Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Protein Sci ; 33(7): e5081, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38924648

ABSTRACT

It has been shown previously that a set of three modifications-termed S1, Crystal Kappa, and elbow-act synergistically to improve the crystallizability of an antigen-binding fragment (Fab) framework. Here, we prepared a phage-displayed library and performed crystallization screenings to identify additional substitutions-located near the heavy-chain elbow region-which cooperate with the S1, Crystal Kappa, and elbow modifications to increase expression and improve crystallizability of the Fab framework even further. One substitution (K141Q) supports the signature Crystal Kappa-mediated Fab:Fab crystal lattice packing interaction. Another substitution (E172G) improves the compatibility of the elbow modification with the Fab framework by alleviating some of the strain incurred by the shortened and bulkier elbow linker region. A third substitution (F170W) generates a split-Fab conformation, resulting in a powerful crystal lattice packing interaction comprising the biological interaction interface between the variable heavy and light chain domains. In sum, we have used K141Q, E172G, and F170W substitutions-which complement the S1, Crystal Kappa, and elbow modifications-to generate a set of highly crystallizable Fab frameworks that can be used as chaperones to enable facile elucidation of Fab:antigen complex structures by x-ray crystallography.


Subject(s)
Immunoglobulin Fab Fragments , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Crystallography, X-Ray , Crystallization , Models, Molecular , Protein Conformation , Humans , Amino Acid Substitution
2.
Elife ; 132024 May 23.
Article in English | MEDLINE | ID: mdl-38780011

ABSTRACT

The receptor tyrosine kinase ROR2 mediates noncanonical WNT5A signaling to orchestrate tissue morphogenetic processes, and dysfunction of the pathway causes Robinow syndrome, brachydactyly B, and metastatic diseases. The domain(s) and mechanisms required for ROR2 function, however, remain unclear. We solved the crystal structure of the extracellular cysteine-rich (CRD) and Kringle (Kr) domains of ROR2 and found that, unlike other CRDs, the ROR2 CRD lacks the signature hydrophobic pocket that binds lipids/lipid-modified proteins, such as WNTs, suggesting a novel mechanism of ligand reception. Functionally, we showed that the ROR2 CRD, but not other domains, is required and minimally sufficient to promote WNT5A signaling, and Robinow mutations in the CRD and the adjacent Kr impair ROR2 secretion and function. Moreover, using function-activating and -perturbing antibodies against the Frizzled (FZ) family of WNT receptors, we demonstrate the involvement of FZ in WNT5A-ROR signaling. Thus, ROR2 acts via its CRD to potentiate the function of a receptor super-complex that includes FZ to transduce WNT5A signals.


Subject(s)
Receptor Tyrosine Kinase-like Orphan Receptors , Signal Transduction , Wnt-5a Protein , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Humans , Animals , Crystallography, X-Ray , Protein Domains , Mice , Protein Conformation , Wnt Proteins/metabolism , Wnt Proteins/genetics
3.
Development ; 151(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38358799

ABSTRACT

The Wnt/ß-catenin signaling governs anterior-posterior neural patterning during development. Current human pluripotent stem cell (hPSC) differentiation protocols use a GSK3 inhibitor to activate Wnt signaling to promote posterior neural fate specification. However, GSK3 is a pleiotropic kinase involved in multiple signaling pathways and, as GSK3 inhibition occurs downstream in the signaling cascade, it bypasses potential opportunities for achieving specificity or regulation at the receptor level. Additionally, the specific roles of individual FZD receptors in anterior-posterior patterning are poorly understood. Here, we have characterized the cell surface expression of FZD receptors in neural progenitor cells with different regional identity. Our data reveal unique upregulation of FZD5 expression in anterior neural progenitors, and this expression is downregulated as cells adopt a posterior fate. This spatial regulation of FZD expression constitutes a previously unreported regulatory mechanism that adjusts the levels of ß-catenin signaling along the anterior-posterior axis and possibly contributes to midbrain-hindbrain boundary formation. Stimulation of Wnt/ß-catenin signaling in hPSCs, using a tetravalent antibody that selectively triggers FZD5 and LRP6 clustering, leads to midbrain progenitor differentiation and gives rise to functional dopaminergic neurons in vitro and in vivo.


Subject(s)
Frizzled Receptors , Glycogen Synthase Kinase 3 , beta Catenin , Humans , beta Catenin/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Glycogen Synthase Kinase 3/metabolism , Mesencephalon , Nervous System/metabolism , Wnt Signaling Pathway , Animals , Rats
4.
Protein Sci ; 33(1): e4824, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37945533

ABSTRACT

The atomic-resolution structural information that X-ray crystallography can provide on the binding interface between a Fab and its cognate antigen is highly valuable for understanding the mechanism of interaction. However, many Fab:antigen complexes are recalcitrant to crystallization, making the endeavor a considerable effort with no guarantee of success. Consequently, there have been significant steps taken to increase the likelihood of Fab:antigen complex crystallization by altering the Fab framework. In this investigation, we applied the surface entropy reduction strategy coupled with phage-display technology to identify a set of surface substitutions that improve the propensity of a human Fab framework to crystallize. In addition, we showed that combining these surface substitutions with previously reported Crystal Kappa and elbow substitutions results in an extraordinary improvement in Fab and Fab:antigen complex crystallizability, revealing a strong synergistic relationship between these sets of substitutions. Through comprehensive Fab and Fab:antigen complex crystallization screenings followed by structure determination and analysis, we defined the roles that each of these substitutions play in facilitating crystallization and how they complement each other in the process.


Subject(s)
Antigen-Antibody Complex , Immunoglobulin Fab Fragments , Humans , Crystallization/methods , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/chemistry , Antigen-Antibody Complex/chemistry , Antigens/chemistry , Crystallography, X-Ray , Protein Conformation
5.
Nat Cancer ; 4(11): 1592-1609, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37904046

ABSTRACT

Safely expanding indications for cellular therapies has been challenging given a lack of highly cancer-specific surface markers. Here we explore the hypothesis that tumor cells express cancer-specific surface protein conformations that are invisible to standard target discovery pipelines evaluating gene or protein expression, and these conformations can be identified and immunotherapeutically targeted. We term this strategy integrating cross-linking mass spectrometry with glycoprotein surface capture 'structural surfaceomics'. As a proof of principle, we apply this technology to acute myeloid leukemia (AML), a hematologic malignancy with dismal outcomes and no known optimal immunotherapy target. We identify the activated conformation of integrin ß2 as a structurally defined, widely expressed AML-specific target. We develop and characterize recombinant antibodies to this protein conformation and show that chimeric antigen receptor T cells eliminate AML cells and patient-derived xenografts without notable toxicity toward normal hematopoietic cells. Our findings validate an AML conformation-specific target antigen and demonstrate a tool kit for applying these strategies more broadly.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Integrins/metabolism , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/genetics
6.
Mol Inform ; 41(9): e2100240, 2022 09.
Article in English | MEDLINE | ID: mdl-35277930

ABSTRACT

There has been a remarkable increase in the number of biologics, especially monoclonal antibodies, in the market over the last decade. In addition to attaining the desired binding to their targets, a crucial aspect is the 'developability' of these drugs, which includes several desirable properties such as high solubility, low viscosity and aggregation, physico-chemical stability, low immunogenicity and low poly-specificity. The lack of any of these desirable properties can lead to significant hurdles in advancing them to the clinic and are often discovered only during late stages of drug development. Hence, in silico methods for early detection of these properties, particularly the ones that affect aggregation and solubility in the earlier stages can be highly beneficial. We have developed a computational framework based on a large and diverse set of protein specific descriptors that is ideal for making liability predictions using a QSPR (quantitative structure-property relationship) approach. This set offers a high degree of feature diversity that may coarsely be classified based on (1) sequence (2) structure and (3) surface patches. We assess the sensitivity and applicability of these descriptors in four dedicated case studies that are believed to be representative of biophysical characterizations commonly employed during the development process of a biologics drug candidate. In addition to data sets obtained from public sources, we have validated the descriptors on novel experimental data sets in order to address antibody developability and to generate prospective predictions on Adnectins. The results show that the descriptors are well suited to assist in the improvement of protein properties of systems that exhibit poor solubility or aggregation.


Subject(s)
Biological Products , Drug Development , Prospective Studies , Quantitative Structure-Activity Relationship , Solubility
7.
Invest Ophthalmol Vis Sci ; 62(13): 15, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34665194

ABSTRACT

Purpose: Integrins play a central role in myofibroblast pathological adhesion, over-contraction, and TGFß activation. Previously, we demonstrated that after corneal wounding, αv integrins are protected from intracellular degradation by upregulation of the deubiquitinase USP10, leading to cell-surface integrin accumulation. Because integrins bind to and internalize extracellular matrix (ECM), we tested whether extracellular fibronectin (FN) accumulation can result from an increase in integrin and matrix recycling in primary human corneal fibroblasts (HCFs). Methods: Primary HCFs were isolated from cadaver eyes. HCFs were transfected with either USP10 cDNA or control cDNA by nucleofection. Internalized FN was quantified with a FN ELISA. Recycled extracellular integrin and FN were detected with streptavidin-488 by live cell confocal microscopy (Zeiss LSM 780). Endogenous FN extra domain A was detected by immunocytochemistry. Cell size and removal of FN from the cell surface was determined by flow cytometry. Results: USP10 overexpression increased α5ß1 (1.9-fold; P < 0.001) and αv (1.7-fold; P < 0.05) integrin recycling, with a concomitant increase in biotinylated FN internalization (2.1-fold; P < 0.05) and recycling over 4 days (1.7-2.2-fold; P < 0.05). The dependence of FN recycling on integrins was demonstrated by α5ß1 and αv integrin blocking antibodies, which, compared with control IgG, decreased biotinylated FN recycling (62% and 84%, respectively; P < 0.05). Overall, we established that extracellular FN was composed of approximately 1/3 recycled biotinylated FN and 2/3 endogenously secreted FN. Conclusions: Our data suggest that reduced integrin degradation with a subsequent increase in integrin/FN recycling after wounding may be a newly identified mechanism for the characteristic accumulation of ECM in corneal scar tissue.


Subject(s)
Cornea/metabolism , Fibronectins/metabolism , Ubiquitin Thiolesterase/biosynthesis , Cell Adhesion , Cell Membrane/metabolism , Cells, Cultured , Cornea/cytology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Flow Cytometry , Humans , Signal Transduction
8.
J Mol Biol ; 433(15): 167090, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34090922

ABSTRACT

Members of the αv family of integrins regulate activation of transforming growth factor beta (TGFß) and are directly involved in pro-tumorigenic phenotypes. Thus, αv integrins may be therapeutic targets for fibrosis and cancer, yet the isolation of selective inhibitors is currently a challenge. We generated synthetic antibodies selective for αv integrins by phage display selections on cell lines that displayed integrin heterodimers. We identified antibodies that targeted two distinct epitopes on cell-surface αv integrins and partially inhibited cell adhesion mediated by interactions between integrins and the latency-associated peptide, part of the pro-form of TGFß. Using the isolated antibody paratope sequences we engineered a bispecific antibody capable of binding to both epitopes simultaneously; this antibody potently and completely inhibited cell adhesion mediated by integrins αvß1, αvß3 and αvß5. In addition, the bispecific antibody inhibited proliferation and migration of lung carcinoma lines, where the highest and lowest potencies observed correlated with integrin-αv cell surface expression levels. Taken together, our results demonstrate that phage display selections with live cells can yield high quality anti-integrin antibodies, which we used as biparatopic building blocks to construct a bispecific antibody that strongly inhibited integrin function and may be a therapeutic candidate for cancer and fibrosis.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Epitopes/metabolism , Integrin alphaV/chemistry , Lung Neoplasms/metabolism , A549 Cells , Animals , Antibodies, Bispecific/chemistry , Antineoplastic Agents, Immunological/chemistry , CHO Cells , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cricetulus , Drug Screening Assays, Antitumor , Humans , Integrin alphaV/metabolism , Lung Neoplasms/drug therapy , Peptide Library
9.
EMBO Mol Med ; 13(7): e13977, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34105895

ABSTRACT

The FZD4:LRP5:TSPAN12 receptor complex is activated by the secreted protein Norrin in retinal endothelial cells and leads to ßcatenin-dependent formation of the blood-retina-barrier during development and its homeostasis in adults. Mutations disrupting Norrin signaling have been identified in several congenital diseases leading to hypovascularization of the retina and blindness. Here, we developed F4L5.13, a tetravalent antibody designed to induce FZD4 and LRP5 proximity in such a way as to trigger ßcatenin signaling. Treatment of cultured endothelial cells with F4L5.13 rescued permeability induced by VEGF in part by promoting surface expression of junction proteins. Treatment of Tspan12-/- mice with F4L5.13 restored retinal angiogenesis and barrier function. F4L5.13 treatment also significantly normalized neovascularization in an oxygen-induced retinopathy model revealing a novel therapeutic strategy for diseases characterized by abnormal angiogenesis and/or barrier dysfunction.


Subject(s)
Endothelial Cells , Retinal Diseases , Animals , Blood-Retinal Barrier , Mice , Retina , Signal Transduction
10.
MAbs ; 13(1): 1933690, 2021.
Article in English | MEDLINE | ID: mdl-34190031

ABSTRACT

In order to direct T cells to specific features of solid cancer cells, we engineered a bispecific antibody format, named Dual Antigen T cell Engager (DATE), by fusing a single-chain variable fragment targeting CD3 to a tumor-targeting antigen-binding fragment. In this format, multiple novel paratopes against different tumor antigens were able to recruit T-cell cytotoxicity to tumor cells in vitro and in an in vivo pancreatic ductal adenocarcinoma xenograft model. Since unique surface antigens in solid tumors are limited, in order to enhance selectivity, we further engineered "double-DATEs" targeting two tumor antigens simultaneously. The double-DATE contains an additional autonomous variable heavy-chain domain, which binds a second tumor antigen without itself eliciting a cytotoxic response. This novel modality provides a strategy to enhance the selectivity of immune redirection through binary targeting of native tumor antigens. The modularity and use of a common, stable human framework for all components enables a pipeline approach to rapidly develop a broad repertoire of tailored DATEs and double-DATEs with favorable biophysical properties and high potencies and selectivities.


Subject(s)
Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , Immunotherapy/methods , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , CD3 Complex/immunology , Carcinoma, Pancreatic Ductal/immunology , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Pancreatic Neoplasms/immunology , Xenograft Model Antitumor Assays
11.
Commun Biol ; 4(1): 561, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980972

ABSTRACT

Synthetic antibody (Ab) technologies are efficient and cost-effective platforms for the generation of monoclonal Abs against human antigens. Yet, they typically depend on purified proteins, which exclude integral membrane proteins that require the lipid bilayers to support their native structure and function. Here, we present an Ab discovery strategy, termed CellectSeq, for targeting integral membrane proteins on native cells in complex environment. As proof of concept, we targeted three transmembrane proteins linked to cancer, tetraspanin CD151, carbonic anhydrase 9, and integrin-α11. First, we performed in situ cell-based selections to enrich phage-displayed synthetic Ab pools for antigen-specific binders. Then, we designed next-generation sequencing procedures to explore Ab diversities and abundances. Finally, we developed motif-based scoring and sequencing error-filtering algorithms for the comprehensive interrogation of next-generation sequencing pools to identify Abs with high diversities and specificities, even at extremely low abundances, which are very difficult to identify using manual sampling or sequence abundances.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Surface Display Techniques/methods , Membrane Proteins/analysis , Antibodies, Monoclonal/biosynthesis , Carbonic Anhydrase IX , Cell Line , Computer Simulation , HEK293 Cells , High-Throughput Nucleotide Sequencing/methods , Humans , Integrin alpha Chains , Membrane Proteins/immunology , Synthetic Biology/methods , Tetraspanins
12.
Protein Sci ; 29(10): 2075-2084, 2020 10.
Article in English | MEDLINE | ID: mdl-32803886

ABSTRACT

Phage-displayed synthetic antibody (Ab) repertoires have become a major source of affinity reagents for basic and clinical research. Specific Abs identified from such libraries are often screened as fragments antigen binding (Fabs) produced in bacteria, and those with desired biochemical characteristics are reformatted for production as full-length immunoglobulin G (IgG) in mammalian cells. The conversion of Fabs to IgGs is a cumbersome and often rate-limiting step in the development of Abs. Moreover, biochemical properties required for lead IgG development are not always shared by the Fabs, and these issues are not uncovered until a significant effort has been spent on Abs that ultimately will not be useful. Thus, there is a need for simple and rapid techniques to convert phage-displayed Fabs to IgGs at an early stage of the Ab screening process. We report the generation of a highly diverse phage-displayed synthetic single-chain Fab (scFab) library, in which the light and heavy chains were tethered with an optimized linker. Following selection, pools of scFabs were converted to single-chain IgGs (scIgGs) en masse, enabling facile screening of hundreds of phage-derived scIgGs. We show that this approach can be used to rapidly screen for and select scIgGs that target cell-surface receptors, and scIgGs behave the same as conventional IgGs.


Subject(s)
Cell Surface Display Techniques , Gene Library , Immunoglobulin Fab Fragments , Immunoglobulin G , Single-Chain Antibodies , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
13.
Elife ; 82019 08 27.
Article in English | MEDLINE | ID: mdl-31452509

ABSTRACT

Secreted Wnt proteins regulate development and adult tissue homeostasis by binding and activating cell-surface Frizzled receptors and co-receptors including LRP5/6. The hydrophobicity of Wnt proteins has complicated their purification and limited their use in basic research and as therapeutics. We describe modular tetravalent antibodies that can recruit Frizzled and LRP5/6 in a manner that phenocopies the activities of Wnts both in vitro and in vivo. The modular nature of these synthetic Frizzled and LRP5/6 Agonists, called FLAgs, enables tailored engineering of specificity for one, two or multiple members of the Frizzled family. We show that FLAgs underlie differentiation of pluripotent stem cells, sustain organoid growth, and activate stem cells in vivo. Activation of Wnt signaling circuits with tailored FLAgs will enable precise delineation of functional outcomes directed by distinct receptor combinations and could provide a new class of therapeutics to unlock the promise of regenerative medicine.


Subject(s)
Antibodies/metabolism , Frizzled Receptors/agonists , Wnt Signaling Pathway/drug effects , Animals , Cell Line , Humans , Low Density Lipoprotein Receptor-Related Protein-5/agonists , Low Density Lipoprotein Receptor-Related Protein-6/agonists , Mice , Organoids/drug effects , Organoids/growth & development , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Protein Binding
14.
Oncoimmunology ; 8(2): e1539613, 2019.
Article in English | MEDLINE | ID: mdl-30713798

ABSTRACT

Epithelial ovarian cancer (EOC) is a leading cause of cancer-related death in women. EOC is often diagnosed at late stages, with peritoneal metastases and ascites production. Current surgery and platinum-based chemotherapy regimes fail to prevent recurrence in most patients. High levels of Transforming growth factor-ß (TGF-ß) within ascites has been linked to poor prognosis. TGF-ß signaling promotes epithelial-mesenchymal transition (EMT) in EOC tumor cells, and immune suppression within the tumor microenvironment, with both contributing to chemotherapy resistance and metastasis. The goal of this study was to develop specific synthetic inhibitory antibodies to the Type II TGF-ß receptor (TGFBR2), and test these antibodies in EOC cell and tumor models. Following screening of a phage-displayed synthetic antigen-binding fragment (Fab) library with the extracellular domain of TGFBR2, we identified a lead inhibitory Fab that suppressed TGF-ß signaling in mouse and human EOC cell lines. Affinity maturation of the lead inhibitory Fab resulted in several derivative Fabs with increased affinity for TGFBR2 and efficacy as suppressors of TGF-ß signaling, EMT and EOC cell invasion. In EOC xenograft and syngeneic tumor models, blockade of TGFBR2 with our lead antibodies led to improved chemotherapy response. This correlated with reversal of EMT and immune exclusion in these tumor models with TGFBR2 blockade. Together, these results describe new inhibitors of the TGF-ß pathway that improve antitumor immunity, and response to chemotherapy in preclinical EOC models.

15.
Methods Mol Biol ; 1869: 155-168, 2019.
Article in English | MEDLINE | ID: mdl-30324522

ABSTRACT

The ability to elucidate the phenotype of brain tumor initiating cell (BTIC) in the context of bulk tumor in glioblastoma multiforme (GBM) provides significant therapeutic benefits for therapeutic evaluation. For the identification of such an elusive and rare subpopulation of cells, a single cell analysis technology with deep profiling capabilities known as Mass Cytometry (CyTOF) can prove to be highly useful. CyTOF circumvents the spectral overlap limitations of traditional flow cytometry by replacing fluorophores with metal isotope tags, allowing the accurate detection of significantly more parameters at the same time. In this chapter, we demonstrate that synthetic antibodies can be conjugated with metal isotope tags for CyTOF analysis, resulting in the development of a highly tailored, custom multi-parameter panel. This toolset was used to stain patient-derived GBM cells, which was analyzed via CyTOF. Analysis software viSNE and SPADE were applied to study the co-expression patterns of the Eph Receptor (EphR) family and several putative BTIC markers in GBM, resulting in the identification of a distinct group of cells consistent with a BTIC subpopulation. This approach can be readily adapted to the detection of cancer stem-like cells in other cancer types.


Subject(s)
Brain Neoplasms/pathology , Ephrins/metabolism , Flow Cytometry/methods , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Antibodies/metabolism , Cell Survival , Humans , Microspheres , Staining and Labeling , Tumor Cells, Cultured
16.
MAbs ; 10(8): 1157-1167, 2018.
Article in English | MEDLINE | ID: mdl-30183492

ABSTRACT

Secreted Wnt ligands play a major role in the development and progression of many cancers by modulating signaling through cell-surface Frizzled receptors (FZDs). In order to achieve maximal effect on Wnt signaling by targeting the cell surface, we developed a synthetic antibody targeting six of the 10 human FZDs. We first identified an anti-FZD antagonist antibody (F2) with a specificity profile matching that of OMP-18R5, a monoclonal antibody that inhibits growth of many cancers by targeting FZD7, FZD1, FZD2, FZD5 and FZD8. We then used combinatorial antibody engineering by phage display to develop a variant antibody F2.A with specificity broadened to include FZD4. We confirmed that F2.A blocked binding of Wnt ligands, but not binding of Norrin, a ligand that also activates FZD4. Importantly, F2.A proved to be much more efficacious than either OMP-18R5 or F2 in inhibiting the growth of multiple RNF43-mutant pancreatic ductal adenocarcinoma cell lines, including patient-derived cells.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Carcinoma, Pancreatic Ductal/immunology , Frizzled Receptors/immunology , Pancreatic Neoplasms/immunology , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Frizzled Receptors/antagonists & inhibitors , Frizzled Receptors/metabolism , HEK293 Cells , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/immunology , Protein Isoforms/metabolism , Sequence Homology, Amino Acid
17.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29945963

ABSTRACT

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Subject(s)
Ephrin-A2/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Ephrin-A2/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Neoplastic Stem Cells/pathology , Prognosis , Radiation , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, EphA3 , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/genetics , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
18.
J Mol Biol ; 430(14): 2139-2152, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29778602

ABSTRACT

Hepatitis C viral infection is the major cause of chronic hepatitis that affects as many as 71 million people worldwide. Rather than target the rapidly shifting viruses and their numerous serotypes, four independent antibodies were made to target the host antigen CD81 and were shown to block hepatitis C viral entry. The single-chain variable fragment of each antibody was crystallized in complex with the CD81 large extracellular loop in order to guide affinity maturation of two distinct antibodies by phage display. Affinity maturation of antibodies using phage display has proven to be critical to therapeutic antibody development and typically involves modification of the paratope for increased affinity, improved specificity, enhanced stability or a combination of these traits. One antibody was engineered for increased affinity for human CD81 large extracellular loop that equated to increased efficacy, while the second antibody was engineered for cross-reactivity with cynomolgus CD81 to facilitate animal model testing. The use of structures to guide affinity maturation library design demonstrates the utility of combining structural analysis with phage display technologies.


Subject(s)
Hepacivirus/physiology , Hepatitis C Antibodies/chemistry , Hepatitis C/immunology , Single-Chain Antibodies/chemistry , Tetraspanin 28/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Antibody Specificity , Binding Sites, Antibody , Cell Line , Hep G2 Cells , Hepacivirus/drug effects , Hepacivirus/immunology , Hepatitis C Antibodies/pharmacology , Humans , Models, Molecular , Peptide Library , Protein Conformation , Single-Chain Antibodies/pharmacology , Structure-Activity Relationship , Tetraspanin 28/chemistry , Virus Internalization/drug effects
19.
Proc Natl Acad Sci U S A ; 115(11): 2836-2841, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29476010

ABSTRACT

Human cells express thousands of different surface proteins that can be used for cell classification, or to distinguish healthy and disease conditions. A method capable of profiling a substantial fraction of the surface proteome simultaneously and inexpensively would enable more accurate and complete classification of cell states. We present a highly multiplexed and quantitative surface proteomic method using genetically barcoded antibodies called phage-antibody next-generation sequencing (PhaNGS). Using 144 preselected antibodies displayed on filamentous phage (Fab-phage) against 44 receptor targets, we assess changes in B cell surface proteins after the development of drug resistance in a patient with acute lymphoblastic leukemia (ALL) and in adaptation to oncogene expression in a Myc-inducible Burkitt lymphoma model. We further show PhaNGS can be applied at the single-cell level. Our results reveal that a common set of proteins including FLT3, NCR3LG1, and ROR1 dominate the response to similar oncogenic perturbations in B cells. Linking high-affinity, selective, genetically encoded binders to NGS enables direct and highly multiplexed protein detection, comparable to RNA-sequencing for mRNA. PhaNGS has the potential to profile a substantial fraction of the surface proteome simultaneously and inexpensively to enable more accurate and complete classification of cell states.


Subject(s)
Antibodies/analysis , Burkitt Lymphoma/genetics , High-Throughput Nucleotide Sequencing/methods , Leukemia/genetics , Membrane Proteins/genetics , Proteomics/methods , Antibodies/genetics , Bacteriophages/genetics , Bacteriophages/metabolism , Burkitt Lymphoma/metabolism , Cell Line, Tumor , Humans , Leukemia/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...