Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(1): 102729, 2023 01.
Article in English | MEDLINE | ID: mdl-36410439

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase that plays a major role in developmental processes and metabolism. The dysregulation of FGFR1 through genetic aberrations leads to skeletal and metabolic diseases as well as cancer. For this reason, FGFR1 is a promising therapeutic target, yet a very challenging one due to potential on-target toxicity. More puzzling is that both agonistic and antagonistic FGFR1 antibodies are reported to exhibit similar toxicity profiles in vivo, namely weight loss. In this study, we aimed to assess and compare the mechanism of action of these molecules to better understand this apparent contradiction. By systematically comparing the binding of these antibodies and the activation or the inhibition of the major FGFR1 signaling events, we demonstrated that the molecules displayed similar properties and can behave either as an agonist or antagonist depending on the presence or the absence of the endogenous ligand. We further demonstrated that these findings translated in xenografts mice models. In addition, using time-resolved FRET and mass spectrometry analysis, we showed a functionally distinct FGFR1 active conformation in the presence of an antibody that preferentially activates the FGFR substrate 2 (FRS2)-dependent signaling pathway, demonstrating that modulating the geometry of a FGFR1 dimer can effectively change the signaling outputs and ultimately the activity of the molecule in preclinical studies. Altogether, our results highlighted how bivalent antibodies can exhibit both agonistic and antagonistic activities and have implications for targeting other receptor tyrosine kinases with antibodies.


Subject(s)
Antibodies, Monoclonal , Receptor, Fibroblast Growth Factor, Type 1 , Signal Transduction , Animals , Humans , Mice , Neoplasms , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology
2.
Nature ; 528(7580): 127-31, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26580007

ABSTRACT

Prevailing dogma holds that cell-cell communication through Notch ligands and receptors determines binary cell fate decisions during progenitor cell divisions, with differentiated lineages remaining fixed. Mucociliary clearance in mammalian respiratory airways depends on secretory cells (club and goblet) and ciliated cells to produce and transport mucus. During development or repair, the closely related Jagged ligands (JAG1 and JAG2) induce Notch signalling to determine the fate of these lineages as they descend from a common proliferating progenitor. In contrast to such situations in which cell fate decisions are made in rapidly dividing populations, cells of the homeostatic adult airway epithelium are long-lived, and little is known about the role of active Notch signalling under such conditions. To disrupt Jagged signalling acutely in adult mammals, here we generate antibody antagonists that selectively target each Jagged paralogue, and determine a crystal structure that explains selectivity. We show that acute Jagged blockade induces a rapid and near-complete loss of club cells, with a concomitant gain in ciliated cells, under homeostatic conditions without increased cell death or division. Fate analyses demonstrate a direct conversion of club cells to ciliated cells without proliferation, meeting a conservative definition of direct transdifferentiation. Jagged inhibition also reversed goblet cell metaplasia in a preclinical asthma model, providing a therapeutic foundation. Our discovery that Jagged antagonism relieves a blockade of cell-to-cell conversion unveils unexpected plasticity, and establishes a model for Notch regulation of transdifferentiation.


Subject(s)
Antibodies/therapeutic use , Cell Transdifferentiation , Lung/cytology , Lung/metabolism , Receptors, Notch/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Death/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Tracking , Cell Transdifferentiation/drug effects , Cilia/metabolism , Disease Models, Animal , Female , Goblet Cells/cytology , Goblet Cells/drug effects , Goblet Cells/pathology , Homeostasis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Jagged-2 Protein , Ligands , Lung/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Serrate-Jagged Proteins , Signal Transduction/drug effects
3.
Biochem J ; 472(2): 169-81, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26385991

ABSTRACT

High temperature requirement A1 (HtrA1) is a trypsin-fold serine protease implicated in the progression of age-related macular degeneration (AMD). Our interest in an antibody therapy to neutralize HtrA1 faces the complication that the target adopts a trimeric arrangement, with three active sites in close proximity. In the present study, we describe antibody 94, obtained from a human antibody phage display library, which forms a distinct macromolecular complex with HtrA1 and inhibits the enzymatic activity of recombinant and native HtrA1 forms. Using biochemical methods and negative-staining EM we were able to elucidate the molecular composition of the IgG94 and Fab94 complexes and the associated inhibition mechanism. The 246-kDa complex between the HtrA1 catalytic domain trimer (HtrA1_Cat) and Fab94 had a propeller-like organization with one Fab bound peripherally to each protomer. Low-resolution EM structures and epitope mapping indicated that the antibody binds to the surface-exposed loops B and C of the catalytic domain, suggesting an allosteric inhibition mechanism. The HtrA1_Cat-IgG94 complex (636 kDa) is a cage-like structure with three centrally located IgG94 molecules co-ordinating two HtrA1_Cat trimers and the six active sites pointing into the cavity of the cage. In both complexes, all antigen-recognition regions (paratopes) are found to bind one HtrA1 protomer and all protomers are bound by a paratope, consistent with the complete inhibition of enzyme activity. Therefore, in addition to its potential therapeutic usefulness, antibody 94 establishes a new paradigm of multimeric serine protease inhibition.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antigen-Antibody Complex/chemistry , Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Allosteric Regulation , Amino Acid Substitution , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Antibody Specificity , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites, Antibody , Catalytic Domain , Cell Line, Tumor , Epitope Mapping , High-Temperature Requirement A Serine Peptidase 1 , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Melanoma/enzymology , Melanoma/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
4.
EBioMedicine ; 2(7): 730-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26288846

ABSTRACT

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/ßKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/ßKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/ßKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/ßKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/ßKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


Subject(s)
Adipose Tissue, Brown/metabolism , Antibodies, Bispecific/pharmacology , Insulin/pharmacology , Membrane Proteins/agonists , Receptor, Fibroblast Growth Factor, Type 1/agonists , Adiponectin/metabolism , Adipose Tissue, Brown/drug effects , Animals , Cell Line , Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , HEK293 Cells , Humans , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Obese , Protein Binding/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Thermogenesis/drug effects , Weight Loss/drug effects
5.
Sci Transl Med ; 5(171): 171ra18, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23390248

ABSTRACT

Although standard chemotherapies are commonly used to treat most types of solid tumors, such treatment often results in inadequate response to, or relapse after, therapy. This is particularly relevant for lung cancer because most patients are diagnosed with advanced-stage disease and are treated with frontline chemotherapy. By studying the residual tumor cells that remain after chemotherapy in several in vivo non-small cell lung cancer models, we found that these cells have increased levels of human epidermal growth factor receptor (HER) signaling due, in part, to the enrichment of a preexisting NRG1(HI) subpopulation. Neuregulin 1 (NRG1) signaling in these models can be mediated by either the HER3 or HER4 receptor, resulting in the differential activation of downstream effectors. Inhibition of NRG1 signaling inhibits primary tumor growth and enhances the magnitude and duration of the response to chemotherapy. Moreover, we show that inhibition of ligand-mediated Her4 signaling impedes disease relapse in cases where NRG1 inhibition is insufficient. These findings demonstrate that ligand-dependent Her4 signaling plays an important role in disease relapse.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Neuregulin-1/antagonists & inhibitors , Signal Transduction , Animals , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Autocrine Communication/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans , Ligands , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm, Residual/drug therapy , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neuregulin-1/metabolism , Receptor, ErbB-4 , Signal Transduction/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Structure ; 20(4): 676-87, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22483114

ABSTRACT

Lacking any discernible sequence similarity, interleukin-34 (IL-34) and colony stimulating factor 1 (CSF-1) signal through a common receptor CSF-1R on cells of mononuclear phagocyte lineage. Here, the crystal structure of dimeric IL-34 reveals a helical cytokine fold homologous to CSF-1, and we further show that the complex architecture of IL-34 bound to the N-terminal immunoglobulin domains of CSF-1R is similar to the CSF-1/CSF-1R assembly. However, unique conformational adaptations in the receptor domain geometry and intermolecular interface explain the cross-reactivity of CSF-1R for two such distantly related ligands. The docking adaptations of the IL-34 and CSF-1 quaternary complexes, when compared to the stem cell factor assembly, draw a common evolutionary theme for transmembrane signaling. In addition, the structure of IL-34 engaged by a Fab fragment reveals the mechanism of a neutralizing antibody that can help deconvolute IL-34 from CSF-1 biology, with implications for therapeutic intervention in diseases with myeloid pathogenic mechanisms.


Subject(s)
Antibodies, Neutralizing/chemistry , Interleukins/chemistry , Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Structural Homology, Protein , Baculoviridae , Binding Sites , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/chemistry , Interleukins/antagonists & inhibitors , Interleukins/genetics , Kinetics , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Macrophage Colony-Stimulating Factor/genetics , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Signal Transduction/genetics , Stem Cell Factor/chemistry , Thermodynamics
7.
Sci Transl Med ; 3(113): 113ra126, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22174314

ABSTRACT

Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5'-monophosphate response element-binding protein), and mRNA expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 2/therapy , Fibroblast Growth Factors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Receptor, Fibroblast Growth Factor, Type 1/genetics , Tissue Distribution , Trans-Activators/metabolism , Transcription Factors
8.
Cancer Cell ; 20(4): 472-86, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-22014573

ABSTRACT

Extensive crosstalk among ErbB/HER receptors suggests that blocking signaling from more than one family member may be essential to effectively treat cancer and limit drug resistance. We generated a conventional IgG molecule MEHD7945A with dual HER3/EGFR specificity by phage display engineering and used structural and mutational studies to understand how a single antigen recognition surface binds two epitopes with high affinity. As a human IgG1, MEHD7945A exhibited dual action by inhibiting EGFR- and HER3-mediated signaling in vitro and in vivo and the ability to engage immune effector functions. Compared with monospecific anti-HER antibodies, MEHD7945A was more broadly efficacious in multiple tumor models, showing that combined inhibition of EGFR and HER3 with a single antibody is beneficial.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Immunoglobulin G/therapeutic use , Receptor, ErbB-3/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/toxicity , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Binding Sites, Antibody , Binding, Competitive , Cetuximab , Crystallography, X-Ray , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , ErbB Receptors/chemistry , ErbB Receptors/immunology , Female , Humans , Immunoglobulin G/adverse effects , Immunoglobulin G/chemistry , MAP Kinase Signaling System , Macaca fascicularis , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/immunology , Signal Transduction
9.
Dev Cell ; 20(1): 33-46, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21238923

ABSTRACT

Robo4 is an endothelial cell-specific member of the Roundabout axon guidance receptor family. To identify Robo4 binding partners, we performed a protein-protein interaction screen with the Robo4 extracellular domain. We find that Robo4 specifically binds to UNC5B, a vascular Netrin receptor, revealing unexpected interactions between two endothelial guidance receptors. We show that Robo4 maintains vessel integrity by activating UNC5B, which inhibits signaling downstream of vascular endothelial growth factor (VEGF). Function-blocking monoclonal antibodies against Robo4 and UNC5B increase angiogenesis and disrupt vessel integrity. Soluble Robo4 protein inhibits VEGF-induced vessel permeability and rescues barrier defects in Robo4(-/-) mice, but not in mice treated with anti-UNC5B. Thus, Robo4-UNC5B signaling maintains vascular integrity by counteracting VEGF signaling in endothelial cells, identifying a novel function of guidance receptor interactions in the vasculature.


Subject(s)
Blood Vessels/metabolism , Blood Vessels/pathology , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Animals , Antibodies, Blocking/pharmacology , Blood Vessels/drug effects , Blood Vessels/enzymology , Capillary Permeability/drug effects , Enzyme Activation/drug effects , Humans , Ligands , Mice , Models, Biological , Netrin Receptors , Protein Binding/drug effects , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction/drug effects , Sus scrofa , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism
10.
PLoS One ; 5(9): e12682, 2010 Sep 13.
Article in English | MEDLINE | ID: mdl-20856934

ABSTRACT

ß-Catenin-dependent Wnt signaling is initiated as Wnt binds to both the receptor FZD and coreceptor LRP5/6, which then assembles a multimeric complex at the cytoplasmic membrane face to recruit and inactivate the kinase GSK3. The large number and sequence diversity of Wnt isoforms suggest the possibility of domain-specific ligand-coreceptor interactions, and distinct binding sites on LRP6 for Wnt3a and Wnt9b have recently been identified in vitro. Whether mechanistically different interactions between Wnts and coreceptors might mediate signaling remains to be determined. It is also not clear whether coreceptor homodimerization induced extracellularly can activate Wnt signaling, as is the case for receptor tyrosine kinases. We generated monoclonal antibodies against LRP6 with the unexpected ability to inhibit signaling by some Wnt isoforms and potentiate signaling by other isoforms. In cell culture, two antibodies characterized further show reciprocal activities on most Wnts, with one antibody antagonizing and the other potentiating. We demonstrate that these antibodies bind to different regions of LRP6 protein, and inhibition of signaling results from blocking Wnt binding. Antibody-mediated dimerization of LRP6 can potentiate signaling only when a Wnt isoform is also able to bind the complex, presumably recruiting FZD. Endogenous autocrine Wnt signaling in different tumor cell lines can be either antagonized or enhanced by the LRP6 antibodies, indicating expression of different Wnt isoforms. As anticipated from the roles of Wnt signaling in cancer and bone development, antibody activities can also be observed in mice for inhibition of tumor growth and in organ culture for enhancement of bone mineral density. Collectively, our results indicate that separate binding sites for different subsets of Wnt isoforms determine the inhibition or potentiation of signaling conferred by LRP6 antibodies. This complexity of coreceptor-ligand interactions may allow for differential regulation of signaling by Wnt isoforms during development, and can be exploited with antibodies to differentially manipulate Wnt signaling in specific tissues or disease states.


Subject(s)
Antibodies/pharmacology , Down-Regulation/drug effects , LDL-Receptor Related Proteins/immunology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Wnt Proteins/metabolism , Animals , Cell Line , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Protein Binding/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/genetics , Species Specificity , Up-Regulation/drug effects , Wnt Proteins/genetics
11.
FASEB J ; 24(11): 4262-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20585027

ABSTRACT

The primary objective of this study was to utilize MR molecular imaging to compare the 3-dimensional spatial distribution of Robo4 and α(V)ß(3)-integrin as biosignatures of angiogenesis, in a rapidly growing, syngeneic tumor. B16-F10 melanoma-bearing mice were imaged with magnetic resonance (MR; 3.0 T) 11 d postimplantation before and after intravenous administration of either Robo4- or α(V)ß(3)-targeted paramagnetic nanoparticles. The percentage of MR signal-enhanced voxels throughout the tumor volume was low and increased in animals receiving α(V)ß(3)- and Robo4-targeted nanoparticles. Neovascular signal enhancement was predominantly associated with the tumor periphery (i.e., outer 50% of volume). Microscopic examination of tumors coexposed to the Robo4- and α(V)ß(3)-targeted nanoparticles corroborated the MR angiogenesis mapping results and further revealed that Robo4 expression generally colocalized with α(V)ß(3)-integrin. Robo4- and α(V)ß(3)-targeted nanoparticles were compared to irrelevant or nontargeted control groups in all modalities. These results suggest that α(V)ß(3)-integrin and Robo4 are useful biomarkers for noninvasive MR molecular imaging in syngeneic mouse tumors, but α(V)ß(3)-integrin expression was more detectable by MR at 3.0 T than Robo4. Noninvasive, neovascular assessments of the MR signal of Robo4, particularly combined with α(V)ß(3)-integrin expression, may help define tumor character prior to and following cancer therapy.


Subject(s)
Biomarkers/metabolism , Integrin alphaVbeta3/metabolism , Melanoma/diagnosis , Molecular Imaging/methods , Nanoparticles , Neovascularization, Pathologic/diagnosis , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Nanoparticles/chemistry , Receptors, Cell Surface , Staining and Labeling
12.
Nature ; 464(7291): 1052-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20393564

ABSTRACT

The four receptors of the Notch family are widely expressed transmembrane proteins that function as key conduits through which mammalian cells communicate to regulate cell fate and growth. Ligand binding triggers a conformational change in the receptor negative regulatory region (NRR) that enables ADAM protease cleavage at a juxtamembrane site that otherwise lies buried within the quiescent NRR. Subsequent intramembrane proteolysis catalysed by the gamma-secretase complex liberates the intracellular domain (ICD) to initiate the downstream Notch transcriptional program. Aberrant signalling through each receptor has been linked to numerous diseases, particularly cancer, making the Notch pathway a compelling target for new drugs. Although gamma-secretase inhibitors (GSIs) have progressed into the clinic, GSIs fail to distinguish individual Notch receptors, inhibit other signalling pathways and cause intestinal toxicity, attributed to dual inhibition of Notch1 and 2 (ref. 11). To elucidate the discrete functions of Notch1 and Notch2 and develop clinically relevant inhibitors that reduce intestinal toxicity, we used phage display technology to generate highly specialized antibodies that specifically antagonize each receptor paralogue and yet cross-react with the human and mouse sequences, enabling the discrimination of Notch1 versus Notch2 function in human patients and rodent models. Our co-crystal structure shows that the inhibitory mechanism relies on stabilizing NRR quiescence. Selective blocking of Notch1 inhibits tumour growth in pre-clinical models through two mechanisms: inhibition of cancer cell growth and deregulation of angiogenesis. Whereas inhibition of Notch1 plus Notch2 causes severe intestinal toxicity, inhibition of either receptor alone reduces or avoids this effect, demonstrating a clear advantage over pan-Notch inhibitors. Our studies emphasize the value of paralogue-specific antagonists in dissecting the contributions of distinct Notch receptors to differentiation and disease and reveal the therapeutic promise in targeting Notch1 and Notch2 independently.


Subject(s)
Antibodies/pharmacology , Antibodies/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Notch/antagonists & inhibitors , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies/adverse effects , Antibodies/immunology , Antibody Specificity/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Goblet Cells/drug effects , Goblet Cells/pathology , Humans , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Peptide Library , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/immunology , Receptor, Notch2/antagonists & inhibitors , Receptor, Notch2/immunology , Receptors, Notch/genetics , Receptors, Notch/immunology , Receptors, Notch/metabolism , Signal Transduction/drug effects
13.
J Clin Invest ; 119(5): 1216-29, 2009 May.
Article in English | MEDLINE | ID: mdl-19381019

ABSTRACT

Overexpression of FGF receptor 3 (FGFR3) is implicated in the development of t(4;14)-positive multiple myeloma. While FGFR3 is frequently overexpressed and/or activated through mutations in bladder cancer, the functional importance of FGFR3 and its potential as a specific therapeutic target in this disease have not been elucidated in vivo. Here we report that inducible knockdown of FGFR3 in human bladder carcinoma cells arrested cell-cycle progression in culture and markedly attenuated tumor progression in xenografted mice. Further, we developed a unique antibody (R3Mab) that inhibited not only WT FGFR3, but also various mutants of the receptor, including disulfide-linked cysteine mutants. Biochemical analysis and 2.1-A resolution crystallography revealed that R3Mab bound to a specific FGFR3 epitope that simultaneously blocked ligand binding, prevented receptor dimerization, and induced substantial conformational changes in the receptor. R3Mab exerted potent antitumor activity against bladder carcinoma and t(4;14)-positive multiple myeloma xenografts in mice by antagonizing FGFR3 signaling and eliciting antibody-dependent cell-mediated cytotoxicity (ADCC). These studies provide in vivo evidence demonstrating an oncogenic role of FGFR3 in bladder cancer and support antibody-based targeting of FGFR3 in hematologic and epithelial cancers driven by WT or mutant FGFR3.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Multiple Myeloma/therapy , Receptor, Fibroblast Growth Factor, Type 3/immunology , Translocation, Genetic/genetics , Urinary Bladder Neoplasms/therapy , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigen-Antibody Complex/chemistry , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Epitopes/chemistry , Epitopes/immunology , Female , Fibroblast Growth Factors/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Nude , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Conformation/drug effects , RNA Interference , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction/drug effects , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
J Biol Chem ; 284(16): 10473-9, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19196712

ABSTRACT

Amplification of the complement cascade through the alternative pathway can lead to excessive inflammation. Targeting C3b, a component central to the alternative pathway of complement, provides a powerful approach to inhibit complement-mediated immune responses and tissue injury. In the present study, phage display technology was employed to generate an antibody that selectively recognizes C3b but not the non-activated molecule C3. The crystal structure of C3b in complex with a Fab fragment of this antibody (S77) illustrates the structural basis for this selectivity. Cleavage of C3 to C3b results in a plethora of structural changes within C3, including the rearrangement of macroglobulin domain 6 enabling binding of S77 to the adjacent macroglobulin domain 7 domain. S77 blocks binding of factor B to C3b inhibiting the first step in the formation of the alternative pathway C3 convertase. In addition, S77 inhibits C5 binding to C3b. This results in significantly reduced formations of anaphylatoxins and membrane-attack complexes. This study for the first time demonstrates the structural basis for complement inhibition by a C3b-selective antibody and provides insights into the molecular mechanisms of alternative pathway complement activation.


Subject(s)
Antibodies , Complement C3b/immunology , Complement Pathway, Alternative/physiology , Immunoglobulin Fab Fragments , Protein Conformation , Animals , Antibodies/chemistry , Antibodies/metabolism , Complement C3 Convertase, Alternative Pathway/metabolism , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement C5 Convertase, Alternative Pathway/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Stability , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Macaca mulatta , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Receptors, Complement 3b/chemistry , Receptors, Complement 3b/metabolism
15.
Science ; 322(5903): 967-70, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18988857

ABSTRACT

A major barrier to regenerating axons after injury in the mammalian central nervous system is an unfavorable milieu. Three proteins found in myelin--Nogo, MAG, and OMgp--inhibit axon regeneration in vitro and bind to the glycosylphosphatidylinositol-anchored Nogo receptor (NgR). However, genetic deletion of NgR has only a modest disinhibitory effect, suggesting that other binding receptors for these molecules probably exist. With the use of expression cloning, we have found that paired immunoglobulin-like receptor B (PirB), which has been implicated in nervous system plasticity, is a high-affinity receptor for Nogo, MAG, and OMgp. Interfering with PirB activity, either with antibodies or genetically, partially rescues neurite inhibition by Nogo66, MAG, OMgp, and myelin in cultured neurons. Blocking both PirB and NgR activities leads to near-complete release from myelin inhibition. Our results implicate PirB in mediating regeneration block, identify PirB as a potential target for axon regeneration therapies, and provide an explanation for the similar enhancements of visual system plasticity in PirB and NgR knockout mice.


Subject(s)
Axons/physiology , Myelin Proteins/metabolism , Nerve Regeneration , Neurons/cytology , Neurons/metabolism , Receptors, Immunologic/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cerebellum/cytology , GPI-Linked Proteins , Ganglia, Spinal/cytology , Growth Cones/physiology , Mice , Molecular Sequence Data , Myelin-Associated Glycoprotein/metabolism , Myelin-Oligodendrocyte Glycoprotein , Neurites/physiology , Nogo Proteins , Nogo Receptor 1 , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
16.
Cancer Cell ; 13(4): 331-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18394556

ABSTRACT

Metastasis, which commonly uses lymphatics, accounts for much of the mortality associated with cancer. The vascular endothelial growth factor (VEGF)-C coreceptor, neuropilin-2 (Nrp2), modulates but is not necessary for developmental lymphangiogenesis, and its significance for metastasis is unknown. An antibody to Nrp2 that blocks VEGFC binding disrupts VEGFC-induced lymphatic endothelial cell migration, but not proliferation, in part independently of VEGF receptor activation. It does not affect established lymphatics in normal adult mice but reduces tumoral lymphangiogenesis and, importantly, functional lymphatics associated with tumors. It also reduces metastasis to sentinel lymph nodes and distant organs, apparently by delaying the departure of tumor cells from the primary tumor. Our results demonstrate that Nrp2, which was originally identified as an axon-guidance receptor, is an attractive target for modulating metastasis.


Subject(s)
Neoplasm Metastasis/prevention & control , Neoplasms/pathology , Neuropilin-2/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Antibody Specificity/drug effects , Bacteriophages , Cell Line , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphangiogenesis/drug effects , Lymphatic Metastasis/prevention & control , Lymphatic System/drug effects , Lymphatic System/pathology , Mice , Neuropilin-2/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
17.
Proc Natl Acad Sci U S A ; 104(50): 19784-9, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077410

ABSTRACT

To better understand how the relatively flat antigen-combining sites of antibodies interact with the concave shaped substrate-binding clefts of proteases, we determined the structures of two antibodies in complex with the trypsin-like hepatocyte growth-factor activator (HGFA). The two inhibitory antibodies, Ab58 and Ab75, were generated from a human Fab phage display library with synthetic diversity in the three complementarity determining regions (H1, H2, and H3) of the heavy chain, mimicking the natural diversity of the human Ig repertoire. Biochemical studies and the structures of the Fab58:HGFA (3.5-A resolution) and the Fab75:HGFA (2.2-A resolution) complexes revealed that Ab58 obstructed substrate access to the active site, whereas Ab75 allosterically inhibited substrate hydrolysis. In both cases, the antibodies interacted with the same protruding element (99-loop), which forms part of the substrate-binding cleft. Ab58 inserted its H1 and H2 loops in the cleft to occupy important substrate interaction sites (S3 and S2). In contrast, Ab75 bound at the backside of the cleft to a region corresponding to thrombin exosite II, which is known to interact with allosteric effector molecules. In agreement with the structural analysis, binding assays with active site inhibitors and enzymatic assays showed that Ab58 is a competitive inhibitor, and Ab75 is a partial competitive inhibitor. These results provide structural insight into antibody-mediated protease inhibition. They suggest that unlike canonical inhibitors, antibodies may preferentially target protruding loops at the rim of the substrate-binding cleft to interfere with the catalytic machinery of proteases without requiring long insertion loops.


Subject(s)
Antibodies/chemistry , Antibodies/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Serine Endopeptidases/immunology , Animals , Antibodies/pharmacology , Binding Sites, Antibody , Binding, Competitive/immunology , Catalysis , Humans , Mice , Protease Inhibitors/pharmacology , Rabbits , Serine Endopeptidases/metabolism
18.
EMBO J ; 26(23): 4902-12, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-17989695

ABSTRACT

Neuropilins (Nrps) are co-receptors for class 3 semaphorins and vascular endothelial growth factors and important for the development of the nervous system and the vasculature. The extracellular portion of Nrp is composed of two domains that are essential for semaphorin binding (a1a2), two domains necessary for VEGF binding (b1b2), and one domain critical for receptor dimerization (c). We report several crystal structures of Nrp1 and Nrp2 fragments alone and in complex with antibodies that selectively block either semaphorin or vascular endothelial growth factor (VEGF) binding. In these structures, Nrps adopt an unexpected domain arrangement in which the a2, b1, and b2 domains form a tightly packed core that is only loosely connected to the a1 domain. The locations of the antibody epitopes together with in vitro experiments indicate that VEGF and semaphorin do not directly compete for Nrp binding. Based upon our structural and functional data, we propose possible models for ligand binding to neuropilins.


Subject(s)
Neuropilins/chemistry , Semaphorin-3A/chemistry , Vascular Endothelial Growth Factor A/chemistry , Amino Acid Sequence , Antibodies/chemistry , Binding Sites , Crystallography, X-Ray/methods , Dimerization , Molecular Conformation , Molecular Sequence Data , Neuropilins/physiology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Semaphorin-3A/metabolism , Semaphorins/metabolism , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A/metabolism
19.
J Mol Biol ; 366(3): 815-29, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17196977

ABSTRACT

Non-immune (naïve) antibody phage libraries have become an important source of human antibodies. The synthetic phage antibody library described here utilizes a single human framework with a template containing human consensus complementarity-determining regions (CDRs). Diversity of the libraries was introduced at select CDR positions using tailored degenerate and trinucleotide codons that mimic natural human antibodies. Neuropilin-1 (NRP1), a cell-surface receptor for both vascular endothelial growth factor (VEGF) and class 3 semaphorins, is expressed on endothelial cells and neurons. NRP1 is required for vascular development and is expressed widely in the developing vasculature. To investigate the possibility of function blocking antibodies to NRP1 as potential therapeutics, and study the consequence of targeting NRP1 in murine tumor models, panels of antibodies that cross-react with human and murine NRP1 were generated from a designed antibody phage library. Antibody (YW64.3) binds to the CUB domains (a1a2) of NRP1 and completely blocks Sema3A induced neuron collapse; antibody (YW107.4.87) binds to the coagulation factor V/VIII domains (b1b2) of NRP1 and blocks VEGF binding and VEGF induced cell migration. YW107.4.87 inhibits tumor growth in animal xenograft models. These antibodies have provided valuable tools to study the roles of NRP1 in vascular and tumor biology.


Subject(s)
Antibodies, Blocking/immunology , Neuropilin-1/immunology , Peptide Library , Amino Acid Sequence , Animals , Antibodies, Blocking/chemistry , Antibody Affinity/drug effects , CHO Cells , Cell Movement/drug effects , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Cricetinae , Cricetulus , Growth Cones/drug effects , Humans , Immunoglobulin G/immunology , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Neoplasms/pathology , Protein Structure, Tertiary/drug effects , Semaphorin-3A/pharmacology
20.
Cancer Cell ; 11(1): 53-67, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17222790

ABSTRACT

Neuropilin-1 (NRP1) guides the development of the nervous and vascular systems. Binding to either semaphorins or VEGF, NRP1 acts with plexins to regulate neuronal guidance, or with VEGFR2 to mediate vascular development. We have generated two monoclonal antibodies that bind to the Sema- and VEGF-binding domains of NRP1, respectively. Both antibodies reduce angiogenesis and vascular remodeling, while having little effect on other VEGFR2-mediated events. Importantly, anti-NRP1 antibodies have an additive effect with anti-VEGF therapy in reducing tumor growth. Vessels from tumors treated with anti-VEGF show a close association with pericytes, while tumors treated with both anti-NRP1 and anti-VEGF lack this organization. We propose that blocking NRP1 function inhibits vascular remodeling, rendering vessels more susceptible to anti-VEGF therapy.


Subject(s)
Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Neuropilin-1/immunology , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies, Monoclonal , Cell Movement , Cells, Cultured , Endothelial Cells/metabolism , Female , Humans , Immunohistochemistry , Mice , Neurons/metabolism , Rats , Semaphorin-3A/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...