Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Nat Commun ; 15(1): 3259, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38627419

The heterogeneity inherent in today's biotherapeutics, especially as a result of heavy glycosylation, can affect a molecule's safety and efficacy. Characterizing this heterogeneity is crucial for drug development and quality assessment, but existing methods are limited in their ability to analyze intact glycoproteins or other heterogeneous biotherapeutics. Here, we present an approach to the molecular assessment of biotherapeutics that uses proton-transfer charge-reduction with gas-phase fractionation to analyze intact heterogeneous and/or glycosylated proteins by mass spectrometry. The method provides a detailed landscape of the intact molecular weights present in biotherapeutic protein preparations in a single experiment. For glycoproteins in particular, the method may offer insights into glycan composition when coupled with a suitable bioinformatic strategy. We tested the approach on various biotherapeutic molecules, including Fc-fusion, VHH-fusion, and peptide-bound MHC class II complexes to demonstrate efficacy in measuring the proteoform-level diversity of biotherapeutics. Notably, we inferred the glycoform distribution for hundreds of molecular weights for the eight-times glycosylated fusion drug IL22-Fc, enabling correlations between glycoform sub-populations and the drug's pharmacological properties. Our method is broadly applicable and provides a powerful tool to assess the molecular heterogeneity of emerging biotherapeutics.


Glycoproteins , Polysaccharides , Glycosylation , Glycoproteins/metabolism , Mass Spectrometry/methods , Polysaccharides/metabolism
2.
Neurobiol Dis ; 177: 105969, 2023 02.
Article En | MEDLINE | ID: mdl-36535551

Parkinson's disease (PD), a neurodegenerative disease affecting dopaminergic (DA) neurons, is characterized by decline of motor function and cognition. Dopaminergic cell loss is associated with accumulation of toxic alpha synuclein aggregates. As DA neuron death occurs late in the disease, therapeutics that block the spread of alpha synuclein may offer functional benefit and delay disease progression. To test this hypothesis, we generated antibodies to the C terminal region of synuclein with high nanomolar affinity and characterized them in in vitro and in vivo models of spread. Interestingly, we found that only antibodies with high affinity to the distal most portion of the C-terminus robustly reduced uptake of alpha synuclein preformed fibrils (PFF) and accumulation of phospho (S129) alpha synuclein in cell culture. Additionally, the antibody treatment blocked the spread of phospho (S129) alpha synuclein associated-pathology in a mouse model of synucleinopathy. Blockade of neuronal PFF uptake by different antibodies was more predictive of in vivo activity than their binding potency to monomeric or oligomeric forms of alpha synuclein. These data demonstrate that antibodies directed to the C-terminus of the alpha synuclein have differential effects on target engagement and efficacy. Furthermore, our data provides additional support for the development of alpha synuclein antibodies as a therapeutic strategy for PD patients.


Neurodegenerative Diseases , Parkinson Disease , Synucleinopathies , Mice , Animals , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Neurodegenerative Diseases/metabolism , Synucleinopathies/pathology , Dopaminergic Neurons/metabolism
3.
Transl Vis Sci Technol ; 11(10): 27, 2022 10 03.
Article En | MEDLINE | ID: mdl-36255358

Purpose: Diabetic macular edema (DME) is the leading cause of vision loss and blindness among working-age adults. Although current intravitreal anti-vascular endothelial growth factor (VEGF) therapies improve vision for many patients with DME, approximately half do not achieve the visual acuity required to drive. We therefore sought additional approaches to resolve edema and improve vision for these patients. Methods: We explored direct agonists of Tie2, a receptor known to stabilize vasculature and prevent leakage. We identified a multivalent PEG-Fab conjugate, Tie2.1-hexamer, that oligomerizes Tie2 and drives receptor activation and characterized its activities in vitro and in vivo. Results: Tie2.1-hexamer normalized and stabilized intercellular junctions of stressed endothelial cell monolayers in vitro, suppressed vascular leak in mice under conditions where anti-VEGF alone was ineffective, and demonstrated extended ocular exposure and robust pharmacodynamic responses in non-human primates. Conclusions: Tie2.1-hexamer directly activates the Tie2 pathway, reduces vascular leak, and is persistent within the vitreal humor. Translational Relevance: Our study presents a promising potential therapeutic for the treatment of DME.


Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Mice , Animals , Macular Edema/drug therapy , Macular Edema/etiology , Diabetic Retinopathy/drug therapy , Endothelial Growth Factors/therapeutic use , Visual Acuity , Vision Disorders/complications , Vision Disorders/drug therapy , Blindness/complications
4.
MAbs ; 14(1): 2115213, 2022.
Article En | MEDLINE | ID: mdl-36206404

T cell-engaging bispecific antibodies (TCEs) are clinically effective treatments for hematological cancers. While the utility of TCEs in solid malignancies is being explored, toxicities arising from antigen expression on normal tissues have slowed or halted several clinical trials. Here, we describe the development of TCEs that preferentially drive T cell-mediated death against target cells co-expressing two tumor-associated antigens. We show that Ly6E and B7-H4 are simultaneously expressed on approximately 50% of breast cancers, whereas normal tissue expression is limited and mostly orthogonal. Traditional bispecific TCEs targeting a singular antigen, either Ly6E or B7-H4, are active when paired with high-affinity CD3-engagers, but normal tissue expression presents a toxicity risk. Treatment with a murine cross-reactive B7-H4-TCE results in rapid and severe weight loss in mice along with damage to B7-H4-expressing tissues. To overcome on-target toxicity, we designed trispecific antibodies co-targeting Ly6E, B7-H4, and CD3 and characterized the impact of dual-antigen binding and the relative placement of each binding domain on tumor killing in vitro and in vivo. In vitro killing of tumor cells co-expressing both antigens correlates to the placement of the higher affinity B7-H4 binding domain, with only modest enhancements seen upon addition of Ly6E binding. In xenograft models, avid binding of appropriately designed trispecific TCEs enables tumor growth inhibition while evading the poor tolerability seen with active bispecific TCEs. Collectively these data highlight the potential for dual-antigen targeting to improve safety and efficacy, and expand the scope of tumors that may effectively be treated by TCEs.Abbreviations: Chimeric antigen receptor T cells (CAR-Ts), dual-antigen targeted T cell engagers (DAT-TCE), Fragment antigen-binding (Fab), Hematoxylin and eosin (H&E), Institutional Animal Care and Use Committee (IACUC), Immunoglobulin G (IgG), immunohistochemistry (IHC), NOD SCID gamma (NSG), peripheral blood mononuclear cells (PBMCs), surface plasmon resonance (SPR), T cell-engagers (TCEs).


Antibodies, Bispecific , Receptors, Chimeric Antigen , Animals , Antibodies, Bispecific/pharmacology , Cell Line, Tumor , Eosine Yellowish-(YS) , Hematoxylin , Humans , Immunoglobulin G , Leukocytes, Mononuclear , Mice , Mice, SCID , T-Lymphocytes , Xenograft Model Antitumor Assays
5.
Sci Transl Med ; 13(593)2021 05 12.
Article En | MEDLINE | ID: mdl-33980574

Tau has become an attractive alternative target for passive immunotherapy efforts for Alzheimer's disease (AD). The anatomical distribution and extent of tau pathology correlate with disease course and severity better than other disease markers to date. We describe here the generation, preclinical characterization, and phase 1 clinical characterization of semorinemab, a humanized anti-tau monoclonal antibody with an immunoglobulin G4 (igG4) isotype backbone. Semorinemab binds all six human tau isoforms and protects neurons against tau oligomer neurotoxicity in cocultures of neurons and microglia. In addition, when administered intraperitoneally once weekly for 13 weeks, murine versions of semorinemab reduced the accumulation of tau pathology in a transgenic mouse model of tauopathy, independent of antibody effector function status. Semorinemab also showed clear evidence of target engagement in vivo, with increases in systemic tau concentrations observed in tau transgenic mice, nonhuman primates, and humans. Higher concentrations of systemic tau were observed after dosing in AD participants compared to healthy control participants. No concerning safety signals were observed in the phase 1 clinical trial at single doses up to 16,800 mg and multiple doses totaling 33,600 mg in a month.


Alzheimer Disease , Tauopathies , Alzheimer Disease/drug therapy , Animals , Brain/metabolism , Disease Models, Animal , Humans , Immunization, Passive , Mice , Mice, Transgenic , Tauopathies/drug therapy , tau Proteins/metabolism
6.
MAbs ; 11(4): 735-746, 2019.
Article En | MEDLINE | ID: mdl-30900945

Traditional hybridoma and B cell cloning antibody discovery platforms have inherent limits in immune repertoire sampling depth. One consequence is that monoclonal antibody (mAb) leads often lack the necessary affinity for therapeutic applications, thus requiring labor-intensive and time-consuming affinity in vitro engineering optimization steps. Here, we show that high-affinity variants of mouse-derived mAbs can be rapidly obtained by testing of somatic sequence variants obtained by deep sequencing of antibody variable regions in immune repertories from immunized mice, even with a relatively sparse sampling of sequence variants from large sequence datasets. Affinity improvements can be achieved for mAbs with a wide range of affinities. The optimized antibody variants derived from immune repertoire mining have no detectable in vitro off-target binding and have in vivo clearance comparable to the parental mAbs, essential properties in therapeutic antibody leads. As generation of antibody variants in vitro is replaced by mining of variants generated in vivo, the procedure can be applied to rapidly identify affinity-optimized mAb variants.


Antibodies, Monoclonal/metabolism , B-Lymphocytes/immunology , Immunoglobulin Variable Region/genetics , Parkinson Disease/therapy , alpha-Synuclein/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Affinity , Clone Cells , High-Throughput Nucleotide Sequencing , Humans , Hybridomas , Immunization , Mice , Mice, Inbred C57BL , Parkinson Disease/immunology , Somatic Hypermutation, Immunoglobulin
7.
MAbs ; 10(7): 1073-1083, 2018 10.
Article En | MEDLINE | ID: mdl-30130444

Developability assessment of therapeutic antibody candidates assists drug discovery by enabling early identification of undesirable instabilities. Rapid chemical stability screening of antibody variants can accelerate the identification of potential solutions. We describe here the development of a high-throughput assay to characterize asparagine deamidation. We applied the assay to identify a mutation that unexpectedly stabilizes a critical asparagine. Ninety antibody variants were incubated under thermal stress in order to induce deamidation and screened for both affinity and total binding capacity. Surprisingly, a mutation five residues downstream from the unstable asparagine greatly reduced deamidation. Detailed assessment by LC-MS analysis confirmed the predicted improvement. This work describes both a high-throughput method for antibody stability screening during the early stages of antibody discovery and highlights the value of broad searches of antibody sequence space.


Antibodies, Monoclonal/chemistry , Antibodies/chemistry , Asparagine/chemistry , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Amides/chemistry , Animals , Antibody Affinity , Drug Discovery , High-Throughput Screening Assays , Humans , Mutation/genetics , Protein Binding , Protein Stability
8.
Sci Rep ; 7(1): 9000, 2017 08 21.
Article En | MEDLINE | ID: mdl-28827556

The growth and motility factor Hepatocyte Growth Factor/Scatter Factor (HGF/SF) and its receptor, the product of the MET proto-oncogene, promote invasion and metastasis of tumor cells and have been considered potential targets for cancer therapy. We generated a new Met-blocking antibody which binds outside the ligand-binding site, and determined the crystal structure of the Fab in complex with its target, which identifies the binding site as the Met Ig1 domain. The antibody, 107_A07, inhibited HGF/SF-induced cell migration and proliferation in vitro and inhibited growth of tumor xenografts in vivo. In biochemical assays, 107_A07 competes with both HGF/SF and its truncated splice variant NK1 for MET binding, despite the location of the antibody epitope on a domain (Ig1) not reported to bind NK1 or HGF/SF. Overlay of the Fab-MET crystal structure with the InternalinB-MET crystal structure shows that the 107_A07 Fab comes into close proximity with the HGF/SF-binding SEMA domain when MET is in the "compact", InternalinB-bound conformation, but not when MET is in the "open" conformation. These findings provide further support for the importance of the "compact" conformation of the MET extracellular domain, and the relevance of this conformation to HGF/SF binding and signaling.


Antibodies, Blocking/isolation & purification , Antibodies, Blocking/metabolism , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/chemistry , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/chemistry , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Disease Models, Animal , Glioblastoma/drug therapy , Heterografts , Humans , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/metabolism , Mice, Nude , Neoplasm Transplantation , Protein Binding , Protein Conformation , Proto-Oncogene Mas , Treatment Outcome
9.
Cancer Cell ; 31(3): 383-395, 2017 03 13.
Article En | MEDLINE | ID: mdl-28262555

The anti-FcRH5/CD3 T cell-dependent bispecific antibody (TDB) targets the B cell lineage marker FcRH5 expressed in multiple myeloma (MM) tumor cells. We demonstrate that TDBs trigger T cell receptor activation by inducing target clustering and exclusion of CD45 phosphatase from the synapse. The dimensions of the target molecule play a key role in the efficiency of the synapse formation. The anti-FcRH5/CD3 TDB kills human plasma cells and patient-derived myeloma cells at picomolar concentrations and results in complete depletion of B cells and bone marrow plasma cells in cynomolgus monkeys. These data demonstrate the potential for the anti-FcRH5/CD3 TDB, alone or in combination with inhibition of PD-1/PD-L1 signaling, in the treatment of MM and other B cell malignancies.


Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , Epitopes , Immunological Synapses/physiology , Multiple Myeloma/drug therapy , Receptors, Fc/immunology , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Humans , Leukocyte Common Antigens/physiology , Lymphocyte Activation , Macaca fascicularis , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/physiology , Receptors, Fc/analysis
10.
Cell Rep ; 16(6): 1690-1700, 2016 08 09.
Article En | MEDLINE | ID: mdl-27475227

The spread of tau pathology correlates with cognitive decline in Alzheimer's disease. In vitro, tau antibodies can block cell-to-cell tau spreading. Although mechanisms of anti-tau function in vivo are unknown, effector function might promote microglia-mediated clearance. In this study, we investigated whether antibody effector function is required for targeting tau. We compared efficacy in vivo and in vitro of two versions of the same tau antibody, with and without effector function, measuring tau pathology, neuron health, and microglial function. Both antibodies reduced accumulation of tau pathology in Tau-P301L transgenic mice and protected cultured neurons against extracellular tau-induced toxicity. Only the full-effector antibody enhanced tau uptake in cultured microglia, which promoted release of proinflammatory cytokines. In neuron-microglia co-cultures, only effectorless anti-tau protected neurons, suggesting full-effector tau antibodies can induce indirect toxicity via microglia. We conclude that effector function is not required for efficacy, and effectorless tau antibodies may represent a safer approach to targeting tau.


Alzheimer Disease/metabolism , Microglia/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Antibodies/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Coculture Techniques/methods , Cytokines/metabolism , Mice, Transgenic , Neurons/metabolism
11.
Mol Cancer Ther ; 15(1): 3-14, 2016 Jan.
Article En | MEDLINE | ID: mdl-26712116

In many cancers, aberrant activation of the Met receptor tyrosine kinase leads to dissociation of cells from the primary tumor, causing metastasis. Accordingly, Met is a high-profile target for the development of cancer therapies, and progress has been made through development of small molecule kinase inhibitors and antibodies. However, both approaches pose significant challenges with respect to either target specificity (kinase inhibitors) or the cost involved in treating large patient cohorts (antibodies). Here, we use a fragment-based approach in order to target the protein-protein interaction (PPI) between the α-chain of hepatocyte growth factor/scatter factor (HGF/SF; the NK1 fragment) and its high-affinity binding site located on the Met Sema domain. Surface plasmon resonance was used for initial fragment library screening and hits were developed into larger compounds using substructure (similarity) searches. We identified compounds able to interfere with NK1 binding to Met, disrupt Met signaling, and inhibit tumorsphere generation and cell migration. Using molecular docking, we concluded that some of these compounds inhibit the PPI directly, whereas others act indirectly. Our results indicate that chemical fragments can efficiently target the HGF/SF-Met interface and may be used as building blocks for generating biologically active lead compounds. This strategy may have broad application for the development of a new class of Met inhibitors, namely receptor antagonists, and in general for the development of small molecule PPI inhibitors of key therapeutic targets when structural information is not available.


Antineoplastic Agents/pharmacology , Drug Discovery/methods , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Antineoplastic Agents/chemistry , Cell Line , Drug Screening Assays, Antitumor/methods , Humans , Mice , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Binding/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Signal Transduction , Small Molecule Libraries , Structure-Activity Relationship
12.
PLoS One ; 8(8): e70452, 2013.
Article En | MEDLINE | ID: mdl-23950939

We have developed a structurally-guided scaffold phage display strategy for identification of ligand mimetic bio-therapeutics. As a proof of concept we used the ligand of integrin αvß6, a tumour cell surface receptor and a major new target for imaging and therapy of many types of solid cancer. NMR structure analysis showed that RGD-helix structures are optimal for αvß6 ligand-interaction, so we designed novel algorithms to generate human single chain fragment variable (scFv) libraries with synthetic VH-CDR3 encoding RGD-helix hairpins with helices of differing pitch, length and amino acid composition. Study of the lead scFv clones D25scFv and D34scFv and their corresponding VH-CDR3 derived peptides, D25p and D34p, demonstrated: specific binding to recombinant and cellular αvß6; inhibition of αvß6-dependent cell and ligand adhesion, αvß6-dependent cell internalisation; and selective retention by αvß6-expressing, but not αvß6-negative, human xenografts. NMR analysis established that both the D25p and D34p retained RGD-helix structures confirming the success of the algorithm. In conclusion, scFv libraries can be engineered based on ligand structural motifs to increase the likelihood of developing powerful bio-therapeutics.


Antigens, Neoplasm/chemistry , Integrins/chemistry , Oligopeptides/chemistry , Peptide Library , Single-Chain Antibodies/chemistry , Algorithms , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Binding Sites , Female , Humans , Integrins/genetics , Integrins/metabolism , Ligands , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Single-Chain Antibodies/metabolism , Single-Chain Antibodies/pharmacology , Structural Homology, Protein , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Ann Rheum Dis ; 72(1): 129-35, 2013 Jan.
Article En | MEDLINE | ID: mdl-22843486

OBJECTIVES: The synovial endothelium targeting peptide (SyETP) CKSTHDRLC has been identified previously and was shown to preferentially localise to synovial xenografts in the human/severe combined immunodeficient (SCID) mouse chimera model of rheumatoid arthritis (RA). The objective of the current work was to generate SyETP-anti-inflammatory-cytokine fusion proteins that would deliver bioactive cytokines specifically to human synovial tissue. METHODS: Fusion proteins consisting of human interleukin (IL)-4 linked via a matrix metalloproteinase (MMP)-cleavable sequence to multiple copies of either SyETP or scrambled control peptide were expressed in insect cells, purified by Ni-chelate chromatography and bioactivity tested in vitro. The ability of SyETP to retain bioactive cytokine in synovial but not control skin xenografts in SCID mice was determined by in vivo imaging using nano-single-photon emission computed tomography-computed tomography (nano-SPECT-CT) and measuring signal transducer and activator of transcription 6 (STAT6) phosphorylation in synovial grafts following intravenous administration of the fusion protein. RESULTS: In vitro assays confirmed that IL-4 and the MMP-cleavable sequence were functional. IL-4-SyETP augmented production of IL-1 receptor antagonist (IL-1ra) by fibroblast-like synoviocytes (FLS) stimulated with IL-1ß  in a dose-dependent manner. In vivo imaging showed that IL-4-SyETP was retained in synovial but not in skin tissue grafts and the period of retention was significantly enhanced through increasing the number of SyETP copies from one to three. Finally, retention correlated with increased bioactivity of the cytokine as quantified by STAT6 phosphorylation in synovial grafts. CONCLUSIONS: The present work demonstrates that SyETP specifically delivers fused IL-4 to human rheumatoid synovium transplanted into SCID mice, thus providing a proof of concept for peptide-targeted tissue-specific immunotherapy in RA. This technology is potentially applicable to other biological treatments providing enhanced potency to inflammatory sites and reducing systemic toxicity.


Arthritis, Rheumatoid , Cytokines/administration & dosage , Drug Delivery Systems/methods , Immunotherapy/methods , Interleukin-4/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Animals , Arthritis, Rheumatoid/drug therapy , Disease Models, Animal , Humans , Mice , Mice, SCID , Multimodal Imaging , Peptides/administration & dosage , Positron-Emission Tomography , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Tomography, X-Ray Computed , Transplantation, Heterologous
14.
RSC Adv ; 2(29): 11019-11028, 2012 Nov 21.
Article En | MEDLINE | ID: mdl-27182435

Integrin αvß6 is an important emerging target for both imaging and therapy of cancer that requires specific ligands based on Arg-Gly-Asp (RGD) peptides. There remains little correlation between integrin-RGD ligand specificity despite studies suggesting an RGD-turn-helix ligand motif is required. Here, we describe the application of 15N NMR relaxation analyses and structure determination of αvß6 peptide ligands in the presence and absence of trifluoroethanol (TFE) to identify their critical molecular nature that influences specificity, interaction and function. Two linear peptides; one known to demonstrate αvß6 specificity (FMDV2) and the other based on a natural RGD ligand (LAP2), were compared to two additional peptides based on FMDV2 but cyclised in different positions using a disulphide bond (DBD1 and DBD2). The cyclic adaptation in DBD1 produces a significant alteration in backbone dynamic properties when compared to FMDV2; a potential driver for the loss in αvß6 specificity by DBD1. The importance of ligand dynamics are highlighted through a comprehensive reduced spectral density and ModelFree analysis of peptide 15N NMR relaxation data and suggest αvß6 specificity requires the formation of a structurally rigid helix preceded by a RGD motif exhibiting slow internal motion. Additional observations include the effect of TFE/water viscosity on global NMR dynamics and the advantages of using spectral density NMR relaxation data to estimate correlation times and motional time regimes for peptides in solution.

15.
J Virol ; 82(3): 1537-46, 2008 Feb.
Article En | MEDLINE | ID: mdl-18045932

The initial stage of foot-and-mouth disease virus (FMDV) infection is virus binding to cell surface integrins via the RGD motif in the GH loop of the VP1 capsid protein. As for all ligand/integrin interactions, the initial contact between FMDV and its integrin receptors is cation dependent and hence inhibited by EDTA. We have investigated this binding process with RGD-containing peptides derived from the VP1 capsid protein of FMDV and discovered that, upon binding, some of these peptides form highly stable, EDTA-resistant associations with integrin alphavbeta6. Peptides containing specific substitutions show that this stable binding is dependent on a helical structure immediately C terminal to the RGD and, specifically, two leucine residues at positions RGD +1 and RGD +4. These observations have a biological consequence, as we show further that stable, EDTA-resistant binding to alphavbeta6 is a property also exhibited by FMDV particles. Thus, the integrin-binding loop of FMDV appears to have evolved to form very stable complexes with the principal receptor of FMDV, integrin alphavbeta6. An ability to induce such stable complexes with its cellular receptor is likely to contribute significantly to the high infectiousness of FMDV.


Antigens, Neoplasm/metabolism , Capsid Proteins/metabolism , Foot-and-Mouth Disease Virus/physiology , Integrins/metabolism , Receptors, Virus/metabolism , Virus Attachment , Protein Binding , Protein Interaction Mapping
16.
Cancer Res ; 67(16): 7833-40, 2007 Aug 15.
Article En | MEDLINE | ID: mdl-17699789

Expression of the epithelial-specific integrin alphavbeta6 is low or undetectable in most adult tissues but may be increased during wound healing and inflammation and is up-regulated dramatically by many different carcinomas, making alphavbeta6 a promising target for the in vivo detection of cancer using noninvasive imaging. In addition, alphavbeta6 is recognized as promoting invasion and correlates with aggressive behavior of human cancers and thus agents that recognize alphavbeta6 specifically in vivo will be an essential tool for the future management of alphavbeta6-positive cancers. Recently, we identified the peptide NAVPNLRGDLQVLAQKVART (A20FMDV2), derived from foot-and-mouth disease virus, as a potent inhibitor of alphavbeta6. Using flow cytometry and ELISA, we show that this peptide is highly selective, inhibiting alphavbeta6-ligand binding with a IC50 of 3 nmol/L, an activity 1,000-fold more selective for alphavbeta6 than for other RGD-directed integrins (alphavbeta3, alphavbeta5, and alpha5beta1). A20FMDV2 was radiolabeled on solid-phase using 4-[18F]fluorobenzoic acid, injected into mice bearing both alphavbeta6-negative and alphavbeta6-positive (DX3puro/DX3purobeta6 cell lines) xenografts and imaged using a small animal positron emission tomography (PET) scanner. Rapid uptake (<30 min) and selective retention (>5 h) of radioactivity in the alphavbeta6-positive versus the alphavbeta6-negative tumor, together with fast renal elimination of nonspecifically bound activity, resulted in specific imaging of the alphavbeta6-positive neoplasm. These data suggest that PET imaging of alphavbeta6-positive tumors is feasible and will provide an important new tool for early detection and improved management of many types of cancers.


Antigens, Neoplasm/analysis , Benzoates , Foot-and-Mouth Disease Virus/chemistry , Integrins/analysis , Melanoma/diagnostic imaging , Peptide Fragments/pharmacokinetics , Radiopharmaceuticals , Viral Envelope Proteins/pharmacokinetics , Amino Acid Sequence , Animals , Antigens, Neoplasm/metabolism , Benzoates/chemistry , Benzoates/pharmacokinetics , Binding, Competitive , Enzyme-Linked Immunosorbent Assay , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , Isotope Labeling , Male , Melanoma/blood , Melanoma/metabolism , Melanoma/urine , Mice , Mice, Nude , Molecular Sequence Data , Peptide Fragments/chemistry , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Viral Envelope Proteins/chemistry
17.
J Biol Chem ; 282(13): 9657-9665, 2007 Mar 30.
Article En | MEDLINE | ID: mdl-17244604

Data relating to the structural basis of ligand recognition by integrins are limited. Here we describe the physical requirements for high affinity binding of ligands to alpha v beta6. By combining a series of structural analyses with functional testing, we show that 20-mer peptide ligands, derived from high affinity ligands of alpha v beta6 (foot-and-mouth-disease virus, latency associated peptide), have a common structure comprising an Arg-Gly-Asp motif at the tip of a hairpin turn followed immediately by a C-terminal helix. This arrangement allows two conserved Leu/Ile residues at Asp(+1) and Asp(+4) to be presented on the outside face of the helix enabling a potential hydrophobic interaction with the alpha v beta6 integrin, in addition to the Arg-Gly-Asp interaction. The extent of the helix determines peptide affinity for alpha v beta6 and potency as an alpha v beta6 antagonist. A major role of this C-terminal helix is likely to be the correct positioning of the Asp(+1) and Asp(+4) residues. These data suggest an explanation for several biological functions of alpha v beta6 and provide a structural platform for design of alpha v beta6 antagonists.


Amino Acid Motifs/physiology , Antigens, Neoplasm/metabolism , Integrins/metabolism , Oligopeptides/physiology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Integrins/chemistry , Ligands , Mice , Molecular Sequence Data , NIH 3T3 Cells , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Fragments/genetics , Protein Structure, Secondary , Structure-Activity Relationship
...