Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(724): eadi0258, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38019931

ABSTRACT

Despite the success of programmed cell death-1 (PD-1) and PD-1 ligand (PD-L1) inhibitors in treating solid tumors, only a proportion of patients respond. Here, we describe a first-in-class bifunctional therapeutic molecule, STAR0602, that comprises an antibody targeting germline Vß6 and Vß10 T cell receptors (TCRs) fused to human interleukin-2 (IL-2) and simultaneously engages a nonclonal mode of TCR activation with costimulation to promote activation and expansion of αß T cell subsets expressing distinct variable ß (Vß) TCR chains. In solution, STAR0602 binds IL-2 receptors in cis with Vß6/Vß10 TCRs on the same T cell, promoting expansion of human Vß6 and Vß10 CD4+ and CD8+ T cells that acquire an atypical central memory phenotype. Monotherapy with a mouse surrogate molecule induced durable tumor regression across six murine solid tumor models, including several refractory to anti-PD-1. Analysis of murine tumor-infiltrating lymphocyte (TIL) transcriptomes revealed that expanded Vß T cells acquired a distinct effector memory phenotype with suppression of genes associated with T cell exhaustion and TCR signaling repression. Sequencing of TIL TCRs also revealed an increased T cell repertoire diversity within targeted Vß T cell subsets, suggesting clonal revival of tumor T cell responses. These immunological and antitumor effects in mice were recapitulated in studies of STAR0602 in nonhuman primates and human ex vivo models, wherein STAR0602 boosted human antigen-specific T cell responses and killing of tumor organoids. Thus, STAR0602 represents a distinct class of T cell-activating molecules with the potential to deliver enhanced antitumor activity in checkpoint inhibitor-refractory settings.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell, alpha-beta , Humans , Animals , Mice , Receptors, Antigen, T-Cell, alpha-beta/metabolism , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Antibodies/pharmacology
2.
MAbs ; 13(1): 1850395, 2021.
Article in English | MEDLINE | ID: mdl-33459147

ABSTRACT

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Enterotoxin/immunology , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , Female , Humans , Hybridomas , Macaca fascicularis/immunology , Macaca fascicularis/metabolism , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Protein Engineering/methods , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Protein Eng Des Sel ; 29(1): 1-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26489878

ABSTRACT

In its basal state, KEAP1 binds the transcription factor NRF2 (Kd = 5 nM) and promotes its degradation by ubiquitylation. Changes in the redox environment lead to modification of key cysteines within KEAP1, resulting in NRF2 protein accumulation and the transcription of genes important for restoring the cellular redox state. Using phage display and a computational loop grafting protocol, we engineered a monobody (R1) that is a potent competitive inhibitor of the KEAP1-NRF2 interaction. R1 bound to KEAP1 with a Kd of 300 pM and in human cells freed NRF2 from KEAP1 resulting in activation of the NRF2 promoter. Unlike cysteine-reactive small molecules that lack protein specificity, R1 is a genetically encoded, reversible inhibitor designed specifically for KEAP1. R1 should prove useful for studying the role of the KEAP1-NRF2 interaction in several disease states. The structure-based phage display strategy employed here is a general approach for engineering high-affinity binders that compete with naturally occurring interactions.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Models, Molecular , NF-E2-Related Factor 2/antagonists & inhibitors , Protein Engineering/methods , Antibodies/chemistry , Antibodies/genetics , Antibodies/metabolism , Cell Surface Display Techniques , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1 , Oxidation-Reduction , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 112(1): 112-7, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25535392

ABSTRACT

The discovery of light-inducible protein-protein interactions has allowed for the spatial and temporal control of a variety of biological processes. To be effective, a photodimerizer should have several characteristics: it should show a large change in binding affinity upon light stimulation, it should not cross-react with other molecules in the cell, and it should be easily used in a variety of organisms to recruit proteins of interest to each other. To create a switch that meets these criteria we have embedded the bacterial SsrA peptide in the C-terminal helix of a naturally occurring photoswitch, the light-oxygen-voltage 2 (LOV2) domain from Avena sativa. In the dark the SsrA peptide is sterically blocked from binding its natural binding partner, SspB. When activated with blue light, the C-terminal helix of the LOV2 domain undocks from the protein, allowing the SsrA peptide to bind SspB. Without optimization, the switch exhibited a twofold change in binding affinity for SspB with light stimulation. Here, we describe the use of computational protein design, phage display, and high-throughput binding assays to create an improved light inducible dimer (iLID) that changes its affinity for SspB by over 50-fold with light stimulation. A crystal structure of iLID shows a critical interaction between the surface of the LOV2 domain and a phenylalanine engineered to more tightly pin the SsrA peptide against the LOV2 domain in the dark. We demonstrate the functional utility of the switch through light-mediated subcellular localization in mammalian cell culture and reversible control of small GTPase signaling.


Subject(s)
Light , Plant Proteins/metabolism , Protein Engineering , Protein Multimerization/radiation effects , Signal Transduction/radiation effects , Amino Acid Sequence , Avena , Cell Surface Display Techniques , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Plant Proteins/chemistry , Protein Structure, Tertiary , Protein Transport/radiation effects , Subcellular Fractions/metabolism
5.
Nat Biotechnol ; 32(2): 191-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24463572

ABSTRACT

Robust generation of IgG bispecific antibodies has been a long-standing challenge. Existing methods require extensive engineering of each individual antibody, discovery of common light chains, or complex and laborious biochemical processing. Here we combine computational and rational design approaches with experimental structural validation to generate antibody heavy and light chains with orthogonal Fab interfaces. Parental monoclonal antibodies incorporating these interfaces, when simultaneously co-expressed, assemble into bispecific IgG with improved heavy chain-light chain pairing. Bispecific IgGs generated with this approach exhibit pharmacokinetic and other desirable properties of native IgG, but bind target antigens monovalently. As such, these bispecific reagents may be useful in many biotechnological applications.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin G/chemistry , Protein Engineering/methods , Animals , Antibodies, Bispecific/metabolism , Biotechnology , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/metabolism , Male , Mice , Models, Biological , Models, Molecular , Protein Binding , Protein Conformation
6.
Proteins ; 81(7): 1245-55, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23504819

ABSTRACT

We computationally designed a de novo protein-protein interaction between wild-type ubiquitin and a redesigned scaffold. Our strategy was to incorporate zinc at the designed interface to promote affinity and orientation specificity. A large set of monomeric scaffold surfaces were computationally engineered with three-residue zinc coordination sites, and the ubiquitin residue H68 was docked to the open coordination site to complete a tetrahedral zinc site. This single coordination bond was intended as a hotspot and polar interaction for ubiquitin binding, and surrounding residues on the scaffold were optimized primarily as hydrophobic residues using a rotamer-based sequence design protocol in Rosetta. From thousands of independent design simulations, four sequences were selected for experimental characterization. The best performing design, called Spelter, binds tightly to zinc (Kd < 10 nM) and binds ubiquitin with a Kd of 20 µM in the presence of zinc and 68 µM in the absence of zinc. Mutagenesis studies and nuclear magnetic resonance chemical shift perturbation experiments indicate that Spelter interacts with H68 and the target surface on ubiquitin; however, H68 does not form a hotspot as intended. Instead, mutation of H68 to alanine results in tighter binding. Although a 3/1 zinc coordination arrangement at an interface cannot be ruled out as a means to improve affinity, our study led us to conclude that 2/2 coordination arrangements or multiple-zinc designs are more likely to promote high-affinity protein interactions.


Subject(s)
Protein Binding , Ubiquitin/chemistry , Zinc/chemistry , Alanine/chemistry , Binding Sites , Magnetic Resonance Spectroscopy , Mutagenesis , Protein Interaction Maps
7.
PLoS One ; 7(4): e35998, 2012.
Article in English | MEDLINE | ID: mdl-22536452

ABSTRACT

Generating diverse protein libraries that contain improved variants at a sufficiently high frequency is critical for improving the properties of proteins using directed evolution. Many studies have illustrated how random mutagenesis, cassette mutagenesis, DNA shuffling and similar approaches are effective diversity generating methods for directed evolution. Very few studies have explored random circular permutation, the intramolecular relocation of the N- and C-termini of a protein, as a diversity-generating step for directed evolution. We subjected a library of random circular permutations of TEM-1 ß-lactamase to selections on increasing concentrations of a variety of ß-lactam antibiotics including cefotaxime. We identified two circularly permuted variants that conferred elevated resistance to cefotaxime but decreased resistance to other antibiotics. These variants were circularly permuted in the Ω-loop proximal to the active site. Remarkably, one variant was circularly permuted such that the key catalytic residue Glu166 was located at the N-terminus of the mature protein.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/drug effects , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , Amino Acid Motifs , Anti-Bacterial Agents/pharmacology , Base Sequence , Catalytic Domain , Cefotaxime/pharmacology , Cloning, Molecular , Directed Molecular Evolution , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Gene Library , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis , Substrate Specificity , beta-Lactamases/chemistry
8.
J Mol Biol ; 418(3-4): 161-6, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22391419

ABSTRACT

Pathways of ubiquitin-like (UBL) molecule transfer regulate a myriad of cellular cascades. Here, we report a high-throughput assay that correlates catalytic human NEDD8 transfer to bacterial survival. The assay was utilized to screen mutant NEDD8 and NAE (NEDD8-activating enzyme) libraries to engineer a more stable NEDD8 and redesign the NEDD8-NAE interface. This approach will be useful in understanding the specificities underlying UBL pathways.


Subject(s)
Escherichia coli/metabolism , Signal Transduction , Ubiquitin-Activating Enzymes/genetics , Ubiquitin/metabolism , Ubiquitins/chemistry , Ubiquitins/metabolism , Amino Acid Sequence , Binding Sites , Escherichia coli/genetics , High-Throughput Screening Assays/methods , Humans , Microbial Viability , Models, Molecular , Mutation , NEDD8 Protein , Protein Engineering , Ubiquitin/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/genetics
9.
Biotechnol Bioeng ; 108(11): 2535-43, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21618478

ABSTRACT

We demonstrate that S1 nuclease converts supercoiled plasmid DNA to unit-length, linear dsDNA through the creation of a single, double-stranded break in a plasmid molecule. These double-stranded breaks occur not only in the origin of replication near inverted repeats but also at a wide variety of locations throughout the plasmid. S1 nuclease exhibits this activity under conditions typically employed for the nuclease's single-stranded nuclease activity. Thus, S1 nuclease digestion of plasmid DNA, unlike analogous digestion with DNaseI, effectively halts after the first double-stranded break. This property makes easier the construction of large domain insertion libraries in which the goal is to insert linear DNA at a variety of locations throughout a plasmid. We used this property to create a library in which a circularly permuted TEM1 ß-lactamase gene was inserted throughout a plasmid containing the gene encoding Escherichia coli ribose binding protein. Gene fusions that encode allosteric switch proteins in which ribose modulates ß-lactamase catalytic activity were isolated from this library using a combination of a genetic selection and a screen.


Subject(s)
DNA/metabolism , Fungal Proteins/metabolism , Mutagenesis, Insertional/methods , Plasmids/metabolism , Proteins/genetics , Proteins/metabolism , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Artificial Gene Fusion , Escherichia coli/genetics , Gene Library , Recombination, Genetic , beta-Lactamases/genetics
10.
Proc Natl Acad Sci U S A ; 107(45): 19296-301, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974935

ABSTRACT

Computational algorithms for protein design can sample large regions of sequence space, but suffer from undersampling of conformational space and energy function inaccuracies. Experimental screening of combinatorial protein libraries avoids the need for accurate energy functions, but has limited access to vast amounts of sequence space. Here, we test if these two traditionally alternative, but potentially complementary approaches can be combined to design a variant of the ubiquitin-ligase E6AP that will bind to a nonnatural partner, the NEDD8-conjugating enzyme Ubc12. Three E6AP libraries were constructed: (i) a naive library in which all 20 amino acids were allowed at every position on the target-binding surface of E6AP (13 positions), (ii) a semidirected library that varied the same residue positions as in the naive library but disallowed mutations computationally predicted to destabilize E6AP, and (iii) a directed library that used docking and sequence optimization simulations to identify mutations predicted to be favorable for binding Ubc12. Both of the directed libraries showed > 30-fold enrichment over the naive library after the first round of screening with a split-dihydrofolate reductase complementation assay and produced multiple tight binders (K(d) < 100 nM) after four rounds of selection. Four rounds of selection with the naive library failed to produce any binders with K(d)'s lower than 50 µM. These results indicate that protein design simulations can be used to create directed libraries that are enriched in tight binders and that in some cases it is sufficient to computationally screen for well-folded sequences without explicit binding calculations.


Subject(s)
Algorithms , Models, Molecular , Peptide Library , Protein Engineering/methods , Protein Interaction Domains and Motifs , Drug Design , Drug Evaluation, Preclinical/methods , Humans , Protein Binding , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
11.
Proc Natl Acad Sci U S A ; 102(32): 11224-9, 2005 Aug 09.
Article in English | MEDLINE | ID: mdl-16061816

ABSTRACT

We describe an iterative approach for creating protein switches involving the in vitro recombination of two nonhomologous genes. We demonstrate this approach by recombining the genes coding for TEM1 beta-lactamase (BLA) and the Escherichia coli maltose binding protein (MBP) to create a family of MBP-BLA hybrids in which maltose is a positive or negative effector of beta-lactam hydrolysis. Some of these MBP-BLA switches were effectively "on-off" in nature, with maltose altering catalytic activity by as much as 600-fold. The ability of these switches to confer an effector-dependent growth/no growth phenotype to E. coli cells was exploited to rapidly identify, from a library of 4 x 10(6) variants, MBP-BLA switch variants that respond to sucrose as the effector. The transplantation of these mutations into wild-type MBP converted MBP into a "sucrose-binding protein," illustrating the switches potential as a tool to rapidly identify ligand-binding proteins.


Subject(s)
Allosteric Regulation/genetics , Directed Molecular Evolution , Escherichia coli Proteins/metabolism , Protein Binding , Protein Engineering/methods , Allosteric Regulation/physiology , Carrier Proteins/metabolism , Catalysis , Escherichia coli , Genes, Switch/genetics , Ligands , Maltose-Binding Proteins , Sucrose/metabolism , beta-Lactamases/metabolism
12.
Chem Biol ; 11(11): 1483-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15555998

ABSTRACT

We have created a molecular switch by the in vitro recombination of nonhomologous genes and subjecting the recombined genes to evolutionary pressure. The gene encoding TEM1 beta-lactamase was circularly permuted in a random fashion and subsequently randomly inserted into the gene encoding Escherichia coli maltose binding protein. From this library, a switch (RG13) was identified in which its beta-lactam hydrolysis activity was compromised in the absence of maltose but increased 25-fold in the presence of maltose. Upon removal of maltose, RG13's catalytic activity returned to its premaltose level, illustrating that the switching is reversible. The modularity of RG13 was demonstrated by increasing maltose affinity while preserving switching activity. RG13 gave rise to a novel cellular phenotype, illustrating the potential of molecular switches to rewire the cellular circuitry.


Subject(s)
Escherichia coli Proteins/genetics , Periplasmic Binding Proteins/genetics , Protein Engineering , Recombination, Genetic , beta-Lactamases/genetics , Animals , Genes, Switch , Models, Molecular , Sequence Homology
13.
J Mol Biol ; 336(1): 263-73, 2004 Feb 06.
Article in English | MEDLINE | ID: mdl-14741221

ABSTRACT

Two allosteric enzymes have been created by the covalent linkage of non-interacting, monomeric proteins with the prerequisite effector-binding and catalytic functionalities, respectively. This was achieved through a combinatorial process called random domain insertion. The fragment of the TEM-1 beta-lactamase gene coding for the mature protein lacking its signal sequence was randomly inserted into the Escherichia coli maltose-binding protein (MBP) gene to create a domain insertion library. This library's diversity derived both from the site of insertion and from a distribution of tandem duplications or deletions of a portion of the MBP gene at the insertion site. From a library of approximately 2 x 10(4) in-frame fusions, approximately 800 library members conferred a phenotype to E.coli cells that was consistent with the presence of bifunctional fusions that could hydrolyze ampicillin and transport maltose in E.coli. Partial screening of this bifunctional sublibrary resulted in the identification of two enzymes in which the presence of maltose modulated the rate of nitrocefin hydrolysis. For one of these enzymes, the presence of maltose increased k(cat) by 70% and k(cat)/K(m) by 80% and resulted in kinetic parameters that were almost identical to TEM-1 beta-lactamase. Such an increase in activity was only observed with maltooligosaccharides whose binding to MBP is known to induce a conformational change. Modulation of the rate of nitrocefin hydrolysis could be detected at maltose concentrations less than 1 microM. Intrinsic protein fluorescence studies were consistent with a conformational change being responsible for the modulation of activity.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Peptide Fragments/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism , Allosteric Regulation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Cephalosporins/metabolism , Gene Library , Maltose-Binding Proteins , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Plasmids/genetics , Plasmids/metabolism , Protein Conformation , Protein Structure, Tertiary , beta-Lactamases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...