Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 10(2)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35214607

ABSTRACT

(1) Influenza viruses constantly change and evade prior immune responses, forcing seasonal re-vaccinations with updated vaccines. Current FDA-approved vaccine manufacturing technologies are too slow and/or expensive to quickly adapt to mid-season changes in the virus or to the emergence of pandemic strains. Therefore, cost-effective vaccine technologies that can quickly adapt to newly emerged strains are desirable. (2) The filamentous fungal host Thermothelomyces heterothallica C1 (C1, formerly Myceliophthora thermophila) offers a highly efficient and cost-effective alternative to reliably produce immunogens of vaccine quality at large scale. (3) We showed the utility of the C1 system expressing hemagglutinin (HA) and a HA fusion protein from different H1N1 influenza A virus strains. Mice vaccinated with the C1-derived HA proteins elicited anti-HA immune responses similar, or stronger than mice vaccinated with HA products derived from prototypical expression systems. A challenge study demonstrated that vaccinated mice were protected against the aggressive homologous viral challenge. (4) The C1 expression system is proposed as part of a set of protein expression systems for plug-and-play vaccine manufacturing platforms. Upon the emergence of pathogens of concern these platforms could serve as a quick solution for producing enough vaccines for immunizing the world population in a much shorter time and more affordably than is possible with current platforms.

2.
Growth Factors ; 32(6): 223-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25413948

ABSTRACT

EphA3 is expressed in solid tumors and leukemias and is an attractive target for the therapy. We have generated a panel of Humaneered® antibodies to the ligand-binding domain using a Fab epitope-focused library that has the same specificity as monoclonal antibody mIIIA4. A high-affinity antibody was selected that competes with the mIIIA4 antibody for binding to EphA3 and has an improved affinity of ∼1 nM. In order to generate an antibody with potent cell-killing activity the variable regions were assembled with human IgG1k constant regions and expressed in a Chinese hamster ovary (CHO) cell line deficient in fucosyl transferase. Non-fucosylated antibodies have been reported to have enhanced binding affinity for the IgG receptor CD16a (FcγRIIIa). The affinity of the antibody for recombinant CD16a was enhanced approximately 10-fold. This resulted in enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity against EphA3-expressing leukemic cells, providing a potent antibody for the evaluation as a therapeutic agent.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Receptor, EphA3/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fc Fragments/immunology , Macaca mulatta , Molecular Sequence Data , Receptors, IgG/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology
3.
Cancer Res ; 74(16): 4470-81, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25125683

ABSTRACT

Eph receptor tyrosine kinases are critical for cell-cell communication during normal and oncogenic tissue patterning and tumor growth. Somatic mutation profiles of several cancer genomes suggest EphA3 as a tumor suppressor, but its oncogenic expression pattern and role in tumorigenesis remain largely undefined. Here, we report unexpected EphA3 overexpression within the microenvironment of a range of human cancers and mouse tumor xenografts where its activation inhibits tumor growth. EphA3 is found on mouse bone marrow-derived cells with mesenchymal and myeloid phenotypes, and activation of EphA3(+)/CD90(+)/Sca1(+) mesenchymal/stromal cells with an EphA3 agonist leads to cell contraction, cell-cell segregation, and apoptosis. Treatment of mice with an agonistic α-EphA3 antibody inhibits tumor growth by severely disrupting the integrity and function of newly formed tumor stroma and microvasculature. Our data define EphA3 as a novel target for selective ablation of the tumor microenvironment and demonstrate the potential of EphA3 agonists for anticancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor Protein-Tyrosine Kinases/agonists , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, EphA3/agonists , Receptor, EphA3/biosynthesis , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3/immunology , Receptor, EphA3/metabolism , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Microenvironment/drug effects
4.
J Allergy Clin Immunol ; 133(5): 1439-47, 1447.e1-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24530099

ABSTRACT

BACKGROUND: Although several novel agents are currently in clinical trials for eosinophilic disorders, none has demonstrated efficacy in reducing blood and tissue eosinophilia in all subjects. Additional approaches are clearly needed. OBJECTIVE: We sought to explore the potential of the human eosinophil surface receptor epidermal growth factor-like module containing mucin-like hormone receptor 1 (EMR1) as a therapeutic target for eosinophilic disorders. METHODS: EMR1 expression was assessed in blood and bone marrow specimens from eosinophilic and healthy subjects, cell lines, CD34(+) cells differentiated in vitro, and tissue biopsy specimens by using flow cytometry, quantitative PCR, and immunostaining. Eosinophil targeting by a novel, humanized, afucosylated anti-EMR1 IgG1 was evaluated in vitro by using a natural killer cell-mediated killing assay and in vivo in cynomolgus monkeys. RESULTS: Analysis of blood and bone marrow cells from healthy and eosinophilic donors and in vitro-differentiated CD34(+) cells confirmed restriction of human EMR1 surface and mRNA expression to mature eosinophils. Tissue eosinophils also expressed EMR1. Although EMR1 was highly expressed on eosinophils from all subjects, surface expression was negatively correlated with absolute eosinophil counts (r = -0.46, P < .001), and soluble plasma levels correlated positively with absolute eosinophil counts (r = 0.69, P < .001), suggesting modulation of EMR1 in vivo. Nevertheless, afucosylated anti-EMR1 mAb dramatically enhanced natural killer cell-mediated killing of eosinophils from healthy and eosinophilic donors and induced a rapid and sustained depletion of eosinophils in monkeys. CONCLUSION: EMR1 expression is restricted to mature blood and tissue eosinophils. Targeting of eosinophils with afucosylated anti-EMR1 antibody shows promise as a treatment for eosinophilic disorders.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Eosinophilia/drug therapy , Eosinophils/immunology , Gene Expression Regulation/drug effects , Immunoglobulin G/pharmacology , Membrane Glycoproteins/immunology , Mucins/immunology , Receptors, G-Protein-Coupled/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Calcium-Binding Proteins , Eosinophilia/immunology , Eosinophilia/pathology , Eosinophils/pathology , Female , Humans , Immunoglobulin G/immunology , K562 Cells , Male , Membrane Glycoproteins/antagonists & inhibitors , Mucins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , U937 Cells
5.
Blood ; 121(25): 5068-77, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23632888

ABSTRACT

Granulocyte-macrophage-colony-stimulating factor (GM-CSF) hypersensitivity is a hallmark of juvenile myelomonocytic leukemia (JMML) but has not been systematically shown in the related human disease chronic myelomonocytic leukemia (CMML). We find that primary CMML samples demonstrate GM-CSF-dependent hypersensitivity by hematopoietic colony formation assays and phospho-STAT5 (pSTAT5) flow cytometry compared with healthy donors. Among CMML patients, the pSTAT5 hypersensitive response positively correlated with high-risk disease, peripheral leukocytes, monocytes, and signaling-associated mutations. When compared with IL-3 and G-CSF, GM-CSF hypersensitivity was cytokine specific and thus a possible target for intervention in CMML. To explore this possibility, we treated primary CMML cells with KB003, a novel monoclonal anti-GM-CSF antibody, and JAK2 inhibitors. We found that an elevated proportion of immature GM-CSF receptor-α(R) subunit-expressing cells were present in the bone marrow myeloid compartment of CMML. In survival assays, we found that myeloid and monocytic progenitors were sensitive to GM-CSF signal inhibition. Our data indicate that a committed myeloid precursor expressing CD38 may represent the progenitor population with enhanced GM-CSF dependence in CMML, consistent with results in JMML. These preclinical data indicate that GM-CSF signaling inhibitors merit further investigation in CMML and that GM-CSFR expression on myeloid progenitors may be a biomarker for this therapy.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Leukemia, Myelomonocytic, Chronic/metabolism , STAT5 Transcription Factor/metabolism , Flow Cytometry , Humans , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Surface Plasmon Resonance
6.
J Cardiovasc Pharmacol ; 57(5): 568-74, 2011 May.
Article in English | MEDLINE | ID: mdl-21326109

ABSTRACT

Granulocyte macrophage colony-stimulating factor (GM-CSF) promotes infarct expansion and inappropriate collagen synthesis in a myocardial infarction (MI). This study was designed to determine if treatment with anti-GM-CSF will inhibit macrophage migration, preserve function, and limit left ventricular (LV) remodeling in the rat coronary artery ligation model. Treatment with a monoclonal antibody to GM-CSF (5 mg/kg) was initiated 24 hours before coronary artery ligation and continued every 3 days for 3 weeks. Left coronary arteries of rats were ligated, animals were recovered, and cardiac function was evaluated 3 weeks postligation. Tissue samples were processed for histochemistry. Anti-GM-CSF treatment increased LV ejection fraction (37 ± 3% vs 47 ± 5%) and decreased LV end systolic diameter (0.75 ± 0.12 vs 0.59 ± 0.05 cm) with no changes in LV systolic pressure (109 ± 4 vs 104 ± 5 mm Hg), LV end diastolic pressure (22 ± 4 vs 21 ± 2 mm Hg), LV end diastolic diameter (0.96 ± 0.04 vs 0.92 ± 0.05 cm), or the time constant of LV relaxation tau (25.4 ± +2.4 vs 22.7 ± 1.4 milliseconds) (P < 0.05). Significantly lower numbers of tissue macrophages and significant reductions in infarct size were found in the myocardium of antibody-treated animals (81 ± 21.24 vs 195 ± 31.7 positive cells per 0.105 mm, compared with controls. These findings suggest that inhibition of macrophage migration may be beneficial in the treatment of heart failure after MI.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophage Activation/drug effects , Macrophages/drug effects , Myocardial Infarction/drug therapy , Ventricular Function, Left/drug effects , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/pharmacology , Cell Count , Cell Movement/drug effects , Coronary Vessels/immunology , Coronary Vessels/pathology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hemodynamics/drug effects , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/immunology , Male , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Rats , Rats, Sprague-Dawley , Ventricular Remodeling/drug effects
7.
Infect Immun ; 77(3): 1083-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19103766

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that can cause acute lung injury and mortality through the delivery of exotoxins by the type III secretion system (TTSS). PcrV is an important structural protein of the TTSS. An engineered human antibody Fab fragment that binds to the P. aeruginosa PcrV protein with high affinity has been identified and has potent in vitro neutralization activity against the TTSS. The instillation of a single dose of Fab into the lungs of mice provided protection against lethal pulmonary challenge of P. aeruginosa and led to a substantial reduction of viable bacterial counts in the lungs. These results demonstrate that blocking of the TTSS by a Fab lacking antibody Fc-mediated effector functions can be sufficient for the effective clearance of pulmonary P. aeruginosa infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunoglobulin Fab Fragments/immunology , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections/immunology , Recombinant Proteins/immunology , ADP Ribose Transferases/immunology , Animals , Antibody Specificity , Cytotoxicity, Immunologic/immunology , Enzyme-Linked Immunosorbent Assay , Exotoxins/immunology , Humans , Immunoglobulin Fab Fragments/therapeutic use , Male , Mice , Mice, Inbred BALB C , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/therapeutic use , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
8.
Anal Biochem ; 359(1): 112-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17027901

ABSTRACT

A Biacore T100 optical biosensor was used to characterize the binding kinetics of a panel of antigen binding fragments (Fabs) directed against the PcrV protein from Pseudomonas aeruginosa. PcrV protein forms part of the type III secretion system complex of this opportunistic pathogen. We demonstrate that the biosensor response data for each Fab collected from three different surface densities of the antigen could be fit globally to a simple 1:1 interaction model. Importantly, we found that the Fabs with the slowest dissociation rate provided the best protection in cell cytotoxicity studies. To further characterize the Fab interactions, binding data were automatically acquired at different temperatures and under different buffer conditions. The comprehensive characterization of these Fabs shows how Biacore T100 can be used to complement protein therapeutic discovery programs from basic research to the selection of therapeutic candidates.


Subject(s)
Antigen-Antibody Reactions/immunology , Antigens, Bacterial/biosynthesis , Bacterial Toxins/biosynthesis , Biosensing Techniques/methods , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/immunology , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas aeruginosa/pathogenicity , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Humans , Mice , Pore Forming Cytotoxic Proteins/biosynthesis , Pore Forming Cytotoxic Proteins/chemistry , Pseudomonas aeruginosa/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Research Design
9.
J Biol Chem ; 279(48): 49995-50003, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15465827

ABSTRACT

Lipopolysaccharide (LPS) induces expression of tumor necrosis factor alpha (TNFalpha) and other pro-inflammatory cytokines in macrophages. Following its induction, TNFalpha gene transcription is rapidly attenuated, in part due to the accumulation of NF-kappaB p50 homodimers that bind to three kappaB sites in the TNFalpha promoter. Here we have investigated the inhibitory role of BCL-3, an IkappaB-like protein that interacts exclusively with p50 and p52 homodimers. BCL-3 was induced by LPS with delayed kinetics and was associated with p50 in the nucleus. Forced expression of BCL-3 suppressed LPS-induced transcription from the TNFalpha promoter and inhibited two artificial promoters composed of TNFalphakappaB sites that preferentially bind p50 dimers. BCL-3-mediated repression was reversed by trichostatin A and was enhanced by overexpression of HDAC-1, indicating that transcriptional attenuation involves recruitment of histone deacetylase. Analysis of macrophages from p50 and BCL-3 knock-out mice revealed that both transcription factors negatively regulate TNFalpha expression and that BCL-3 inhibits IL-1alpha and IL-1beta. In contrast, induction of the anti-inflammatory cytokine IL-10 was reduced in BCL-3 null macrophages. BCL-3 was not required for the production of p50 homodimers but BCL-3 expression was severely diminished in p50-deficient cells. Together, these findings indicate that p50 and BCL-3 function as anti-inflammatory regulators in macrophages by attenuating transcription of pro-inflammatory cytokines and activating IL-10 expression.


Subject(s)
Lipopolysaccharides/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Animals , B-Cell Lymphoma 3 Protein , Gene Expression Regulation/physiology , Interleukin-10/biosynthesis , Interleukin-10/genetics , Lipopolysaccharides/immunology , Macrophages/immunology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B p50 Subunit , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Transcription Factors , Tumor Necrosis Factor-alpha/genetics
10.
J Biol Chem ; 277(26): 23563-72, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-11980905

ABSTRACT

The CCAAT/enhancer-binding proteins (C/EBPs) are basic leucine zipper transcription factors that play important roles in regulating cell growth and differentiation. C/EBP proteins form leucine zipper-mediated homodimers but are also capable of heterodimerizing with other C/EBPs in vitro. Here we show that C/EBPbeta occurs predominantly as a heterodimer that displays rapid mobility in gel shift assays. Biochemical fractionation and antibody supershift assays demonstrate that the C/EBPbeta heterodimeric partner is C/EBPgamma (Ig/EBP), a C/EBP protein that has been implicated as an inhibitor of other family members. Although most cell types express C/EBPbeta.C/EBPgamma heterodimers, macrophages contain a C/EBPbeta partner that is serologically distinct from C/EBPgamma. We found that C/EBPgamma blocked the ability of C/EBPbeta and C/EBPgamma to activate a reporter gene in L cell fibroblasts but did not inhibit a chimeric C/EBPbeta protein containing the GCN4 leucine zipper. Repression by C/EBPgamma occurs at the level of transactivation and requires heterodimerization with the C/EBP partner. C/EBPgamma was an ineffective repressor in HepG2 hepatoma cells despite forming C/EBP heterodimers, and C/EBPalpha was not effectively inhibited in either L or HepG2 cells. Our findings demonstrate that C/EBPgamma modulates C/EBP activity in a cell- and isoform-specific manner.


Subject(s)
CCAAT-Enhancer-Binding Proteins/chemistry , Transcription Factors , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/chemistry , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/physiology , Cell Line , DNA/metabolism , Dimerization , Humans , Mice , Repressor Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...