smallest binding domain components and have concomitantly driven the requirement for devising strategies to increase serum half-life to optimise drug exposure, thereby increasing therapeutic efficacy. In this study, we adopted an immunization route to raise picomolar affinity shark immunoglobulin new antigen receptors (IgNARs) to target human serum albumin (HSA). From our model shark species, Squalus acanthias, a phage display library encompassing the variable binding domain of IgNAR (VNAR) was constructed, screened against target, and positive clones were characterized for affinity and specificity. N-terminal and C-terminal molecular fusions of our lead hit in complex with a naïve VNAR domain were expressed, purified and exhibited the retention of high affinity binding to HSA, but also cross-selectivity to mouse, rat and monkey serum albumin both in vitro and in vivo. Furthermore, the naïve VNAR had enhanced pharmacokinetic (PK) characteristics in both N- and C-terminal orientations and when tested as a three domain construct with naïve VNAR flanking the HSA binding domain at both the N and C termini. Molecules derived from this platform technology also demonstrated the potential for clinical utility by being available via the subcutaneous route of delivery. This study thus demonstrates the first in vivo functional efficacy of a VNAR binding domain with the ability to enhance PK properties and support delivery of multifunctional therapies.


Subject(s)
Biological Products/pharmacokinetics , Receptors, Antigen/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Single-Domain Antibodies/metabolism , Animals , Antibody Affinity , Antibody Specificity , Drug Design , Haplorhini , Humans , Mice , Protein Engineering/methods , Rats , Receptors, Antigen/genetics , Recombinant Fusion Proteins/genetics , Serum Albumin/immunology , Sharks , Single-Domain Antibodies/genetics
6.
Appl Biochem Biotechnol ; 159(1): 178-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18931951

ABSTRACT

Low yields, poor folding efficiencies and improper disulfide bridge formation limit large-scale production of cysteine-rich proteins in Escherichia coli. Human renal dipeptidase (MDP), the only human beta-lactamase known to date, is a homodimeric enzyme, which contains six cysteine residues per monomer. It hydrolyses penem and carbapenem beta-lactam antibiotics and can cleave dipeptides containing amino acids in both D: - and L: -configurations. In this study, MDP accumulated in inactive form in high molecular weight, disulfide-linked aggregates when produced in the E. coli periplasm. Mutagenesis of Cys361 that mediates dimer formation and Cys93 that is unpaired in the native MDP led to production of soluble recombinant enzyme, with no change in activity compared with the wild-type enzyme. The removal of unpaired or structurally inessential cysteine residues in this manner may allow functional production of many multiply disulfide-linked recombinant proteins in E. coli.


Subject(s)
Cysteine/chemistry , Cysteine/metabolism , Dipeptidases/chemistry , Dipeptidases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Enhancement/methods , Mutagenesis, Site-Directed/methods , Protein Engineering/methods , Cysteine/genetics , Dipeptidases/genetics , Humans
7.
J Biotechnol ; 120(1): 38-45, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16019098

ABSTRACT

Domoic acid is a potent neuroexcitatory toxin that causes amnesic shellfish poisoning in humans through ingestion of contaminated shellfish. The variable regions of the heavy chain (V(H)) and light chain (V(L)) of an antibody specific for domoic acid were cloned from a mouse hybridoma cell line and used to construct single-chain antibody fragments (scFvs) in a variety of formats. V(H)-linker-V(L) scFvs were expressed better in Escherichia coli than the V(L)-linker-V(H) format, while use of the commonly used (Gly4Ser)3 inter-domain linker resulted in higher yields than a longer (Gly4Ser)6 linker variant. Higher soluble protein yields were achieved in E. coli TOP 10 than in E. coli XL1-Blue cells and co-production of the E. coli disulfide bond isomerase enzyme DsbC allowed higher cell densities to be attained during scFv production, leading to increased yields of recombinant protein. The purified scFv exhibited binding similar to the parent monoclonal antibody and is being used to develop an immunosensor to detect domoic acid in contaminated shellfish samples.


Subject(s)
Antibodies, Monoclonal/immunology , Cloning, Molecular/methods , Escherichia coli/immunology , Escherichia coli/metabolism , Immunoglobulin Fragments/immunology , Kainic Acid/analogs & derivatives , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Escherichia coli/genetics , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Kainic Acid/immunology , Mice , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...